Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Innate Immun ; 11(6): 469-480, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30884482

RESUMO

To investigate the role of fatty acid-binding protein 5 (FABP5) in infectious diseases, FABP5-deficient mice were challenged with Listeria monocytogenes, a facultative intracellular bacterial pathogen. Interestingly, FABP5-deficient animals were able to clear the infection within 3 days whereas control wild-type (WT) animals showed comparatively higher bacterial burdens in the liver and spleen. Sections of infected tissues showed an increase in inflammatory foci in WT mice compared to FABP5-deficient mice. FABP5-deficient mice had lower circulating inflammatory cytokines and increased inducible nitric oxide synthase production. FABP5-deficient mouse bone marrow-derived macrophages produced higher levels of nitrite anion than their WT counterparts in response to various stimuli. Additionally, in contrast to FABP5-/- mice, transgenic mice overexpressing FABP5 in myeloid cells (LysM-Cre driven) showed decreased survival rates and increased bacterial burden and inflammatory cytokines. Overall, these findings suggest that increased FABP5 levels correlate with a higher L. monocytogenes bacterial burden and elevated subsequent inflammation.


Assuntos
Proteínas de Ligação a Ácido Graxo/metabolismo , Inflamação/metabolismo , Listeria monocytogenes/fisiologia , Listeriose/metabolismo , Macrófagos/fisiologia , Proteínas de Neoplasias/metabolismo , Animais , Carga Bacteriana , Sobrevivência Celular , Células Cultivadas , Citocinas/metabolismo , Proteínas de Ligação a Ácido Graxo/genética , Humanos , Inflamação/genética , Mediadores da Inflamação/metabolismo , Listeriose/genética , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/genética
2.
Radiat Environ Biophys ; 58(1): 89-98, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30483886

RESUMO

Radiodermatitis is a painful side effect for cancer patients undergoing radiotherapy. Irradiation of the skin causes inflammation and breakdown of the epidermis and can lead to significant morbidity and mortality in severe cases, as seen in exposure from accidents or weapons such as "dirty bombs" and ultimately leads to tissue fibrosis. However, the pathogenesis of radiodermatitis is not fully understood. Using a mouse model of radiodermatitis, we showed that the Transient Receptor Potential Melastatin 2 (TRPM2) ion channel plays a significant role in the development of dermatitis following exposure to ionizing radiation. Irradiated TRPM2-deficient mice developed less inflammation, fewer severe skin lesions and decreased fibrosis when compared to wild type mice. The TRPM2-deficient mice also showed a faster recovery period as seen by their increased weight gain post irradiation. Finally, TRPM2-deficient mice exhibited lower systemic inflammation with a reduction in inflammatory cytokines present in the serum. These findings suggest that TRPM2 may be a potential therapeutic target for reducing the severity of radiodermatitis.


Assuntos
Radiodermite/etiologia , Radiodermite/metabolismo , Canais de Cátion TRPM/metabolismo , Animais , Relação Dose-Resposta à Radiação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Radiodermite/patologia , Pele/patologia , Pele/efeitos da radiação
3.
Sci Signal ; 11(533)2018 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-29871912

RESUMO

Members of the transient receptor potential (TRP) family of ion channels are cellular sensors involved in numerous physiological and pathological processes. We identified the TRP subfamily M member 7 (TRPM7) channel-kinase as a previously uncharacterized regulator of B cell activation. We showed that TRPM7 played a critical role in the early events of B cell activation through both its ion channel and kinase functions. DT40 B cells deficient in TRPM7 or expressing a kinase-deficient mutant of TRPM7 showed defective gathering of antigen and prolonged B cell receptor (BCR) signaling. We showed that lipid metabolism was altered in TRPM7-deficient cells and in cells expressing a kinase-deficient mutant of TRPM7 and suggest that PLC-γ2 may be a target of the kinase activity of TRPM7. Primary B cells that expressed less TRPM7 or were treated with a pharmacological inhibitor of TRPM7 also displayed defective antigen gathering and increased BCR signaling. Finally, we demonstrated that blocking TRPM7 function compromised antigen internalization and presentation to T cells. These data suggest that TRPM7 controls an essential process required for B cell affinity maturation and the production of high-affinity antibodies.


Assuntos
Apresentação de Antígeno , Linfócitos B/metabolismo , Canais de Cátion TRPM/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Linfócitos B/citologia , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfolipase C gama/genética , Fosfolipase C gama/metabolismo , Fosforilação , Transdução de Sinais
4.
Biochim Biophys Acta Mol Basis Dis ; 1864(1): 126-132, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28970008

RESUMO

Trisomy 21 (Down Syndrome, DS) is the most common chromosomal anomaly. Although DS is mostly perceived as affecting cognitive abilities and cardiac health, individuals with DS also exhibit dysregulated immune functions. Levels of pro-inflammatory cytokines are increased, but intrinsic alterations of innate immunity are understudied in DS. Furthermore, elevated Reactive Oxygen Species (ROS) are well documented in individuals with DS, further exacerbating inflammatory processes. Chronic inflammation and oxidative stress are often precursors of subsequent tissue destruction and pathologies, which affect a majority of persons with DS. Together with ROS, the second messenger ion Ca2+ plays a central role in immune regulation. TRPM2 (Transient Receptor Potential Melastatin 2) is a Ca2+-permeable ion channel that is activated under conditions of oxidative stress. The Trpm2 gene is located on human Chromosome 21 (Hsa21). TRPM2 is strongly represented in innate immune cells, and numerous studies have documented its role in modulating inflammation. We have previously found that as a result of suboptimal cytokine production, TRPM2-/- mice are highly susceptible to the bacterial pathogen Listeria monocytogenes (Lm). We therefore used Lm infection to trigger and characterize immune responsiveness in the DS mouse model Dp10(yey), and to investigate the potential contribution of TRPM2. In comparison to wildtype (WT), Dp10(yey) mice show an increased resistance against Lm infection and higher IFNγ serum concentrations. Using a gene elimination approach, we show that these effects correlate with Trpm2 gene copy number, supporting the notion that Trpm2 might promote hyperinflammation in DS.


Assuntos
Citocinas/metabolismo , Síndrome de Down/patologia , Canais de Cátion TRPM/fisiologia , Animais , Modelos Animais de Doenças , Síndrome de Down/genética , Síndrome de Down/metabolismo , Feminino , Imunidade Inata/genética , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Listeria monocytogenes/imunologia , Listeriose/genética , Listeriose/imunologia , Listeriose/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Espécies Reativas de Oxigênio/metabolismo , Canais de Cátion TRPM/genética
5.
Arterioscler Thromb Vasc Biol ; 38(2): 344-352, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29146750

RESUMO

OBJECTIVE: TRPM7 (transient receptor potential cation channel, subfamily M, member 7) is a ubiquitously expressed bifunctional protein comprising a transient receptor potential channel segment linked to a cytosolic α-type serine/threonine protein kinase domain. TRPM7 forms a constitutively active Mg2+ and Ca2+ permeable channel, which regulates diverse cellular processes in both healthy and diseased conditions, but the physiological role of TRPM7 kinase remains largely unknown. APPROACH AND RESULTS: Here we show that point mutation in TRPM7 kinase domain deleting the kinase activity in mice (Trpm7R/R ) causes a marked signaling defect in platelets. Trpm7R/R platelets showed an impaired PIP2 (phosphatidylinositol-4,5-bisphosphate) metabolism and consequently reduced Ca2+ mobilization in response to stimulation of the major platelet receptors GPVI (glycoprotein VI), CLEC-2 (C-type lectin-like receptor), and PAR (protease-activated receptor). Altered phosphorylation of Syk (spleen tyrosine kinase) and phospholipase C γ2 and ß3 accounted for these global platelet activation defects. In addition, direct activation of STIM1 (stromal interaction molecule 1) with thapsigargin revealed a defective store-operated Ca2+ entry mechanism in the mutant platelets. These defects translated into an impaired platelet aggregate formation under flow and protection of the mice from arterial thrombosis and ischemic stroke in vivo. CONCLUSIONS: Our results identify TRPM7 kinase as a key modulator of phospholipase C signaling and store-operated Ca2+ entry in platelets. The protection of Trpm7R/R mice from acute ischemic disease without developing intracranial hemorrhage indicates that TRPM7 kinase might be a promising antithrombotic target.


Assuntos
Arteriopatias Oclusivas/sangue , Plaquetas/metabolismo , Sinalização do Cálcio , Cálcio/sangue , Infarto da Artéria Cerebral Média/sangue , Canais de Cátion TRPM/sangue , Trombose/sangue , Animais , Arteriopatias Oclusivas/genética , Arteriopatias Oclusivas/patologia , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/patologia , Lectinas Tipo C/sangue , Camundongos Mutantes , Fosfatidilinositol 4,5-Difosfato/sangue , Fosfolipase C beta/sangue , Fosfolipase C gama/sangue , Fosforilação , Glicoproteínas da Membrana de Plaquetas/metabolismo , Mutação Puntual , Receptores Ativados por Proteinase/sangue , Molécula 1 de Interação Estromal/sangue , Sinaptofisina/sangue , Canais de Cátion TRPM/deficiência , Canais de Cátion TRPM/genética , Trombose/genética , Trombose/patologia
6.
PLoS One ; 12(5): e0178021, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28542209

RESUMO

Cigarette smoking is the primary cause of chronic obstructive pulmonary disease (COPD) with repeated and sustained infections linked to disease pathogenesis and exacerbations. The airway epithelium constitutes the first line of host defense against infection and is known to be impaired in COPD. We have previously identified Fatty Acid Binding Protein 5 (FABP5) as an important anti-inflammatory player during respiratory infections and showed that overexpression of FABP5 in primary airway epithelial cells protects against bacterial infection and inflammation. While cigarette smoke down regulates FABP5 expression, its mechanism remains unknown. In this report, we have identified three putative c-Jun binding sites on the FABP5 promoter and show that cigarette smoke inhibits the binding of c-Jun to its consensus sequence and prevents LPS-induced FABP5 expression. Using chromatin immunoprecipitation, we have determined that c-Jun binds the FABP5 promoter when stimulated with LPS but the presence of cigarette smoke greatly reduces this binding. Furthermore, cigarette smoke or a mutation in the c-Jun binding site inhibits LPS-induced FABP5 promoter activity. These data demonstrate that cigarette smoke interferes with FABP5 expression in response to bacterial infection. Thus, functional activation of FABP5 may be a new therapeutic strategy when treating COPD patients suffering from exacerbations.


Assuntos
Proteínas de Ligação a Ácido Graxo/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Fumaça/efeitos adversos , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Imunoprecipitação da Cromatina , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Proteínas de Ligação a Ácido Graxo/genética , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/química , Lipopolissacarídeos/toxicidade , Mutagênese Sítio-Dirigida , Regiões Promotoras Genéticas , Ligação Proteica , RNA Mensageiro/metabolismo , Nicotiana/química , Nicotiana/metabolismo
7.
Exp Hematol ; 44(7): 596-602.e3, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27033163

RESUMO

Transient potential receptor melastatin-2 (TRPM2) is a nonselective cationic, Ca(2+)-permeable transmembrane pore that is preferentially expressed in cells of the myeloid lineage and modulates signaling pathways converging into NF-kB. This is of potential interest for acute myeloid leukemia (AML) therapy, as NF-κB signaling is emerging as a key pathway, mediating drug resistance and leukemia-initiating cell survival in AML. Inhibition of NF-κB signaling has been found to be synergistic with chemotherapy. TRPM2 is overexpressed in AML compared with normal bone marrow, with the highest levels in the FAB M3-6 subtypes. To determine the effect of TRPM2 depletions in a defined genetic model, we established MLL-AF9-driven AML on a Trpm2(-/-) genetic background. Trpm2(-/-) MLL-AF9 leukemias displayed reduced NF-κB phosphorylation as well as nuclear translocation. In vivo, primary and secondary recipients of Trpm2(-/-) MLL-AF9 leukemias exhibit increased latency compared with recipients of wild-type leukemia cells. However, the difference in latency was small and was lost in tertiary transplants. The effect of loss of Trpm2 in a BCR-ABL/NUP98-HOXA9 fusion model was even smaller. Given reports that loss or inhibition of TRPM2 enhanced killing by DNA-damaging agents in neuroblastoma, breast cancer, and prostate cancer cell lines, we exposed Trpm2(-/-) and Trpm2(wt) primary MLL-AF9 leukemias to doxorubicin, cytarabine, and etoposide, but found no difference in IC50 values. The in vitro response to decitabine was also unaffected. In summary, Trpm2 does not seem to play a major role in myeloid leukemogenesis. Additionally, loss of Trpm2 does not augment the cytotoxicity of standard AML chemotherapeutic agents.


Assuntos
Transformação Celular Neoplásica/genética , Resistencia a Medicamentos Antineoplásicos/genética , Leucemia Mieloide Aguda/genética , Canais de Cátion TRPM/genética , Animais , Transformação Celular Neoplásica/metabolismo , Modelos Animais de Doenças , Regulação Leucêmica da Expressão Gênica , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/mortalidade , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Fosforilação , Prognóstico , Canais de Cátion TRPM/metabolismo
8.
Front Immunol ; 6: 375, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26257741

RESUMO

Although the concept of Ca(2+) as a universal messenger is well established, it was assumed that the regulatory mechanisms of Ca(2+)-signaling were divided along the line of electric excitability. Recent advances in molecular biology and genomics have, however, provided evidence that non-excitable cells such as immunocytes also express a wide and diverse pool of ion channels that does not differ as significantly from that of excitable cells as originally assumed. Ion channels and transporters are involved in virtually all aspects of immune response regulation, from cell differentiation and development to activation, and effector functions such as migration, antibody-secretion, phagosomal maturation, or vesicular delivery of bactericidal agents. This comprises TRP channel family members, voltage- and Ca(2+)-gated K(+)- and Na(+)-channels, as well as unexpectedly, components of the CaV1-subfamily of voltage-gated L-type Ca(2+)-channels, originally thought to be signature molecules of excitability. This article provides an overview of recent observations made in the field of CaV1 L-type channel function in the immune context, as well as presents results we obtained studying these channels in B-lymphocytes.

9.
Cell Mol Life Sci ; 71(24): 4853-67, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24858416

RESUMO

The channel kinases TRPM6 and TRPM7 are both members of the melastatin-related transient receptor potential (TRPM) subfamily of ion channels and the only known fusions of an ion channel pore with a kinase domain. TRPM6 and TRPM7 form functional, tetrameric channel complexes at the plasma membrane by heteromerization. TRPM6 was previously shown to cross-phosphorylate TRPM7 on threonine residues, but not vice versa. Genetic studies demonstrated that TRPM6 and TRPM7 fulfill non-redundant functions and that each channel contributes uniquely to the regulation of Mg(2+) homeostasis. Although there are indications that TRPM6 and TRPM7 can influence each other's cellular distribution and activity, little is known about the functional relationship between these two channel-kinases. In the present study, we examined how TRPM6 kinase activity influences TRPM7 serine phosphorylation, intracellular trafficking, and cell surface expression of TRPM7, as well as Mg(2+)-dependent cellular growth. We found TRPM7 serine phosphorylation via the TRPM6 kinase, but no TRPM6 serine phosphorylation via the TRPM7 kinase. Intracellular trafficking of TRPM7 was altered in HEK-293 epithelial kidney cells and DT40 B cells in the presence of TRPM6 with intact kinase activity, independently of the availability of extracellular Mg(2+), but TRPM6/7 surface labeling experiments indicate comparable levels of the TRPM6/7 channels at the plasma membrane. Furthermore, using a complementation approach in TRPM7-deficient DT40 B-cells, we demonstrated that wild-type TRPM6 inhibited cell growth under hypomagnesic cell culture conditions in cells co-expressing TRPM6 and TRPM7; however, co-expression of a TRPM6 kinase dead mutant had no effect-a similar phenotype was also observed in TRPM6/7 co-expressing HEK-293 cells. Our results provide first clues about how heteromer formation between TRPM6 and TRPM7 influences the biological activity of these ion channels. We show that TRPM6 regulates TRPM7 intracellular trafficking and TRPM7-dependent cell growth. All these effects are dependent upon the presence of an active TRPM6 kinase domain. Dysregulated Mg(2+)-homeostasis causes or exacerbates many pathologies. As TRPM6 and TRPM7 are expressed simultaneously in numerous cell types, understanding how their relationship impacts regulation of Mg(2+)-uptake is thus important knowledge.


Assuntos
Proliferação de Células , Magnésio/metabolismo , Proteínas Quinases/metabolismo , Canais de Cátion TRPM/metabolismo , Animais , Linfócitos B/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Células HEK293 , Homeostase , Humanos , Immunoblotting , Microscopia Confocal , Modelos Moleculares , Mutação , Fosforilação , Proteínas Quinases/química , Proteínas Quinases/genética , Multimerização Proteica , Proteínas Serina-Treonina Quinases , Estrutura Quaternária de Proteína , Transporte Proteico , Serina/genética , Serina/metabolismo , Canais de Cátion TRPM/química , Canais de Cátion TRPM/genética
10.
Magnes Res ; 27(1): 9-15, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24752033

RESUMO

Ion homeostasis dysregulations have severe effects on human health, impairing the effectiveness and appropriateness of major cellular events, including immune responses. The adverse effects of Mg(2+) deficiency on cellular physiology are well known and documented, but mechanistic insights into Mg(2+) sensitive signal transduction are still lacking. TRPM7 and its sister channel TRPM6 stand out as the only known fusions of an ion pore with a Ser/Thr kinase domain. Both channels are permeable to divalent cations and are central regulators of Mg(2+) homeostasis. One crucial aspect of TRPM7 function we have extensively studied is the relationship between its ion channel portion and its C-terminal Ser/Thr kinase domain. The modulation of ion channels by phosphorylation through exogenous kinases is common, however the covalent bound between the TRPM7 channel and its kinase suggests a novel kind of link between ion-entry and signal transduction events. Current knowledge supports a reciprocal "two-way street" model where TRPM7-kinase modulates ion transport function through Ser/Thr phosphorylation, and in turn, channel gating and ionic conditions in close proximity to the pore regulate TRPM7-kinase mediated signaling. We have shown that TRPM7 acts as a sensor of Mg(2+)-availability, adjusting key cellular functions such as the rate of cellular protein translation to the Mg(2+) nutritional status. Since molecular mechanisms controlling rates of protein translation are critical for cell growth and division in response to nutrient availability, this could have relevance for example for therapies targeted at molecules shaping the cancerous translational apparatus. In our quest to understand the biology of Mg(2+) in the context of immune responses, we found that TRPM7 associates with, and phosphorylates phospholipase C gamma 2 (PLCγ2), a pivotal molecule in the signaling pathway following B-cell receptor (BCR) activation. This contributes to the Mg(2+)-dependent modulation of the Ca(2+) response elicited by BCR ligation, and provides the first molecular pathway underlying the Mg(2+)-sensitivity of immune responses. Expanding our knowledge about the modulation of immunoreceptor signaling in response to Mg(2+) availability could allow for the development of unexplored strategies for therapeutic intervention in autoimmune diseases, immunodeficiencies, and lymphoma.


Assuntos
Magnésio/metabolismo , Transdução de Sinais , Canais de Cátion TRPM/metabolismo , Animais , Humanos
11.
Cell Signal ; 25(11): 2163-75, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23838006

RESUMO

We demonstrated a role for the Mg(2+) transporter TRPM7, a bifunctional protein with channel and α-kinase domains, in aldosterone signaling. Molecular mechanisms underlying this are elusive. Here we investigated the function of TRPM7 and its α-kinase domain on Mg(2+) and pro-inflammatory signaling by aldosterone. Kidney cells (HEK-293) expressing wild-type human TRPM7 (WThTRPM7) or constructs in which the α-kinase domain was deleted (ΔKinase) or rendered inactive with a point mutation in the ATP binding site of the α-kinase domain (K1648R) were studied. Aldosterone rapidly increased [Mg(2+)]i and stimulated NADPH oxidase-derived generation of reactive oxygen species (ROS) in WT hTRPM7 and TRPM7 kinase dead mutant cells. Translocation of annexin-1 and calpain-II and spectrin cleavage (calpain target) were increased by aldosterone in WT hTRPM7 cells but not in α-kinase-deficient cells. Aldosterone stimulated phosphorylation of MAP kinases and increased expression of pro-inflammatory mediators ICAM-1, Cox-2 and PAI-1 in Δkinase and K1648R cells, effects that were inhibited by eplerenone (mineralocorticoid receptor (MR) blocker). 2-APB, a TRPM7 channel inhibitor, abrogated aldosterone-induced Mg(2+) responses in WT hTRPM7 and mutant cells. In 2-APB-treated ΔKinase and K1648R cells, aldosterone-stimulated inflammatory responses were unchanged. These data indicate that aldosterone stimulates Mg(2+) influx and ROS production in a TRPM7-sensitive, kinase-insensitive manner, whereas activation of annexin-1 requires the TRPM7 kinase domain. Moreover TRPM7 α-kinase modulates inflammatory signaling by aldosterone in a TRPM7 channel/Mg(2+)-independent manner. Our findings identify novel mechanisms for non-genomic actions of aldosterone involving differential signaling through MR-activated TRPM7 channel and α-kinase.


Assuntos
Aldosterona/metabolismo , Magnésio/metabolismo , Proteínas Quinases/genética , Transdução de Sinais , Canais de Cátion TRPM/genética , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Anexina A1/metabolismo , Sítios de Ligação , Compostos de Boro/farmacologia , Calpaína/metabolismo , Eplerenona , Regulação da Expressão Gênica , Células HEK293 , Humanos , Transporte de Íons , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Fosforilação , Ligação Proteica , Proteínas Quinases/deficiência , Proteínas Serina-Treonina Quinases , Estrutura Terciária de Proteína , Espécies Reativas de Oxigênio/metabolismo , Espectrina/metabolismo , Espironolactona/análogos & derivados , Espironolactona/farmacologia , Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/metabolismo
12.
J Cereb Blood Flow Metab ; 33(10): 1549-55, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23801245

RESUMO

The calcium-permeable transient receptor potential M2 (TRPM2) ion channel was recently demonstrated to have a sexually dimorphic contribution to ischemic brain injury, with inhibition or knockdown of the channel protecting male brain preferentially. We tested the hypothesis that androgen signaling is required for this male-specific cell-death pathway. Additionally, we tested the hypothesis that differential activation of the enzyme poly (ADP-ribose) polymerase-1 (PARP-1) is responsible for male-specific TRPM2 channel activation and neuronal injury. We observed that administration of the TRPM2 inhibitor clotrimazole (CTZ) 2 hours after onset of ischemia reduced infarct volume in male mice and that protection from ischemic damage by CTZ was abolished by removal of testicular androgens (castration; CAST) and rescued by androgen replacement. Male PARP-1 knockout mice had reduced ischemic damage compared with WT mice and inhibition of TRPM2 with CTZ failed to reduce infarct size. Lastly, we observed that ischemia increased PARP activity in the peri-infarct region of male mice to a greater extent than female mice and the difference was abolished in CAST male mice. Data presented in the current study indicate that TRPM2-mediated neuronal death in the male brain requires intact androgen signaling and PARP-1 activity.


Assuntos
Androgênios/metabolismo , Isquemia Encefálica/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Caracteres Sexuais , Canais de Cátion TRPM/metabolismo , Animais , Isquemia Encefálica/enzimologia , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Morte Celular , Células Cultivadas , Di-Hidrotestosterona/administração & dosagem , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/enzimologia , Neurônios/metabolismo , Neurônios/patologia , Orquiectomia , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/genética , Cultura Primária de Células , Reação em Cadeia da Polimerase em Tempo Real , Receptores Androgênicos/metabolismo , Transdução de Sinais , Canais de Cátion TRPM/antagonistas & inibidores , Canais de Cátion TRPM/genética
13.
J Immunol ; 190(6): 2835-43, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23378430

RESUMO

MPYS (also known as STING, MITA, and TMEM173) is a type I IFN stimulator that is essential for host defense against DNA virus infection and appears important in defense against certain bacteria. The in vivo significance and mechanisms by which MPYS mediates host defense against nonviral pathogens are unknown. Using an MPYS-deficient mouse (Tmem173()), we determined that, distinct from the IFNAR(-/-) mice, MPYS deficiency leads to increased bacterial burden in the liver upon Listeria monocytogenes infection. The increase was correlated with the diminished MCP-1 and MCP-3 chemokine production and decreased blood and liver Ly6C(hi) monocyte frequency. We further demonstrate that MPYS-deficient Ly6C(hi) monocytes are intrinsically defective in migration to the liver. Lastly, adoptive transfer of wild-type Ly6C(hi) monocyte into MPYS-deficient mice decreases their liver bacterial burden. Our findings reveal a novel in vivo function of MPYS that is distinct from its role in activating type I IFN production.


Assuntos
Antígenos Ly/biossíntese , Movimento Celular/imunologia , Listeriose/imunologia , Listeriose/patologia , Proteínas de Membrana/fisiologia , Monócitos/imunologia , Animais , Movimento Celular/genética , Células Cultivadas , Modelos Animais de Doenças , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Listeriose/genética , Fígado/imunologia , Fígado/microbiologia , Fígado/patologia , Proteínas de Membrana/deficiência , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Monócitos/microbiologia , Monócitos/patologia , Baço/imunologia , Baço/microbiologia
14.
Immunol Res ; 55(1-3): 241-8, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22975787

RESUMO

TRPM2 (transient receptor potential melastatin 2) is the unique fusion of a Ca(2+)-permeable pore with an enzymatic domain that binds the NAD(+)-metabolite ADP-ribose (ADPR), resulting in channel opening. ADPR formation is a metabolic corollary of cellular stress, but can also be elicited enzymatically through NAD glycohydrolases like CD38. TRPM2 thus functions as a metabolic and oxidative stress sensor and translates this information into ion fluxes that can affect Ca(2+) signaling and the membrane potential. TRPM2 is strongly represented in immune cells of the phagocytic lineage, themselves professional generators of oxidants. The recent characterization of TRPM2-deficient mouse models has revealed the involvement of this channel in various aspects of immunity. Monocytes lacking TRPM2 show reduced production of the CXCL2 chemokine, resulting in diminished neutrophilic influx to the colon in chemically induced colitis, and thus protection against tissue ulceration in TRPM2(-/-) mice. However, the insufficient production of proinflammatory cytokines leads to high morbidity and lethality of the TRPM2(-/-) mice following infection with the bacterial pathogen Listeria monocytogenes. In the context of endotoxin-induced pulmonary inflammation, TRPM2's absence was found to promote inflammation and ROS production. TRPM2 acts thereby as a negative feedback loop by interfering through membrane depolarization with ROS generation by NADPH oxidases. In dendritic cells, TRPM2 is a lysosomal Ca(2+)-release channel that promotes chemokine responsiveness and cell migration, which is reminiscent of CD38-mediated functions. The discovery of TRPM2 has unveiled an unsuspected signaling pathway and established ADPR as a novel second messenger. Understanding TRPM2's complex involvement in inflammation is crucial to evaluating the potential of manipulating TRPM2 activity and ADPR metabolism for therapeutic intervention.


Assuntos
Canais de Cátion TRPM/imunologia , Animais , Citocinas/imunologia , Células Dendríticas/imunologia , Humanos , Imunidade Inata , Inflamação , Neurônios/imunologia , Estresse Oxidativo
15.
Immunol Res ; 55(1-3): 261-9, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22990458

RESUMO

The physiological and clinical relevance of Mg(2+) has evolved over the last decades. The molecular identification of multiple Mg(2+) transporters (Acdp2, MagT1, Mrs2, Paracellin-1, SLC41A1, SLC41A2, TRPM6 and TRPM7) and their biophysical characterization in recent years has improved our understanding of Mg(2+) homeostasis regulation and has provided a basis for investigating the role of Mg(2+) in the immune system. Deletions and mutations of Mg(2+) transporters produce severe phenotypes with more systemic symptoms than those seen with Ca(2+) channel deletions, which tend to be more specific and less profound. Deficiency of the Mg(2+) permeable ion channels TRPM6 or TRPM7 in mice is lethal at embryonic day 12.5 or at day 6.5, respectively, and, even more surprisingly, chicken DT40 B cells lacking TRPM7 die after 24-48 h. Recent progress made in Mg(2+) research has helped to define underlying mechanisms of two hereditary diseases, human Hypomagnesemia (TRPM6 deletion) and X-chromosomal immunodeficiency (MagT1 deletion), and has revealed a potential new role for Mg(2+) as a second messenger. Future elucidation of human Mg(2+) transporters (Mrs2, SLC41A1, SLC41A2, TRPM7) expressed in immunocytes, beyond MagT1 and TRPM6, will widen our knowledge about the potential role of Mg(2+) in the activation of the immune response.


Assuntos
Magnésio/imunologia , Animais , Linfócitos B/imunologia , Proteínas de Transporte de Cátions/imunologia , Humanos , Linfócitos T/imunologia , Canais de Cátion TRPM/imunologia
16.
Cell Signal ; 24(11): 2070-5, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22759789

RESUMO

PLC-isozymes are central elements of cellular signaling downstream of numerous receptors. PLCγ2 is a pivotal component of B cell receptor (BCR) signaling. The regulation of PLCγ2-dependent signaling functions by Tyr-phosphorylation is well characterized, however, the potential role of Ser/Thr phosphorylation events remains undefined. TRPM7 is the fusion of a Ser/Thr kinase with an ion channel, and an essential component of Mg(2+)-homeostasis regulation. Although the interaction between the C2 domain of several PLC-isozymes and TRPM7 is well established, previous studies have focused on the effect of PLC-activity on TRPM7. Here, we investigated whether Ser/Thr phosphorylation sites in the C2 domain of PLCγ2 could be identified using TRPM7-kinase. We show that TRPM7-kinase phosphorylates PLCγ2 in its C2-domain at position Ser1164 and in the linker region preceding the C2-domain at position Thr1045. Using a complementation approach in PLCγ2(-/-) DT40 cells, we found that the PLCγ2-S1164A mutant fully restores BCR mediated Ca(2+)-responses under standard growth conditions. However, under hypomagnesic conditions, PLCγ2-S1164A fails to reach Ca(2+)-levels seen in cells expressing PLCγ2 wildtype. These results suggest that Mg(2+)-sensitivity of the BCR signaling pathway may be regulated by Ser/Thr phosphorylation of PLCγ2.


Assuntos
Fosfolipase C gama/metabolismo , Canais de Cátion TRPM/metabolismo , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Linhagem Celular , Galinhas , Humanos , Magnésio/metabolismo , Dados de Sequência Molecular , Fosfolipase C gama/química , Fosforilação , Proteínas Serina-Treonina Quinases , Receptores de Antígenos de Linfócitos B/metabolismo , Serina/metabolismo , Transdução de Sinais , Treonina/metabolismo
17.
J Immunol ; 187(5): 2595-601, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21813776

RESUMO

Cyclic-di-GMP and cyclic-di-AMP are second messengers produced by bacteria and influence bacterial cell survival, differentiation, colonization, biofilm formation, virulence, and bacteria-host interactions. In this study, we show that in both RAW264.7 macrophage cells and primary bone marrow-derived macrophages, the production of IFN-ß and IL-6, but not TNF, in response to cyclic-di-AMP and cyclic-di-GMP requires MPYS (also known as STING, MITA, and TMEM173). Furthermore, expression of MPYS was required for IFN response factor 3 but not NF-κB activation in response to these bacterial metabolites. We also confirm that MPYS is required for type I IFN production by cultured macrophages infected with the intracellular pathogens Listeria monocytogenes and Francisella tularensis. However, during systemic infection with either pathogen, MPYS deficiency did not impact bacterial burdens in infected spleens. Serum IFN-ß and IL-6 concentrations in the infected control and MPYS(-/-) mice were also similar at 24 h postinfection, suggesting that these pathogens stimulate MPYS-independent cytokine production during in vivo infection. Our findings indicate that bifurcating MPYS-dependent and -independent pathways mediate sensing of cytosolic bacterial infections.


Assuntos
AMP Cíclico/imunologia , GMP Cíclico/análogos & derivados , Fator Regulador 3 de Interferon/imunologia , Interferon Tipo I/imunologia , Macrófagos/imunologia , Proteínas de Membrana/imunologia , Animais , Infecções Bacterianas/imunologia , Infecções Bacterianas/metabolismo , Linhagem Celular , AMP Cíclico/metabolismo , GMP Cíclico/imunologia , GMP Cíclico/metabolismo , Citocinas/biossíntese , Citocinas/imunologia , Ensaio de Imunoadsorção Enzimática , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/biossíntese , Macrófagos/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagócitos , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
Proc Natl Acad Sci U S A ; 108(28): 11578-83, 2011 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-21709234

RESUMO

The generation of reactive oxygen species (ROS) is inherent to immune responses. ROS are crucially involved in host defense against pathogens by promoting bacterial killing, but also as signaling agents coordinating the production of cytokines. Transient Receptor Potential Melastatin 2 (TRPM2) is a Ca(2+)-permeable channel gated via binding of ADP-ribose, a metabolite formed under conditions of cellular exposure to ROS. Here, we show that TRPM2-deficient mice are extremely susceptible to infection with Listeria monocytogenes (Lm), exhibiting an inefficient innate immune response. In a comparison with IFNγR-deficient mice, TRPM2(-/-) mice shared similar features of uncontrolled bacterial replication and reduced levels of inducible (i)NOS-expressing monocytes, but had intact IFNγ responsiveness. In contrast, we found that levels of cytokines IL-12 and IFNγ were diminished in TRPM2(-/-) mice following Lm infection, which correlated with their reduced innate activation. Moreover, TRPM2(-/-) mice displayed a higher degree of susceptibility than IL-12-unresponsive mice, and supplementation with recombinant IFNγ was sufficient to reverse the unrestrained bacterial growth and ultimately the lethal phenotype of Lm-infected TRPM2(-/-) mice. The severity of listeriosis we observed in TRPM2(-/-) mice has not been reported for any other ion channel. These findings establish an unsuspected role for ADP-ribose and ROS-mediated cation flux for innate immunity, opening up unique possibilities for immunomodulatory intervention through TRPM2.


Assuntos
Imunidade Inata/fisiologia , Listeria monocytogenes/imunologia , Canais de Cátion TRPM/imunologia , Adjuvantes Imunológicos/farmacologia , Animais , Citocinas/biossíntese , Feminino , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/genética , Interferon gama/farmacologia , Interleucina-12/deficiência , Interleucina-12/genética , Interleucina-12/imunologia , Subunidade beta 2 de Receptor de Interleucina-12/deficiência , Subunidade beta 2 de Receptor de Interleucina-12/genética , Subunidade beta 2 de Receptor de Interleucina-12/imunologia , Listeria monocytogenes/patogenicidade , Listeriose/imunologia , Listeriose/prevenção & controle , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/imunologia , Receptores de Interferon/deficiência , Receptores de Interferon/genética , Receptores de Interferon/imunologia , Proteínas Recombinantes , Canais de Cátion TRPM/deficiência , Canais de Cátion TRPM/genética , Receptor de Interferon gama
19.
FEBS Lett ; 585(14): 2275-8, 2011 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-21627970

RESUMO

Magnesium (Mg(2+)) transport across membranes plays an essential role in cellular growth and survival. TRPM7 is the unique fusion of a Mg(2+) permeable pore with an active cytosolic kinase domain, and is considered a master regulator of cellular Mg(2+) homeostasis. We previously found that the genetic deletion of TRPM7 in DT40 B cells results in Mg(2+) deficiency and severe growth impairment, which can be rescued by supplementation with excess extracellular Mg(2+). Here, we show that gene expression of the Mg(2+) selective transporter MagT1 is upregulated in TRPM7(-/-) cells. Furthermore, overexpression of MagT1 in TRPM7(-/-) cells augments their capacity to uptake Mg(2+), and improves their growth behavior in the absence of excess Mg(2+).


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Magnésio/metabolismo , Canais de Cátion TRPM/metabolismo , Animais , Proteínas de Transporte de Cátions/genética , Linhagem Celular , Galinhas , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Proteínas Serina-Treonina Quinases , Canais de Cátion TRPM/genética
20.
Cell Signal ; 23(3): 586-93, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21112387

RESUMO

Protein translation is an essential but energetically expensive process, which is carefully regulated in accordance to the cellular nutritional and energy status. Eukaryotic elongation factor 2 (eEF2) is a central regulation point since it mediates ribosomal translocation and can be inhibited by phosphorylation at Thr56. TRPM7 is the unique fusion of an ion channel with a functional Ser/Thr-kinase. While TRPM7's channel function has been implicated in regulating vertebrate Mg(2+) uptake required for cell growth, the function of its kinase domain remains unclear. Here, we show that under conditions where cell growth is limited by Mg(2+) availability, TRPM7 via its kinase mediates enhanced Thr56 phosphorylation of eEF2. TRPM7-kinase does not appear to directly phosphorylate eEF2, but rather to influence the amount of eEF2's cognate kinase eEF2-k, involving its phosphorylation at Ser77. These findings suggest that TRPM7's structural duality ensures ideal positioning of its kinase in close proximity to channel-mediated Mg(2+) uptake, allowing for the adjustment of protein translational rates to the availability of Mg(2+).


Assuntos
Quinase do Fator 2 de Elongação/metabolismo , Fator 2 de Elongação de Peptídeos/metabolismo , Canais de Cátion TRPM/fisiologia , Animais , Linfócitos B/enzimologia , Linhagem Celular , Galinhas , Humanos , Magnésio/metabolismo , Camundongos , Elongação Traducional da Cadeia Peptídica , Fosforilação , Biossíntese de Proteínas , Proteínas Serina-Treonina Quinases , Proteínas Recombinantes/biossíntese , Canais de Cátion TRPM/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...