Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
1.
Front Immunol ; 13: 801120, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35154114

RESUMO

An increase in mast cells (MCs) and MCs mediators has been observed in malaria-associated bacteremia, however, the role of these granulocytes in malarial immunity is poorly understood. Herein, we studied the role of mouse MC protease (Mcpt) 4, an ortholog of human MC chymase, in malaria-induced bacteremia using Mcpt4 knockout (Mcpt4-/-) mice and Mcpt4+/+ C57BL/6J controls, and the non-lethal mouse parasite Plasmodium yoelii yoelii 17XNL. Significantly lower parasitemia was observed in Mcpt4-/- mice compared with Mcpt4+/+ controls by day 10 post infection (PI). Although bacterial 16S DNA levels in blood were not different between groups, increased intestinal permeability to FITC-dextran and altered ileal adherens junction E-cadherin were observed in Mcpt4-/- mice. Relative to infected Mcpt4+/+ mice, ileal MC accumulation in Mcpt4-/- mice occurred two days earlier and IgE levels were higher by days 8-10 PI. Increased levels of circulating myeloperoxidase were observed at 6 and 10 days PI in Mcpt4+/+ but not Mcpt4-/- mice, affirming a role for neutrophil activation that was not predictive of parasitemia or bacterial 16S copies in blood. In contrast, early increased plasma levels of TNF-α, IL-12p40 and IL-3 were observed in Mcpt4-/- mice, while levels of IL-2, IL-10 and MIP1ß (CCL4) were increased over the same period in Mcpt4+/+ mice, suggesting that the host response to infection was skewed toward a type-1 immune response in Mcpt4-/- mice and type-2 response in Mcpt4+/+ mice. Spearman analysis revealed an early (day 4 PI) correlation of Mcpt4-/- parasitemia with TNF-α and IFN-γ, inflammatory cytokines known for their roles in pathogen clearance, a pattern that was observed in Mcpt4+/+ mice much later (day 10 PI). Transmission success of P. y. yoelii 17XNL to Anopheles stephensi was significantly higher from infected Mcpt4-/- mice compared with infected Mcpt4+/+ mice, suggesting that Mcpt4 also impacts transmissibility of sexual stage parasites. Together, these results suggest that early MCs activation and release of Mcpt4 suppresses the host immune response to P. y. yoelii 17XNL, perhaps via degradation of TNF-α and promotion of a type-2 immune response that concordantly protects epithelial barrier integrity, while limiting the systemic response to bacteremia and parasite transmissibility.


Assuntos
Anopheles/parasitologia , Permeabilidade da Membrana Celular/imunologia , Quimases/genética , Quimases/imunologia , Interações Hospedeiro-Parasita/imunologia , Malária/imunologia , Mastócitos/enzimologia , Plasmodium yoelii/imunologia , Animais , Feminino , Íleo/citologia , Íleo/patologia , Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fator de Necrose Tumoral alfa/metabolismo
2.
Front Immunol ; 12: 650522, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33897698

RESUMO

Proinflammatory stimuli lead to endothelial injury, which results in pathologies such as cardiovascular diseases, autoimmune diseases, and contributes to alloimmune responses after organ transplantation. Both mesenchymal stromal cells (MSC) and the extracellular vesicles (EV) released by them are widely studied as regenerative therapy for the endothelium. However, for therapeutic application, the manipulation of living MSC and large-scale production of EV are major challenges. Membrane particles (MP) generated from MSC may be an alternative to the use of whole MSC or EV. MP are nanovesicles artificially generated from the membranes of MSC and possess some of the therapeutic properties of MSC. In the present study we investigated whether MP conserve the beneficial MSC effects on endothelial cell repair processes under inflammatory conditions. MP were generated by hypotonic shock and extrusion of MSC membranes. The average size of MP was 120 nm, and they showed a spherical shape. The effects of two ratios of MP (50,000; 100,000 MP per target cell) on human umbilical vein endothelial cells (HUVEC) were tested in a model of inflammation induced by TNFα. Confocal microscopy and flow cytometry showed that within 24 hours >90% of HUVEC had taken up MP. Moreover, MP ended up in the lysosomes of the HUVEC. In a co-culture system of monocytes and TNFα activated HUVEC, MP did not affect monocyte adherence to HUVEC, but reduced the transmigration of monocytes across the endothelial layer from 138 ± 61 monocytes per microscopic field in TNFα activated HUVEC to 61 ± 45 monocytes. TNFα stimulation induced a 2-fold increase in the permeability of the HUVEC monolayer measured by the translocation of FITC-dextran to the lower compartment of a transwell system. At a dose of 1:100,000 MP significantly decreased endothelial permeability (1.5-fold) respect to TNFα Stimulated HUVEC. Finally, MP enhanced the angiogenic potential of HUVEC in an in vitro Matrigel assay by stimulating the formation of angiogenic structures, such as percentage of covered area, total tube length, total branching points, total loops. In conclusion, MP show regenerative effects on endothelial cells, opening a new avenue for treatment of vascular diseases where inflammatory processes damage the endothelium.


Assuntos
Tecido Adiposo/citologia , Vesículas Extracelulares/imunologia , Células Endoteliais da Veia Umbilical Humana/imunologia , Células-Tronco Mesenquimais/imunologia , Monócitos/imunologia , Adesão Celular/imunologia , Permeabilidade da Membrana Celular/imunologia , Células Cultivadas , Técnicas de Cocultura , Microscopia Crioeletrônica , DNA/genética , DNA/isolamento & purificação , Vesículas Extracelulares/genética , Vesículas Extracelulares/ultraestrutura , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Microscopia Eletrônica de Transmissão , Monócitos/citologia , Tamanho da Partícula , RNA/genética , RNA/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa
3.
Poult Sci ; 99(11): 5407-5414, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33142457

RESUMO

In recent years, several studies emphasize the deleterious effects of Campylobacter jejuni on the chicken intestine. In this context, it was shown that C. jejuni, contrary to the general belief, has a negative influence on the gut barrier in chickens. More precisely, we demonstrated that C. jejuni affects gut physiology characterized by changes in ion transport and transepithelial ion conductance, but the underlying mechanism is yet to be investigated. In the actual study, to determine epithelial paracellular permeability, the mucosal to serosal flux of 14C-mannitol in the small and large intestine was measured applying Ussing chamber. A total of seventy-five 1-day-old Ross 308 broiler chickens were housed in floor pens on wood shavings with feed and water provided ad libitum. Birds were randomly allocated to 3 different groups (n = 25 with 5 replicates/group) and infected at 14 d of age with a high (108 colony forming units [CFU]) or a low (104 CFU) dose of C. jejuni and a third group kept as noninfected control. Infection with the low dose of C. jejuni resulted in delayed cecal colonization but equalized at 21 d postinfection, independent of the dose. Invasion of liver and spleen with C. jejuni was only noticed in birds infected with 108 (CFU). Body weight (BW) and body weight gain of all birds infected with C. jejuni were lower than in the control group and varied with the dose of infection, confirming a negative correlation between the infection dose and birds BW. Mannitol flux in jejunum and cecum was significantly (P < 0.05) higher in all C. jejuni infected birds compared with control birds. Likewise, significant differences in mannitol flux of both jejunum and cecum were detected depending on the infection dose of C. jejuni. The correlation analyses revealed a positive relationship between Campylobacter dose and mannitol flux of both jejunum and cecum. Altogether, the actual results emphasize that the adverse effect of C. jejuni on gut permeability arises in a dose-dependent manner.


Assuntos
Infecções por Campylobacter , Campylobacter jejuni , Doenças das Aves Domésticas , Animais , Infecções por Campylobacter/imunologia , Infecções por Campylobacter/veterinária , Permeabilidade da Membrana Celular/imunologia , Galinhas , Feminino , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Masculino , Doenças das Aves Domésticas/imunologia , Doenças das Aves Domésticas/microbiologia
4.
Nat Immunol ; 21(7): 746-755, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32514064

RESUMO

Plasma membranes of animal cells are enriched for cholesterol. Cholesterol-dependent cytolysins (CDCs) are pore-forming toxins secreted by bacteria that target membrane cholesterol for their effector function. Phagocytes are essential for clearance of CDC-producing bacteria; however, the mechanisms by which these cells evade the deleterious effects of CDCs are largely unknown. Here, we report that interferon (IFN) signals convey resistance to CDC-induced pores on macrophages and neutrophils. We traced IFN-mediated resistance to CDCs to the rapid modulation of a specific pool of cholesterol in the plasma membrane of macrophages without changes to total cholesterol levels. Resistance to CDC-induced pore formation requires the production of the oxysterol 25-hydroxycholesterol (25HC), inhibition of cholesterol synthesis and redistribution of cholesterol to an esterified cholesterol pool. Accordingly, blocking the ability of IFN to reprogram cholesterol metabolism abrogates cellular protection and renders mice more susceptible to CDC-induced tissue damage. These studies illuminate targeted regulation of membrane cholesterol content as a host defense strategy.


Assuntos
Infecções Bacterianas/imunologia , Toxinas Bacterianas/imunologia , Hidroxicolesteróis/metabolismo , Interferons/isolamento & purificação , Fagócitos/imunologia , Estreptolisinas/imunologia , Animais , Bactérias/imunologia , Bactérias/metabolismo , Proteínas de Bactérias/administração & dosagem , Proteínas de Bactérias/imunologia , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular/imunologia , Células Cultivadas , Modelos Animais de Doenças , Suscetibilidade a Doenças/imunologia , Feminino , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Microscopia Intravital , Masculino , Camundongos , Camundongos Transgênicos , Fagócitos/citologia , Fagócitos/metabolismo , Cultura Primária de Células , Esteroide Hidroxilases/genética , Esteroide Hidroxilases/metabolismo , Estreptolisinas/administração & dosagem , Estreptolisinas/metabolismo
5.
Nat Commun ; 11(1): 1995, 2020 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-32332732

RESUMO

Gut microbial dysbiosis is associated with the development of autoimmune disease, but the mechanisms by which microbial dysbiosis affects the transition from asymptomatic autoimmunity to inflammatory disease are incompletely characterized. Here, we identify intestinal barrier integrity as an important checkpoint in translating autoimmunity to inflammation. Zonulin family peptide (zonulin), a potent regulator for intestinal tight junctions, is highly expressed in autoimmune mice and humans and can be used to predict transition from autoimmunity to inflammatory arthritis. Increased serum zonulin levels are accompanied by a leaky intestinal barrier, dysbiosis and inflammation. Restoration of the intestinal barrier in the pre-phase of arthritis using butyrate or a cannabinoid type 1 receptor agonist inhibits the development of arthritis. Moreover, treatment with the zonulin antagonist larazotide acetate, which specifically increases intestinal barrier integrity, effectively reduces arthritis onset. These data identify a preventive approach for the onset of autoimmune disease by specifically targeting impaired intestinal barrier function.


Assuntos
Artrite Reumatoide/prevenção & controle , Permeabilidade da Membrana Celular/efeitos dos fármacos , Disbiose/complicações , Haptoglobinas/antagonistas & inibidores , Mucosa Intestinal/efeitos dos fármacos , Oligopeptídeos/administração & dosagem , Precursores de Proteínas/antagonistas & inibidores , Adulto , Animais , Artrite Experimental/sangue , Artrite Experimental/imunologia , Artrite Experimental/microbiologia , Artrite Experimental/prevenção & controle , Artrite Reumatoide/sangue , Artrite Reumatoide/imunologia , Artrite Reumatoide/microbiologia , Translocação Bacteriana/efeitos dos fármacos , Translocação Bacteriana/imunologia , Células CACO-2 , Permeabilidade da Membrana Celular/imunologia , Estudos de Coortes , Estudos Transversais , Disbiose/imunologia , Disbiose/microbiologia , Feminino , Microbioma Gastrointestinal/imunologia , Haptoglobinas/metabolismo , Voluntários Saudáveis , Humanos , Íleo/citologia , Íleo/efeitos dos fármacos , Íleo/microbiologia , Íleo/patologia , Mucosa Intestinal/citologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Masculino , Camundongos , Pessoa de Meia-Idade , Precursores de Proteínas/sangue , Precursores de Proteínas/metabolismo , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo
6.
Methods Enzymol ; 629: 115-150, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31727237

RESUMO

The P2X7 receptor [P2X7R or P2RX7 in National Center for Biotechnology Information (NCBI) gene nomenclature] is a member of the P2X receptor (P2XR) subfamily of P2 receptors (P2Rs). The P2X7R is an extracellular ATP-gated ion channel with peculiar permeability properties expressed by most cell types, mainly in the immune system, where it has a leading role in cytokine release, oxygen radical generation, T lymphocyte differentiation and proliferation. A role in cancer cell growth and tumor progression has also been demonstrated. These features make the P2X7R an appealing target for drug development in inflammation and cancer. The functional P2X7R, recently (partially) crystallized and 3-D solved, is formed by the assembly of three identical subunits (homotrimer). The P2X7R is preferentially permeable to small cations (Ca2+, Na+, K+), and in most (but not all) cell types also to large positively charged molecules of molecular mass up to 900Da. Permeability to negatively charged species of comparable molecular mass (e.g., Lucifer yellow) is debated. Several highly selective P2X7R pharmacological blockers have been developed over the years, thus providing powerful tools for P2X7R studies. Biophysical properties and coupling to several different physiological responses make the P2X7R amenable to investigation by electrophysiology and cell biology techniques, which allow its identification and characterization in many different cell types and tissues. A careful description of the physiological features of the P2X7R is a prerequisite for an effective therapeutic development. Here we describe the most common techniques to asses P2X7R functions, including patch-clamp, intracellular calcium measurements, and membrane permeabilization to large fluorescent dyes in a selection of different cell types. In addition, we also describe common toxicity assays used to verify the effects of P2X7R stimulation on cell viability.


Assuntos
Ensaios de Seleção de Medicamentos Antitumorais/métodos , Receptores Purinérgicos P2X7/análise , Trifosfato de Adenosina/imunologia , Trifosfato de Adenosina/metabolismo , Regulação Alostérica , Animais , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/imunologia , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/imunologia , Desenho de Fármacos , Corantes Fluorescentes/química , Corantes Fluorescentes/farmacologia , Técnicas de Inativação de Genes/instrumentação , Técnicas de Inativação de Genes/métodos , Células HEK293 , Humanos , Microscopia de Fluorescência/métodos , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/patologia , Técnicas de Patch-Clamp/instrumentação , Técnicas de Patch-Clamp/métodos , Cultura Primária de Células , Antagonistas do Receptor Purinérgico P2X/farmacologia , Antagonistas do Receptor Purinérgico P2X/uso terapêutico , Receptores Purinérgicos P2X7/química , Receptores Purinérgicos P2X7/genética , Receptores Purinérgicos P2X7/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Análise de Célula Única/métodos , Relação Estrutura-Atividade
7.
Methods Enzymol ; 629: 81-102, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31727258

RESUMO

The efficacy of cancer chemotherapy is enhanced by induction of sustainable anti-tumor immune responses. Such responses involve accumulation of immunogenic mediators, such as extracellular ATP and ATP metabolites, within the tumor microenvironment. Recent studies have identified nucleotide-permeable plasma membrane channels or pores that are activated as early downstream consequences of different regulated cell death pathways: pannexin-1 channels in apoptosis, MLKL pores in necroptosis, and gasdermin-family pores in pyroptosis. This chapter describes the use of highly quantitative and semi-high-throughput methods based on the ATP sensor luciferase to measure dynamic changes in extracellular ATP, ADP, and AMP in tissue/cell culture models of cancer cells during various modes of regulated cell death in response to chemotherapeutic drugs, death receptors, or metabolic perturbation.


Assuntos
Trifosfato de Adenosina/análise , Luciferases/química , Neoplasias/tratamento farmacológico , Difosfato de Adenosina/análise , Difosfato de Adenosina/imunologia , Difosfato de Adenosina/metabolismo , Monofosfato de Adenosina/análise , Monofosfato de Adenosina/imunologia , Monofosfato de Adenosina/metabolismo , Trifosfato de Adenosina/imunologia , Trifosfato de Adenosina/metabolismo , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/imunologia , Espaço Extracelular/imunologia , Espaço Extracelular/metabolismo , Humanos , Morte Celular Imunogênica/efeitos dos fármacos , Proteínas de Membrana Transportadoras/metabolismo , Camundongos , Neoplasias/imunologia , Neoplasias/patologia , Cultura Primária de Células , Piroptose/efeitos dos fármacos , Piroptose/imunologia , Ratos
8.
Cells ; 8(10)2019 09 21.
Artigo em Inglês | MEDLINE | ID: mdl-31546582

RESUMO

Approximately 40% of global HIV-1 transmission occurs in the female genital tract (FGT) through heterosexual transmission. Epithelial cells lining the FGT provide the first barrier to HIV-1 entry. Previous studies have suggested that certain hormonal contraceptives or a dysbiosis of the vaginal microbiota can enhance HIV-1 acquisition in the FGT. We examined the effects of lactobacilli and female sex hormones on the barrier functions and innate immune responses of primary endometrial genital epithelial cells (GECs). Two probiotic strains, Lactobacillus reuteri RC-14 and L. rhamnosus GR-1, were tested, as were sex hormones estrogen (E2), progesterone (P4), and the hormonal contraceptive medroxyprogesterone acetate (MPA). Our results demonstrate that probiotic lactobacilli enhance barrier function without affecting cytokines. Treatment of GECs with MPA resulted in reduced barrier function. In contrast, E2 treatment enhanced barrier function and reduced production of proinflammatory cytokines. Comparison of hormones plus lactobacilli as a pre-treatment prior to HIV exposure revealed a dominant effect of lactobacilli in preventing loss of barrier function by GECs. In summary, the combination of E2 and lactobacilli had the best protective effect against HIV-1 seen by enhancement of barrier function and reduction in proinflammatory cytokines. These studies provide insights into how probiotic lactobacilli in the female genital microenvironment can alter HIV-1-mediated barrier disruption and how the combination of E2 and lactobacilli may decrease susceptibility to primary HIV infection.


Assuntos
Permeabilidade da Membrana Celular/efeitos dos fármacos , Citoproteção/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Estradiol/farmacologia , Genitália Feminina/efeitos dos fármacos , HIV-1/fisiologia , Probióticos/farmacologia , Adulto , Antibiose/efeitos dos fármacos , Antibiose/fisiologia , Permeabilidade da Membrana Celular/imunologia , Células Cultivadas , Citoproteção/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Feminino , Genitália Feminina/metabolismo , Genitália Feminina/patologia , Genitália Feminina/virologia , Infecções por HIV/prevenção & controle , HIV-1/efeitos dos fármacos , HIV-1/patogenicidade , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/fisiologia , Lactobacillus/fisiologia , Pessoa de Meia-Idade , Cultura Primária de Células , Progesterona/farmacologia
9.
Front Immunol ; 10: 926, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31105711

RESUMO

Pituitary adenylate cyclase-activating polypeptide (PACAP) is a multifunctional neuropeptide that is widely distributed in mammals and is capable of performing roles as a neurotransmitter, neuromodulator, and vasodilator. This polypeptide belongs to the glucagon/secretin superfamily, of which some members have been shown to act as antimicrobial peptides in both mammalian and aquatic organisms. In teleosts, PACAP has been demonstrated to have direct antimicrobial activity against several aquatic pathogens, yet this phenomenon has never been studied throughout a live bacterial challenge. The present study focuses on the influence of synthetic Clarias gariepinus 38 amino acid PACAP on the rainbow trout monocyte/macrophage-like cell line, RTS11, when exposed to the coldwater bacterial pathogen Flavobacterium psychrophilum. PACAP was shown to have direct antimicrobial activity on F. psychrophilum when grown in both cytophaga broth and cell culture media (L-15). Further, the ability of teleostean PACAP to permeabilize the membrane of an aquatic pathogen, F. psychrophilum, was demonstrated for the first time. The viability of RTS11 when exposed to PACAP was also observed using a trypan blue exclusion assay to determine optimal experimental doses of the antimicrobial peptide. This displayed that only concentrations higher than 0.1 µM negatively impacted RTS11 survival. Interestingly, when RTS11 was pre-treated with PACAP for 24 h before experiencing infection with live F. psychrophilum, growth of the pathogen was severely inhibited in a dose-dependent manner when compared to cells receiving no pre-treatment with the polypeptide. Relative expression of pro-inflammatory cytokines (IL-1ß, TNFα, and IL-6) and PACAP receptors (VPAC1 and PAC1) was also analyzed in RTS11 following PACAP exposure alone and in conjunction with live F. psychrophilum challenge. These qRT-PCR findings revealed that PACAP may have a synergistic effect on RTS11 immune function. The results of this study provide evidence that PACAP has immunostimulatory activity on rainbow trout immune cells as well as antimicrobial activity against aquatic bacterial pathogens such as F. psychrophilum. As there are numerous pathogens that plague the aquaculture industry, PACAP may stimulate the teleost immune system while also providing an efficacious alternative to antibiotic use.


Assuntos
Permeabilidade da Membrana Celular/imunologia , Doenças dos Peixes/imunologia , Flavobacterium/imunologia , Macrófagos/imunologia , Oncorhynchus mykiss/imunologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/imunologia , Animais , Aquicultura/métodos , Linhagem Celular
10.
Inflamm Bowel Dis ; 25(7): 1196-1207, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-30794280

RESUMO

BACKGROUND: Inflammatory bowel disease (IBD) has a remitting and relapsing disease course; however, relatively little is understood regarding how inflammatory damage in acute colitis influences the microbiota, epithelial barrier, and immune function in subsequent colitis. METHODS: Mice were administered trinitrobenzene sulphonic acid (TNBS) via enema, and inflammation was assessed 2 days (d2) or 28 days (d28) later. Colitis was reactivated in some mice by re-treating at 28 days with TNBS and assessing 2 days later (d30). Epithelial responsiveness to secretagogues, microbiota composition, colonic infiltration, and immune activation was compared between all groups. RESULTS: At day 28, the distal colon had healed, mucosa was restored, and innate immune response had subsided, but colonic transepithelial transport (P = 0.048), regulatory T-cell (TREG) infiltration (P = 0.014), adherent microbiota composition (P = 0.0081), and responsiveness of stimulated innate immune bone marrow cells (P < 0.0001 for IL-1ß) differed relative to health. Two days after subsequent instillation of TNBS (d30 mice), the effects on inflammatory damage (P < 0.0001), paracellular permeability (P < 0.0001), and innate immune infiltration (P < 0.0001 for Ly6C+ Ly6G- macrophages) were reduced relative to d2 colitis. However, TREG infiltration was increased (P < 0.0001), and the responsiveness of stimulated T cells in the mesenteric lymph nodes shifted from pro-inflammatory at d2 to immune-suppressive at d30 (P < 0.0001 for IL-10). These effects were observed despite similar colonic microbiota composition and degradation of the mucosal layer between d2 and d30. CONCLUSIONS: Collectively, these results indicate that acute colitis chronically alters epithelial barrier function and both innate and adaptive immune responses. These effects reduce the consequences of a subsequent colitis event, warranting longitudinal studies in human IBD subjects.


Assuntos
Imunidade Adaptativa/imunologia , Permeabilidade da Membrana Celular/imunologia , Colite/complicações , Epitélio/patologia , Tolerância Imunológica/imunologia , Imunidade Inata/imunologia , Inflamação/etiologia , Doença Aguda , Animais , Colite/induzido quimicamente , Colite/patologia , Modelos Animais de Doenças , Epitélio/imunologia , Epitélio/lesões , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ácido Trinitrobenzenossulfônico/toxicidade
11.
Arthritis Res Ther ; 21(1): 34, 2019 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-30674349

RESUMO

BACKGROUND: Endothelial activation and damage is commonly observed in patients with systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) and is related to development of atherosclerosis and cardiovascular diseases. Different components of the immune system seem to participate in the endothelial injury, such as generation of autoantibodies and formation of immune complexes (ICs). Microparticles (MPs) and their immune complexes (MPs-ICs) are increased in the circulation of patients with SLE and RA; therefore, we propose these extracellular vesicles could interact and modulate the function of endothelial cells. Hence, the effect of MPs and MPs-ICs from patients with SLE and RA in endothelial cells was evaluated. METHODS: Macrovascular and microvascular endothelial cells were exposed to MPs and MPs-ICs from healthy donors and patients with SLE and RA. Vesicles uptake/binding, expression of adhesion molecules, cytokine and chemokine production, monocyte adherence, and alterations of endothelial monolayer were evaluated by flow cytometry and fluorescence microscopy. RESULTS: Endothelial cells internalized MPs and MPs-ICs and increased CD54 and CD102 expression and CCL2, CCL5, and IL-6 production after the treatment with these extracellular vesicles, which led to an increase in the adherence of classic monocytes. These vesicles also induced low expression of VE-cadherin in membrane, depolymerization of actin filaments, and formation of intercellular spaces, which led to endothelial death and increased permeability after MPs and MPs-ICs exposure. CONCLUSIONS: MPs and MPs-ICs from patients with SLE and RA increase adhesion molecules expression, chemokine production, and structural alterations in macrovascular and microvascular endothelial cells. Therefore, high counts of these vesicles in patients would promote endothelial alterations and secondary tissue leukocyte infiltration.


Assuntos
Artrite Reumatoide/imunologia , Micropartículas Derivadas de Células/imunologia , Células Endoteliais/imunologia , Endotélio/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Antígenos CD/imunologia , Antígenos CD/metabolismo , Artrite Reumatoide/metabolismo , Artrite Reumatoide/patologia , Caderinas/imunologia , Caderinas/metabolismo , Adesão Celular/imunologia , Permeabilidade da Membrana Celular/imunologia , Micropartículas Derivadas de Células/metabolismo , Células Cultivadas , Quimiocinas/imunologia , Quimiocinas/metabolismo , Citocinas/imunologia , Citocinas/metabolismo , Células Endoteliais/metabolismo , Endotélio/metabolismo , Endotélio/patologia , Citometria de Fluxo , Humanos , Lúpus Eritematoso Sistêmico/metabolismo , Lúpus Eritematoso Sistêmico/patologia , Microscopia de Fluorescência , Monócitos/imunologia , Monócitos/metabolismo
12.
Schizophr Res ; 208: 414-419, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30685393

RESUMO

About one third of people with schizophrenia have elevated IgG antibodies to gliadin (AGA IgG) and increased inflammation. Understanding the mechanism by which this immune response occurs is critical to the development of personalized treatments. We examined gut permeability and mimicry to the glutamate receptor as possible mechanisms related to high gliadin antibodies (AGA IgG) seen in some people with schizophrenia. The Glutamate Ionotropic Receptor NMDA type Subunit Associated with protein 1 (GRINA) has a similar protein structure to gliadin representing a potential target for cross reactivity or mimicry. In a population of schizophrenia subjects (N = 160) and healthy controls (N = 80) we analyzed serum samples for both GRINA and Anti-Saccharomyces Cerevisiae antibodies (ASCA), related to gut permeability. Schizophrenia patients compared to controls had a higher prevalence of positivity to ASCA IgA (p = 0.004) and IgG (p < 0.001). Multinomial logistic regression showed an association between AGA IgG and ASCA IgG in schizophrenia (p = 0.05 for the estimated regression coefficient) but not in healthy controls (p = 0.13). GRINA IgG was higher in schizophrenia patients than in healthy controls (0.43 ±â€¯0.30 vs. 0.22 ±â€¯0.24, p < 0.001). Logistic regressions showed an association between AGA IgG and GRINA IgG in schizophrenia (p = 0.016 for the estimated regression coefficient) but not for the controls (p = 0.471). Thus, we propose that mimicry through the presence of cross-reactivity between gliadin and GRINA might disrupt the functions of the glutamate system and relate to illness pathophysiology in those with schizophrenia and elevated AGA IgG.


Assuntos
Autoanticorpos/sangue , Permeabilidade da Membrana Celular/imunologia , Gliadina/imunologia , Mucosa Intestinal/imunologia , Receptores de N-Metil-D-Aspartato/sangue , Esquizofrenia/imunologia , Doença Celíaca/imunologia , Reações Cruzadas , Humanos , Imunoglobulina G/sangue , Mimetismo Molecular
13.
Nat Commun ; 9(1): 1962, 2018 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-29773800

RESUMO

Defensins are cationic antimicrobial peptides expressed throughout the plant and animal kingdoms as a first line of defense against pathogens. Membrane targeting and disruption is a crucial function of many defensins, however the precise mechanism remains unclear. Certain plant defensins form dimers that specifically bind the membrane phospholipids phosphatidic acid (PA) and phosphatidylinositol 4,5-bisphosphate, thereby triggering the assembly of defensin-lipid oligomers that permeabilize cell membranes. To understand this permeabilization mechanism, here we determine the crystal structure of the plant defensin NaD1 bound to PA. The structure reveals a 20-mer that adopts a concave sheet- or carpet-like topology where NaD1 dimers form one face and PA acyl chains form the other face of the sheet. Furthermore, we show that Arg39 is critical for PA binding, oligomerization and fungal cell killing. These findings identify a putative defensin-phospholipid membrane attack configuration that supports a longstanding proposed carpet mode of membrane disruption.


Assuntos
Membrana Celular/metabolismo , Defensinas/química , Ácidos Fosfatídicos/química , Proteínas de Plantas/química , Candida albicans/patogenicidade , Candida albicans/fisiologia , Permeabilidade da Membrana Celular/imunologia , Cristalografia por Raios X , Defensinas/fisiologia , Imunidade Inata/fisiologia , Testes de Sensibilidade Microbiana , Mutagênese , Ácidos Fosfatídicos/metabolismo , Doenças das Plantas/imunologia , Doenças das Plantas/microbiologia , Proteínas de Plantas/fisiologia , Ligação Proteica , Multimerização Proteica/fisiologia , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Nicotiana/microbiologia , Nicotiana/fisiologia
14.
Biochem Biophys Res Commun ; 495(1): 1432-1439, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29158087

RESUMO

Functional abnormal airway epithelial cells, along with activated inflammatory cells, resulting in chronic airway inflammation, are considered as the characteristic of asthma. Fatty Acid Binding Protein 4 (FABP4) takes part in glucose and lipid homeostasis, and also have an important role in allergic airway inflammation. However, whether FABP4 influence barrier function of airway epithelial cells is unknown. In vivo, a HDM-induced murine model of asthma was obtained to assessed airway inflammation and protein expression of E-cadherin and Forkhead Box M1 (FoxM1). In vitro, 16-HBE was cultured and was treated with hrFABP4, siFABP4, FABPF4 inhibitor BMS, or FoxM1 inhibitor RCM-1. IL-4, IL-5, and IL-13 level was determined by ELISA. Transepithelial electrical resistance (TER), paracellular permeability and E-cadherin-special immunofluorescence were measured to value airway epithelial barrier function. Intracellular ROS production was determined by DCF-DA fluorescence. FABP4 inhibitor BMS alleviate airway inflammation and destruction of E-cad in allergic mouse. Treatment with HDM or hrFABP4 aggravated inflammatory response, damaged airway epithelial barrier, which could be inhibited by siFABP4 and BMS. Treatment with HDM or hrFABP4 also enhanced levels of FoxM1, and Inhibited FoxM1 suppressed HDM- and hrFABP4-induced inflammation and airway epithelial barrier dysfunction. In addition, H2O2 promoted FoxM1 expression, HDM and hrFABP4 induced-FoxM1 could be inhibited by NAC, leading to decreased inflammation and improved airway epithelial barrier. Upregulated ROS induced by FABP4 was of significance in activating FoxM1 leading to airway inflammation and epithelial barrier dysfunction.


Assuntos
Células Epiteliais Alveolares/imunologia , Asma/imunologia , Permeabilidade da Membrana Celular/imunologia , Proteínas de Ligação a Ácido Graxo/imunologia , Proteína Forkhead Box M1/imunologia , Espécies Reativas de Oxigênio/imunologia , Mucosa Respiratória/imunologia , Animais , Asma/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Mucosa Respiratória/patologia
15.
Brain Behav Immun ; 65: 68-89, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28676349

RESUMO

When related to central nervous system (CNS) health and disease, brain mast cells (MCs) can be a source of either beneficial or deleterious signals acting on neural cells. We review the current state of knowledge about molecular interactions between MCs and glia in neurodegenerative diseases such as Multiple Sclerosis, Alzheimer's disease, Amyotrophic Lateral Sclerosis, Parkinson's disease, Epilepsy. We also discuss the influence on MC actions evoked by the host microbiota, which has a profound effect on the host immune system, inducing important consequences in neurodegenerative disorders. Gut dysbiosis, reduced intestinal motility and increased intestinal permeability, that allow bacterial products to circulate and pass through the blood-brain barrier, are associated with neurodegenerative disease. There are differences between the microbiota of neurologic patients and healthy controls. Distinguishing between cause and effect is a challenging task, and the molecular mechanisms whereby remote gut microbiota can alter the brain have not been fully elucidated. Nevertheless, modulation of the microbiota and MC activation have been shown to promote neuroprotection. We review this new information contributing to a greater understanding of MC-microbiota-neural cells interactions modulating the brain, behavior and neurodegenerative processes.


Assuntos
Mastócitos/imunologia , Mastócitos/fisiologia , Doenças Neurodegenerativas/imunologia , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/microbiologia , Barreira Hematoencefálica/fisiologia , Encéfalo/imunologia , Comunicação Celular , Permeabilidade da Membrana Celular/imunologia , Sistema Nervoso Central/imunologia , Microbioma Gastrointestinal/imunologia , Microbioma Gastrointestinal/fisiologia , Motilidade Gastrointestinal/imunologia , Humanos , Intestinos/microbiologia , Mastócitos/microbiologia , Microbiota , Doenças Neurodegenerativas/microbiologia , Neuroglia/imunologia
16.
Virology ; 496: 9-20, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27236741

RESUMO

We examined the effects of complement factors on primary-cultured neurons infected with prions. The amount of protease K (PK)-resistant abnormal form of prion protein (PrP(Sc)) reached a maximum level at 12 and 16 days post exposure (dpe) in 22L- and Chandler-infected neurons, respectively. In Chandler-infected neurons, the reaction of complement factors C1q, C3 and C9 significantly increased membrane permeability. This was followed by a decrease of PK-resistant PrP(Sc) at 16 and 20dpe. In contrast, in 22L-infected neurons, the effects of complement factors were observed at 12 and 16dpe, but not at 20dpe. Membrane permeability also increased in 22L-infected neurons by reaction of complement factor C3, but interestingly, the amount of PK-resistant PrP(Sc) initially decreased, and then increased. These results suggest that the reactivity of complement factors in prion-infected neurons depends on the amount of PrP(Sc) and the prion strain.


Assuntos
Permeabilidade da Membrana Celular , Proteínas do Sistema Complemento/imunologia , Neurônios/metabolismo , Proteínas PrPSc/metabolismo , Animais , Morte Celular , Permeabilidade da Membrana Celular/imunologia , Células Cultivadas , Córtex Cerebral/citologia , Córtex Cerebral/imunologia , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Camundongos , Príons/imunologia , Príons/metabolismo
17.
Respir Res ; 17: 2, 2016 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-26739349

RESUMO

BACKGROUND: COPD patients have a higher risk of pneumonia when treated with fluticasone propionate (FP) than with placebo, and a lower risk with budesonide (BUD). We hypothesized that BUD and FP differentially affect the mucosal barrier in response to viral infection and/or cigarette smoke. METHODS: We assessed protective effects of equivalent concentrations of BUD and FP on cytokine production and barrier function (electrical resistance) in human bronchial epithelial 16HBE cells and primary bronchial epithelial cells (PBECs) upon exposure to viral mimetic poly-(I:C) and/or cigarette smoke extract (CSE) or epidermal growth factor (EGF). RESULTS: BUD and FP were equally effective in suppressing poly-(I:C)- and/or CSE-induced IL-8 secretion in 16HBE and PBECs. Poly-(I:C) substantially decreased electrical resistance in 16HBE cells and both BUD and FP fully counteracted this effect. However, FP hardly affected 16HBE barrier dysfunction induced by CSE with/without poly-(I:C), whereas BUD (16 nM) provided full protection, an effect likely mediated by affecting EGFR-downstream target GSK-3ß. Similarly, BUD, but not FP, significantly improved CSE-induced barrier dysfunction in PBECs. Finally, BUD, but not FP, exerted a modest but significant protective effect against Streptococcus Pneumoniae-induced barrier dysfunction, and BUD, but not FP, prevented cellular adhesion and/or internalization of these bacteria induced by poly-(I:C) in 16HBE. CONCLUSIONS: Collectively, both BUD and FP efficiently control epithelial pro-inflammatory responses and barrier function upon mimicry of viral infection. Of potential clinical relevance, BUD more effectively counteracted CSE-induced barrier dysfunction, reinforcing the epithelial barrier and potentially limiting access of pathogens upon smoking in vivo.


Assuntos
Brônquios/imunologia , Budesonida/administração & dosagem , Células Epiteliais/imunologia , Células Epiteliais/virologia , Fluticasona/administração & dosagem , Poli C/imunologia , Brônquios/efeitos dos fármacos , Brônquios/virologia , Broncodilatadores/administração & dosagem , Linhagem Celular , Permeabilidade da Membrana Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/imunologia , Citocinas/imunologia , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Humanos , Rhinovirus/efeitos dos fármacos , Rhinovirus/fisiologia , Alcatrões
18.
Xenotransplantation ; 22(4): 295-301, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26031609

RESUMO

Endothelial cell activation and injury by the terminal pathway of complement is important in various pathobiological processes, including xenograft rejection. Protection against injury by human complement can be induced in porcine endothelial cells (ECs) with IL-4 and IL-13 through metabolic activation. However, despite this resistance, the complement-treated ECs were found to lose membrane permeability control assessed with the small molecule calcein. Therefore, to define the apparent discrepancy of permeability changes vis-à-vis the protection from killing, we now investigated whether IL-4 and IL-13 influence the release of the large cytoplasmic protein lactate dehydrogenase (LDH) in ECs incubated with complement or the pore-forming protein melittin. Primary cultures of ECs were pre-treated with IL-4 or IL-13 and then incubated with human serum as source of antibody and complement or melittin. Cell death was assessed using neutral red. Membrane permeability was quantitated measuring LDH release. We found that IL-4-/IL-13-induced protection of ECs from killing by complement or melittin despite loss of LDH in amounts similar to control ECs. However, the cytokine-treated ECs that were protected from killing rapidly regained effective control of membrane permeability. Moreover, the viability of the protected ECs was maintained for at least 2 days. We conclude that the protection induced by IL-4/IL-13 in ECs against lethal attack by complement or melittin is effective and durable despite severe initial impairment of membrane permeability. The metabolic changes responsible for protection allow the cells to repair the membrane injury caused by complement or melittin.


Assuntos
Proteínas do Sistema Complemento/toxicidade , Células Endoteliais/imunologia , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Interleucina-13/administração & dosagem , Interleucina-4/administração & dosagem , Meliteno/toxicidade , Animais , Permeabilidade da Membrana Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/imunologia , Citoplasma/metabolismo , Citotoxicidade Imunológica , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Humanos , L-Lactato Desidrogenase/metabolismo , Suínos , Transplante Heterólogo/efeitos adversos , Transplante Heterólogo/métodos
19.
Acta Biomater ; 19: 66-75, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25818949

RESUMO

Hyaluronic acid (HA) has received a lot of attention recently as a biomaterial with applications in wound healing, drug delivery, vascular repair and cell and/or gene delivery. Interstitial cystitis (IC) is characterised by an increase in the permeability of the bladder wall urothelium due to loss of the glycosaminoglycan (GAG) layer. The degradation of the urothelium leads to chronic pain and urinary dysfunction. The aetiology of the degradation of the GAG layer in this instance is currently unknown. At a clinical level, GAG replacement therapy using a HA solution is currently utilised as a treatment for IC. However, there is a significant lack of data on the mechanism of action of HA in IC. The current study investigates the mechanistic effect of clinically relevant HA treatment on an in vitro model of IC using urothelial cells, examining cytokine secretion, GAG secretion and trans-epithelial permeability. This study demonstrates that HA can significantly decrease induced cytokine secretion (4-5 fold increase), increase sulphated GAG production (2-fold increase) and without altering tight junction expression, decrease trans-epithelial permeability, suggesting that the HA pathway is a clinical target and potential treatment vector.


Assuntos
Cistite Intersticial/tratamento farmacológico , Cistite Intersticial/imunologia , Ácido Hialurônico/administração & dosagem , Interleucina-6/imunologia , Interleucina-8/imunologia , Urotélio/imunologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Permeabilidade da Membrana Celular/imunologia , Cistite Intersticial/patologia , Relação Dose-Resposta a Droga , Glicosaminoglicanos/imunologia , Humanos , Teste de Materiais , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/imunologia , Urotélio/efeitos dos fármacos , Urotélio/patologia
20.
JAMA Surg ; 149(5): 451-7, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24671426

RESUMO

IMPORTANCE: In conjunction with chemotherapy, immunotherapy with dendritic cells (DCs) may eliminate minimal disease burden by generating cytotoxic T lymphocytes. Enhanced cytosolic bioavailability of tumor-specific antigens improves access to human leukocyte antigen (HLA) class I molecules for more efficient cytotoxic T lymphocyte generation. Various cell-penetrating domains (CPDs) are known to ferry covalently linked heterologous antigens to the intracellular compartment by traversing the plasma membrane. OBJECTIVE: To determine whether generating melanoma antigen family A, 3 (MAGE-A3), a tumor-specific cancer-testis antigen, as a fusion protein with CPD will enhance the cytosolic bioavailability of MAGE-A3. DESIGN: MAGE-A3 was amplified by polymerase chain reaction using complementary DNA from renal tissue and cloned in frame with a CPD (YARKARRQARR) at the amino-terminal end and hexahistidine at the carboxy-terminal end to generate CPD-MAGE-A3 in a pQE-70 expression vector. Cultures were grown in Escherichia coli BL21 Star (DE3-pLysS) cells followed by nickel-nitrilotriacetic acid affinity purification of recombinant proteins. MAIN OUTCOMES AND MEASURES: Measurement of DC membrane penetration of CPD-MAGE-A3 vs MAGE-A3 and determination of the effect of CPD-MAGE-A3 pulsing on DC phenotypic expression of cell-surface antigens. RESULTS: Media composition and isopropyl-d-thiogalactosidase induction were optimized to achieve high levels of protein expression followed by purification. Western blot analysis with MAGE-A3 antibodies recognized both MAGE-A3 and CPD-MAGE-A3 proteins, while CPD antibodies recognized only CPD-MAGE-A3. Purified CPD-MAGE-A3 exhibited more efficient DC membrane penetration than did MAGE-A3 alone as confirmed by immunofluorescence analysis. High-level expression of several unique DC markers (CD80, CD83, CD86, and HLA-DR) by flow cytometry was consistent with a mature DC phenotype, indicating that pulsing with CPD-MAGE-A3 did not alter specific cell-surface antigens required for T-cell activation. CONCLUSIONS AND RELEVANCE: We have demonstrated for the first time, to our knowledge, that cloning and purification of MAGE-A3 with CPD enhances its cytosolic bioavailability in DCs without altering cell-surface antigens, potentially making it a more potent therapeutic cancer vaccine compared with existing MAGE-A3 protein and peptide vaccines.


Assuntos
Antígenos de Neoplasias/uso terapêutico , Vacinas Anticâncer/farmacocinética , Vacinas Anticâncer/uso terapêutico , Permeabilidade da Membrana Celular/efeitos dos fármacos , Citosol/efeitos dos fármacos , Citosol/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Proteínas de Neoplasias/farmacocinética , Proteínas de Neoplasias/uso terapêutico , Antígenos de Neoplasias/imunologia , Disponibilidade Biológica , Vacinas Anticâncer/imunologia , Permeabilidade da Membrana Celular/imunologia , Peptídeos Penetradores de Células , Clonagem Molecular , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Proteínas de Neoplasias/imunologia , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...