Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
1.
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
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.
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
4.
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
5.
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
6.
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
7.
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
8.
Proc Natl Acad Sci U S A ; 105(46): 18029-34, 2008 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-19004782

RESUMO

Two related neurodegenerative disorders, Western Pacific amyotrophic lateral sclerosis (ALS) and parkinsonism-dementia (PD), originally occurred at a high incidence on Guam, in the Kii peninsula of Japan, and in southern West New Guinea more than 50 years ago. These three foci shared a unique mineral environment characterized by the presence of severely low levels of Ca(2+) and Mg(2+), coupled with high levels of bioavailable transition metals in the soil and drinking water. Epidemiological studies suggest that genetic factors also contribute to the etiology of these disorders. Here, we report that a variant of the transient receptor potential melastatin 2 (TRPM2) gene may confer susceptibility to these diseases. TRPM2 encodes a calcium-permeable cation channel highly expressed in the brain that has been implicated in mediating cell death induced by oxidants. We found a heterozygous variant of TRPM2 in a subset of Guamanian ALS (ALS-G) and PD (PD-G) cases. This variant, TRPM2(P1018L), produces a missense change in the channel protein whereby proline 1018 (Pro(1018)) is replaced by leucine (Leu(1018)). Functional studies revealed that, unlike WT TRPM2, P1018L channels inactivate. Our results suggest that the ability of TRPM2 to maintain sustained ion influx is a physiologically important function and that its disruption may, under certain conditions, contribute to disease states.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Proteínas Mutantes/metabolismo , Transtornos Parkinsonianos/metabolismo , Canais de Cátion TRPM/metabolismo , Adenosina Difosfato Ribose/farmacologia , Sequência de Aminoácidos , Diamino Aminoácidos/farmacologia , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Sequência Conservada , Toxinas de Cianobactérias , Evolução Molecular , Guam , Humanos , Peróxido de Hidrogênio/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Leucina/genética , Magnésio/farmacologia , Modelos Moleculares , Dados de Sequência Molecular , Mutação/genética , Estresse Oxidativo/efeitos dos fármacos , Prolina/genética , Canais de Cátion TRPM/química , Canais de Cátion TRPM/genética , Temperatura
9.
J Cell Physiol ; 225(2): 444-53, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20458742

RESUMO

Although most signaling responses initiated by tumor necrosis factor-alpha (TNF-alpha) occur in a Ca(2+)-independent fashion, TNF-alpha receptor signaling augments Ca(2+) entry induced by Galpha(q/11) G-protein coupled receptors (GPCRs) in endothelial cells and increases trans-endothelial permeability. The signaling events involved in GPCR-induced Ca(2+) influx have been characterized and involve store-operated Ca(2+) entry facilitated by the Ca(2+) permeable ion channel, transient receptor potential canonical 4 (TRPC4). Little is known about the mechanisms by which TNF-alpha receptor signaling augments GPCR-induced Ca(2+) entry. TNF-alpha Receptor Ubiquitous Signaling and Scaffolding protein (TRUSS) is a tumor necrosis factor receptor-1 (TNF-R1)-associated protein whose gene name is TRPC4-associated protein (TRPC4AP). The goal of our study was to test the hypothesis that TRUSS serves to link TNF-R1 and GPCR-signaling pathways at the level of TRPC4 by: (i) determining if TRUSS and TNF-R1 interact with TRPC4, and (ii) investigating the role of TRUSS, TNF-R1, and TRPC4 in GPCR-induced Ca(2+) signaling. Here, we show that TRUSS and TNF-R1 interact with a sub-family of TRPC channels (TRPC1, 4, and 5). In addition, we show that TRUSS and TNF-R1 function together with TRPC4 to elevate endoplasmic reticulum Ca(2+) filling in the context of reduced endoplasmic reticulum Ca(2+) storage initiated by G-protein coupled m1 muscarinic acetylcholine receptor (m1AchR) signaling. Together, these findings suggest that TNF-R1, TRUSS, and TRPC4 augment Ca(2+) loading of endoplasmic reticulum Ca(2+) stores in the context of m1AchR stimulation and provide new insights into the mechanisms that connect TNF-R1 to GPCR-induced Ca(2+) signaling.


Assuntos
Cálcio/metabolismo , Receptor Muscarínico M1/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Transdução de Sinais/fisiologia , Canais de Cátion TRPC/metabolismo , Linhagem Celular , Membrana Celular/fisiologia , Retículo Endoplasmático/metabolismo , Regulação da Expressão Gênica/fisiologia , Humanos , Permeabilidade , Receptor Muscarínico M1/genética , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Canais de Cátion TRPC/genética
10.
Am J Respir Crit Care Med ; 179(9): 816-26, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19201925

RESUMO

RATIONALE: Modulation of the activity of sarcoendoplasmic reticulum calcium ATPase (SERCA) can profoundly affect Ca(2+) homeostasis. Although altered calcium homeostasis is a characteristic of cystic fibrosis (CF), the role of SERCA is unknown. OBJECTIVES: This study provides a comprehensive investigation of expression and activity of SERCA in CF airway epithelium. A detailed study of the mechanisms underlying SERCA changes and its consequences was also undertaken. METHODS: Lung tissue samples (bronchus and bronchiole) from subjects with and without CF were evaluated by immunohistochemistry. Protein and mRNA expression in primary non-CF and CF cells was determined by Western and Northern blots. MEASUREMENTS AND MAIN RESULTS: SERCA2 expression was decreased in bronchial and bronchiolar epithelia of subjects with CF. SERCA2 expression in lysates of polarized tracheobronchial epithelial cells from subjects with CF was decreased by 67% as compared with those from subjects without CF. Several non-CF and CF airway epithelial cell lines were also probed. SERCA2 expression and activity were consistently decreased in CF cell lines. Adenoviral expression of mutant F508 cystic fibrosis transmembrane regulator gene (CFTR), inhibition of CFTR function pharmacologically (CFTR(inh)172), or stable expression of antisense oligonucleotides to inhibit CFTR expression caused decreased SERCA2 expression. In CF cells, SERCA2 interacted with Bcl-2, leading to its displacement from caveolae-related domains of endoplasmic reticulum membranes, as demonstrated in sucrose density gradient centrifugation and immunoprecipitation studies. Knockdown of SERCA2 using siRNA enhanced epithelial cell death due to ozone, hydrogen peroxide, and TNF-alpha. CONCLUSIONS: Reduced SERCA2 expression may alter calcium signaling and apoptosis in CF. These findings decrease the likelihood of therapeutic benefit of SERCA inhibition in CF.


Assuntos
Apoptose , Fibrose Cística/metabolismo , Fibrose Cística/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Estudos de Casos e Controles , Células Cultivadas , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Retículo Endoplasmático/metabolismo , Células Epiteliais/metabolismo , Humanos , Imuno-Histoquímica , Mutação , Mucosa Respiratória/metabolismo
11.
Am J Respir Cell Mol Biol ; 40(4): 433-42, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18931329

RESUMO

The multifunctional surface protein CD38 acts as a receptor with ecto-enzymatic activity, hydrolyzing NAD to generate several products known to exhibit Ca2+-mobilizing properties. Although CD38 is a convenient marker of immune cell development, and an indicator of progression for several diseases, it is not restricted to the immune compartment. To determine the potentially multilayered involvement of CD38 in allergen-induced airway inflammation and hyperreactivity, we dissected the potential role of CD38 as a regulator of immunity, but also pulmonary function. CD38-deficient and wild-type (WT) mice were sensitized and airway challenged with ovalbumin, and subsequently analyzed regarding their level of airway hyperresponsiveness (AHR) in response to methacholine. Parameters of lung inflammation were also analyzed. Similar sets of measurements were obtained from reciprocal bone marrow swapping experiments between CD38(-/-) and WT mice. Mice lacking CD38 exhibit strongly reduced AHR, which is accompanied by a decrease in typical hallmarks of pulmonary inflammation, including eosinophilia and lymphocytic lung infiltrates, as well as Th2-cytokine levels (IL-4, -5, and -13). Antigen-specific immunoglobulin (Ig)E and IgG1 antibody titers are substantially reduced, consistent with CD38 being crucial for mounting a primary humoral systemic immune response. Reconstitution of lethally irradiated, lung-shielded, CD38-deficient mice with WT bone marrow does not restore WT levels of airway hyperreactivity, nor mucus secretion. The opposite experiment, transferring CD38(-/-) bone marrow into WT mice, also shows reduced AHR levels. These studies demonstrate that CD38 not only acts as a key modulator of the immune response, but also plays an equally important role as an intrinsic pulmonary component.


Assuntos
ADP-Ribosil Ciclase 1/imunologia , Alérgenos/imunologia , Hipersensibilidade Respiratória/imunologia , ADP-Ribosil Ciclase 1/deficiência , Animais , Formação de Anticorpos , Medula Óssea/imunologia , Células Cultivadas , Quimera , Citocinas/biossíntese , Imunização , Imunoglobulina E/sangue , Imunoglobulina G/sangue , Chumbo , Pulmão/imunologia , Pulmão/patologia , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia , Pneumonia/sangue , Pneumonia/imunologia , Pneumonia/patologia , Proteção Radiológica , Hipersensibilidade Respiratória/sangue , Hipersensibilidade Respiratória/patologia , Células Th2/imunologia
12.
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
13.
J Biomol Screen ; 13(1): 54-61, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18057180

RESUMO

TRPM2 is a member of the transient receptor potential melastatin (TRPM)-related ion channel family. The activation of TRPM2 induced by oxidative/nitrosative stress leads to an increase in intracellular free Ca(2+). Although further progress in understanding TRPM2's role in cell and organism physiology would be facilitated by isolation of compounds able to specifically modulate its function in primary cells or animal models, no cell-based assays for TRPM2 function well suited for high-throughput screening have yet been described. Here, a novel suspension B lymphocyte cell line stably expressing TRPM2 was used to develop a cell-based assay. The assay uses the Ca(2+)-sensitive fluorescence dye, Fluo-4 NW (no wash), to measure TRPM2-dependent Ca(2+) transients induced by H(2)O(2) and N-methyl-N'-nitrosoguanidine in a 96-well plate format. Assay performance was evaluated by statistical analysis of the Z' factor value and was consistently greater than 0.5 under optimal conditions, suggesting that the assay is very robust. For assay validation, the effects of known inhibitors of TRPM2 and TRPM2 gating secondary messenger production were determined. Overall, the authors have developed a cell-based assay that may be used to identify TRPM2 ion channel modulators from large compound libraries.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Canais de Cátion TRPM/efeitos dos fármacos , Compostos de Anilina , Animais , Linhagem Celular , Galinhas , Corantes Fluorescentes , Humanos , Cinética , Proteínas Recombinantes/efeitos dos fármacos , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Canais de Cátion TRPM/genética , Canais de Cátion TRPM/metabolismo , Transfecção , Xantenos
14.
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
15.
Subcell Biochem ; 40: 257-70, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17623910

RESUMO

The DT40 B-lymphocyte cell line is a chicken bursal lymphocyte tumor cell line which grows rapidly, expresses a variety of types of constitutive and signal dependent ion transport systems., and supports the efficient use of stable and conditional genetic manipulations. Below, we review the use of DT40 cells in dissecting molecular mechanisms involved in Ca2+, Mg2+, and Zn2+ transport physiology. These studies highlight the flexibility and advantages the DT40 environment offers to investigators interested in the study of basic vertebrate ion transport physiology.


Assuntos
Sinalização do Cálcio , Homeostase , Canais Iônicos/fisiologia , Animais , Linfócitos B/citologia , Linhagem Celular , Galinhas , Zinco/fisiologia
16.
Subcell Biochem ; 40: 373-8, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17623923

RESUMO

Study of Mg2+ homeostasis in continuously growing cells requires the capacity to measure total cellular Mg2+ and net Mg2+ fluxes per unit time. Our laboratory's protocols for measurement of total cellular Mg2+ by atomic absorption spectrophotometry and measurement of net Mg2+ fluxes using the stable Mg2+ isotope 26Mg are described below.


Assuntos
Linfócitos B/metabolismo , Magnésio/metabolismo , Animais , Linhagem Celular , Galinhas , Espectrometria de Massas/métodos , Espectrofotometria Atômica
17.
Magnes Res ; 20(1): 6-18, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17536484

RESUMO

Over the past decades, the clinical relevance and biological significance of Mg2+ have been thoroughly documented. Although multiple Mg2+-transport pathways have been biophysically characterized, the molecular identity of the postulated components of Mg2+-homeostasis regulation in vertebrates remain undefined. Recent advances in the fields of genetics, genomics and proteomics, and novel technologies such as cDNA microarrays have allowed for substantial progress in this area. The mitochondrial Mrs2 protein was the first human Mg2+ transporter characterized as such, and an important element for future analyses of the role of mitochondria in managing intracellular Mg2+. Several molecules with Mg2+ transport capabilities have been identified through a screen designed to find genes upregulated under hypomagnesic conditions. This includes SLC41A1 and 2, ACDP2 and MagT1. Finally, the elucidation of the molecular cause underlying two different hereditary diseases leading to hypomagnesemia resulted in the cloning and characterization of claudin 16 (paracellin-1), and TRPM6. Whereas claudin 16 plays a crucial role in paracellular Mg2+ transport, TRPM6 is involved in the transcellular pathway. TRPM6 and its closest relative TRPM7 are both puzzling ion channel-kinase fusions, and perhaps the most unexpected newly identified players in the regulation of Mg2+-homeostasis in vertebrates.


Assuntos
Cátions/metabolismo , Canais Iônicos/metabolismo , Deficiência de Magnésio/sangue , Magnésio/metabolismo , Proteínas Quinases/metabolismo , Canais de Cátion TRPM/fisiologia , Animais , Transporte Biológico/fisiologia , Claudinas , Homeostase , Humanos , Canais Iônicos/química , Proteínas de Membrana/fisiologia , Mitocôndrias/química , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Proteínas Quinases/química , Proteínas Serina-Treonina Quinases , Vertebrados
18.
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
19.
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
20.
J Mol Biol ; 332(2): 385-98, 2003 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-12948489

RESUMO

Human ADP-ribose pyrophosphatase NUDT9 belongs to a superfamily of Nudix hydrolases that catabolize potentially toxic compounds in the cell. The enzyme hydrolyzes ADP-ribose (ADPR) to AMP and ribose 5'-phosphate. NUDT9 shares 39% sequence identity with the C-terminal cytoplasmic domain of the ADPR-gated calcium channel TRPM2, which exhibits low but specific enzyme activity. We determined crystal structures of NUDT9 in the presence and in the absence of the reaction product ribose 5'-phosphate. On the basis of these structures and comparison with a bacterial homologue, a model of the substrate complex was built. The structure and activity of a double point mutant (R(229)E(230)F(231) to R(229)I(230)L(231)), which mimics the Nudix signature of the ion channel domain, was determined. Finally, the activities of a pair of additional mutated constructs were compared to the wild-type enzyme. The first corresponds to a minimal Nudix domain missing an N-terminal domain and C-terminal tail; the second disrupts two potential general bases in the active site. NUDT9 contains an N-terminal domain with a novel fold and a catalytic C-terminal Nudix domain. Unlike its closest functional homologue (homodimeric Escherichia coli ADPRase), it is active as a monomer, and the substrate is bound in a cleft between the domains. The structure of the RIL mutant provides structural basis for the reduced activity of the TRPM2 ion channel. The conformation and binding interactions of ADPR substrate are predicted to differ from those observed for E.coli ADPRase; mutation of structurally aligned acidic residues in their active sites produce significantly different effects on catalytic efficiency, indicating that their reaction pathways and mechanisms may have diverged.


Assuntos
Estrutura Terciária de Proteína , Pirofosfatases/química , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Domínio Catalítico , Cristalografia por Raios X , Humanos , Magnésio/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Ligação Proteica , Pirofosfatases/genética , Pirofosfatases/metabolismo , Ribosemonofosfatos/metabolismo , Alinhamento de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA