Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 269
Filtrar
1.
Theranostics ; 12(2): 842-858, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34976216

RESUMO

Background: FcγRIIB, the sole inhibitory receptor of the Fc gamma receptor family, plays pivotal roles in innate and adaptive immune responses. However, the expression and function of FcγRIIB in myeloid-derived suppressor cells (MDSCs) remains unknown. This study aimed to investigate whether and how FcγRIIB regulates the immunosuppressive activity of MDSCs during cancer development. Methods: The MC38 and B16-F10 tumor-bearing mouse models were established to investigate the role of FcγRIIB during tumor progression. FcγRIIB-deficient mice, adoptive cell transfer, mRNA-sequencing and flow cytometry analysis were used to assess the role of FcγRIIB on immunosuppressive activity and differentiation of MDSCs. Results: Here we show that FcγRIIB was upregulated in tumor-infiltrated MDSCs. FcγRIIB-deficient mice showed decreased accumulation of MDSCs in the tumor microenvironment (TME) compared with wild-type mice. FcγRIIB was required for the differentiation and immunosuppressive activity of MDSCs. Mechanistically, tumor cell-derived granulocyte-macrophage colony stimulating factor (GM-CSF) increased the expression of FcγRIIB on hematopoietic progenitor cells (HPCs) by activating specificity protein 1 (Sp1), subsequently FcγRIIB promoted the generation of MDSCs from HPCs via Stat3 signaling. Furthermore, blockade of Sp1 dampened MDSC differentiation and infiltration in the TME and enhanced the anti-tumor therapeutic efficacy of gemcitabine. Conclusion: These results uncover an unrecognized regulatory role of the FcγRIIB in abnormal differentiation of MDSCs during cancer development and suggest a potential therapeutic target for anti-tumor therapy.


Assuntos
Carcinogênese , Diferenciação Celular , Células Supressoras Mieloides/citologia , Receptores de IgG/fisiologia , Evasão Tumoral , Adulto , Animais , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células Supressoras Mieloides/imunologia , Receptores de IgG/deficiência , Receptores de IgG/metabolismo , Transdução de Sinais
2.
Front Immunol ; 12: 669162, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34248948

RESUMO

Renal ischemia is the most common cause of acute kidney injury (AKI) that might be exacerbate lupus activity through neutrophil extracellular traps (NETs) and apoptosis. Here, the renal ischemia reperfusion injury (I/R) was performed in Fc gamma receptor 2b deficient (Fcgr2b-/-) lupus mice and the in vitro experiments. At 24 h post-renal I/R injury, NETs in peripheral blood neutrophils and in kidneys were detected using myeloperoxidase (MPO), neutrophil elastase (NE) and citrullinated histone H3 (CitH3), as well as kidney apoptosis (activating caspase-3), which were prominent in Fcgr2b-/- mice more compared to wild-type (WT). After 120 h renal-I/R injury, renal NETs (using MPO and NE) were non-detectable, whereas glomerular immunoglobulin (Ig) deposition and serum anti-dsDNA were increased in Fcgr2b-/- mice. These results imply that renal NETs at 24 h post-renal I/R exacerbated the lupus nephritis at 120 h post-renal I/R injury in Fcgr2b-/- lupus mice. Furthermore, a Syk inhibitor attenuated NETs, that activated by phorbol myristate acetate (PMA; a NETs activator) or lipopolysaccharide (LPS; a potent inflammatory stimulator), more prominently in Fcgr2b-/- neutrophils than the WT cells as determined by dsDNA, PAD4 and MPO. In addition, the inhibitors against Syk and PAD4 attenuated lupus characteristics (serum creatinine, proteinuria, and anti-dsDNA) in Fcgr2b-/- mice at 120 h post-renal I/R injury. In conclusion, renal I/R in Fcgr2b-/- mice induced lupus exacerbation at 120 h post-I/R injury partly because Syk-enhanced renal NETs led to apoptosis-induced anti-dsDNA, which was attenuated by a Syk inhibitor.


Assuntos
Injúria Renal Aguda/metabolismo , Apoptose , Armadilhas Extracelulares/metabolismo , Rim/metabolismo , Nefrite Lúpica/metabolismo , Neutrófilos/metabolismo , Receptores de IgG/deficiência , Traumatismo por Reperfusão/metabolismo , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/imunologia , Injúria Renal Aguda/patologia , Animais , Apoptose/efeitos dos fármacos , Modelos Animais de Doenças , Progressão da Doença , Feminino , Rim/efeitos dos fármacos , Rim/imunologia , Rim/patologia , Nefrite Lúpica/tratamento farmacológico , Nefrite Lúpica/imunologia , Nefrite Lúpica/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Neutrófilos/patologia , Inibidores da Fosfodiesterase 4/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Receptores de IgG/genética , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/patologia , Quinase Syk/antagonistas & inibidores , Quinase Syk/metabolismo , Fatores de Tempo
3.
Front Immunol ; 11: 532684, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33117338

RESUMO

We generated an NOD/Shi-scid-IL2Rγ null (NOG) mouse deficient for the Fcer1g and Fcgr2b genes (NOG-FcγR-/- mice), in which monocytes/macrophages do not express activating (FcγRI, III, and IV) or inhibitory (FcγRIIB) Fcγ receptors. Antibody-dependent cellular cytotoxicity (ADCC) by innate immune cells was strongly reduced in this strain. Thus, while the growth of xenogeneic human tumors engrafted in conventional NOG mice was suppressed by innate cells upon specific antibody treatment, such growth inhibition was abrogated in NOG-FcγR-/- mice. Using this novel strain, we further produced NOG-FcγR-/--mice expressing human IL-15 (NOG-FcγR-/--hIL-15 Tg). The mice inherited unique features from each strain, i.e., the long-term sustenance of human natural killer (NK) cells, and the elimination of mouse innate cell-mediated ADCC. As a result, segregation of human NK cell-mediated ADCC from mouse cell-mediated ADCC was possible in the NOG-FcγR-/--hIL-15 Tg mice. Our results suggest that NOG-FcγR-/--hIL-15 Tg mice are useful for validating the in vivo function of antibody drug candidates.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos , Proteínas de Transporte/genética , Interleucina-15/imunologia , Células Matadoras Naturais/imunologia , Receptores de IgG/deficiência , Animais , Proteínas de Transporte/imunologia , Humanos , Interleucina-15/genética , Camundongos , Camundongos Knockout , Receptores de IgG/imunologia
4.
Lupus ; 29(10): 1248-1262, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32700597

