Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
PLoS Pathog ; 19(2): e1011168, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36812267

RESUMO

Angiotensin-converting enzyme 2 (ACE2), part of the renin-angiotensin system (RAS), serves as an entry point for SARS-CoV-2, leading to viral proliferation in permissive cell types. Using mouse lines in which the Ace2 locus has been humanized by syntenic replacement, we show that regulation of basal and interferon induced ACE2 expression, relative expression levels of different ACE2 transcripts, and sexual dimorphism in ACE2 expression are unique to each species, differ between tissues, and are determined by both intragenic and upstream promoter elements. Our results indicate that the higher levels of expression of ACE2 observed in the lungs of mice relative to humans may reflect the fact that the mouse promoter drives expression of ACE2 in populous airway club cells while the human promoter drives expression in alveolar type 2 (AT2) cells. In contrast to transgenic mice in which human ACE2 is expressed in ciliated cells under the control of the human FOXJ1 promoter, mice expressing ACE2 in club cells under the control of the endogenous Ace2 promoter show a robust immune response after infection with SARS-CoV-2, leading to rapid clearance of the virus. This supports a model in which differential expression of ACE2 determines which cell types in the lung are infected, and this in turn modulates the host response and outcome of COVID-19.


Assuntos
Enzima de Conversão de Angiotensina 2 , COVID-19 , Receptores Virais , Animais , Humanos , Camundongos , Enzima de Conversão de Angiotensina 2/genética , COVID-19/genética , Camundongos Transgênicos , Receptores Virais/genética , SARS-CoV-2 , Tropismo Viral
2.
J Immunol ; 188(8): 4093-102, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22412193

RESUMO

Elevated PGE(2) is a hallmark of most inflammatory lesions. This lipid mediator can induce the cardinal signs of inflammation, and the beneficial actions of nonsteroidal anti-inflammatory drugs are attributed to inhibition of cyclooxygenase (COX)-1 and COX-2, enzymes essential in the biosynthesis of PGE(2) from arachidonic acid. However, both clinical studies and rodent models suggest that, in the asthmatic lung, PGE(2) acts to restrain the immune response and limit physiological change secondary to inflammation. To directly address the role of PGE(2) in the lung, we examined the development of disease in mice lacking microsomal PGE(2) synthase-1 (mPGES1), which converts COX-1/COX-2-derived PGH(2) to PGE(2). We show that mPGES1 determines PGE(2) levels in the naive lung and is required for increases in PGE(2) after OVA-induced allergy. Although loss of either COX-1 or COX-2 increases the disease severity, surprisingly, mPGES1(-/-) mice show reduced inflammation. However, an increase in serum IgE is still observed in the mPGES1(-/-) mice, suggesting that loss of PGE(2) does not impair induction of a Th2 response. Furthermore, mPGES1(-/-) mice expressing a transgenic OVA-specific TCR are also protected, indicating that PGE(2) acts primarily after challenge with inhaled Ag. PGE(2) produced by the lung plays the critical role in this response, as loss of lung mPGES1 is sufficient to protect against disease. Together, this supports a model in which mPGES1-dependent PGE(2) produced by populations of cells native to the lung contributes to the effector phase of some allergic responses.


Assuntos
Ciclo-Oxigenase 1/imunologia , Ciclo-Oxigenase 2/imunologia , Dinoprostona/imunologia , Hipersensibilidade/imunologia , Oxirredutases Intramoleculares/imunologia , Pulmão/imunologia , Proteínas de Membrana/imunologia , Animais , Proliferação de Células , Ciclo-Oxigenase 1/deficiência , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 2/deficiência , Ciclo-Oxigenase 2/genética , Citocinas/biossíntese , Citocinas/imunologia , Dinoprostona/metabolismo , Feminino , Hipersensibilidade/metabolismo , Hipersensibilidade/patologia , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Oxirredutases Intramoleculares/deficiência , Oxirredutases Intramoleculares/genética , Pulmão/metabolismo , Pulmão/patologia , Masculino , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovalbumina , Prostaglandina-E Sintases , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Células Th2/imunologia , Células Th2/metabolismo
3.
J Immunol ; 189(4): 2006-16, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22753929

RESUMO

Acute inflammation in response to both exogenous and endogenous danger signals can lead to the assembly of cytoplasmic inflammasomes that stimulate the activation of caspase-1. Subsequently, caspase-1 facilitates the maturation and release of cytokines and also, under some circumstances, the induction of cell death by pyroptosis. Using a mouse line lacking expression of NLRP1, we show that assembly of this inflammasome in cells is triggered by a toxin from anthrax and that it initiates caspase-1 activation and release of IL-1ß. Furthermore, NLRP1 inflammasome activation also leads to cell death, which escalates over 3 d following exposure to the toxin and culminates in acute lung injury and death of the mice. We show that these events are not dependent on production of IL-1ß by the inflammasome but are dependent on caspase-1 expression. In contrast, muramyl dipeptide-mediated inflammasome formation is not dependent on NLRP1 but NLRP3. Taken together, our findings show that assembly of the NLRP1 inflammasome is sufficient to initiate pyroptosis, which subsequently leads to a self-amplifying cascade of cell injury within the lung from which the lung cannot recover, eventually resulting in catastrophic consequences for the organism.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Inflamassomos/metabolismo , Lesão Pulmonar Aguda/imunologia , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Animais , Apoptose/imunologia , Proteínas Reguladoras de Apoptose/imunologia , Caspase 1/metabolismo , Citometria de Fluxo , Inflamassomos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout
4.
Immunol Cell Biol ; 90(7): 733-42, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22249203

RESUMO

ONZIN is abundantly expressed in immune cells of both the myeloid and lymphoid lineage. Expression by lymphoid cells has been reported to further increase after cutaneous exposure of mice to antigens and haptens capable of inducing contact hypersensitivity (CHS), suggesting that ONZIN has a critical role in this response. Here, we report that indeed ONZIN-deficient mice develop attenuated CHS to a number of different haptens. Dampened CHS responses correlated with a significant reduction in pro-inflammatory IL-6 at the challenge site in ONZIN-deficient animals, compared with wild-type controls. Together the study of these animals indicates that loss of ONZIN impacts the effector phase of the CHS response through the regulation of pro-inflammatory factors.


Assuntos
Dermatite de Contato/imunologia , Haptenos/imunologia , Interleucina-6/imunologia , Proteínas Oncogênicas/imunologia , Transferência Adotiva/métodos , Animais , Apresentação de Antígeno/imunologia , Western Blotting , Transplante de Medula Óssea/métodos , Quimiocina CXCL1/genética , Quimiocina CXCL1/imunologia , Quimiocina CXCL1/metabolismo , Dermatite de Contato/genética , Dermatite de Contato/metabolismo , Orelha Externa/imunologia , Orelha Externa/metabolismo , Ensaio de Imunoadsorção Enzimática , Feminino , Expressão Gênica/imunologia , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Interleucina-6/genética , Interleucina-6/metabolismo , Linfonodos/imunologia , Linfonodos/metabolismo , Linfonodos/transplante , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
5.
Am J Physiol Renal Physiol ; 301(5): F1035-41, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21835766

RESUMO

Prostaglandins have been implicated as paracrine regulators of renin secretion, but the specific pathways and receptor(s) carrying out these functions have not been fully elucidated. To examine the contributions of prostanoid synthetic pathways and receptors to regulation of renin in the intact animal, we used a panel of mice with targeted disruption of several key genes: cyclooxygenase-2 (COX-2), microsomal PGE synthases 1 and 2 (mPGES1, mPGES2), EP2 and EP4 receptors for PGE(2), and the IP receptor for PGI(2). To activate the macula densa signal for renin stimulation, mice were treated with furosemide over 5 days and renin mRNA levels were determined by real-time RT-PCR. At baseline, there were no differences in renin mRNA levels between wild-type and the various strains of mutant mice. Furosemide caused marked stimulation of renin mRNA expression across all groups of wild-type control mice. This response was completely abrogated in the absence of COX-2, but was unaffected in mice lacking mPGES1 or mPGES2. The absence of G(s)/cAMP-linked EP2 receptors had no effect on stimulation of renin by furosemide and there was only a modest, insignificant reduction in renin responses in mice lacking the IP receptor. By contrast, renin stimulation in EP4(-/-) mice was significantly reduced by ∼70% compared with wild-type controls. These data suggest that stimulation of renin by the macula densa mechanism is mediated by PGE(2) through a pathway requiring COX-2 and the EP4 receptor, but not EP2 or IP receptors. Surprisingly, mPGES1 or mPGES2 are not required, suggesting other alternative mechanisms for generating PGE(2) in response to macula densa stimulation.


Assuntos
Receptores de Prostaglandina E Subtipo EP4/fisiologia , Renina/fisiologia , Animais , Pressão Sanguínea , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , DNA Complementar/biossíntese , DNA Complementar/genética , Dinoprostona/fisiologia , Diuréticos/farmacologia , Furosemida/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Endogâmicos , Camundongos Knockout , RNA/biossíntese , RNA/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real , Renina/biossíntese , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto , Estimulação Química , Telemetria
6.
ACS Infect Dis ; 7(12): 3303-3313, 2021 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-34752055

RESUMO

Multidrug-resistant bacterial infections have become a global threat. We recently disclosed that the known IKK-ß inhibitor IMD-0354 and subsequent analogues abrogate colistin resistance in several Gram-negative strains. Herein, we report the activity of a second-generation library of IMD-0354 analogues incorporating a benzimidazole moiety as an amide isostere. We identified several analogues that show increased colistin potentiation activity against Gram-negative bacteria.


Assuntos
Colistina , Salicilanilidas , Antibacterianos/farmacologia , Benzimidazóis/farmacologia , Colistina/farmacologia , Testes de Sensibilidade Microbiana
7.
Chem Biol Drug Des ; 96(5): 1180-1186, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32562384

RESUMO

The mounting threat of multi-drug-resistant (MDR) bacteria places a tremendous strain on the antimicrobial clinical arsenal, forcing physicians to revert to near-obsolete antibiotics to treat otherwise intractable infections. Antibiotic adjuvant therapy has emerged as a viable alternative to the development of novel antimicrobial agents. This method uses combinations of an existing antibiotic and a non-antimicrobial small molecule, where the combination either breaks drug resistance or further potentiates antibiotic activity. Through a high-content screen of eukaryotic kinase inhibitors, our group previously identified two highly potent adjuvants that synergize with colistin, a cyclic, polycationic antimicrobial peptide that serves as a drug of last resort for the treatment of MDR Gram-negative bacterial infections. Cell signaling proteins implicated in colistin resistance mechanisms display both kinase and phosphatase activities. Herein, we explore the potential for eukaryotic phosphatase inhibitors to be repurposed as colistin adjuvants. From a panel of 48 unique structures, we discovered that the natural product kuwanon G breaks colistin resistance, while the non-antimicrobial macrolide ascomycin potentiates colistin in polymyxin-susceptible bacteria.


Assuntos
Antibacterianos/farmacologia , Colistina/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Monoéster Fosfórico Hidrolases/antagonistas & inibidores , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Sinergismo Farmacológico , Eucariotos/enzimologia , Flavonoides/farmacologia , Bactérias Gram-Negativas/crescimento & desenvolvimento , Testes de Sensibilidade Microbiana , Tacrolimo/análogos & derivados , Tacrolimo/farmacologia
8.
Environ Health Perspect ; 128(8): 87003, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32779937

RESUMO

BACKGROUND: Chronic exposure to inorganic arsenic (iAs) is a significant public health problem. Methylation of iAs by arsenic methyltransferase (AS3MT) controls iAs detoxification and modifies risks of iAs-induced diseases. Mechanisms underlying these diseases have been extensively studied using animal models. However, substantive differences between humans and laboratory animals in efficiency of iAs methylation have hindered the translational potential of the laboratory studies. OBJECTIVES: The goal of this study was to determine whether humanization of the As3mt gene confers a human-like pattern of iAs metabolism in mice. METHODS: We generated a mouse strain in which the As3mt gene along with the adjacent Borcs7 gene was humanized by syntenic replacement. We compared expression of the mouse As3mt and the human AS3MT and the rate and pattern of iAs metabolism in the wild-type and humanized mice. RESULTS: AS3MT expression in mouse tissues closely modeled that of human and differed substantially from expression of As3mt. Detoxification of iAs was much less efficient in the humanized mice than in wild-type mice. Profiles for iAs and its methylated metabolites in tissues and excreta of the humanized mice were consistent with those reported in humans. Notably, the humanized mice expressed both the full-length AS3MT that catalyzes iAs methylation and the human-specific AS3MTd2d3 splicing variant that has been linked to schizophrenia. CONCLUSIONS: These results suggest that AS3MT is the primary genetic locus responsible for the unique pattern of iAs metabolism in humans. Thus, the humanized mouse strain can be used to study the role of iAs methylation in the pathogenesis of iAs-induced diseases, as well as to evaluate the role of AS3MTd2d3 in schizophrenia. https://doi.org/10.1289/EHP6943.


Assuntos
Arsênio/metabolismo , Metiltransferases/metabolismo , Animais , Arsenicais , Humanos , Metiltransferases/genética , Camundongos
9.
ChemMedChem ; 15(2): 210-218, 2020 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-31756025

RESUMO

Infections caused by multidrug-resistant (MDR) bacteria, particularly Gram-negative bacteria, are an escalating global health threat. Often clinicians are forced to administer the last-resort antibiotic colistin; however, colistin resistance is becoming increasingly prevalent, giving rise to the potential for a situation in which there are no treatment options for MDR Gram-negative infections. The development of adjuvants that circumvent bacterial resistance mechanisms is a promising orthogonal approach to the development of new antibiotics. We recently disclosed that the known IKK-ß inhibitor IMD-0354 potently suppresses colistin resistance in several Gram-negative strains. In this study, we explore the structure-activity relationship (SAR) between the IMD-0354 scaffold and colistin resistance suppression, and identify several compounds with more potent activity than the parent against highly colistin-resistant strains of Acinetobacter baumannii and Klebsiella pneumoniae.


Assuntos
Acinetobacter baumannii/efeitos dos fármacos , Adjuvantes Farmacêuticos/farmacologia , Antibacterianos/farmacologia , Benzamidas/farmacologia , Klebsiella pneumoniae/efeitos dos fármacos , Adjuvantes Farmacêuticos/síntese química , Adjuvantes Farmacêuticos/química , Antibacterianos/síntese química , Antibacterianos/química , Benzamidas/síntese química , Benzamidas/química , Colistina/farmacologia , Relação Dose-Resposta a Droga , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Humanos , Testes de Sensibilidade Microbiana , Estrutura Molecular , Relação Estrutura-Atividade
10.
Prostaglandins Other Lipid Mediat ; 88(3-4): 73-81, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19010439

RESUMO

Prostaglandin E(2) (PGE(2)) plays an important role in the normal physiology of many organ systems. Increased levels of this lipid mediator are associated with many disease states, and it potently regulates inflammatory responses. Three enzymes capable of in vitro synthesis of PGE(2) from the cyclooxygenase metabolite PGH(2) have been described. Here, we examine the contribution of one of these enzymes to PGE(2) production, mPges-2, which encodes microsomal prostaglandin synthase-2 (mPGES-2), by generating mice homozygous for the null allele of this gene. Loss of mPges-2 expression did not result in a measurable decrease in PGE(2) levels in any tissue or cell type examined from healthy mice. Taken together, analysis of the mPGES-2 deficient mouse lines does not substantiate the contention that mPGES-2 is a PGE(2) synthase.


Assuntos
Dinoprostona/biossíntese , Oxirredutases Intramoleculares/fisiologia , Animais , Northern Blotting , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Interferon gama/farmacologia , Oxirredutases Intramoleculares/genética , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Mutantes , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase , Prostaglandina-E Sintases
11.
ACS Med Chem Lett ; 10(5): 828-833, 2019 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-31098007

RESUMO

Antibiotic resistance has significantly increased since the beginning of the 21st century. Currently, the polymyxin colistin is typically viewed as the antibiotic of last resort for the treatment of multidrug resistant Gram-negative bacterial infections. However, increased colistin usage has resulted in colistin-resistant bacterial isolates becoming more common. The recent dissemination of plasmid-borne colistin resistance genes (mcr 1-8) into the human pathogen pool is further threatening to render colistin therapy ineffective. New methods to combat antibiotic resistant pathogens are needed. Herein, the utilization of a colistin-adjuvant combination that is effective against colistin-resistant bacteria is described. At 5 µM, the lead adjuvant, which is nontoxic to the bacteria alone, increases colistin efficacy 32-fold against bacteria containing the mcr-1 gene and effects a 1024-fold increase in colistin efficacy against bacteria harboring chromosomally encoded colistin resistance determinants; these combinations lower the colistin minimum inhibitory concentration (MIC) to or below clinical breakpoint levels (≤2 µg/mL).

12.
PLoS One ; 14(11): e0225449, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31747445

RESUMO

Environmental and endogenous electrophiles cause tissue damage through their high reactivity with endogenous nucleophiles such as DNA, proteins, and lipids. Protection against damage is mediated by glutathione (GSH) conjugation, which can occur spontaneously or be facilitated by the glutathione S-transferase (GST) enzymes. To determine the role of GST enzymes in protection against electrophiles as well as the role of specific GST families in mediating this protection, we exposed mutant mouse lines lacking the GSTP, GSTM, and/or GSTT enzyme families to the model electrophile acrylamide, a ubiquitous dietary contaminant known to cause adverse effects in humans. An analysis of urinary metabolites after acute acrylamide exposure identified the GSTM family as the primary mediator of GSH conjugation to acrylamide. However, surprisingly, mice lacking only this enzyme family did not show increased toxicity after an acute acrylamide exposure. Therefore, GSH conjugation is not the sole mechanism by which GSTs protect against the toxicity of this substrate. Given the prevalence of null GST polymorphisms in the human population (approximately 50% for GSTM1 and 20-50% for GSTT1), a substantial portion of the population may also have impaired acrylamide metabolism. However, our study also defines a role for GSTP and/or GSTT in protection against acrylamide mediated toxicity. Thus, while the canonical detoxification function of GSTs may be impaired in GSTM null individuals, disease risk secondary to acrylamide exposure may be mitigated through non-canonical pathways involving members of the GSTP and/or GSTT families.


Assuntos
Acrilamida/toxicidade , Compostos de Epóxi/toxicidade , Deleção de Genes , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Fígado/patologia , Animais , Modelos Animais de Doenças , Feminino , Glutationa/urina , Humanos , Inativação Metabólica , Isoenzimas/genética , Isoenzimas/metabolismo , Fígado/enzimologia , Masculino , Camundongos , Testes de Mutagenicidade
13.
ACS Infect Dis ; 5(10): 1764-1771, 2019 10 11.
Artigo em Inglês | MEDLINE | ID: mdl-31434474

RESUMO

Kinase inhibitors comprise a diverse cohort of chemical scaffolds that are active in multiple biological systems. Currently, thousands of eukaryotic kinase inhibitors are commercially available, have well-characterized targets, and often carry pharmaceutically favorable toxicity profiles. Recently, our group disclosed that derivatives of the natural product meridianin D, a known inhibitor of eukaryotic kinases, modulated behaviors of both Gram-positive and Gram-negative bacteria. Herein, we expand our exploration of kinase inhibitors in Gram-negative bacilli utilizing three commercially available kinase inhibitor libraries and, ultimately, identify two chemical structures that potentiate colistin (polymyxin E) in multiple strains. We report IMD-0354, an inhibitor of IKK-ß, as a markedly effective adjuvant in colistin-resistant bacteria and also describe AR-12 (OSU-03012), an inhibitor of pyruvate dehydrogenase kinase-1 (PDK-1), as a potentiator in colistin-sensitive strains. This report comprises the first description of the novel cross-reactivity of these molecules.


Assuntos
Adjuvantes Farmacêuticos/farmacologia , Colistina/farmacologia , Bactérias Gram-Negativas/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Adjuvantes Farmacêuticos/química , Benzamidas/farmacologia , Linhagem Celular , Colistina/química , Combinação de Medicamentos , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Eucariotos , Bactérias Gram-Positivas/efeitos dos fármacos , Quinase I-kappa B/efeitos dos fármacos , Lipídeo A , Testes de Sensibilidade Microbiana , Pirazóis/farmacologia , Piruvato Desidrogenase Quinase de Transferência de Acetil/efeitos dos fármacos , Sulfonamidas/farmacologia
14.
Cell Rep ; 24(5): 1254-1265, 2018 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-30067980

RESUMO

Lysosomes play a critical role in maintenance of the integrity of neuronal function, and mutations in genes that contribute to lysosome formation, transport, and activity are associated with neurodegenerative disorders. Recently, the multisubunit complex, BLOC-one-related complex (BORC), has been shown to be involved in positioning lysosomes within the cytoplasm, although the consequences of altered BORC function in adult animals have not been established. We show that a spontaneous truncation mutation in the mouse Borcs7 gene, identified through whole-genome sequencing followed by genetic complementation, results in progressive axonal dystrophy with dramatic impairment of motor function. Furthermore, mice homozygous for deletion of the entire Borcs7 coding sequence die shortly after birth, and neurons cultured from these animals show impaired centrifugal transport of lysosomes. This identifies BORCS7 as a central factor in axonal transport of lysosomes and a possible target for improving disease-related disturbances in this important function.


Assuntos
Transporte Axonal , Proteínas de Membrana Lisossomal/genética , Distrofias Neuroaxonais/genética , Animais , Feminino , Locomoção , Mutação com Perda de Função , Lisossomos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neurônios/metabolismo
15.
J Immunol ; 178(4): 2065-74, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17277110

RESUMO

Francisella tularensis is a highly infectious bacterial pathogen, and is likely to have evolved strategies to evade and subvert the host immune response. In this study, we show that F. tularensis infection of macrophages alters T cell responses in vitro, by blocking T cell proliferation and promoting a Th2-like response. We demonstrate that a soluble mediator is responsible for this effect and identify it as PGE(2). Supernatants from F. tularensis-infected macrophages inhibited IL-2 secretion from both MHC class I and MHC class II-restricted T cell hybridomas, as well as enhanced a Th2-like response by inducing increased production of IL-5. Furthermore, the soluble mediator blocked proliferation of naive MHC class I-restricted T cells when stimulated with cognate tetramer. Indomethacin treatment partially restored T cell proliferation and lowered IL-5 production to wild-type levels. Macrophages produced PGE(2) when infected with F. tularensis, and treatment of infected macrophages with indomethacin, a cyclooxygenase-1/cyclooxygenase-2 inhibitor, blocked PGE(2) production. To further demonstrate that PGE(2) was responsible for skewing of T cell responses, we infected macrophages from membrane PGE synthase 1 knockout mice (mPGES1(-/-)) that cannot produce PGE(2). Supernatants from F. tularensis-infected membrane PGE synthase 1(-/-) macrophages did not inhibit T cell proliferation. Furthermore, treatment of T cells with PGE(2) recreated the effects seen with infected supernatant. From these data, we conclude that F. tularensis can alter host T cell responses by causing macrophages to produce PGE(2). This study defines a previously unknown mechanism used by F. tularensis to modulate adaptive immunity.


Assuntos
Proliferação de Células , Dinoprostona/imunologia , Francisella tularensis/imunologia , Macrófagos/imunologia , Células Th2/imunologia , Tularemia/imunologia , Animais , Proliferação de Células/efeitos dos fármacos , Ciclo-Oxigenase 1/imunologia , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/imunologia , Ciclo-Oxigenase 2/metabolismo , Inibidores de Ciclo-Oxigenase/farmacologia , Dinoprostona/metabolismo , Francisella tularensis/metabolismo , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Hibridomas , Imunidade Inata/efeitos dos fármacos , Indometacina/farmacologia , Interleucina-2/imunologia , Interleucina-2/metabolismo , Interleucina-5/imunologia , Interleucina-5/metabolismo , Macrófagos/metabolismo , Macrófagos/microbiologia , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Células Th2/metabolismo , Tularemia/metabolismo
16.
J Immunol ; 178(5): 3208-18, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17312169

RESUMO

The retinoid-related orphan receptors (ROR) comprise a distinct subfamily of nuclear receptors with the capacity to act as both repressors and activators of transcription. RORgamma, the most recently identified member of the ROR family, has been shown to be important for the development of normal lymphocyte compartments as well as organogenesis of some lymphoid organs. In this report, we examine the capacity of RORgamma-deficient mice to develop an adaptive immune response to Ag using OVA-induced inflammation in mice as a model for allergic airway disease. In sham-treated mice lacking RORgamma, low-grade pulmonary inflammation was observed and characterized by the perivascular accumulation of B and T lymphocytes, increased numbers of inflammatory cells in the lung lavage fluid, and polyclonal Ig activation. Following sensitization and challenge, the capacity of these animals to develop the allergic phenotype was severely impaired as evidenced by attenuated eosinophilic pulmonary inflammation, reduced numbers of CD4+ lymphocytes, and lower Th2 cytokines/chemokine protein and mRNA expression in the lungs. IFN-gamma and IL-10 production was markedly greater in splenocytes from RORgamma-deficient mice following in vitro restimulation with OVA compared with wild-type splenocytes, and a shift toward a Th1 immune response was observed in sensitized/challenged RORgamma-deficient animals in vivo. These data reveal a critical role for RORgamma in the regulation of Ig production and Th1/Th2 balance in adaptive immunity.


Assuntos
Alérgenos/imunologia , Formação de Anticorpos/imunologia , Receptores do Ácido Retinoico/imunologia , Receptores dos Hormônios Tireóideos/imunologia , Hipersensibilidade Respiratória/imunologia , Células Th1/imunologia , Células Th2/imunologia , Alérgenos/toxicidade , Animais , Formação de Anticorpos/genética , Interferon gama/imunologia , Interleucina-10/imunologia , Camundongos , Camundongos Mutantes , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares , Receptores do Ácido Retinoico/deficiência , Receptores dos Hormônios Tireóideos/deficiência , Hipersensibilidade Respiratória/induzido quimicamente , Hipersensibilidade Respiratória/genética , Hipersensibilidade Respiratória/patologia , Células Th1/patologia , Células Th2/patologia
17.
Am J Physiol Lung Cell Mol Physiol ; 290(1): L105-13, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16113047

RESUMO

Prostaglandin E2 (PGE2), similar to beta-adrenergic receptor agonists, can protect airways from bronchoconstriction and resulting increase in airway resistance induced by a number of agents, including cholinergic receptor agonists and antigen. We examined the impact of sustained alterations in PGE2 pathways on changes in airway resistance. Genetic methods were utilized to alter PGE2 metabolism and signal transduction in the murine lung. PGE2 levels were elevated by generating mice lacking 15-hydroxyprostaglandin (Hpgd-/-), the major catabolic enzyme of PGE2, and by generating a transgenic line in which mouse PGE2 synthase (Ptges) expression is driven by a human lung-specific promoter, hSP-C. Conversely, to determine the impact of loss of PGE2 on airway reactivity, we examined mice lacking this synthase (Ptges-/-) and receptors that mediate the actions of PGE2, particularly the PGE2 EP2 receptor (Ptger2). Diminished capacity to produce and respond to PGE2 did not alter the response of mice to cholinergic stimuli. In contrast, the responsiveness to cholinergic stimulation was dramatically altered in animals with elevated PGE2 levels. The Hpgd-/- and hSP-C-Ptges transgenic lines both showed attenuated airway responsiveness to methacholine as measured by lung resistance. Thus, whereas compromise of the Ptges/PGE2/Ptger2 pathway does not alter airway responsiveness, genetic modulation that elevates PGE2 levels in the lung attenuates airway responsiveness.


Assuntos
Resistência das Vias Respiratórias/fisiologia , Brônquios/metabolismo , Broncoconstrição/fisiologia , Dinoprostona/fisiologia , Animais , Brônquios/fisiologia , Humanos , Hidroxiprostaglandina Desidrogenases/deficiência , Oxirredutases Intramoleculares/deficiência , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/metabolismo , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Camundongos Transgênicos , Regiões Promotoras Genéticas , Prostaglandina-E Sintases
18.
Am J Physiol Lung Cell Mol Physiol ; 291(2): L144-56, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16473862

RESUMO

Prostacyclin is one of a number of lipid mediators elaborated from the metabolism of arachidonic acid by the cyclooxygenase (COX) enzymes. This prostanoid is a potent inhibitor of platelet aggregation, and its production by endothelial cells and protective role in the vasculature are well established. In contrast, much less is known regarding the function of this prostanoid in other disease processes. We show here that COX-2-dependent production of prostacyclin plays an important role in the development of fibrotic lung disease, limiting both the development of fibrosis and the consequential alterations in lung mechanics. In stark contrast, loss of prostaglandin E(2) synthesis and signaling through the G(s)-coupled EP2 and EP4 receptors had no effect on the development of disease. These findings suggest that prostacyclin analogs will protect against bleomycin-induced pulmonary fibrosis in COX-2(-/-) mice. If such protection is observed, investigation of these agents as a novel therapeutic approach to pulmonary fibrosis in humans may be warranted.


Assuntos
Antibióticos Antineoplásicos/toxicidade , Bleomicina/toxicidade , Ciclo-Oxigenase 2/metabolismo , Epoprostenol/metabolismo , Fibrose Pulmonar , Animais , Ciclo-Oxigenase 1/genética , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/genética , Pulmão/citologia , Pulmão/patologia , Pulmão/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Prostaglandinas/metabolismo , Fibrose Pulmonar/induzido quimicamente , Fibrose Pulmonar/metabolismo
19.
Am J Physiol Lung Cell Mol Physiol ; 291(5): L1005-17, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16829631

RESUMO

A genetic contribution to asthma susceptibility is well recognized, and linkage studies have identified a large number of genes associated with asthma pathogenesis. Recently, a locus encoding a seven-transmembrane protein was shown to be associated with asthma in founder populations. The expression of the protein GPRA (G protein-coupled receptor for asthma susceptibility) in human airway epithelia and smooth muscle, and its increased expression in a mouse model of asthma, suggested that a gain-of-function mutation in this gene increased the disease risk. However, we report here that the development of allergic lung disease in GPRA-deficient mice is unaltered. A possible explanation for this finding became apparent upon reexamination of the expression of this gene. In contrast to initial studies, our analyses failed to detect expression of GPRA in human lung tissue or in mice with allergic lung disease. We identify a single parameter that distinguishes GPRA-deficient and wild-type mice. Whereas the change in airway resistance in response to methacholine was identical in control and GPRA-deficient mice, the mutant animals showed an attenuated response to thromboxane, a cholinergic receptor-dependent bronchoconstricting agent. Together, our studies fail to support a direct contribution of GPRA to asthma pathogenesis. However, our data suggest that GPRA may contribute to the asthmatic phenotype by altering the activity of other pathways, such as neurally mediated mechanisms, that contribute to disease. This interpretation is supported by high levels of GPRA expression in the brain and its recent identification as the neuropeptide S receptor.


Assuntos
Asma/fisiopatologia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Doença Aguda , Anafilaxia/imunologia , Anafilaxia/metabolismo , Anafilaxia/fisiopatologia , Animais , Asma/imunologia , Asma/metabolismo , Broncoconstritores/farmacologia , Modelos Animais de Doenças , Expressão Gênica/imunologia , Humanos , Hipotálamo/fisiologia , Lipopolissacarídeos/farmacologia , Pulmão/citologia , Pulmão/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Músculo Liso/fisiologia , Ovalbumina/imunologia , Ovalbumina/farmacologia , Fenótipo , Pneumonia/imunologia , Pneumonia/metabolismo , Pneumonia/fisiopatologia , Mecânica Respiratória , Retina/fisiologia
20.
J Lipid Res ; 46(12): 2636-48, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16204198

RESUMO

The mammary gland, like most tissues, produces measurable amounts of prostaglandin E2 (PGE2), a metabolite of arachidonic acid produced by sequential actions of two cyclooxygenases (COX-1 and COX-2) and three terminal PGE synthases: microsomal prostaglandin E2 synthase-1 (mPGES1), mPGES2, and cytosolic prostaglandin E2 synthase (cPGES). High PGE2 levels and COX-2 overexpression are frequently detected in mammary tumors and cell lines. However, less is known about PGE2 metabolic enzymes in the context of normal mammary development. Additionally, the primary COX partnerships of terminal PGE synthases and their contribution to normal mammary PGE2 biosynthesis are poorly understood. We demonstrate that expression of COX-1, generally considered constitutive, increases dramatically with lactogenic differentiation of the murine mammary gland. Concordantly, total PGE2 levels increase throughout mammary development, with highest levels measured in lactating tissue and breast milk. In contrast, COX-2 expression is extremely low, with only a modest increase detected during mammary involution. Expression of the G(s)-coupled PGE2 receptors, EP2 and EP4, is also temporally regulated, with highest levels detected at stages of maximal proliferation. PGE2 production is dependent on COX-1, as PGE2 levels are nearly undetectable in COX-1-deficient mammary glands. Interestingly, PGE2 levels are similarly reduced in lactating glands of mPGES1-deficient mice, indicating that PGE2 biosynthesis results from the coordinated activity of COX-1 and mPGES1. We thus provide evidence for the first time of functional coupling between COX-1 and mPGES1 in the murine mammary gland in vivo.


Assuntos
Ciclo-Oxigenase 1/metabolismo , Dinoprostona/biossíntese , Glândulas Mamárias Animais/metabolismo , Proteínas de Membrana/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Animais , Ciclo-Oxigenase 1/deficiência , Ciclo-Oxigenase 1/genética , Feminino , Regulação Enzimológica da Expressão Gênica/genética , Glândulas Mamárias Animais/crescimento & desenvolvimento , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Gravidez , Prostaglandina-Endoperóxido Sintases/deficiência , Prostaglandina-Endoperóxido Sintases/genética , Receptores de Prostaglandina E/genética , Receptores de Prostaglandina E Subtipo EP2 , Receptores de Prostaglandina E Subtipo EP4
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA