Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Cell Sci ; 134(21)2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34622926

RESUMO

Protein kinase C (PKC)-ε is required for membrane addition during IgG-mediated phagocytosis, but its role in this process is ill defined. Here, we performed high-resolution imaging, which reveals that PKC-ε exits the Golgi and enters phagosomes on vesicles that then fuse. TNF and PKC-ε colocalize at the Golgi and on vesicles that enter the phagosome. Loss of PKC-ε and TNF delivery upon nocodazole treatment confirmed vesicular transport on microtubules. That TNF+ vesicles were not delivered in macrophages from PKC-ε null mice, or upon dissociation of the Golgi-associated pool of PKC-ε, implies that Golgi-tethered PKC-ε is a driver of Golgi-to-phagosome trafficking. Finally, we established that the regulatory domain of PKC-ε is sufficient for delivery of TNF+ vesicles to the phagosome. These studies reveal a novel role for PKC-ε in focal exocytosis - its regulatory domain drives Golgi-derived vesicles to the phagosome, whereas catalytic activity is required for their fusion. This is one of the first examples of a PKC requirement for vesicular trafficking and describes a novel function for a PKC regulatory domain. This article has an associated First Person interview with the first author of the paper.


Assuntos
Fagocitose , Proteína Quinase C-épsilon , Animais , Exocitose , Imunoglobulina G , Camundongos , Fagossomos
2.
Cell Immunol ; 345: 103962, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31582169

RESUMO

Previous in vivo studies established that inactivated Francisella tularensis immune complexes (mAb-iFt) are a more protective vaccine against lethal tularemia than iFt alone. Subsequent in vitro studies revealed enhanced DC maturation marker expression with mAb-iFt stimulation. The goal of this study was to determine the mechanism of enhanced DC maturation. Multiparameter analysis of surface marker expression and cytokine secretion demonstrates a requirement for FcγR signaling in enhanced DC maturation. MyD88 was also found to be essential for heightened DC maturation, implicating MyD88-dependent TLRs in DC maturation. Upon further study, we discovered that TLRs 2 & 4 drive cytokine secretion, but surprisingly TLR9 is required for DC maturation marker upregulation. These studies reveal a separation of DC cytokine and maturation marker induction pathways and demonstrate that FcγR-TLR/MyD88 synergy underlies the enhanced dendritic cell maturation in response to the mAb-iFt vaccine.


Assuntos
Diferenciação Celular/imunologia , Células Dendríticas/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Receptores de IgG/imunologia , Receptor Toll-Like 9/imunologia , Animais , Anticorpos Monoclonais/imunologia , Vacinas Bacterianas/imunologia , Citocinas/imunologia , Citocinas/metabolismo , Células Dendríticas/metabolismo , Francisella tularensis/imunologia , Humanos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fator 88 de Diferenciação Mieloide/metabolismo , Receptores de IgG/genética , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/imunologia , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia , Receptor 4 Toll-Like/metabolismo , Receptor Toll-Like 9/genética , Receptor Toll-Like 9/metabolismo , Tularemia/imunologia , Tularemia/microbiologia
3.
J Immunol ; 199(1): 271-277, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28539432

RESUMO

Protein kinase C-ε (PKC-ε) at phagocytic cups mediates the membrane fusion necessary for efficient IgG-mediated phagocytosis. The C1B and pseudosubstrate (εPS) domains are necessary and sufficient for this concentration. C1B binds diacylglycerol; the docking partner for εPS is unknown. Liposome assays revealed that the εPS binds phosphatidylinositol 4-phosphate (PI4P) and PI(3,5)P2 Wortmannin, but not LY294002, inhibits PKC-ε concentration at cups and significantly reduces the rate of phagocytosis. As Wortmannin inhibits PI4 kinase, we hypothesized that PI4P mediates the PKC-ε concentration at cups and the rate of phagocytosis. PKC-ε colocalizes with the trans-Golgi network (TGN) PI4P reporter, P4M, suggesting it is tethered at the TGN. Real-time imaging of GFP-PKC-ε-expressing macrophages revealed a loss of Golgi-associated PKC-ε during phagocytosis, consistent with a Golgi-to-phagosome translocation. Treatment with PIK93, a PI4 kinase inhibitor, reduces PKC-ε at both the TGN and the cup, decreases phagocytosis, and prevents the increase in capacitance that accompanies membrane fusion. Finally, expression of the Golgi-directed PI4P phosphatase, hSac1-K2A, recapitulates the PIK93 phenotype, confirming that Golgi-associated PI4P is critical for efficient phagocytosis. Together these data are consistent with a model in which PKC-ε is tethered to the TGN via an εPS-PI4P interaction. The TGN-associated pool of PKC-ε concentrates at the phagocytic cup where it mediates the membrane fusion necessary for phagocytosis. The novelty of these data lies in the demonstration that εPS binds PI4P and PI(3,5)P2 and that PI4P is necessary for PKC-ε localization at the TGN, its translocation to the phagocytic cup, and the membrane fusion required for efficient Fc [γ] receptor-mediated phagocytosis.


Assuntos
Fagocitose , Fagossomos/imunologia , Fosfatos de Fosfatidilinositol/metabolismo , Proteína Quinase C-épsilon/metabolismo , Animais , Membrana Celular/metabolismo , Complexo de Golgi/metabolismo , Proteínas de Fluorescência Verde , Fusão de Membrana , Camundongos , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Fagossomos/metabolismo , Proteína Quinase C/imunologia , Proteína Quinase C/metabolismo , Transdução de Sinais
4.
Arterioscler Thromb Vasc Biol ; 35(5): 1101-12, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25792447

RESUMO

OBJECTIVE: Fcγ receptors (FcγRs) are classified as activating (FcγRI, III, and IV) and inhibitory (FcγRII) receptors. We have reported that deletion of activating FcγRs in apolipoprotein E (apoE) single knockout mice attenuated atherosclerosis. In this report, we investigated the hypothesis that deficiency of inhibitory FcγRIIb exacerbates atherosclerosis. APPROACH AND RESULTS: ApoE-FcγRIIb double knockout mice, congenic to the C57BL/6 (apoE-FcγRIIbB6 (-/-)), were generated and atherosclerotic lesions were assessed. In contrary to our hypothesis, when compared with apoE single knockout mice, arterial lesions were significantly decreased in apoE-FcγRIIbB6 (-/-) male and female mice fed chow or high-fat diets. Chimeric mice generated by transplanting apoE-FcγRIIbB6 (-/-) marrow into apoE single knockout mice also developed reduced lesions. CD4(+) T cells from apoE-FcγRIIbB6 (-/-) mice produced higher levels of interleukin-10 and transforming growth factor-ß than their apoE single knockout counterparts. As our findings conflict with a previous report using apoE-FcγRIIb129/B6 (-/-) mice on a mixed genetic background, we investigated whether strain differences contributed to the anti-inflammatory response. Macrophages from FcγRIIb129/B6 (-/-) mice on a mixed genetic background produced more interleukin-1ß and MCP-1 (monocyte chemoattractant protein-1) in response to immune complexes, whereas congenic FcγRIIbB6 (-/-) mice generated more interleukin-10 and significantly less interleukin-1ß. Interestingly, the expression of lupus-associated slam genes, located in proximity to fcgr2b in mouse chromosome 1, is upregulated only in mixed FcγRIIb129/B6 (-/-) mice. CONCLUSIONS: Our findings demonstrate a detrimental role for FcγRIIb signaling in atherosclerosis and the contribution of anti-inflammatory cytokine responses in the attenuated lesions observed in apoE-FcγRIIbB6 (-/-) mice. As 129/sv genome-derived lupus-associated genes have been implicated in lupus phenotype in FcγRIIb129/B6 (-/-) mice, our findings suggest possible epistatic mechanism contributing to the decreased lesions.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/metabolismo , Mediadores da Inflamação/metabolismo , Macrófagos/metabolismo , Receptores de IgG/metabolismo , Animais , Aterosclerose/imunologia , Aterosclerose/fisiopatologia , Antígenos CD4/imunologia , Antígenos CD4/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Distribuição Aleatória , Receptores de IgG/imunologia , Sensibilidade e Especificidade , Linfócitos T/imunologia , Linfócitos T/metabolismo
5.
J Biol Chem ; 288(19): 13676-94, 2013 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-23532844

RESUMO

BACKGROUND: Missense mutations in AHI1 result in the neurodevelopmental ciliopathy called Joubert syndrome. RESULTS: Mutations in AHI1 decrease cilia formation, alter its localization and stability, and change its binding to HAP1 and NPHP1. CONCLUSION: Mutations in AHI1 affect ciliogenesis, AHI1 protein localization, and AHI1-protein interactions. SIGNIFICANCE: This study begins to describe how missense mutations in AHI1 can cause Joubert syndrome. Mutations in AHI1 cause Joubert syndrome (JBTS), a neurodevelopmental ciliopathy, characterized by midbrain-hindbrain malformations and motor/cognitive deficits. Here, we show that primary cilia (PC) formation is decreased in fibroblasts from individuals with JBTS and AHI1 mutations. Most missense mutations in AHI1, causing JBTS, occur in known protein domains, however, a common V443D mutation in AHI1 is found in a region with no known protein motifs. We show that cells transfected with AHI1-V443D, or a new JBTS-causing mutation, AHI1-R351L, have aberrant localization of AHI1 at the basal bodies of PC and at cell-cell junctions, likely through decreased binding of mutant AHI1 to NPHP1 (another JBTS-causing protein). The AHI1-V443D mutation causes decreased AHI1 stability because there is a 50% reduction in AHI1-V443D protein levels compared with wild type AHI1. Huntingtin-associated protein-1 (Hap1) is a regulatory protein that binds Ahi1, and Hap1 knock-out mice have been reported to have JBTS-like phenotypes, suggesting a role for Hap1 in ciliogenesis. Fibroblasts and neurons with Hap1 deficiency form PC with normal growth factor-induced ciliary signaling, indicating that the Hap1 JBTS phenotype is likely not through effects at PC. These results also suggest that the binding of Ahi1 and Hap1 may not be critical for ciliary function. However, we show that HAP1 has decreased binding to AHI1-V443D indicating that this altered binding could be responsible for the JBTS-like phenotype through an unknown pathway. Thus, these JBTS-associated missense mutations alter their subcellular distribution and protein interactions, compromising functions of AHI1 in cell polarity and cilium-mediated signaling, thereby contributing to JBTS.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Doenças Cerebelares/genética , Anormalidades do Olho/genética , Doenças Renais Císticas/genética , Mutação de Sentido Incorreto , Anormalidades Múltiplas , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transporte Vesicular , Sequência de Aminoácidos , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Polaridade Celular , Células Cultivadas , Doenças Cerebelares/metabolismo , Doenças Cerebelares/patologia , Cerebelo/anormalidades , Cílios/metabolismo , Cílios/patologia , Sequência Conservada , Proteínas do Citoesqueleto , Anormalidades do Olho/metabolismo , Anormalidades do Olho/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Humanos , Junções Intercelulares/metabolismo , Doenças Renais Císticas/metabolismo , Doenças Renais Císticas/patologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapas de Interação de Proteínas , Estabilidade Proteica , Transporte Proteico , Retina/anormalidades , Retina/metabolismo , Retina/patologia , Transdução de Sinais , Técnicas do Sistema de Duplo-Híbrido
6.
Arterioscler Thromb Vasc Biol ; 32(11): 2662-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22922963

RESUMO

OBJECTIVE: Abnormal proliferation and migration of vascular smooth muscle cells (SMCs) are the key events in the progression of neointima formation in response to vascular injury. The goal of this study is to investigate the functional role of a potent oncogene yes-associated protein (YAP) in SM phenotypic modulation in vitro and in vivo. METHODS AND RESULTS: In vitro cell culture and in vivo in both mouse and rat arterial injury models YAP expression is significantly induced and correlated with the vascular SMC synthetic phenotype. Overexpression of YAP promotes SMC migration and proliferation while attenuating SM contractile gene expression. Conversely, knocking down endogenous YAP in SMCs upregulates SM gene expression but attenuates SMC proliferation and migration. Consistent with this, knocking down YAP expression in a rat carotid balloon injury model and genetic deletion of YAP, specifically, in vascular SMCs in mouse after carotid artery ligation injury attenuates injury-induced SM phenotypic switch and neointima formation. CONCLUSIONS: YAP plays a novel integrative role in SM phenotypic modulation by inhibiting SM-specific gene expression while promoting SM proliferation and migration in vitro and in vivo. Blocking the induction of YAP would be a potential therapeutic approach for ameliorating vascular occlusive diseases.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/metabolismo , Lesões das Artérias Carótidas/enzimologia , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Fosfoproteínas/metabolismo , Lesões do Sistema Vascular/enzimologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Aorta/enzimologia , Aorta/patologia , Proteínas Reguladoras de Apoptose/genética , Artérias Carótidas/enzimologia , Artérias Carótidas/patologia , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/patologia , Lesões das Artérias Carótidas/prevenção & controle , Proteínas de Ciclo Celular , Movimento Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Genótipo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Neointima , Fenótipo , Fosfoproteínas/genética , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Transfecção , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/patologia , Lesões do Sistema Vascular/prevenção & controle , Proteínas de Sinalização YAP
7.
Front Immunol ; 11: 269, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32153579

RESUMO

Macrophages are a heterogeneous and plastic population of cells whose phenotype changes in response to their environment. Macrophage biologists utilize peritoneal (pMAC) and bone marrow-derived macrophages (BMDM) for in vitro studies. Given that pMACs mature in vivo while BMDM are ex vivo differentiated from stem cells, it is likely that their responses differ under experimental conditions. Surprisingly little is known about how BMDM and pMACs responses compare under the same experimental conditionals. While morphologically similar with respect to forward and side scatter by flow cytometry, reports in the literature suggest that pMACs are more mature than their BMDM counterparts. Given the dearth of information comparing BMDM and pMACs, this work was undertaken to test the hypothesis that elicited pMACs are more responsive to defined conditions, including phagocytosis, respiratory burst, polarization, and cytokine and chemokine release. In all cases, our hypothesis was disproved. At steady state, BMDM are more phagocytic (both rate and extent) than elicited pMACs. In response to polarization, they upregulate chemokine and cytokine gene expression and release more cytokines. The results demonstrate that BMDM are generally more responsive and poised to respond to their environment, while pMAC responses are, in comparison, less pronounced. BMDM responses are a function of intrinsic differences, while pMAC responses reflect their differentiation in the context of the whole animal. This distinction may be important in knockout animals, where the pMAC phenotype may be influenced by the absence of the gene of interest.


Assuntos
Macrófagos Peritoneais/imunologia , Macrófagos/imunologia , Animais , Diferenciação Celular , Células Cultivadas , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fagocitose , Transcriptoma
8.
Anal Chem ; 81(24): 9961-71, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-19904964

RESUMO

Matrix metalloproteinases (MMPs) are a family of endoproteases that break down extracellular matrix and whose upregulation contributes to several diseases. A liquid chromatography/tandem mass spectrometry (LC/MS/MS) method was developed to quantify MMP-1 and MMP-9 substrates and their N-terminal peptide products in samples obtained from implanted microdialysis sampling probes. In vitro studies with purified human MMP-1 and MMP-9 were used to optimize the assay and determine the effectiveness of the local delivery of a broad-spectrum MMP inhibitor, GM 6001. Localized delivery of GM 6001 at 10 microM was sufficient to completely inhibit product formation in vitro. In vivo studies in male Sprague-Dawley rats were performed with microdialysis probes implanted into the subcutaneous tissue. Directly after microdialysis probe implantation, infusions of the MMP-1 and MMP-9 substrates (50 microM each) resulted in recovered product concentrations of approximately 2 microM. During a 50 microM GM 6001 coinfusion with the substrates, a 30% and 25% reduction in product formation for the MMP-1 and MMP-9 substrates was obtained, respectively. Blank dialysates were negative for enzymatic activity that could cleave the MMP substrates. This method allowed for the activity of different MMPs surrounding the microdialysis probe to be observed during in vivo sampling.


Assuntos
Dipeptídeos/análise , Metaloproteinase 1 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Microdiálise , Animais , Cromatografia Líquida de Alta Pressão , Dipeptídeos/farmacologia , Humanos , Masculino , Metaloproteinase 1 da Matriz/química , Metaloproteinase 9 da Matriz/química , Inibidores de Metaloproteinases de Matriz , Ratos , Ratos Sprague-Dawley , Espectrometria de Massas em Tandem
9.
J Cell Biol ; 159(6): 939-44, 2002 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-12499353

RESUMO

Protein kinase C (PKC) plays a prominent role in immune signaling, and the paradigms for isoform selective signaling are beginning to be elucidated. Real-time microscopy was combined with molecular and biochemical approaches to demonstrate a role for PKC- epsilon in Fc gamma receptor (Fc gammaR)-dependent phagocytosis. RAW 264.7 macrophages were transfected with GFP-conjugated PKC isoforms, and GFP movement was followed during phagocytosis of fluorescent IgG-opsonized beads. PKC- epsilon, but not PKC-delta, concentrated around the beads. PKC- epsilon accumulation was transient; apparent as a "flash" on target ingestion. Similarly, endogenous PKC- epsilon was specifically recruited to the nascent phagosomes in a time-dependent manner. Overexpression of PKC- epsilon, but not PKC-alpha, PKC-delta, or PKC-gamma enhanced bead uptake 1.8-fold. Additionally, the rate of phagocytosis in GFP PKC- epsilon expressors was twice that of cells expressing GFP PKC-delta. Expression of the regulatory domain ( epsilon RD) and the first variable region ( epsilon V1) of PKC- epsilon inhibited uptake, whereas the corresponding PKC-delta region had no effect. Actin polymerization was enhanced on expression of GFP PKC- epsilon and epsilon RD, but decreased in cells expressing epsilon V1, suggesting that the epsilon RD and epsilon V1 inhibition of phagocytosis is not due to effects on actin polymerization. These results demonstrate a role for PKC- epsilon in Fc gammaR-mediated phagocytosis that is independent of its effects on actin assembly.


Assuntos
Isoenzimas/fisiologia , Proteína Quinase C/fisiologia , Receptores de IgG/metabolismo , Animais , Linhagem Celular , Dextranos/farmacologia , Genes Dominantes , Proteínas de Fluorescência Verde , Imunoglobulina G/metabolismo , Isoenzimas/metabolismo , Proteínas Luminescentes/metabolismo , Macrófagos , Camundongos , Fagocitose , Faloidina/farmacologia , Isoformas de Proteínas , Proteína Quinase C/metabolismo , Proteína Quinase C-delta , Proteína Quinase C-épsilon , Estrutura Terciária de Proteína , Rodaminas/farmacologia , Fatores de Tempo , Transfecção
10.
Mol Biol Cell ; 17(2): 799-813, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16319178

RESUMO

Protein kinase C-epsilon (PKC-epsilon) translocates to phagosomes and promotes uptake of IgG-opsonized targets. To identify the regions responsible for this concentration, green fluorescent protein (GFP)-protein kinase C-epsilon mutants were tracked during phagocytosis and in response to exogenous lipids. Deletion of the diacylglycerol (DAG)-binding epsilonC1 and epsilonC1B domains, or the epsilonC1B point mutant epsilonC259G, decreased accumulation at phagosomes and membrane translocation in response to exogenous DAG. Quantitation of GFP revealed that epsilonC259G, epsilonC1, and epsilonC1B accumulation at phagosomes was significantly less than that of intact PKC-epsilon. Also, the DAG antagonist 1-hexadecyl-2-acetyl glycerol (EI-150) blocked PKC-epsilon translocation. Thus, DAG binding to epsilonC1B is necessary for PKC-epsilon translocation. The role of phospholipase D (PLD), phosphatidylinositol-specific phospholipase C (PI-PLC)-gamma1, and PI-PLC-gamma2 in PKC-epsilon accumulation was assessed. Although GFP-PLD2 localized to phagosomes and enhanced phagocytosis, PLD inhibition did not alter target ingestion or PKC-epsilon localization. In contrast, the PI-PLC inhibitor U73122 decreased both phagocytosis and PKC-epsilon accumulation. Although expression of PI-PLC-gamma2 is higher than that of PI-PLC-gamma1, PI-PLC-gamma1 but not PI-PLC-gamma2 consistently concentrated at phagosomes. Macrophages from PI-PLC-gamma2-/- mice were similar to wild-type macrophages in their rate and extent of phagocytosis, their accumulation of PKC-epsilon at the phagosome, and their sensitivity to U73122. This implicates PI-PLC-gamma1 as the enzyme that supports PKC-epsilon localization and phagocytosis. That PI-PLC-gamma1 was transiently tyrosine phosphorylated in nascent phagosomes is consistent with this conclusion. Together, these results support a model in which PI-PLC-gamma1 provides DAG that binds to epsilonC1B, facilitating PKC-epsilon localization to phagosomes for efficient IgG-mediated phagocytosis.


Assuntos
Fagocitose , Fosfolipase C gama/fisiologia , Proteína Quinase C-épsilon/metabolismo , Receptores de IgG/fisiologia , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , Diglicerídeos/antagonistas & inibidores , Diglicerídeos/metabolismo , Diglicerídeos/farmacologia , Proteínas de Fluorescência Verde/análise , Imunoglobulina G/metabolismo , Macrófagos/fisiologia , Camundongos , Modelos Biológicos , Fagossomos/metabolismo , Fagossomos/ultraestrutura , Fosfolipase D/fisiologia , Proteína Quinase C-épsilon/química , Estrutura Terciária de Proteína/fisiologia , Transporte Proteico , Proteínas Recombinantes de Fusão/análise
11.
ACS Appl Bio Mater ; 2(4): 1498-1508, 2019 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-31061988

RESUMO

Macrophages are immune cells involved in wound healing and tissue regeneration; however, the sustained presence of proinflammatory macrophages in wound sites impairs healing. In this study, we shifted peritoneal macrophage polarization away from a proinflammatory (M1) phenotype through exposure to stabilized interleukin-4 (IL-4) in poly(lactic-co-glycolic acid) films in combination with topographical guidance from electrospun poly-L-lactic acid fibers. To our knowledge, this was the first study to stabilize IL-4 with bovine serum albumin (BSA) within a biomaterial. When IL-4 was coloaded with BSA for stabilization, we saw increased IL-4 bioactivity compared to no added stabilization, trehalose stabilization, or murine serum albumin stabilization. We observed increased elongation of peritoneal macrophages, increased RNA expression of anti-inflammatory marker arginase-1, increased ratio of interleukin-10/interleukin- 12 p40 RNA, and decreased protein expression of proinflammatory markers (interleukin-12 p40 and RANTES) compared to controls. Taken together, these results suggest the macrophages were less proinflammatory and were a more pro-resolving phenotype. When stabilized with BSA, IL-4-loaded films effectively shift macrophage polarization state and are thus promising scaffolds to reduce inflammation within in vivo injury models.

12.
Cytokine ; 43(1): 15-9, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18519165

RESUMO

Cytokines are important cellular signaling proteins involved in inflammation, wound healing and are thought to direct the foreign body response to implanted materials. In this work, polyurethane tubes (25 mm length, 1.02 mm i.d., and 1.65 mm o.d.) were implanted into subcutaneous tissue of male Sprague-Dawley rats. The tubes served as the biomaterial and a means to collect the interstitial fluid that would be exchanged within the tube lumen and the surrounding tissue. After 3 and 7 days, the tubes were explanted and cytokines in the fluid were quantified with a multiplexed cytokine immunoassay. Six cytokines, interleukin-1beta (IL-1beta), IL-4, IL-6, IL-10, macrophage chemoattractant protein-1 (MCP-1), and tumor necrosis factor-alpha (TNF-alpha), were simultaneously quantified. All cytokine concentrations with the exception of IL-4 and TNF-alpha ranged between low pg/mL to mid ng/mL levels. Neither TNF-alpha nor IL-4 was detected from any sample. These results illustrate the potential of using the tube materials combined with bead-based immunoassays as a direct method for in vivo collection of multiple cytokines in low microliter sample volumes for fixed day biomaterial implant studies.


Assuntos
Citocinas/metabolismo , Líquido Extracelular/metabolismo , Próteses e Implantes , Animais , Quimiocina CCL2/análise , Citocinas/análise , Líquido Extracelular/química , Líquido Extracelular/imunologia , Estudos de Viabilidade , Imunoensaio , Mediadores da Inflamação/análise , Interleucina-10/análise , Interleucina-1beta/análise , Interleucina-4/análise , Interleucina-6/análise , Masculino , Poliuretanos , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/análise
13.
J Immunol Sci ; 2(2): 26-32, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30112519

RESUMO

During phagocytosis, internal membranes are recruited to the site of pathogen binding and fuse with the plasma membrane, providing the membrane needed for pseudopod extension and target uptake. The mechanism by which vesicles destined for the phagosome are generated, targeted, and fuse is unknown. We established that Golgi-associated protein kinase C-epsilon (PKC-ε) is necessary for the addition of membrane during FcyR-mediated phagocytosis. PKC-ε is tethered to the Golgi through interactions between its' regulatory domain and the Golgi lipids PI4P and diacylglycerol; disruption of these interactions prevents PKC-ε concentration at phagosomes and decreases phagocytosis. The accumulated evidence suggests that PKC-ε orchestrates vesicle formation at the Golgi by a mechanism requiring lipid binding but not enzymatic activity. This review discusses how PKC-ε might mediate vesicle formation at the level of budding and fission. Specifically, we discuss PKC-ε binding partners, the formation of lipid subdomains to generate membrane curvature, and PKC-ε mediated links to the actin and microtubule cytoskeleton to provide tension for vesicle fission. Assimilating information from several model systems, we propose a model for PKC-ε mediated vesicle formation for exocytosis during phagocytosis that may be applicable to other processes that require directed membrane delivery and fusion.

14.
J Leukoc Biol ; 79(2): 408-15, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16330529

RESUMO

Ligation of Fc receptors for immunoglobulin G (FcgammaRs) inhibits lipopolysaccharide (LPS)-stimulated secretion of interleukin (IL)-12 by macrophages. FcgammaR activation of protein kinase C (PKC) contributes to several functions of this receptor including phagocytosis, activation of the reduced nicotinamide adenine dinucleotide phosphate oxidase, and secretion of certain cytokines. Therefore, we tested the hypothesis that PKC mediates the FcgammaR inhibition of IL-12 secretion by macrophages. In murine macrophages, FcgammaR ligation augmented LPS-stimulated activation of PKC-alpha and PKC-delta but reduced IL-12p40 secretion. Similarly, activation of PKC with phorbol 12-myristate 13-acetate (PMA) depressed LPS-stimulated IL-12p40 secretion, and depletion of PKC augmented LPS-stimulated IL-12p40 secretion. Antisense down-regulation of PKC-delta increased LPS-stimulated IL-12p40 secretion and fully prevented the effects of FcgammaR ligation or PMA on IL-12p40 secretion. In contrast, down-regulation of PKC-epsilon blocked LPS-stimulated secretion of IL-12p40. Down-regulation of PKC-alpha had no effect on LPS-stimulated IL-12p40 secretion. The results suggest a negative role for PKC-delta and a positive role for PKC-epsilon in the regulation of LPS-stimulated IL-12p40 secretion.


Assuntos
Interleucina-12/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/efeitos dos fármacos , Proteína Quinase C/fisiologia , Receptores de IgG/imunologia , Animais , Células Cultivadas , Regulação para Baixo , Interleucina-12/antagonistas & inibidores , Isoenzimas/efeitos dos fármacos , Isoenzimas/fisiologia , Ligantes , Lipopolissacarídeos/antagonistas & inibidores , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase C/efeitos dos fármacos , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacologia , Fator de Necrose Tumoral alfa/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo
15.
ACS Chem Neurosci ; 8(4): 752-758, 2017 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-28140557

RESUMO

Peritoneal macrophages (PMACs) and spinal cord astrocytes were exposed to varying concentrations of soluble sophorolipid butyl ester diacetate (SLBEDA) in vitro. Macrophages and astrocytes demonstrated no decrease in viability in response to SLBEDA. Studying pro- and anti-inflammatory genes, PMACs did not show a shift toward a pro-inflammatory phenotype. However, at higher concentrations (3 and 30 µM), astrocytes showed an increase in their expression of glial acidic fibrillary protein. This novel category of compounds poses low risk to PMAC and astrocyte viability; however, the effect on PMAC polarization and astrocyte reactivity requires more elucidation.


Assuntos
Astrócitos/metabolismo , Polaridade Celular/efeitos dos fármacos , Proteína Glial Fibrilar Ácida/biossíntese , Glicolipídeos/farmacologia , Macrófagos/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL
16.
J Biomater Sci Polym Ed ; 28(13): 1303-1323, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28420296

RESUMO

Currently, it is unknown how the mechanical properties of electrospun fibers, and the presentation of surface nanotopography influence macrophage gene expression and protein production. By further elucidating how specific fiber properties (mechanical properties or surface properties) alter macrophage behavior, it may be possible to create electrospun fiber scaffolds capable of initiating unique cellular and tissue responses. In this study, we determined the elastic modulus and rigidity of fibers with varying topographies created by finely controlling humidity and including a non-solvent during electrospinning. In total,five fiber scaffold types were produced. Analysis of fiber physical properties demonstrated no change in fiber diameter amongst the five different fiber groups. However, the four different fibrous scaffolds with nanopits or divots each possessed different numbers of pits with different nanoscale dimensions. Unpolarized bone marrow derived murine macrophages (M0), macrophages polarized towards a pro-inflammatory phenotype (M1), or macrophages polarized towards anti-inflammatory phenotype (M2b) were placed onto each of the scaffolds and cytokine RNA expression and protein production were analyzed. Specific nanotopographies did not appreciably alter cytokine production from undifferentiated macrophages (M0) or anti-inflammatory macrophages (M2b), but a specific fiber (with many small pits) did increase IL-12 transcript and IL-12 protein production compared to fibers with small divots. When analyzing the mechanical properties between fibers with divots or with many small pits,divoted fibers possessed similar elastic moduli but different stiffness values. In total,we present techniques capable of creating unique electrospun fibers. These unique fibers have varying fiber mechanical characteristics and modestly modulate macrophage cytokine expression.


Assuntos
Citocinas/biossíntese , Eletricidade , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Nanotecnologia/métodos , Alicerces Teciduais/química , Animais , Células da Medula Óssea/citologia , Macrófagos/citologia , Fenômenos Mecânicos , Camundongos , Células RAW 264.7 , Propriedades de Superfície
17.
J Pharm Biomed Anal ; 40(4): 915-21, 2006 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-16242281

RESUMO

Microdialysis sampling probes were used to collect cytokine samples from lipopolysaccharide (LPS)-stimulated macrophages. The probes were immersed into cell culture wells containing either RAW 264.7 or isolated peritoneal macrophages. Dialysates (15 microL) from these wells were subjected to a multiplexed cytokine sandwich immunoassay platform analyzed by flow cytometry that measures up to six separate cytokines, interleukin-6 (IL-6), interleukin-10 (IL-10), interleukin-12p70 (IL-12p70), interferon-gamma (IFN-gamma), macrophage chemoattractant protein-1 (MCP-1), and tumor necrosis factor-alpha (TNF-alpha) in a single 15-muL sample. In vitro microdialysis sampling relative recovery experiments showed that only IFN-gamma, IL-6, MCP-1, and TNF-alpha could be recovered across a commercially-available 100-kDa MWCO microdialysis membrane. Eleven hours after LPS addition (1 microg/mL), RAW 264.7 macrophages secreted greater than 8000 pg/mL of TNF-alpha and greater than 1000 pg/mL MCP-1. With the peritoneal macrophages, greater than 6000 pg/mL of IL-6, MCP-1, and TNF-alpha were obtained. The maximum dialysate concentrations obtained from the RAW macrophages were 1300 pg/mL for TNF-alpha and 55 pg/mL for MCP-1. Maximum cytokine concentrations from peritoneal macrophage dialysates reached approximately 2000 pg/mL, 1100 pg/mL and 500 pg/mL for TNF-alpha, MCP-1 and IL-6, respectively. Microdialysis sampling allowed for 20-min samples to be collected during the cytokine release from the activated macrophages. These results demonstrate that microdialysis sampling can be used for collection of selected cytokines with improved temporal resolution.


Assuntos
Citocinas/análise , Ativação de Macrófagos , Macrófagos Peritoneais/metabolismo , Animais , Linhagem Celular , Quimiocina CCL2/análise , Quimiocina CCL2/metabolismo , Meios de Cultura/metabolismo , Citocinas/metabolismo , Citometria de Fluxo , Imunoensaio/métodos , Interleucina-6/análise , Interleucina-6/metabolismo , Lipopolissacarídeos , Macrófagos Peritoneais/imunologia , Masculino , Camundongos , Microdiálise , Peso Molecular , Fator de Necrose Tumoral alfa/análise , Fator de Necrose Tumoral alfa/metabolismo
18.
Mol Cancer Ther ; 3(11): 1355-64, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15542774

RESUMO

Resveratrol, a naturally occurring stilbene with antitumor properties, caused mitogen-activated protein kinase [MAPK, extracellular signal-regulated kinase 1/2 (ERK1/2)] activation, nuclear translocation of Ser15-phosphorylated p53, and p53-dependent apoptosis in hormone-insensitive DU145 prostate cancer cells. Exposure of these cells to epidermal growth factor (EGF) for up to 4 hours resulted in brief activation of MAPK followed by inhibition of resveratrol-induced signal transduction, p53 phosphorylation, and apoptosis. Resveratrol stimulated c-fos and c-jun expression in DU145 cells, an effect also suppressed by EGF. An inhibitor of protein kinase C (PKC)-alpha, -beta, and -gamma (CGP41251) enhanced Ser15 phosphorylation of p53 by resveratrol in the absence of EGF and blocked EGF inhibition of the resveratrol effect. EGF caused PKC-alpha/beta phosphorylation in DU145 cells, an effect reversed by CGP41251. Activation of PKC by phorbol ester (phorbol 12-myristate 13-acetate) enhanced EGF action on ERK1/2 phosphorylation without significantly altering p53 phosphorylation by resveratrol. DU145 cells transfected with a dominant-negative PKC-alpha construct showed resveratrol-induced ERK1/2 phosphorylation and Ser15 phosphorylation of p53 but were unresponsive to EGF. Thus, resveratrol and EGF activate MAPK by discrete mechanisms in DU145 cells. The stilbene promoted p53-dependent apoptosis, whereas EGF opposed induction of apoptosis by resveratrol via a PKC-alpha-mediated mechanism. Resveratrol also induced p53 phosphorylation in LNCaP prostate cancer cells, an effect also inhibited by EGF. Inhibition of PKC activation in LNCaP cells, however, resulted in a reduction, rather than increase, in p53 activation and apoptosis, suggesting that resveratrol-induced apoptosis in these two cell lines occurs through different PKC-mediated and MAPK-dependent pathways.


Assuntos
Apoptose/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Proteína Quinase C/metabolismo , Estilbenos/antagonistas & inibidores , Estilbenos/farmacologia , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C-alfa , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Resveratrol , Transdução de Sinais/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo
19.
Inflammation ; 28(1): 23-31, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15072227

RESUMO

Fc gamma receptor (Fc gammaR) signaling mediates several important macrophage functions including cytokine secretion and respiratory burst. The present study describes the development of a model using the macrophage cell line, RAW 264.7 for studying Fc gammaR-stimulated tumor necrosis factor-alpha (TNF-alpha) secretion and hydrogen peroxide (H2O2) production. In unprimed cells these functions were low but pretreatment with interferon-gamma augmented Fc gammaR-stimulated TNF-alpha secretion and H2O2 production to levels that were about half that caused by lipopolysaccharide (LPS) and zymosan, respectively. Studies on the signaling pathways found that TNF-alpha secretion stimulated by either Fc gammaR or LPS was decreased by inhibitors of PKC, MAPK p42/p44, and MAPK p38. TNF-alpha secretion was also reduced by the combination of PLC and PLD inhibitors but not by the individual inhibitors alone. H2O2 production stimulated by either Fc gammaR or zymosan was blocked by inhibitors of PKC, PLC, PLD, and MAPK p42/44 but not by MAPK p38. Thus, interferon-gamma treated RAW 264.7 cells are a model of inflammatory macrophages and are well suited for further study of these signaling pathways.


Assuntos
Macrófagos/metabolismo , Receptores de IgG/fisiologia , Explosão Respiratória/fisiologia , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Linhagem Celular , Estrenos/farmacologia , Macrófagos/efeitos dos fármacos , Camundongos , Pirrolidinonas/farmacologia , Explosão Respiratória/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
20.
Inflammation ; 26(6): 305-10, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12546140

RESUMO

Lipopolysaccharide (LPS) increases serum TNF-alpha levels due to TNF-alpha secretion by macrophages. The serum TNF-alpha response to LPS was augmented 10x when FcgammaR ligation was induced by the intravenous injection of Gig-coated erythrocytes (IgG) prior to the administration of LPS. The macrophage population responsible for the augmented TNF-alpha secretion was determined by isolating Kupffer cells, splenic macrophages and peritoneal macrophages from mice that had been given ElgG prior to LPS and determining TNF-alpha secretion ex vivo. The intravenous injection of ElgG augmented LPS-stimulate TNF-alpha secretion by Kupffer cells and splenic macrophages. In contrast, LPS-stimulated TNF-alpha secretion by peritoneal macrophages was not altered by either the intravenous or intraperitoneal injection of ElgG. In vitro phagocytosis of ElgG by isolated peritoneal macrophages also did not augment LPS-stimulated TNF-alpha secretion. These results show that FcgammaR ligation augments LPS-stimulated TNF-alpha secretion by Kupffer cells and splenic macrophages but not by peritoneal macrophages. Thus, the ability of FcgammaR ligation to influence TNF-alpha secretion may be specific to the tissue source of the macrophages.


Assuntos
Fígado/metabolismo , Macrófagos/metabolismo , Receptores de IgG/metabolismo , Baço/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Lipopolissacarídeos/farmacologia , Fígado/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Baço/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA