Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Glycobiology ; 32(2): 136-147, 2022 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-34939104

RESUMO

Polysaccharide A (PSA) is the immunodominant capsular carbohydrate from the gram negative commensal microbe Bacteroides fragilis that has shown remarkable potency in ameliorating many rodent models of inflammatory disease by eliciting downstream suppressive CD4+ T cells. PSA is composed of a zwitterionic repeating unit that allows it to be processed by antigen presenting cells (APCs) and presented by MHCII in a glycosylation-dependent manner. While previous work has uncovered much about the interactions between MHCII and PSA, as well as the downstream T cell response, little is known about how PSA affects the phenotype of MHCII+ APCs, including macrophages. Here, we utilized an unbiased systems approach consisting of RNAseq transcriptomics, high-throughput flow cytometry, Luminex analysis and targeted validation experiments to characterize the impact of PSA-mediated stimulation of splenic MHCII+ cells. The data revealed that PSA potently elicited the upregulation of an alternatively activated M2 macrophage transcriptomic and cell surface signature. Cell-type-specific validation experiments further demonstrated that PSA-exposed bone marrow-derived macrophages (BMDMs) induced cell surface and intracellular markers associated with M2 macrophages compared with conventional peptide ovalbumin (ova)-exposed BMDMs. In contrast to macrophages, we also found that CD11c+ dendritic cells (DCs) upregulated the pro-T cell activation costimulatory molecule CD86 following PSA stimulation. Consistent with the divergent BMDM and DC changes, PSA-exposed DCs elicited an antigen-experienced T cell phenotype in co-cultures, whereas macrophages did not. These findings collectively demonstrate that the PSA-induced immune response is characterized by both T cell stimulation via presentation by DCs, and a previously unrecognized anti-inflammatory polarization of macrophages.


Assuntos
Células Apresentadoras de Antígenos , Antígeno Prostático Específico , Animais , Anti-Inflamatórios/metabolismo , Células Apresentadoras de Antígenos/metabolismo , Células Dendríticas , Humanos , Macrófagos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Polissacarídeos/metabolismo , Antígeno Prostático Específico/metabolismo
2.
Gastroenterology ; 162(1): 166-178, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34606847

RESUMO

BACKGROUND & AIMS: Fecal microbiota transplantation (FMT) is an emerging treatment modality for ulcerative colitis (UC). Several randomized controlled trials have shown efficacy for FMT in the treatment of UC, but a better understanding of the transferable microbiota and their immune impact is needed to develop more efficient microbiome-based therapies for UC. METHODS: Metagenomic analysis and strain tracking was performed on 60 donor and recipient samples receiving FMT for active UC. Sorting and sequencing of immunoglobulin (Ig) A-coated microbiota (called IgA-seq) was used to define immune-reactive microbiota. Colonization of germ-free or genetically engineered mice with patient-derived strains was performed to determine the mechanism of microbial impact on intestinal immunity. RESULTS: Metagenomic analysis defined a core set of donor-derived transferable bacterial strains in UC subjects achieving clinical response, which predicted response in an independent trial of FMT for UC. IgA-seq of FMT recipient samples and gnotobiotic mice colonized with donor microbiota identified Odoribacter splanchnicus as a transferable strain shaping mucosal immunity, which correlated with clinical response and the induction of mucosal regulatory T cells. Colonization of mice with O splanchnicus led to an increase in Foxp3+/RORγt+ regulatory T cells, induction of interleukin (IL) 10, and production of short chain fatty acids, all of which were required for O splanchnicus to limit colitis in mouse models. CONCLUSIONS: This work provides the first evidence of transferable, donor-derived strains that correlate with clinical response to FMT in UC and reveals O splanchnicus as a key component promoting both metabolic and immune cell protection from colitis. These mechanistic features will help enable strategies to enhance the efficacy of microbial therapy for UC. Clinicaltrials.gov ID NCT02516384.


Assuntos
Bacteroidetes/imunologia , Colite/terapia , Colo/microbiologia , Transplante de Microbiota Fecal , Microbioma Gastrointestinal , Imunoglobulina A/imunologia , Mucosa Intestinal/microbiologia , Animais , Bacteroidetes/genética , Bacteroidetes/metabolismo , Ensaios Clínicos como Assunto , Colite/imunologia , Colite/metabolismo , Colite/microbiologia , Colite Ulcerativa/diagnóstico , Colite Ulcerativa/imunologia , Colite Ulcerativa/metabolismo , Colite Ulcerativa/microbiologia , Colo/imunologia , Colo/metabolismo , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/metabolismo , Microbioma Gastrointestinal/genética , Microbioma Gastrointestinal/imunologia , Vida Livre de Germes , Humanos , Imunidade nas Mucosas , Imunoglobulina A/genética , Imunoglobulina A/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Linfócitos Intraepiteliais/imunologia , Linfócitos Intraepiteliais/metabolismo , Linfócitos Intraepiteliais/microbiologia , Metagenoma , Metagenômica , Camundongos Endogâmicos C57BL , Camundongos Knockout , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/microbiologia , Resultado do Tratamento
3.
Front Immunol ; 11: 556813, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33193325

RESUMO

Polysaccharide A (PSA), a capsular carbohydrate from the commensal gut bacteria Bacteroides fragilis, has been shown to possess both potent T cell-dependent pro- and anti-inflammatory properties. PSA is able to induce abscess and adhesion formation in sepsis models, but can also inhibit asthma, inflammatory bowel disease (IBD) and experimental autoimmune encephalomyelitis (EAE) through MHCII-dependent activation of CD4+ T cells. Yet, despite decades of study, the ability of PSA to balance both these pro- and anti-inflammatory responses remains poorly understood. Here, we utilized an unbiased systems immunology approach consisting of RNAseq transcriptomics, high-throughput flow cytometry, and Luminex analysis to characterize the full impact of PSA-mediated stimulation of CD4+ T cells. We found that exposure to PSA resulted in the upregulation and secretion of IFNγ, TNFα, IL-6, and CXCL10, consistent with an interferon responsive gene (IRG) signature. Importantly, PSA stimulation also led to expression of immune checkpoint markers Lag3, Tim3, and, especially, PD1, which were also enriched and sustained in the gut associated lymphoid tissue of PSA-exposed mice. Taken together, PSA responding cells display an unusual mixture of pro-inflammatory cytokines and anti-inflammatory surface receptors, consistent with the ability to both cause and inhibit inflammatory disease.


Assuntos
Biomarcadores , Imunomodulação/genética , Interferons/metabolismo , Polissacarídeos Bacterianos/imunologia , Transcriptoma , Animais , Perfilação da Expressão Gênica , Ativação Linfocitária , Tecido Linfoide/imunologia , Tecido Linfoide/metabolismo , Camundongos , Transdução de Sinais , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
4.
Nature ; 581(7809): 475-479, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32461639

RESUMO

Intestinal health relies on the immunosuppressive activity of CD4+ regulatory T (Treg) cells1. Expression of the transcription factor Foxp3 defines this lineage, and can be induced extrathymically by dietary or commensal-derived antigens in a process assisted by a Foxp3 enhancer known as conserved non-coding sequence 1 (CNS1)2-4. Products of microbial fermentation including butyrate facilitate the generation of peripherally induced Treg (pTreg) cells5-7, indicating that metabolites shape the composition of the colonic immune cell population. In addition to dietary components, bacteria modify host-derived molecules, generating a number of biologically active substances. This is epitomized by the bacterial transformation of bile acids, which creates a complex pool of steroids8 with a range of physiological functions9. Here we screened the major species of deconjugated bile acids for their ability to potentiate the differentiation of pTreg cells. We found that the secondary bile acid 3ß-hydroxydeoxycholic acid (isoDCA) increased Foxp3 induction by acting on dendritic cells (DCs) to diminish their immunostimulatory properties. Ablating one receptor, the farnesoid X receptor, in DCs enhanced the generation of Treg cells and imposed a transcriptional profile similar to that induced by isoDCA, suggesting an interaction between this bile acid and nuclear receptor. To investigate isoDCA in vivo, we took a synthetic biology approach and designed minimal microbial consortia containing engineered Bacteroides strains. IsoDCA-producing consortia increased the number of colonic RORγt-expressing Treg cells in a CNS1-dependent manner, suggesting enhanced extrathymic differentiation.


Assuntos
Bactérias/metabolismo , Ácidos e Sais Biliares/química , Ácidos e Sais Biliares/metabolismo , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Sequência de Aminoácidos , Animais , Bacteroides/metabolismo , Colo/microbiologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Feminino , Fermentação , Microbioma Gastrointestinal , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Consórcios Microbianos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo
5.
Cell Host Microbe ; 24(3): 353-363.e5, 2018 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-30122655

RESUMO

Colonic wound repair is an orchestrated process, beginning with barrier re-establishment and followed by wound channel formation and crypt regeneration. Elevated levels of prostaglandin E2 (PGE2) promote barrier re-establishment; however, we found that persistently elevated PGE2 hinders subsequent repair phases. The bacterial metabolite deoxycholate (DCA) promotes transition through repair phases via PGE2 regulation. During barrier re-establishment, DCA levels are locally diminished in the wound, allowing enhanced PGE2 production and barrier re-establishment. However, during transition to the wound channel formation phase, DCA levels increase to inhibit PGE2 production and promote crypt regeneration. Altering DCA levels via antibiotic treatment enhances PGE2 levels but impairs wound repair, which is rescued with DCA treatment. DCA acts via its receptor, farnesoid X receptor, to inhibit the enzyme cPLA2 required for PGE2 synthesis. Thus, colonic wound repair requires temporally regulated signals from microbial metabolites to coordinate host-associated signaling cascades. VIDEO ABSTRACT.


Assuntos
Bactérias/metabolismo , Colo/lesões , Colo/fisiologia , Ácido Desoxicólico/metabolismo , Microbioma Gastrointestinal/fisiologia , Mucosa Intestinal/lesões , Cicatrização , Animais , Biópsia , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Hidroxiprostaglandina Desidrogenases/farmacologia , Mucosa Intestinal/fisiologia , Camundongos , Camundongos Knockout , Nitrobenzenos/farmacologia , Cultura Primária de Células , Sulfonamidas/farmacologia , Vancomicina/farmacologia
6.
Stem Cells Dev ; 22(16): 2326-40, 2013 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-23517237

RESUMO

Clinically available red blood cells (RBCs) for transfusions are at high demand, but in vitro generation of RBCs from hematopoietic stem cells requires significant quantities of growth factors. Here, we describe the production of four human growth factors: erythropoietin (EPO), stem cell factor (SCF), interleukin 3 (IL-3), and insulin-like growth factor-1 (IGF-1), either as non-fused proteins or as fusions with a carrier molecule (lichenase), in plants, using a Tobacco mosaic virus vector-based transient expression system. All growth factors were purified and their identity was confirmed by western blotting and peptide mapping. The potency of these plant-produced cytokines was assessed using TF1 cell (responsive to EPO, IL-3 and SCF) or MCF-7 cell (responsive to IGF-1) proliferation assays. The biological activity estimated here for the cytokines produced in plants was slightly lower or within the range cited in commercial sources and published literature. By comparing EC50 values of plant-produced cytokines with standards, we have demonstrated that all four plant-produced growth factors stimulated the expansion of umbilical cord blood-derived CD34+ cells and their differentiation toward erythropoietic precursors with the same potency as commercially available growth factors. To the best of our knowledge, this is the first report on the generation of all key bioactive cytokines required for the erythroid development in a cost-effective manner using a plant-based expression system.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Eritropoetina/farmacologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/farmacologia , Interleucina-3/farmacologia , Nicotiana/genética , Fator de Células-Tronco/farmacologia , Agrobacterium tumefaciens/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Clonagem Molecular/métodos , Eritrócitos/citologia , Eritrócitos/metabolismo , Eritropoetina/biossíntese , Eritropoetina/genética , Eritropoetina/isolamento & purificação , Sangue Fetal/citologia , Sangue Fetal/efeitos dos fármacos , Sangue Fetal/metabolismo , Expressão Gênica , Vetores Genéticos , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Fator de Crescimento Insulin-Like I/biossíntese , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/isolamento & purificação , Interleucina-3/biossíntese , Interleucina-3/genética , Interleucina-3/isolamento & purificação , Plantas Geneticamente Modificadas , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/farmacologia , Fator de Células-Tronco/biossíntese , Fator de Células-Tronco/genética , Fator de Células-Tronco/isolamento & purificação , Nicotiana/metabolismo , Nicotiana/virologia , Vírus do Mosaico do Tabaco/genética , Transgenes
7.
J Biol Chem ; 285(22): 17209-17, 2010 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-20212047

RESUMO

Stem cell biology offers advantages to investigators seeking to identify new therapeutic molecules. Specifically, stem cells are genetically stable, scalable for molecular screening, and function in cellular assays for drug efficacy and safety. A key hurdle for drug discoverers of central nervous system disease is a lack of high quality neuronal cells. In the central nervous system, alpha-amino-3-hydroxyl-5-methyl-4-isoxazolepropionate (AMPA) subtype glutamate receptors mediate the vast majority of excitatory neurotransmissions. Embryonic stem (ES) cell protocols were developed to differentiate into neuronal subtypes that express AMPA receptors and were pharmacologically responsive to standard compounds for AMPA potentiation. Therefore, we hypothesized that stem cell-derived neurons should be predictive in high-throughput screens (HTSs). Here, we describe a murine ES cell-based HTS of a 2.4 x 10(6) compound library, the identification of novel chemical "hits" for AMPA potentiation, structure function relationship of compounds and receptors, and validation of chemical leads in secondary assays using human ES cell-derived neurons. This reporting of murine ES cell derivatives being formatted to deliver HTS of greater than 10(6) compounds for a specific drug target conclusively demonstrates a new application for stem cells in drug discovery. In the future new molecular entities may be screened directly in human ES or induced pluripotent stem cell derivatives.


Assuntos
Células-Tronco Embrionárias/citologia , Neurônios/metabolismo , Receptores de AMPA/química , Receptores de Glutamato/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/farmacologia , Animais , Química Farmacêutica/métodos , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Fluorometria/métodos , Humanos , Imuno-Histoquímica/métodos , Camundongos , Modelos Biológicos , Mutação , Tecnologia Farmacêutica/métodos
8.
Am J Physiol Cell Physiol ; 293(4): C1209-15, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17652426

RESUMO

Cellular growth and differentiation are readouts of multiple signaling pathways from the intercellular and/or extracellular milieu. The extracellular matrix through the activation of cellular receptors transmits these signals. Therefore, extracellular matrix proteolysis could affect cell fate in a variety of biological events. However, the biological consequence of inadequate extracellular matrix degradation in vivo is not clear. We developed a mouse model expressing human collagenase (matrix metalloproteinase-1, MMP-1) under the control of Col2a1 promoter. The mice showed significant growth retardation during embryogenesis and a loss of the demarcation of zonal structure and columnar array of the cartilage. Immunological examination revealed increased degradation of type II collagen and upregulation of fibronectin and alpha(5)-integrin subunit in the transgenic cartilage. The resting zone and proliferating zone of the growth plate cartilage exhibited a simultaneous increase in bromodeoxyuridine (BrdU)-incorporated proliferating cells and terminal deoxynucleotidyl transferase-mediated X-dUTP nick-end labeling-positive apoptotic cells, respectively. Chondrocyte differentiation was not disturbed in the transgenic mice as evidenced by normal expression of the Ihh and type X collagen expression. These data demonstrate that type II collagen proteolysis is an important determinant for the skeletal outgrowth through modulation of chondrocyte survival and cartilagenous growth.


Assuntos
Desenvolvimento Ósseo/fisiologia , Colágeno Tipo II/metabolismo , Embrião de Mamíferos/metabolismo , Retardo do Crescimento Fetal/metabolismo , Metaloproteinase 1 da Matriz/metabolismo , Animais , Animais Recém-Nascidos , Apoptose/genética , Apoptose/fisiologia , Northern Blotting , Western Blotting , Desenvolvimento Ósseo/genética , Cartilagem/anormalidades , Cartilagem/metabolismo , Condrócitos/metabolismo , Condrócitos/patologia , Colágeno Tipo II/genética , Embrião de Mamíferos/anormalidades , Retardo do Crescimento Fetal/genética , Retardo do Crescimento Fetal/patologia , Fibronectinas/metabolismo , Expressão Gênica , Lâmina de Crescimento/anormalidades , Lâmina de Crescimento/metabolismo , Humanos , Integrina alfa5/metabolismo , Metaloproteinase 1 da Matriz/genética , Camundongos , Camundongos Endogâmicos , Camundongos Transgênicos , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
9.
Proc Natl Acad Sci U S A ; 104(25): 10619-24, 2007 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-17537919

RESUMO

Bone is accrued and maintained primarily through the coupled actions of bone-forming osteoblasts and bone-resorbing osteoclasts. Cumulative in vitro studies indicated that proline-rich tyrosine kinase 2 (PYK2) is a positive mediator of osteoclast function and activity. However, our investigation of PYK2-/- mice did not reveal evidence supporting an essential function for PYK2 in osteoclasts either in vivo or in culture. We find that PYK2-/- mice have high bone mass resulting from an unexpected increase in bone formation. Consistent with the in vivo findings, mouse bone marrow cultures show that PYK2 deficiency enhances differentiation and activity of osteoprogenitor cells, as does expressing a PYK2-specific short hairpin RNA or dominantly interfering proteins in human mesenchymal stem cells. Furthermore, the daily administration of a small-molecule PYK2 inhibitor increases bone formation and protects against bone loss in ovariectomized rats, an established preclinical model of postmenopausal osteoporosis. In summary, we find that PYK2 regulates the differentiation of early osteoprogenitor cells across species and that inhibitors of the PYK2 have potential as a bone anabolic approach for the treatment of osteoporosis.


Assuntos
Quinase 2 de Adesão Focal/fisiologia , Osteoblastos/fisiologia , Osteoclastos/fisiologia , Osteogênese/fisiologia , Osteoporose/terapia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/fisiologia , Diferenciação Celular , Células Cultivadas , Inibidores Enzimáticos/uso terapêutico , Feminino , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Camundongos , Camundongos Knockout , Ovariectomia , Ratos , Ratos Sprague-Dawley
10.
Toxicol Sci ; 90(1): 149-58, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16357009

RESUMO

Embryonic stem (ES) cells offer unprecedented opportunities for in vitro drug discovery and safety assessment of compounds. Cardiomyocytes derived from ES cells enable development of predictive cardiotoxicity models to increase the safety of novel drugs. Heterogeneity of differentiated ES cells limits the development of reliable in vitro models for compound screening. We report an innovative and robust approach to isolate ES-derived cardiomyocytes using laser microdissection and pressure catapulting (LMPC). LMPC cells were readily applied onto 96-well format in vitro pharmacology assays. The expression of developmental and functional cardiac markers, Nkx 2.5, MLC2V, GATA-4, Connexin 43, Connexin 45, Serca-2a, cardiac alpha actin, and phospholamban, among others, was confirmed in LMPC ES-derived cardiomyocytes. Functional assays exhibited cardiac-like response to increased extracellular calcium (5.4 mM extracellular Ca2+) and L-type calcium channel antagonist (1 microM nifedipine). In conclusion, laser microdissection and pressure catapulting is a robust technology to isolate homogeneous ES-derived cell types from heterogeneous populations applicable to assay development.


Assuntos
Cardiopatias/induzido quimicamente , Microscopia Confocal/métodos , Miócitos Cardíacos/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Xenobióticos/toxicidade , Animais , Bioensaio/métodos , Biomarcadores/metabolismo , Cálcio/metabolismo , Cálcio/farmacologia , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos/métodos , Coração Fetal/citologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Cardiopatias/patologia , Lasers , Camundongos , Camundongos Endogâmicos DBA , Microdissecção/métodos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Nifedipino/farmacologia , Análise de Sequência com Séries de Oligonucleotídeos , Células-Tronco/metabolismo , Células-Tronco/patologia
11.
Proc Natl Acad Sci U S A ; 100(15): 9044-9, 2003 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-12835414

RESUMO

Prostaglandin (PG)E2 is a potent mediator of pain and inflammation, and high levels of this lipid mediator are observed in numerous disease states. The inhibition of PGE2 production to control pain and to treat diseases such as rheumatoid arthritis to date has depended on nonsteroidal antiinflammatory agents such as aspirin. However, these agents inhibit the synthesis of all prostanoids. To produce biologically active PGE2, PGE synthases catalyze the isomerization of PGH2 into PGE2. Recently, several PGE synthases have been identified and cloned, but their role in inflammation is not clear. To study the physiological role of the individual PGE synthases, we have generated by targeted homologous recombination a mouse line deficient in microsomal PGE synthase 1 (mPGES1) on the inbred DBA/1lacJ background. mPGES1-deficient (mPGES1-/-) mice are viable and fertile and develop normally compared with wild-type controls. However, mPGES1-/- mice displayed a marked reduction in inflammatory responses compared with mPGES1+/+ mice in multiple assays. Here, we identify mPGES1 as the PGE synthase that contributes to the pathogenesis of collagen-induced arthritis, a disease model of human rheumatoid arthritis. We also show that mPGES1 is responsible for the production of PGE2 that mediates acute pain during an inflammatory response. These findings suggest that mPGES1 provides a target for the treatment of inflammatory diseases and pain associated with inflammatory states.


Assuntos
Inflamação/fisiopatologia , Oxirredutases Intramoleculares/deficiência , Dor/fisiopatologia , Animais , Artrite Experimental/etiologia , Artrite Experimental/patologia , Artrite Experimental/fisiopatologia , Artrite Reumatoide/etiologia , Artrite Reumatoide/patologia , Artrite Reumatoide/fisiopatologia , Dinoprostona/biossíntese , Feminino , Humanos , Hipersensibilidade Tardia , Mediadores da Inflamação/metabolismo , Oxirredutases Intramoleculares/genética , Oxirredutases Intramoleculares/fisiologia , Macrófagos/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos DBA , Camundongos Knockout , Dor/tratamento farmacológico , Prostaglandina-E Sintases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...