RESUMO

Macrophages are responsible for the recognition of pathogen molecules. The downstream signalling of the innate immune responses against pathogen molecules, lipopolysaccharide (LPS) and (1→3)-ß-D-glucan (BG), and the adaptive immune response to antibodies, Fc gamma receptor (FcgR), is spleen tyrosine kinase (Syk). Because pathogen molecules and antibodies could be presented in lupus, impact of Syk and macrophages in lupus is explored. FcgR-IIb deficient (FcgRIIb-/-) mice, a model of inhibitory signalling loss, at 40 weeks old, but not pristane mice (a chemical induction lupus model) demonstrated spontaneous elevation of LPS and BG in serum from gut translocation despite the similarity in faecal microbiome analysis. Syk abundance in FcgRIIb-/- mice was higher than in pristane mice, possibly due to several Syk activators (anti-dsDNA, LPS and BG), and Syk inhibitor-attenuated proteinuria and serum cytokines only in FcgRIIb-/- mice. In addition, LPS + BG enhanced the expression of activating FcgRs, NF-κB and Syk, together with supernatant TNF-α predominantly in FcgRIIb-/- compared to wild-type macrophages. The inhibitors against Dectin-1, Syk and nuclear factor kappa B, but not anti-Raf-1, reduced supernatant TNF-α in LPS+BG-activated macrophages, implying Syk-dependent signalling. The pathogen molecules enhanced activating-FcgRs, without inhibition, through Syk, a shared downstream innate and adaptive signalling, is responsible for the hyper-responsiveness in FcgRIIb-/- macrophages. In conclusion, Syk inhibitor attenuated inflammation in FcgRIIb-/- but not in pristane mice, implying the influence of a lupus genetic background in treatment modalities.


Assuntos
Endotoxinas/toxicidade , Lúpus Eritematoso Sistêmico/genética , Animais , Citocinas/sangue , Modelos Animais de Doenças , Humanos , Inflamação , Camundongos , Camundongos Endogâmicos C57BL , Receptores de IgG/deficiência , Receptores de IgG/genética , Quinase Syk/antagonistas & inibidores
5.
Front Immunol ; 11: 905, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32582142

RESUMO

Anti-factor VIII (fVIII) alloantibodies, which can develop in patients with hemophilia A, limit the therapeutic options and increase morbidity and mortality of these patients. However, the factors that influence anti-fVIII antibody development remain incompletely understood. Recent studies suggest that Fc gamma receptors (FcγRs) may facilitate recognition and uptake of fVIII by recently developed or pre-existing naturally occurring anti-fVIII antibodies, providing a mechanism whereby the immune system may recognize fVIII following infusion. However, the role of FcγRs in anti-fVIII antibody formation remains unknown. In order to define the influence of FcγRs on the development of anti-fVIII antibodies, fVIII was injected into WT or FcγR knockout recipients, followed by evaluation of anti-fVIII antibodies. Anti-fVIII antibodies were readily observed following fVIII injection into FcγR knockouts, with similar anti-fVIII antibody levels occurring in FcγR knockouts as detected in WT mice injected in parallel. As antibodies can also fix complement, providing a potential mechanism whereby anti-fVIII antibodies may influence anti-fVIII antibody formation independent of FcγRs, fVIII was also injected into complement component 3 (C3) knockout recipients in parallel. Similar to FcγR knockouts, C3 knockout recipients developed a robust response to fVIII, which was likewise similar to that observed in WT recipients. As FcγRs or C3 may compensate for each other in recipients only deficient in FcγRs or C3 alone, we generated mice deficient in both FcγRs and C3 to test for potential antibody effector redundancy in anti-fVIII antibody formation. Infusion of fVIII into FcγRs and C3 (FcγR × C3) double knockouts likewise induced anti-fVIII antibodies. However, unlike individual knockouts, anti-fVIII antibodies in FcγRs × C3 knockouts were initially lower than WT recipients, although anti-fVIII antibodies increased to WT levels following additional fVIII exposure. In contrast, infusion of RBCs expressing distinct alloantigens into FcγRs, C3 or FcγR × C3 knockout recipients either failed to change anti-RBC levels when compared to WT recipients or actually increased antibody responses, depending on the target antigen. Taken together, these results suggest FcγRs and C3 can differentially impact antibody formation following exposure to distinct alloantigens and that FcγRs and C3 work in concert to facilitate early anti-fVIII antibody formation.


Assuntos
Complemento C3/metabolismo , Fator VIII/imunologia , Hemofilia A/imunologia , Isoanticorpos/sangue , Isoantígenos/imunologia , Receptores de IgG/metabolismo , Animais , Formação de Anticorpos , Complemento C3/deficiência , Complemento C3/genética , Modelos Animais de Doenças , Fator VIII/administração & dosagem , Feminino , Hemofilia A/sangue , Hemofilia A/tratamento farmacológico , Hemofilia A/genética , Isoantígenos/administração & dosagem , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de IgG/deficiência , Receptores de IgG/genética
6.
Front Immunol ; 11: 959, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32582149

RESUMO

FcgRIIB dysfunction is commonly found in patients with lupus, especially in Asia. LPS-tolerance is prominent in FcgRIIB-/- lupus mice. LPS-tolerant macrophages demonstrate cell energy depletion, which might affect lipid metabolism. Therefore, to explore lipid metabolism, LPS-tolerance was induced twice by LPS administration in macrophages and in mice. LPS-tolerant FcgRIIB-/- macrophages demonstrated lesser mitochondrial DNA (mtDNA), more severe ATP depletion, lower cytokine production, and higher lipid accumulation (oil red O staining) compared to LPS-tolerant WT cells. Mass-spectrometry-based lipidomic analysis demonstrated a higher abundance of phosphatidylethanolamine (PE) phospholipid in LPS-tolerant FcgRIIB-/- macrophages than WT cells. This was at least in part due to the lower expression of phosphatidylethanolamine N-methyltransferase (pemt), an enzyme that converts PE to phosphatidylcholine (PC). Aminoimidazole-4-carboxamide ribonucleotide (AICAR), a pemt inhibitor, worsens LPS-tolerance in WT macrophages and supports the impact of pemt upon LPS-tolerant FcgRIIB-/- macrophages. Additionally, phosphorylated AMP-activated protein kinase (AMPK-p), a molecule for ATP-restoration associated with pemt, and phosphorylated acetyl CoA carboxylase, a downstream signaling of AMPK-p, were higher in LPS-tolerant FcgRIIB-/- macrophages than WT. Furthermore, Compound C, an AMPK inhibitor, attenuated LPS-tolerance in both FcgRIIB-/- macrophages and mice. Taken together, the intense decrease in cytokine production after the second LPS stimulation (LPS-tolerance) in FcgRIIB-/- macrophages was possibly due to the impact of an immense cytokine synthesis after the first dose of LPS. This includes using up PEMT, an enzyme of phospholipid synthesis during cytokine production, and AMPK-p induction in response to profound ATP-depletion. Therefore, the manipulation of the AMPK/PEMT axis provides a novel therapeutic candidate for the treatment of severe LPS-tolerance in lupus.


Assuntos
Endotoxemia/metabolismo , Metabolismo Energético , Metabolismo dos Lipídeos , Lúpus Eritematoso Sistêmico/metabolismo , Macrófagos/metabolismo , Receptores de IgG/deficiência , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Endotoxemia/induzido quimicamente , Endotoxemia/genética , Feminino , Lipopolissacarídeos , Lúpus Eritematoso Sistêmico/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Fagocitose , Fosfatidiletanolamina N-Metiltransferase/metabolismo , Fosforilação , Receptores de IgG/genética
7.
Sci Rep ; 10(1): 7757, 2020 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-32385351

RESUMO

T cell non-Hodgkin lymphoma (T-NHL) is highly invasive and heterogeneous without accurate prognosis prediction. We proposed peripheral CD16-/CD16 + monocytes the additional indicators for T-NHL prognosis. We prospectively recruited 31 T-NHL patients without previous treatment. The CD16-/CD16 + monocyte ratio before chemotherapy was calculated and regular follow up was performed to calculate prognostic prediction value. Tumor associated macrophages (TAM) in tumor tissue were counted and transcriptome sequencing of CD16- and CD16 + monocytes was applied to explore potential mechanisms. We found that T-NHL patients had higher ratio of total monocytes especially the CD16 + monocytes along with a decreased ratio of CD16-/CD16 + monocytes, compared to the health control. The 1-year overall survival rate was 0.492 and 0.755 for CD16- monocyte/CD16 + monocyte ratio of <11 and ≥11(p < 0.05), respectively. The peripheral CD16-/CD16 + monocyte ratio was significantly relevant with the pathological CD68/CD206 macrophage ratio. The differently expressed genes in CD16- and CD16 + monocytes from T-NHL patients were mainly involved in signaling molecules related to tumor microenvironment. Pro-tumor genes were identified in monocyte subsets especially in CD16 + monocytes. In conclusion, the ratio of peripheral CD16-/CD16 + monocyte helps to stratify the prognosis of T-NHL. The relatively increased CD16 + monocytes may contribute to the pro-tumor microenvironment of T-NHL.


Assuntos
Linfoma de Células T/diagnóstico , Linfoma de Células T/imunologia , Monócitos/citologia , Monócitos/metabolismo , Receptores de IgG/deficiência , Receptores de IgG/metabolismo , Adulto , Contagem de Células , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Prognóstico
8.
Front Immunol ; 11: 118, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32117269

RESUMO

Antibodies are essential mediators of immunological defense mechanisms, are clinically used as therapeutic agents, but are also functionally involved in various immune-mediated disorders. Whereas IgG antibodies accomplish some of their biological tasks autonomously, many functions depend on their binding to activating and inhibitory Fcγ receptors (FcγR). From a qualitative point of view expression patterns of FcγR on immunologically relevant cell types are well-characterized both for mice and humans. Surprisingly, however, there is only quite limited information available on actual quantities of FcγR expressed by the different leukocyte populations. In this study we provide a comprehensive data set assessing quantitatively how many individual human and mouse FcγRs are expressed on B cells, NK cells, eosinophils, neutrophils, basophils and both classical, and non-classical monocytes under steady state conditions. Moreover, among human donors we found two groups with different expression levels of the inhibitory FcγRIIb on monocytes which appears to correlate with haplotypes of the activating FcγRIIIa.


Assuntos
Leucócitos/metabolismo , Receptores de IgG/genética , Receptores de IgG/metabolismo , Animais , Linfócitos B/metabolismo , Basófilos/metabolismo , Plaquetas/metabolismo , Eosinófilos/metabolismo , Feminino , Haplótipos , Voluntários Saudáveis , Humanos , Imunoglobulina G/imunologia , Células Matadoras Naturais/metabolismo , Leucócitos Mononucleares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/metabolismo , Neutrófilos/metabolismo , Receptores de IgG/deficiência , Receptores de IgG/imunologia
9.
Sci Rep ; 10(1): 777, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31964918

RESUMO

The influence of gut-leakage or gut-microbiota upon lupus progression was explored in 2 lupus mouse models. Pristane, administered in 4-wk-old wild-type (WT) female mice, induced lupus characteristics at 24-wk-old similar to the lupus-onset in FcGRIIb-/- mice. Gut-microbiota alteration was induced by co-housing together with the gavage of feces from 40-wk-old FcGRIIb-/- mice (symptomatic lupus). On the other hand,  gut-leakage was induced  by dextran sulfate solution (DSS). DSS and gut-microbiota alteration induced high serum anti-dsDNA immunoglobulin (Ig) as early as 30 days post-DSS only in FcGRIIb-/- mice. DSS, but not gut-microbiota alteration, enhanced lupus characteristics (serum creatinine and proteinuria) in both lupus models (but not in WT) at 60 days post-DSS. Indeed, DSS induced the translocation of molecular components of gut-pathogens as determined by bacterial burdens in mesenteric lymph node (MLN), endotoxemia (gut-bacterial molecule) and serum (1→3)-ß-D-glucan (BG) (gut-fungal molecule) as early as 15 days post-DSS together with enhanced MLN apoptosis in both WT and lupus mice. However, DSS induced spleen apoptosis in FcGRIIb-/- and WT mice at 30 and 60 days post-DSS, respectively, suggesting the higher impact of gut-leakage against spleen of lupus mice. In addition, macrophages preconditioning with LPS plus BG were susceptible to starvation-induced apoptosis, predominantly in FcGRIIb-/- cell, implying the influence of gut-leakage upon cell stress. In summary, gut-leakage induced gut-translocation of organismal-molecules then enhanced the susceptibility of stress-induced apoptosis, predominantly in lupus. Subsequently, the higher burdens of apoptosis in lupus mice increased anti-dsDNA Ig and worsen lupus severity through immune complex deposition. Hence, therapeutic strategies addressing gut-leakage in lupus are interesting.


Assuntos
Sulfato de Dextrana/efeitos adversos , Lúpus Eritematoso Sistêmico/genética , Receptores de IgG/deficiência , Terpenos/efeitos adversos , Animais , Citocinas/sangue , Modelos Animais de Doenças , Progressão da Doença , Fezes/microbiologia , Feminino , Lúpus Eritematoso Sistêmico/induzido quimicamente , Lúpus Eritematoso Sistêmico/microbiologia , Camundongos , beta-Glucanas/sangue
10.
Front Immunol ; 10: 2452, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31681326

RESUMO

Systemic lupus erythematosus (SLE) is an autoimmune disorder characterized by increased production of autoantibodies, which commonly target nuclear antigens, and concomitant deposition of immune complexes that cause inflammation in tissues. SLE is often associated with increased systemic expression of type I interferons, in some cases due to dysregulation in nucleic acid-sensing innate pathways. There is strong genetic evidence for a link between cytoplasmic RNA sensing pathways (RIG-I/MDA5) and SLE, both in human patients and murine models, however questions still remain regarding pathway initiation, cell types involved and downstream effects. Here we show that MAVS, an essential adaptor for RIG-I/MDA5 signaling, is necessary for all symptoms of autoimmune disease that develop spontaneously in the lupus model FcγRIIB-/- mice. This effect was independent of type I interferon signaling, TLR7 expression or STING, all three factors that have been connected to autoimmunity. Mixed bone marrow reconstitution experiments showed reduced occurrence in autoimmune germinal centers and diminished autoantibody production by MAVS-deficient B cells. Thus, MAVS plays a B cell intrinsic role in autoreactive B cell activation that is independent of its anti-viral functions and independent of elevated type I interferon expression.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Autoimunidade , Linfócitos B/imunologia , Linfócitos B/metabolismo , Interações Hospedeiro-Patógeno , Lúpus Eritematoso Sistêmico/etiologia , Lúpus Eritematoso Sistêmico/metabolismo , Animais , Autoanticorpos/imunologia , Células da Medula Óssea/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Centro Germinativo/imunologia , Centro Germinativo/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Lúpus Eritematoso Sistêmico/patologia , Camundongos , Mutação , Receptores de IgG/deficiência , Receptor 7 Toll-Like/metabolismo
11.
Biomed Res Int ; 2019: 3514574, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31534958

RESUMO

The immune system is involved in the development of diabetes complications and IgG Fc gamma receptors (FcgRs) are key immune receptors responsible for the effective control of both humoral and innate immunity. We investigated the effects of members of the FcgR superfamily into both the streptozotocin plus high fat-induced type 2 diabetes and high fat diet (HFD) models. FcgRIII-/- diabetic mice and FcgRIIb-/- diabetic mice had elevated levels of serum creatinine compared with wildtype (WT) diabetic mice. Renal histology of diabetic FcgRIII knockout and FcgRIIb knockout mice showed mesangial expansion and GBM thickening; the mechanistic study indicated a higher expression of TGF-ß1, TNF-α, and p-NFκB-p65 compared with wild type mouse. The HFD mouse with FcgRIII knockout or FcgRIIb knockout had increased biochemical and renal injury factors, but oxLDL deposition was higher than in FcgRIII-/- diabetic mice and FcgRIIb-/- diabetic mice. In vitro we further examined the mechanism by which the Fc gamma receptor promoted renal injury and transfected glomerular mesangial cells (GMCs) with FcgRI siRNA attenuated the level of TGF-ß1, TNF-α expression. In summary, FcgRI knockdown downregulated kidney inflammation and fibrosis and FcgRIIb knockout accelerated inflammation, fibrosis, and the anomalous deposition of oxLDL whereas FcgRIII deficiency failed to protect kidney from diabetic renal injury. These observations suggested that FcgRs might represent a novel target for the therapeutic intervention of diabetic nephropathy.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Nefropatias Diabéticas/metabolismo , Receptores de IgG/deficiência , Animais , Diabetes Mellitus Tipo 2/induzido quimicamente , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Nefropatias Diabéticas/induzido quimicamente , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/patologia , Gorduras na Dieta/efeitos adversos , Gorduras na Dieta/farmacologia , Lipoproteínas LDL/genética , Lipoproteínas LDL/metabolismo , Camundongos , Camundongos Knockout , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
12.
J Exp Med ; 216(8): 1904-1924, 2019 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-31196979

RESUMO

Rheumatoid arthritis-associated joint pain is frequently observed independent of disease activity, suggesting unidentified pain mechanisms. We demonstrate that antibodies binding to cartilage, specific for collagen type II (CII) or cartilage oligomeric matrix protein (COMP), elicit mechanical hypersensitivity in mice, uncoupled from visual, histological and molecular indications of inflammation. Cartilage antibody-induced pain-like behavior does not depend on complement activation or joint inflammation, but instead on tissue antigen recognition and local immune complex (IC) formation. smFISH and IHC suggest that neuronal Fcgr1 and Fcgr2b mRNA are transported to peripheral ends of primary afferents. CII-ICs directly activate cultured WT but not FcRγ chain-deficient DRG neurons. In line with this observation, CII-IC does not induce mechanical hypersensitivity in FcRγ chain-deficient mice. Furthermore, injection of CII antibodies does not generate pain-like behavior in FcRγ chain-deficient mice or mice lacking activating FcγRs in neurons. In summary, this study defines functional coupling between autoantibodies and pain transmission that may facilitate the development of new disease-relevant pain therapeutics.


Assuntos
Anticorpos Monoclonais/imunologia , Complexo Antígeno-Anticorpo/metabolismo , Artralgia/imunologia , Artrite Reumatoide/imunologia , Autoanticorpos/imunologia , Cartilagem/imunologia , Neurônios/metabolismo , Animais , Anticorpos Monoclonais/uso terapêutico , Artralgia/tratamento farmacológico , Artrite Reumatoide/tratamento farmacológico , Autoanticorpos/uso terapêutico , Comportamento Animal/efeitos dos fármacos , Proteína de Matriz Oligomérica de Cartilagem/imunologia , Colágeno Tipo II/imunologia , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Receptores de IgG/deficiência , Receptores de IgG/genética
13.
Am J Physiol Lung Cell Mol Physiol ; 317(1): L49-L56, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30969811

RESUMO

Bronchopulmonary dysplasia (BPD) remains a devastating consequence of prematurity. Repeated inflammatory insults worsen lung injury, but there are no predictors for BPD-related respiratory outcomes or targeted therapies. We sought to understand inflammatory mechanisms in evolving BPD through molecular characterization of monocytes in tracheal aspirates from infants at risk for developing BPD. We performed flow cytometry targeting myeloid cell populations on prospectively collected tracheal aspirates from intubated patients born before 29 wk of gestation and <30 days old. We identified CD14+CD16+ (double-positive) and CD14+CD16- (single-positive) monocytes and characterized their gene expression profiles by RNA sequencing and quantitative PCR. We further analyzed differential gene expression between time points to evaluate changes in monocyte function over the first weeks of life. Expression of IL-1A, IL-1B, and IL-1 receptor antagonist mRNA was increased in monocytes collected at day of life (DOL) 7, DOL 14, and DOL 28 compared with those collected at DOL 3. This study suggests that early changes in monocyte-specific IL-1 cytokine pathways may be associated with evolving BPD.


Assuntos
Displasia Broncopulmonar/genética , Proteína Antagonista do Receptor de Interleucina 1/genética , Interleucina-1alfa/genética , Interleucina-1beta/genética , Monócitos/imunologia , RNA Mensageiro/genética , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/imunologia , Displasia Broncopulmonar/imunologia , Displasia Broncopulmonar/patologia , Feminino , Proteínas Ligadas por GPI/deficiência , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/imunologia , Regulação da Expressão Gênica , Idade Gestacional , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Proteína Antagonista do Receptor de Interleucina 1/imunologia , Interleucina-1alfa/imunologia , Interleucina-1beta/imunologia , Receptores de Lipopolissacarídeos/genética , Receptores de Lipopolissacarídeos/imunologia , Masculino , Monócitos/patologia , Projetos Piloto , Estudos Prospectivos , RNA Mensageiro/imunologia , Receptores de IgG/deficiência , Receptores de IgG/genética , Receptores de IgG/imunologia , Análise de Sequência de RNA , Transdução de Sinais , Traqueia/imunologia , Traqueia/patologia
14.
Int J Mol Sci ; 20(6)2019 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-30889825

RESUMO

Dysfunction of FcGRIIb, the only inhibitory receptor of the FcGR family, is commonly found in the Asian population and is possibly responsible for the extreme endotoxin exhaustion in lupus. Here, the mechanisms of prominent endotoxin (LPS) tolerance in FcGRIIb-/- mice were explored on bone marrow-derived macrophages using phosphoproteomic analysis. As such, LPS tolerance decreased several phosphoproteins in the FcGRIIb-/- macrophage, including protein kinase C-ß type II (PRKCB), which was associated with phagocytosis function. Overexpression of PRKCB attenuated LPS tolerance in RAW264.7 cells, supporting the role of this gene in LPS tolerance. In parallel, LPS tolerance in macrophages and in mice was attenuated by phorbol 12-myristate 13-acetate (PMA) administration. This treatment induced several protein kinase C families, including PRKCB. However, PMA attenuated the severity of mice with cecal ligation and puncture on LPS tolerance preconditioning in FcGRIIb-/- but not in wild-type cells. The significant reduction of PRKCB in the FcGRIIb-/- macrophage over wild-type cell possibly induced the more severe LPS-exhaustion and increased the infection susceptibility in FcGRIIb-/- mice. PMA induced PRKCB, improved LPS-tolerance, and attenuated sepsis severity, predominantly in FcGRIIb-/- mice. PRKCB enhancement might be a promising strategy to improve macrophage functions in lupus patients with LPS-tolerance from chronic infection.


Assuntos
Endotoxinas/metabolismo , Lúpus Eritematoso Sistêmico/patologia , Macrófagos/metabolismo , Fosfoproteínas/metabolismo , Proteína Quinase C beta/metabolismo , Proteômica , Receptores de IgG/deficiência , Animais , Citocinas/sangue , Lipopolissacarídeos , Lúpus Eritematoso Sistêmico/sangue , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de IgG/metabolismo , Sepse/sangue , Sepse/patologia , Índice de Gravidade de Doença , Acetato de Tetradecanoilforbol/farmacologia
15.
J Bone Miner Res ; 34(7): 1352-1365, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30779858

RESUMO

Next to proinflammatory cytokines, autoimmunity has been identified as a key trigger for osteoclast activation and bone loss. IgG-rheumatoid factor (IgG-RF) immune complexes, which are present in patients with rheumatoid arthritis, were shown to boost osteoclast differentiation. To date, the regulation of IgG-RF production in the absence of inflammatory triggers is unknown. Herein, we describe Fra1 as a key checkpoint that controls IgG-RF production by plasma cells and regulates autoimmune-mediated bone loss. Fra1 deficiency in B cells (Fra1ΔBcell ) led to increased IgG1-producing bone marrow plasma cells, enhanced IgG-RF production, and increased bone loss associated with elevated osteoclast numbers after immunization. The effect of IgG-RF on osteoclasts in vitro and on osteoclasts associated with bone loss in vivo was dependent on FcγR, especially FcγR3. Furthermore, immunization of WT mice with T-cell-dependent antigens induced a significant and robust decrease in Fra1 expression in bone marrow B cells, which was followed by increased IgG1 production and the induction of osteoclast-mediated bone loss. Overall, these data identify Fra1 as a key mediator of IgG-RF production and autoimmune-mediated bone loss. © 2019 American Society for Bone and Mineral Research.


Assuntos
Autoanticorpos/biossíntese , Células da Medula Óssea/metabolismo , Reabsorção Óssea/imunologia , Reabsorção Óssea/patologia , Plasmócitos/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Fator Reumatoide/metabolismo , Animais , Osso e Ossos/patologia , Contagem de Células , Diferenciação Celular , Deleção de Genes , Imunidade Humoral , Imunização , Imunoglobulina G/metabolismo , Camundongos Endogâmicos C57BL , Osteoclastos/patologia , Osteogênese , Osteoporose/imunologia , Fenótipo , Proteínas Proto-Oncogênicas c-fos/deficiência , Receptores de IgG/deficiência , Receptores de IgG/metabolismo , Linfócitos T/imunologia
16.
J Microbiol ; 57(1): 45-53, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30456753

RESUMO

The severity of cryptococcosis in lupus from varying genetic-backgrounds might be different due to the heterogeneity of lupus-pathogenesis. This study explored cryptococcosis in lupus mouse models of pristane-induction (normal genetic-background) and FcGRIIb deficiency (genetic defect). Because the severity of lupus nephritis, as determined by proteinuria and serum creatinine, between pristane and FcGRIIb-/- mice were similar at 6-month-old, Cryptococcus neoformans was intravenously administered in 6-month-old mice and were age-matched with wild-type. Indeed, the cryptococcosis disease severity, as evaluated by mortality rate, internal-organ fungal burdens and serum cytokines, between pristane and FcGRIIb-/- mice was not different. However, the severity of cryptococcosis in wild-type was less severe than the lupus mice. On the other hand, phagocytosis activity of peritoneal macrophages from lupus mice (pristane and FcGRIIb-/-) was more predominant than the wild-type without the difference in macrophage killing-activity among these groups. In addition, the number of active T helper cells (Th-cell) in the spleen, including Th-cells with intracellular IFN-γ, from lupus mice (pristane and FcGRIIb-/-) was higher than wildtype. Moreover, these active Th-cells were even higher after 2 weeks of cryptococcal infection. These data support enhanced macrophage activation through prominent Th-cells in both lupus models. In conclusion, an increased susceptibility of cryptococcosis in both lupus models was independent to genetic background. This might due to Th-cell enhanced macrophage phagocytosis with the interference of macrophage killing activity from Cryptococcal immune-evasion properties.


Assuntos
Criptococose/microbiologia , Cryptococcus neoformans/fisiologia , Lúpus Eritematoso Sistêmico/complicações , Macrófagos/imunologia , Terpenos/efeitos adversos , Animais , Criptococose/etiologia , Criptococose/genética , Criptococose/imunologia , Citocinas/genética , Citocinas/imunologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Feminino , Humanos , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/metabolismo , Ativação de Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fagocitose , Receptores de IgG/deficiência , Receptores de IgG/genética , Baço/imunologia , Células Th1/imunologia
17.
Arthritis Res Ther ; 21(1): 303, 2019 12 30.
Artigo em Inglês | MEDLINE | ID: mdl-31888754

RESUMO

BACKGROUND: Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by the production of various autoantibodies. This disease causes disabling neuropsychiatric symptoms even in the absence of apparent inflammation in the central nervous system (CNS), but the mechanisms involved remain unknown. Innate immune-mediated inflammation has attracted attention as a pathogenic mechanism in neuropsychiatric diseases. METHODS: We investigated the CNS of lupus-prone mice focusing on innate immunity. Three strains of lupus-prone mice, FcγRIIB-/-Yaa, an F1 hybrid of NZB and NZW (NZB/NZW) mice, and MRL/Faslpr (MRL/lpr) mice were used to analyze CNS immunopathology. RESULTS: Flow cytometry analysis demonstrated the numbers of brain CD45+ cells were increased compared with controls in lupus-prone mice. Upregulation of MHC class I and PDCA1 was observed in microglia and CD11b+ myeloid cells of lupus-prone mice, indicating they were activated in response to interferons (IFN). Microglial gene expression analysis of FcγRIIB-/-Yaa mice revealed the upregulation of IFN-responsive genes and inflammation-related genes including Axl, Clec7a, and Itgax, which were previously reported in neurodegenerative conditions and primed conditions. Upregulated chemokine gene expressions including Ccl5 and Cxcl10 were concurrent with increased numbers of T cells and monocytes, especially Ly6Clo monocytes in the CNS. Upregulation of Axl, Clec7a, Itgax, Ccl5, and Cxcl10 was also observed in NZB/NZW mice, indicating common lupus pathology. The primed status of microglia in FcγRIIB-/-Yaa mice was also demonstrated by morphological changes such as enlarged cell bodies with hypertrophic processes, and hyperreactivity to lipopolysaccharide. Immunohistochemistry of FcγRIIB-/-Yaa mice indicated reactive responses of astrocytes and vascular endothelium. Behavioral studies of FcγRIIB-/-Yaa mice revealed depressive-like behavior and heat hyperalgesia in the forced swim test and the tail-flick test, respectively. CONCLUSIONS: Our data indicated that microglia in lupus exhibit a unique primed phenotype characterized by the upregulated expressions of neurodegeneration-related genes and IFN-responsive genes. Interaction with peripheral cells and brain resident cells was presumed to orchestrate neuroinflammation. Targeting innate immune cells, such as microglia and monocytes, may be a promising therapeutic approach for neuropsychiatric SLE.


Assuntos
Autoimunidade/imunologia , Sistema Nervoso Central/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Microglia/imunologia , Animais , Autoimunidade/genética , Antígenos CD11/genética , Antígenos CD11/imunologia , Antígenos CD11/metabolismo , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/patologia , Expressão Gênica/imunologia , Lectinas Tipo C/genética , Lectinas Tipo C/imunologia , Lectinas Tipo C/metabolismo , Lúpus Eritematoso Sistêmico/genética , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos MRL lpr , Camundongos Endogâmicos NZB , Camundongos Endogâmicos , Camundongos Knockout , Microglia/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Células Mieloides/imunologia , Células Mieloides/metabolismo , Receptores de IgG/deficiência , Receptores de IgG/genética , Receptores de IgG/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
18.
PLoS One ; 13(9): e0204168, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30212590

RESUMO

BACKGROUND: Antigen presentation on monocyte surface to T-cells by Major Histocompatibility Complex, Class II (MHC-II) molecules is fundamental for pathogen recognition and efficient host response. Accordingly, loss of Major Histocompatibility Complex, Class II, DR (HLA-DR) surface expression indicates impaired monocyte functionality in patients suffering from sepsis-induced immunosuppression. Besides the impact of Class II Major Histocompatibility Complex Transactivator (CIITA) on MHC-II gene expression, X box-like (XL) sequences have been proposed as further regulatory elements. These elements are bound by the DNA-binding protein CCCTC-Binding Factor (CTCF), a superordinate modulator of gene transcription. Here, we hypothesized a differential interaction of CTCF with the MHC-II locus contributing to an altered monocyte response in immunocompromised septic patients. METHODS: We collected blood from six patients diagnosed with sepsis and six healthy controls. Flow cytometric analysis was used to identify sepsis-induced immune suppression, while inflammatory cytokine levels in blood were determined via ELISA. Isolation of CD14++ CD16-monocytes was followed by (i) RNA extraction for gene expression analysis and (ii) chromatin immunoprecipitation to assess the distribution of CTCF and chromatin modifications in selected MHC-II regions. RESULTS: Compared to healthy controls, CD14++ CD16-monocytes from septic patients with immune suppression displayed an increased binding of CTCF within the MHC-II locus combined with decreased transcription of CIITA gene. In detail, enhanced CTCF enrichment was detected on the intergenic sequence XL9 separating two subregions coding for MHC-II genes. Depending on the relative localisation to XL9, gene expression of both regions was differentially affected in patients with sepsis. CONCLUSION: Our experiments demonstrate for the first time that differential CTCF binding at XL9 is accompanied by uncoupled MHC-II expression as well as transcriptional and epigenetic alterations of the MHC-II regulator CIITA in septic patients. Overall, our findings indicate a sepsis-induced enhancer blockade mediated by variation of CTCF at the intergenic sequence XL9 in altered monocytes during immunosuppression.


Assuntos
Fator de Ligação a CCCTC/imunologia , DNA Intergênico/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Hospedeiro Imunocomprometido , Sepse/imunologia , Adulto , Apresentação de Antígeno , Sequência de Bases , Fator de Ligação a CCCTC/genética , Estudos de Casos e Controles , Cromatina/química , Cromatina/imunologia , DNA Intergênico/genética , Feminino , Proteínas Ligadas por GPI/deficiência , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/imunologia , Regulação da Expressão Gênica , Loci Gênicos , Antígenos de Histocompatibilidade Classe II/genética , Humanos , Receptores de Lipopolissacarídeos/genética , Receptores de Lipopolissacarídeos/imunologia , Masculino , Monócitos/imunologia , Monócitos/patologia , Proteínas Nucleares/genética , Proteínas Nucleares/imunologia , Ligação Proteica , Receptores de IgG/deficiência , Receptores de IgG/genética , Receptores de IgG/imunologia , Elementos Reguladores de Transcrição , Sepse/genética , Sepse/patologia , Transdução de Sinais , Linfócitos T/imunologia , Linfócitos T/patologia , Transativadores/genética , Transativadores/imunologia , Transcrição Gênica
19.
Calcif Tissue Int ; 103(6): 686-697, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30116830

RESUMO

Patients with systemic lupus erythematosus (SLE), a chronic inflammatory disease characterized by loss of T- and B-cell tolerance to autoantigens, are at increased risk for osteoporosis and fractures. Mice deficient in Fc gamma receptor IIb (FcγRIIB) exhibit spontaneous SLE and its restoration rescues the disease. To determine whether deleting FcγRIIB affects cortical bone mass and mechanical properties, we analyzed cortical bone phenotype of FcγRIIB knockouts at different ages. FACS analysis revealed that 6-month-old FcγRIIB-/- mice had increased B220lowCD138+ cells, markers of plasma cells, indicating active SLE disease. In contrast, 3-month-old FcγRIIB-/- mice did not develop the active SLE disease. µCT analysis indicated that FcγRIIB deletion did not affect cortical bone in 3-month-old mutants. However, 6- and 10-month-old FcγRIIB-/- males and females had osteopenic cortical bone and the severity of bone loss increased with disease duration. FcγRIIB deletion decreased cross-sectional area, cortical area, and marrow area in 6-month-old males. Cortical area and cortical thickness were decreased in 10-month-old FcγRIIB-/- males. Lack of FcγRIIB decreased cortical thickness without affecting cortical area in females. However, deletion of a single FcγRIIB allele was insufficient to induce cortical bone loss. The bending strength was decreased in 6- and 10-month-old FcγRIIB-deficient males compared to WT controls. A microindentation analysis demonstrated significantly decreased hardness in both 10-month-old FcγRIIB-/- males and females. Our data indicate that FcγRIIB contributes to the regulation of cortical bone homeostasis subsequent to SLE development and that deletion of FcγRIIB in mice leads to SLE-like disease associated with cortical bone loss and decreased bending strength and hardness.


Assuntos
Osso Cortical/patologia , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/patologia , Receptores de IgG/deficiência , Animais , Doenças Ósseas Metabólicas/genética , Doenças Ósseas Metabólicas/metabolismo , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de IgG/genética
20.
J Virol ; 92(19)2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30021906

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) induces B cell hyperplasia and neoplasia, such as multicentric Castleman's disease (MCD) and primary effusion lymphoma (PEL). To explore KSHV-induced B cell reprogramming in vivo, we expressed the KSHV latency locus, inclusive of all viral microRNAs (miRNAs), in B cells of transgenic mice in the absence of the inhibitory FcγRIIB receptor. The BALB/c strain was chosen as this is the preferred model to study B cell differentiation. The mice developed hyperglobulinemia, plasmacytosis, and B lymphoid hyperplasia. This phenotype was ameliorated by everolimus, which is a rapamycin derivative used for the treatment of mantle cell lymphoma. KSHV latency mice exhibited hyperresponsiveness to the T-dependent (TD) antigen mimic anti-CD40 and increased incidence of pristane-induced inflammation. Lastly, the adaptive immunity against a secondary infection with Zika virus (ZIKV) was markedly enhanced. These phenotypes are consistent with KSHV lowering the activation threshold of latently infected B cells, which may be beneficial in areas of endemicity, where KSHV is acquired in childhood and infections are common.IMPORTANCE Kaposi's sarcoma-associated herpesvirus (KSHV) establishes latency in B cells and is stringently linked to primary effusion lymphoma (PEL) and the premalignant B cell hyperplasia multicentric Castleman's disease (MCD). To investigate potential genetic background effects, we expressed the KSHV miRNAs in BALB/c transgenic mice. BALB/c mice are the preferred strain for B cell hybridoma development because of their propensity to develop predictable B cell responses to antigen. The BALB/c latency mice exhibited a higher incidence of B cell hyperplasia as well as sustained hyperglobulinemia. The development of neutralizing antibodies against ZIKV was augmented in BALB/c latency mice. Hyperglobulinemia was dampened by everolimus, a derivative of rapamycin, suggesting a role for mTOR inhibitors in managing immune activation, which is hallmark of KSHV infection as well as HIV infection.


Assuntos
Linfócitos B/virologia , Resistência à Doença/genética , Herpesvirus Humano 8/imunologia , Receptores de IgG/imunologia , Sarcoma de Kaposi/imunologia , Latência Viral , Infecção por Zika virus/imunologia , Animais , Antineoplásicos/farmacologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Diferenciação Celular/efeitos dos fármacos , Coinfecção , Everolimo/farmacologia , Herpesvirus Humano 8/efeitos dos fármacos , Herpesvirus Humano 8/genética , Humanos , Hipergamaglobulinemia/genética , Hipergamaglobulinemia/imunologia , Hipergamaglobulinemia/virologia , Imunossupressores/farmacologia , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Nus , MicroRNAs/genética , MicroRNAs/imunologia , Plasmocitoma/genética , Plasmocitoma/imunologia , Plasmocitoma/virologia , RNA Viral/genética , RNA Viral/imunologia , Receptores de IgG/deficiência , Receptores de IgG/genética , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/virologia , Terpenos/farmacologia , Zika virus/efeitos dos fármacos , Zika virus/genética , Zika virus/imunologia , Infecção por Zika virus/genética , Infecção por Zika virus/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...