Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 306
Filtrar
1.
Inflammation ; 45(1): 116-128, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34494203

RESUMO

Angioedema is characterized by swelling of the skin or mucous membranes. Overproduction of the vasodilator bradykinin (BK) is an important contributor to the disease pathology, which causes rapid increase in vascular permeability. BK formation on endothelial cells results from high molecular weight kininogen (HK) interacting with gC1qR, the receptor for the globular heads of C1q, the first component of the classical pathway of complement. Endothelial cells are sensitive to blood-flow-induced shear stress and it has been shown that shear stress can modulate gC1qR expression. This study aimed to determine the following: (1) how BK or angioedema patients' (HAE) plasma affected endothelial cell permeability and gC1qR expression under shear stress, and (2) if monoclonal antibody (mAb) 74.5.2, which recognizes the HK binding site on gC1qR, had an inhibitory effect in HK binding to endothelial cells. Human dermal microvascular endothelial cells (HDMECs) grown on Transwell inserts were exposed to shear stress in the presence of HAE patients' plasma. Endothelial cell permeability was measured using FITC-conjugated bovine serum albumin. gC1qR expression and HK binding to endothelial cell surface was measured using solid-phase ELISA. Cell morphology was quantified using immunofluorescence microscopy. The results demonstrated that BK at 1 µg/mL, but not HAE patients' plasma and/or shear stress, caused significant increases in HDMEC permeability. The mAb 74.5.2 could effectively inhibit HK binding to recombinant gC1qR, and reduce HAE patients' plasma-induced HDMEC permeability change. These results suggested that monoclonal antibody to gC1qR, i.e., 74.5.2, could be potentially used as an effective therapeutic reagent to prevent angioedema.


Assuntos
Angioedema/tratamento farmacológico , Anticorpos Monoclonais/farmacologia , Bradicinina/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Fármacos Cardiovasculares/farmacologia , Proteínas de Transporte/imunologia , Células Endoteliais/efeitos dos fármacos , Proteínas Mitocondriais/imunologia , Angioedema/imunologia , Angioedema/metabolismo , Angioedema/fisiopatologia , Anticorpos Monoclonais/uso terapêutico , Biomarcadores/metabolismo , Permeabilidade Capilar/imunologia , Fármacos Cardiovasculares/uso terapêutico , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Endotélio Vascular/fisiopatologia , Humanos , Permeabilidade/efeitos dos fármacos , Resistência ao Cisalhamento/efeitos dos fármacos
2.
Front Immunol ; 12: 698249, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34456910

RESUMO

As the first inflammatory cell recruited to the site of spinal cord injury (SCI), neutrophils were reported to be detrimental to SCI. However, the precise mechanisms as to how neutrophils exacerbate SCI remain largely obscure. In the present study, we demonstrated that infiltrated neutrophils produce neutrophil extracellular traps (NETs), which subsequently promote neuroinflammation and blood-spinal cord barrier disruption to aggravate spinal cord edema and neuronal apoptosis following SCI in rats. Both inhibition of NETs formation by peptidylarginine deiminase 4 (PAD4) inhibitor and disruption of NETs by DNase 1 alleviate secondary damage, thus restraining scar formation and promoting functional recovery after SCI. Furthermore, we found that NETs exacerbate SCI partly via elevating transient receptor potential vanilloid type 4 (TRPV4) level in the injured spinal cord. Therefore, our results indicate that NETs might be a promising therapeutic target for SCI.


Assuntos
Armadilhas Extracelulares/imunologia , Doenças Neuroinflamatórias/etiologia , Doenças Neuroinflamatórias/patologia , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/patologia , Animais , Permeabilidade Capilar/imunologia , Feminino , Doenças Neuroinflamatórias/imunologia , Ratos , Ratos Sprague-Dawley , Traumatismos da Medula Espinal/imunologia
3.
Int Immunopharmacol ; 100: 108077, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34464887

RESUMO

BACKGROUND: Disruption of alveolar endothelial barrier caused by inflammation drives the progression of septic acute lung injury (ALI). Pravastatin, an inhibitor of HMG Co-A reductase, has potent anti-inflammatory effects. In the present study, we aim to explore the beneficial role of pravastatin in sepsis-induced ALI and its related mechanisms. METHODS: A septic ALI model was established by cecal ligation and puncture (CLP) in mice. The pulmonary microvascular endothelial cells (PMVECs) were challenged with lipopolysaccharide (LPS). The pathological changes in lung tissues were examined by HE staining. The pulmonary microvascular permeability was determined by lung wet-to-dry (W/D) weight ratio and Evans blue staining. The total protein concentration in bronchoalveolar lavage fluid (BALF) was detected by BCA assay. The levels of TNF-α, IL-1ß, and IL-6 were assessed by qRT-PCR and ELISA. Apoptosis was determined by flow cytometry and TUNEL. Western blotting was performed for detection of target protein levels. The expression of VE-Cadherin in lung tissues was evaluated by immunohistochemical staining. RESULTS: Pravastatin improved survival rate, attenuated lung pathological changes and reduced pulmonary microvascular permeability in septic mice. In addition, pravastatin restrained sepsis-induced inflammatory response and apoptosis in the lung tissues and PMVECs. Moreover, pravastatin up-regulated the levels of junction proteins ZO-1, JAM-C, and VE-Cadherin. Finally, pravastatin suppressed inflammation, apoptosis and enhanced the expression of junction proteins via regulating Cav-1/eNOS signaling pathway in LPS-exposed PMVECs. CONCLUSION: Pravastatin ameliorates sepsis-induced ALI through improving alveolar endothelial barrier disruption via modulating Cav-1/eNOS pathway, which may be an effective candidate for treating septic ALI.


Assuntos
Lesão Pulmonar Aguda/tratamento farmacológico , Células Endoteliais/efeitos dos fármacos , Pravastatina/farmacologia , Sepse/tratamento farmacológico , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/patologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/imunologia , Líquido da Lavagem Broncoalveolar/imunologia , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/imunologia , Caveolina 1/metabolismo , Modelos Animais de Doenças , Células Endoteliais/imunologia , Células Endoteliais/patologia , Humanos , Masculino , Camundongos , Óxido Nítrico Sintase Tipo III/metabolismo , Pravastatina/uso terapêutico , Alvéolos Pulmonares/irrigação sanguínea , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/imunologia , Alvéolos Pulmonares/patologia , Sepse/complicações , Sepse/imunologia , Sepse/patologia
4.
JCI Insight ; 6(14)2021 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-34128840

RESUMO

BACKGROUNDThe fungal cell wall constituent 1,3-ß-d-glucan (BDG) is a pathogen-associated molecular pattern that can stimulate innate immunity. We hypothesized that BDG from colonizing fungi in critically ill patients may translocate into the systemic circulation and be associated with host inflammation and outcomes.METHODSWe enrolled 453 mechanically ventilated patients with acute respiratory failure (ARF) without invasive fungal infection and measured BDG, innate immunity, and epithelial permeability biomarkers in serially collected plasma samples.RESULTSCompared with healthy controls, patients with ARF had significantly higher BDG levels (median [IQR], 26 pg/mL [15-49 pg/mL], P < 0.001), whereas patients with ARF with high BDG levels (≥40 pg/mL, 31%) had higher odds for assignment to the prognostically adverse hyperinflammatory subphenotype (OR [CI], 2.88 [1.83-4.54], P < 0.001). Baseline BDG levels were predictive of fewer ventilator-free days and worse 30-day survival (adjusted P < 0.05). Integrative analyses of fungal colonization and epithelial barrier disruption suggested that BDG may translocate from either the lung or gut compartment. We validated the associations between plasma BDG and host inflammatory responses in 97 hospitalized patients with COVID-19.CONCLUSIONBDG measurements offered prognostic information in critically ill patients without fungal infections. Further research in the mechanisms of translocation and innate immunity recognition and stimulation may offer new therapeutic opportunities in critical illness.FUNDINGUniversity of Pittsburgh Clinical and Translational Science Institute, COVID-19 Pilot Award and NIH grants (K23 HL139987, U01 HL098962, P01 HL114453, R01 HL097376, K24 HL123342, U01 HL137159, R01 LM012087, K08HK144820, F32 HL142172, K23 GM122069).


Assuntos
COVID-19 , Candida , Imunidade Inata/imunologia , Respiração Artificial , beta-Glucanas/sangue , Biomarcadores/sangue , COVID-19/imunologia , COVID-19/terapia , Candida/imunologia , Candida/isolamento & purificação , Permeabilidade Capilar/imunologia , Estado Terminal/terapia , Feminino , Microbioma Gastrointestinal/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Prognóstico , Respiração Artificial/efeitos adversos , Respiração Artificial/métodos , Insuficiência Respiratória/etiologia , Insuficiência Respiratória/terapia , Sistema Respiratório/imunologia , Sistema Respiratório/microbiologia , SARS-CoV-2 , Índice de Gravidade de Doença , Análise de Sobrevida
5.
Curr Opin Immunol ; 72: 94-106, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33932709

RESUMO

Dysregulated mast cell-mediated inflammation and/or activation have been linked to a number of human diseases, including asthma, anaphylaxis, chronic spontaneous urticaria, and mast cell activation syndromes. As a major mast cell granule protein, tryptase is a biomarker commonly used in clinical practice to diagnose mast cell-associated disorders and -mediated reactions, but its mechanistic roles in disease pathogenesis remains incompletely understood. Here, we summarize recent advances in the understanding of human tryptase genetics and the effects that different genetic composition may have on the quaternary structure of tetrameric mature tryptases. We also discuss how these differences may impact clinical phenotypes including allergic inflammation, immediate hypersensitivity, and others seen in patients with mast cell-associated disorders. With the increased application of next-generation sequencing, we foresee that human genetic approaches will be a major focus of understanding human tryptase functions in various human mast cell disorders and in new therapeutic development.


Assuntos
Suscetibilidade a Doenças/imunologia , Hipersensibilidade/etiologia , Hipersensibilidade/metabolismo , Inflamação/etiologia , Inflamação/metabolismo , Mastócitos/imunologia , Mastócitos/metabolismo , Triptases/metabolismo , Alelos , Animais , Permeabilidade Capilar/genética , Permeabilidade Capilar/imunologia , Ativação Enzimática , Regulação da Expressão Gênica , Predisposição Genética para Doença , Genótipo , Humanos , Hipersensibilidade/diagnóstico , Hipersensibilidade Imediata/diagnóstico , Hipersensibilidade Imediata/etiologia , Hipersensibilidade Imediata/metabolismo , Inflamação/diagnóstico , Isoenzimas , Mutação , Fenótipo , Relação Estrutura-Atividade , Especificidade por Substrato , Triptases/genética
6.
Scand J Immunol ; 93(6): e13024, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33523532

RESUMO

Early airway responses to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection are of interest since they could decide whether coronavirus disease-19 (COVID-19) will proceed to life-threatening pulmonary disease stages. Here I discuss endothelial-epithelial co-operative in vivo responses producing first-line, humoral innate defence opportunities in human airways. The pseudostratified epithelium of human nasal and tracheobronchial airways are prime sites of exposure and infection by SARS-CoV-2. Just beneath the epithelium runs a profuse systemic microcirculation. Its post-capillary venules respond conspicuously to mucosal challenges with autacoids, allergens and microbes, and to mere loss of epithelium. By active venular endothelial gap formation, followed by transient yielding of epithelial junctions, non-sieved plasma macromolecules move from the microcirculation to the mucosal surface. Hence, plasma-derived protein cascade systems and antimicrobial peptides would have opportunity to operate jointly on an unperturbed mucosal lining. Similarly, a plasma-derived, dynamic gel protects sites of epithelial sloughing-regeneration. Precision for this indiscriminate humoral molecular response lies in restricted location and well-regulated duration of plasma exudation. Importantly, the endothelial responsiveness of the airway microcirculation differs distinctly from the relatively non-responsive, low-pressure pulmonary microcirculation that non-specifically, almost irreversibly, leaks plasma in life-threatening COVID-19. Observations in humans of infections with rhinovirus, coronavirus 229E, and influenza A and B support a general but individually variable early occurrence of plasma exudation in human infected nasal and tracheobronchial airways. Investigations are warranted to elucidate roles of host- and drug-induced airway plasma exudation in restriction of viral infection and, specifically, whether it contributes to variable disease responses following exposure to SARS-CoV-2.


Assuntos
COVID-19/imunologia , COVID-19/virologia , Interações Hospedeiro-Patógeno/imunologia , Imunidade Humoral , Mucosa Respiratória/imunologia , Mucosa Respiratória/virologia , SARS-CoV-2/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Biomarcadores/sangue , Proteínas Sanguíneas , COVID-19/diagnóstico , COVID-19/metabolismo , Permeabilidade Capilar/imunologia , Ativação do Complemento/imunologia , Proteínas do Sistema Complemento/imunologia , Proteínas do Sistema Complemento/metabolismo , Exsudatos e Transudatos , Humanos , Imunidade Inata , Imunoglobulina M/sangue , Imunoglobulina M/imunologia , Microvasos/imunologia , Microvasos/metabolismo , Mucosa Respiratória/metabolismo
7.
Int J Mol Sci ; 22(3)2021 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-33573189

RESUMO

Hyperglycemia alters the function of cerebral endothelial cells from the blood-brain barrier, increasing the risk of cerebrovascular complications during diabetes. This study evaluated the protective effect of polyphenols on inflammatory and permeability markers on bEnd3 cerebral endothelial cells exposed to high glucose concentration. Results show that hyperglycemic condition increased nuclear factor kappa B (NFκB) activity, deregulated the expression of interleukin-1 beta (IL-1ß), interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-α), cyclooxygenase-2 (COX-2), inducible nitric oxide synthase (iNOS), interleukin-10 (IL-10) and endothelial-leukocyte adhesion molecule (E-selectin) genes, raised MCP-1 secretion and elevated monocyte adhesion and transendothelial migration. High glucose decreased occludin, claudin-5, zona occludens-1 (ZO-1) and zona occludens-2 (ZO-2) tight junctions production and altered the endothelial permeability. Characterized polyphenolic extracts from the French medicinal plants Antirhea borbonica, Ayapana triplinervis, Dodonaea viscosa and Terminalia bentzoe, and their major polyphenols quercetin, caffeic, chlorogenic and gallic acids limited the pro-inflammatory and permeability alterations caused by high glucose. Peroxisome proliferator-activated receptor gamma (PPARγ) agonist also attenuated these damages while PPARγ antagonist aggravated them, suggesting PPARγ protective action. Interestingly, polyphenols improved PPARγ gene expression lowered by high glucose. Moreover, polyphenols were detected at the intracellular level or membrane-bound to cells, with evidence for breast cancer resistance protein (BCRP) efflux transporter role. Altogether, these findings emphasize the ability of polyphenols to protect cerebral endothelial cells in hyperglycemic condition and their relevance for pharmacological strategies aiming to limit cerebrovascular disorders in diabetes.


Assuntos
Barreira Hematoencefálica/efeitos dos fármacos , Transtornos Cerebrovasculares/prevenção & controle , Hiperglicemia/imunologia , Extratos Vegetais/farmacologia , Polifenóis/farmacologia , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Glicemia/metabolismo , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/patologia , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/imunologia , Linhagem Celular , Transtornos Cerebrovasculares/etiologia , Transtornos Cerebrovasculares/imunologia , Transtornos Cerebrovasculares/patologia , Avaliação Pré-Clínica de Medicamentos , Células Endoteliais/imunologia , Células Endoteliais/patologia , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Humanos , Hiperglicemia/sangue , Hiperglicemia/complicações , Camundongos , Monócitos/efeitos dos fármacos , Monócitos/imunologia , NF-kappa B/metabolismo , PPAR gama/agonistas , PPAR gama/antagonistas & inibidores , PPAR gama/metabolismo , Extratos Vegetais/uso terapêutico , Polifenóis/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/imunologia , Junções Íntimas/patologia
8.
Cell Mol Gastroenterol Hepatol ; 11(3): 683-696, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33075564

RESUMO

BACKGROUND & AIMS: Increased vascular permeability (VP) has been indicated to play an important role in the pathogenesis of inflammatory bowel disease (IBD). However, the pathological causes of increased intestinal VP in IBD remain largely unknown. METHOD: Fibrinogen level was measured in dextran sulphate sodium (DSS)-induced colitis and patients with ulcerative colitis. Gly-Pro-Arg-Pro acetate (GPRP), an Fg inhibitor, was used to detect the effect of Fg inhibition on the pathogenesis of DSS-induced colitis, as indicated by tissue damage, cytokine release and inflammatory cell infiltration. Miles assay was used to detect vascular permeability. RESULTS: Through tandem mass tag-based quantitative proteomics, fibrinogen (Fg) was found to be upregulated in the colon of DSS-treated mice, which was consistent with increased Fg level in colon sample of patients with ulcerative colitis. Gly-Pro-Arg-Pro acetate (GPRP), an Fg inhibitor, significantly alleviated DSS-induced colitis as indicated by improvement of body weight loss and mortality. GPRP decreased colonic inflammation and VP in DSS-treated mice. In vivo, Fg enhanced VP as indicated by Miles assay, which was significantly inhibited by GRPR, AKT (serine/threonine kinase 1) inhibitors and low doses of Jasplakinolide which induced actin polymerization, while was dramatically enhanced by Cytochalasin D (an actin polymerization inhibitor). Moreover, activation of AKT was found in vessels of DSS-treated mice. In vitro, Fg induced activation of AKT and depolymerization of microfilament and promoted cell-to-cell disaggregation. Furthermore, inhibition of AKT decreased Fg-induced microfilament depolymerization. CONCLUSIONS: Our findings highlight the importance of Fg in regulating colitis by modulation of VP via activating AKT and subsequent depolymerization of microfilament and suggest Fg as an attractive target for anti-colitis treatment.


Assuntos
Permeabilidade Capilar/imunologia , Colite Ulcerativa/imunologia , Fibrinogênio/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Biópsia , Permeabilidade Capilar/efeitos dos fármacos , Estudos de Casos e Controles , Linhagem Celular , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/patologia , Colo/irrigação sanguínea , Colo/imunologia , Colo/patologia , Sulfato de Dextrana/administração & dosagem , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Fibrinogênio/antagonistas & inibidores , Voluntários Saudáveis , Humanos , Mucosa Intestinal/irrigação sanguínea , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Masculino , Camundongos , Oligopeptídeos/administração & dosagem , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia
9.
Curr Drug Discov Technol ; 18(4): 525-531, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32357814

RESUMO

INTRODUCTION: The present study deals with the effect of Nectaroscordum koelzi fruit extract on acute and chronic inflammation. METHODS: A total of 84 NMRI mice were used in this study. The effect of the extract on acute inflammation was analyzed by increasing vascular permeability via acetic acid and xylene induced ear edema among mice. The extract was evaluated in terms of effects on chronic inflammation by means of the cotton pellet test among mice. For the assessment of inflammation degree, the mice paw edema volume was measured by the plethysmometric test. RESULTS: The findings showed that the extract was effective on acute inflammation induced by acetic acid in mice. In the xylene ear edema, N. koelzi extract indicated a significant activity in mice. In the cotton pellet method, the methanol extract produced a significant reduction in comparison with the control and dexamethasone. Mice paw edema volume decreased with the extract. CONCLUSION: In general, the data from the experiments indicated that the methanol extract of N. koelzi has an anti-inflammatory effect on acute and chronic inflammation. However, the exact contributing mechanisms have not been investigated for the pharmacological effects.


Assuntos
Allium/química , Anti-Inflamatórios/farmacologia , Edema/tratamento farmacológico , Inflamação/tratamento farmacológico , Extratos Vegetais/farmacologia , Ácido Acético/administração & dosagem , Ácido Acético/toxicidade , Animais , Anti-Inflamatórios/isolamento & purificação , Anti-Inflamatórios/uso terapêutico , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/imunologia , Modelos Animais de Doenças , Orelha/irrigação sanguínea , Edema/induzido quimicamente , Edema/imunologia , Edema/patologia , Humanos , Inflamação/imunologia , Masculino , Metanol/química , Camundongos , Extratos Vegetais/isolamento & purificação , Extratos Vegetais/uso terapêutico , Xilenos/administração & dosagem , Xilenos/toxicidade
10.
J Invest Dermatol ; 141(4): 787-799, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32888954

RESUMO

Neutrophil infiltration and papillary vessel dilation are hallmarks of the initiation phase of psoriatic lesions. However, how neutrophils aggravate psoriasis development during transendothelial migration and the interaction between neutrophils and cutaneous vascular endothelial cells are less well-understood. In this study, we reported that neutrophils and cutaneous vascular endothelial cells activated each other when neutrophils migrated through the cutaneous endothelial barrier. In addition, neutrophil infiltration into skin lesions caused vascular remodeling including cutaneous vasodilation and enhanced vascular permeability in vivo and in vitro. Microarray gene profile data showed that matrix metallopeptidase (MMP)-9 was overexpressed in psoriatic neutrophils, and zymography assay further validated the bioactivity of MMP-9 secreted by psoriatic neutrophils. Moreover, MMP-9 activated vascular endothelial cells through the extracellular signal‒regulated kinase 1/2 and p38-MAPK signaling pathways, enhancing CD4+ T-cell transmigration in vitro. Correspondingly, an MMP-9 inhibitor significantly reduced cutaneous vasodilation, vascular permeability, and psoriatic symptoms in an imiquimod- or IL-23‒induced psoriasiform mouse model. Overall, our study demonstrates that neutrophil-derived MMP-9 induces cutaneous vasodilation and hyperpermeability by activating cutaneous vascular endothelial cells, thus facilitating psoriatic lesion development, which increases our knowledge on the role of neutrophils in the pathogenesis of psoriasis.


Assuntos
Metaloproteinase 9 da Matriz/metabolismo , Inibidores de Metaloproteinases de Matriz/farmacologia , Neutrófilos/imunologia , Psoríase/imunologia , Animais , Biópsia , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/imunologia , Linhagem Celular , Quimiotaxia/imunologia , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/imunologia , Células Endoteliais/patologia , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Feminino , Imiquimode/imunologia , Interleucina-23/imunologia , Sistema de Sinalização das MAP Quinases/imunologia , Inibidores de Metaloproteinases de Matriz/uso terapêutico , Camundongos , Infiltração de Neutrófilos , Neutrófilos/metabolismo , Cultura Primária de Células , Psoríase/tratamento farmacológico , Psoríase/patologia , Proteínas Recombinantes/metabolismo , Pele/irrigação sanguínea , Pele/imunologia , Migração Transendotelial e Transepitelial/imunologia , Vasodilatação/imunologia
12.
Front Immunol ; 11: 2091, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33072072

RESUMO

Macrophages play a central role in dictating the tissue response to infection and orchestrating subsequent repair of the damage. In this context, macrophages residing in the lungs continuously sense and discriminate among a wide range of insults to initiate the immune responses important to host-defense. Inflammatory tissue injury also leads to activation of proteases, and thereby the coagulation pathway, to optimize injury and repair post-infection. However, long-lasting inflammatory triggers from macrophages can impair the lung's ability to recover from severe injury, leading to increased lung vascular permeability and neutrophilic injury, hallmarks of Acute Lung Injury (ALI). In this review, we discuss the roles of toll-like receptor 4 (TLR4) and protease activating receptor 2 (PAR2) expressed on the macrophage cell-surface in regulating lung vascular inflammatory signaling.


Assuntos
Lesão Pulmonar Aguda/imunologia , Vasos Sanguíneos/imunologia , Pulmão/imunologia , Macrófagos/imunologia , Receptor PAR-2/imunologia , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/imunologia , Lesão Pulmonar Aguda/patologia , Animais , Vasos Sanguíneos/lesões , Vasos Sanguíneos/patologia , Permeabilidade Capilar/imunologia , Humanos , Pulmão/irrigação sanguínea , Macrófagos/patologia
13.
Theranostics ; 10(16): 7163-7177, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32641985

RESUMO

Rationale: Kupffer cells (KCs) play a crucial role in liver immune homeostasis through interacting with other immune cells and liver sinusoidal endothelial cells (LSECs). However, how KCs exactly interact with these cells for maintaining the homeostasis still require the further investigation. CXCL10 is a chemokine that has been implicated in chemoattraction of monocytes, T cells, NK cells, and dendritic cells, and promotion of T cell adhesion to endothelial cells. Although CXCL10 is also known to participate in the pathogenesis of hepatic inflammation, the degree to which it is functionally involved in the crosstalk between immune cells and regulation of immune response is still unclear. Methods: To dynamically investigate the function of KCs, we used our recently developed rapid cell ablation model, intermedilysin (ILY)/human CD59 (hCD59)-mediated cell ablation tool, to selectively ablate KC pool under normal condition or concanavalin A (Con A)- induced hepatitis. At certain time points after KCs ablation, we performed flow cytometry to monitor the amount of hepatic infiltrating immune cells. mRNA array was used to detect the change of hepatic cytokines and chemokines levels. Cytokines and chemokines in the serum were further measured by LEGENDplexTM mouse proinflammatory chemokine panel and inflammation panel. Evans blue staining and transmission electron microscopy were used to investigate the interaction between KCs and LSECs in steady condition. CXCL10 neutralizing antibody and CXCL10 deficient mouse were used to study the role of CXCL10 in immune cell migration and pathogenesis of Con A-induced hepatitis. Results: At steady state, elimination of KCs results in a reduction of hepatic infiltrating monocytes, T, B, and NK cells and a list of cytokines and chemokines at transcriptional level. In the meantime, the depletion of KCs resulted in increased sinusoidal vascular permeability. In the pathological condition, the KCs elimination rescues Con A-induced acute hepatitis through suppressing proinflammatory immune responses by down-regulation of hepatitis-associated cytokines/chemokines in serum such as CXCL10, and recruitment of infiltrating immune cells (monocytes, T, B, and NK cells). We further documented that deficiency or blockade of CXCL10 attenuated the development of Con A-induced hepatitis associated with reduction of the infiltrating monocytes, especially inflammatory Ly6Chi monocytes. Conclusions: This study supports the notion that KCs actively interact with immune cells and LSECs for maintaining immune response and liver homeostasis. Our data indicate that the interplay between KCs and infiltrated monocytes via CXCL10 contribute to Con A-induced hepatitis.


Assuntos
Quimiocina CXCL10/metabolismo , Hepatite C/imunologia , Hepatite/imunologia , Células de Kupffer/imunologia , Linfócitos T/imunologia , Animais , Permeabilidade Capilar/imunologia , Comunicação Celular/imunologia , Quimiocina CXCL10/análise , Quimiocina CXCL10/genética , Concanavalina A/administração & dosagem , Concanavalina A/imunologia , Modelos Animais de Doenças , Células Endoteliais/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/patologia , Hepatite/patologia , Hepatite C/patologia , Hepatite C/cirurgia , Hepatite C/virologia , Humanos , Células de Kupffer/metabolismo , Fígado/irrigação sanguínea , Fígado/imunologia , Fígado/patologia , Fígado/cirurgia , Cirrose Hepática/imunologia , Cirrose Hepática/patologia , Cirrose Hepática/cirurgia , Cirrose Hepática/virologia , Transplante de Fígado , Camundongos , Camundongos Knockout , Microvasos/citologia , Microvasos/patologia
14.
Front Immunol ; 11: 1028, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32536926

RESUMO

B cell adaptor molecule of 32 kDa (Bam32), known as dual adapter for phosphotyrosine and 3-phosphoinositides 1 (DAPP1), has been implicated in regulating lymphocyte proliferation and recruitment during inflammation. However, its role in neutrophils during inflammation remains unknown. Using intravital microscopy, we examined the role of Bam32 in formyl peptide receptor agonist WKYMVm-induced permeability changes in post-capillary venules and assessed simultaneously neutrophil adhesion and emigration in cremaster muscles of Bam32-deficient (Bam32-/-) and wild-type (WT) control mice. We observed significantly reduced WKYMVm-induced microvascular hyperpermeability accompanied by markedly decreased neutrophil emigration in Bam32-/- mice. The Bam32-specific decrease in WKYMVm-induced hyperpermeability was neutrophil-dependent as this was verified in bone marrow transplanted chimeric mice. We discovered that Bam32 was critically required for WKYMVm-induced intracellular and extracellular production of reactive oxygen species (ROS) in neutrophils. Pharmacological scavenging of ROS eliminated the differences in WKYMVm-induced hyperpermeability between Bam32-/- and WT mice. Deficiency of Bam32 decreased WKYMVm-induced ERK1/2 but not p38 or JNK phosphorylation in neutrophils. Inhibition of ERK1/2 signaling cascade suppressed WKYMVm-induced ROS generation in WT neutrophils and microvascular hyperpermeability in WT mice. In conclusion, our study reveals that Bam32-dependent, ERK1/2-involving ROS generation in neutrophils is critical in WKYMVm-induced microvascular hyperpermeability during neutrophil recruitment.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Permeabilidade Capilar/fisiologia , Lipoproteínas/metabolismo , Neutrófilos/metabolismo , Oligopeptídeos/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Transplante de Medula Óssea , Permeabilidade Capilar/imunologia , Adesão Celular/efeitos dos fármacos , Adesão Celular/imunologia , Adesão Celular/fisiologia , Lipoproteínas/deficiência , Lipoproteínas/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos/efeitos dos fármacos , Infiltração de Neutrófilos/imunologia , Infiltração de Neutrófilos/fisiologia , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Espécies Reativas de Oxigênio/metabolismo , Receptores de Formil Peptídeo/agonistas , Quimeras de Transplante/imunologia , Quimeras de Transplante/fisiologia , Vênulas/efeitos dos fármacos , Vênulas/imunologia , Vênulas/fisiologia
15.
Aging (Albany NY) ; 12(12): 11781-11793, 2020 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-32568100

RESUMO

Excess salt (NaCl) intake is closely related to a variety of central nervous system (CNS) diseases characterized by increased cerebral microvascular permeability. However, the link between a high salt diet (HSD) and the breakdown of tight junctions (TJs) remains unclear. In the present study, we found that high salt does not directly influence the barrier between endothelial cells, but it suppresses expression of TJ proteins when endothelial cells are co-cultured with astrocytes. This effect is independent of blood pressure, but depends on the astrocyte activation via the NFκB/MMP-9 signaling pathway, resulting in a marked increase in VEGF expression. VEGF, in turn, induces disruption of TJs by inducing phosphorylation and activation of ERK and eNOS. Correspondingly, the HSD-induced disruption of TJ proteins is attenuated by blocking VEGF using the specific monoclonal antibody Bevacizumab. These results reveal a new axis linking a HSD to increased cerebral microvascular permeability through a VEGF-initiated inflammatory response, which may be a potential target for preventing the deleterious effects of HSD on the CNS.


Assuntos
Astrócitos/metabolismo , Barreira Hematoencefálica/patologia , Permeabilidade Capilar/efeitos dos fármacos , Cloreto de Sódio na Dieta/efeitos adversos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Bevacizumab , Barreira Hematoencefálica/citologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/imunologia , Permeabilidade Capilar/imunologia , Células Cultivadas , Técnicas de Cocultura , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Células Endoteliais , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/imunologia , Endotélio Vascular/patologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/imunologia , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Transgênicos , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo III/genética , Óxido Nítrico Sintase Tipo III/metabolismo , Fosforilação , Cultura Primária de Células , Ratos , Cloreto de Sódio na Dieta/administração & dosagem , Organismos Livres de Patógenos Específicos , Proteínas de Junções Íntimas/metabolismo , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/patologia , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
16.
J Neuroinflammation ; 17(1): 153, 2020 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-32386505

RESUMO

BACKGROUND: Multiple sclerosis (MS) is an autoimmune disease which results from the invasion of the brain by activated immune cells across the endothelial cells (ECs) of the blood-brain barrier (BBB), due to loss of immune self-tolerance. Many reports define the metabolic profile of immune cells in MS, however little is known about the metabolism of the BBB ECs during the disease. We aim to determine whether circulating factors in MS induce metabolic alterations of the BBB ECs compared to a healthy state, which can be linked with disruption of BBB integrity and subsequent immune cell extravasation. METHODS AND RESULTS: In this report, we used an in vitro model to study the effect of sera from naïve-to-treatment, relapsing-remitting MS (RRMS) patients on the human brain microvascular endothelium, comparing effects to age/sex-matched healthy donor (HD) sera. Our data show that RRMS serum components affect brain endothelial cells by impairing intercellular tightness through the down-modulation of occludin and VE-cadherin, and facilitating immune cell extravasation through upregulation of intercellular adhesion molecules (ICAM-1) and P-glycoprotein (P-gp). At a metabolic level, the treatment of the endothelial cells with RRMS sera reduced their glycolytic activity (measured through the extracellular acidification rate-ECAR) and oxygen consumption rate (oxidative phosphorylation rate-OCR). Such changes were associated with the down-modulation of endothelial glucose transporter 1 (GLUT-1) expression and by altered mitochondrial membrane potential. Higher level of reactive oxygen species released from the endothelial cells treated with RRMS sera indicate a pro-inflammatory status of the cells together with the higher expression of ICAM-1, endothelial cell cytoskeleton perturbation (stress fibres) as well as disruption of the cytoskeleton signal transduction MSK1/2 and ß-catenin phosphorylation. CONCLUSIONS: Our data suggest that circulating factors present in RRMS patient serum induce physiological and biochemical alterations to the BBB, namely reducing expression of essential tightness regulators, as well as reduced engagement of glycolysis and alteration of mitochondrial potential. As these last changes have been linked with alterations in nutrient usage and metabolic function in immune cells; we propose that the BBB endothelium of MS patients may similarly undergo metabolic dysregulation, leading to enhanced permeability and increased disease susceptibility.


Assuntos
Barreira Hematoencefálica/metabolismo , Endotélio Vascular/metabolismo , Esclerose Múltipla Recidivante-Remitente/sangue , Esclerose Múltipla Recidivante-Remitente/imunologia , Adulto , Permeabilidade Capilar/imunologia , Células Cultivadas , Feminino , Humanos , Masculino , Migração Transendotelial e Transepitelial/imunologia
17.
Basic Clin Pharmacol Toxicol ; 127(2): 102-110, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32162446

RESUMO

Cerebrovascular endothelial cells (CECs) are integral components of both the blood-brain barrier (BBB) and the neurovascular unit (NVU). As the primary cell type of the BBB, CECs are responsible for the tight regulation of molecular transport between the brain parenchyma and the periphery. Additionally, CECs are essential in neurovascular coupling where they help regulate cerebral blood flow in response to regional increases in cellular demand in the NVU. CEC dysfunction occurs during both normative ageing and in cerebrovascular disease, which leads to increased BBB permeability and neurovascular uncoupling. This MiniReview compiles what is known about the molecular changes underlying CEC dysfunction, many of which are reminiscent of cells that have become senescent. In general, cellular senescence is defined as an irreversible growth arrest characterized by the acquisition of a pro-inflammatory secretory phenotype in response to DNA damage or other cellular stresses. We discuss evidence for endothelial cell senescence in ageing and cardiovascular disease, and how CEC senescence may contribute to age-related cerebrovascular dysfunction.


Assuntos
Envelhecimento/patologia , Encéfalo/irrigação sanguínea , Senescência Celular/fisiologia , Transtornos Cerebrovasculares/patologia , Células Endoteliais/patologia , Acoplamento Neurovascular/fisiologia , Envelhecimento/imunologia , Envelhecimento/metabolismo , Barreira Hematoencefálica/patologia , Barreira Hematoencefálica/fisiopatologia , Permeabilidade Capilar/imunologia , Permeabilidade Capilar/fisiologia , Senescência Celular/imunologia , Transtornos Cerebrovasculares/imunologia , Transtornos Cerebrovasculares/metabolismo , Transtornos Cerebrovasculares/fisiopatologia , Citocinas/metabolismo , Células Endoteliais/imunologia , Células Endoteliais/metabolismo , Humanos , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo III/metabolismo
18.
Am J Respir Crit Care Med ; 202(3): 361-370, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32101446

RESUMO

Endothelial cells (ECs) are vascular, nonconventional immune cells that play a major role in the systemic response after bacterial infection to limit its dissemination. Triggered by exposure to pathogens, microbial toxins, or endogenous danger signals, EC responses are polymorphous, heterogeneous, and multifaceted. During sepsis, ECs shift toward a proapoptotic, proinflammatory, proadhesive, and procoagulant phenotype. In addition, glycocalyx damage and vascular tone dysfunction impair microcirculatory blood flow, leading to organ injury and, potentially, life-threatening organ failure. This review aims to cover the current understanding of the EC adaptive or maladaptive response to acute inflammation or bacterial infection based on compelling recent basic research and therapeutic clinical trials targeting microvascular and endothelial alterations during septic shock.


Assuntos
Apoptose/imunologia , Permeabilidade Capilar/imunologia , Citocinas/imunologia , Células Endoteliais/imunologia , Endotélio Vascular/fisiopatologia , Inflamação/imunologia , Sepse/imunologia , Alarminas/metabolismo , Animais , Apoptose/fisiologia , Coagulação Sanguínea/fisiologia , Moléculas de Adesão Celular/metabolismo , Citocinas/metabolismo , Células Endoteliais/metabolismo , Endotelinas/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/imunologia , Endotélio Vascular/metabolismo , Epoprostenol/metabolismo , Fibrinólise/fisiologia , Glicocálix/metabolismo , Humanos , Inflamação/metabolismo , Microcirculação , Óxido Nítrico/metabolismo , Moléculas com Motivos Associados a Patógenos/metabolismo , Fenótipo , Adesividade Plaquetária , Sepse/metabolismo , Tromboplastina/metabolismo , Vasoconstrição , Vasodilatação
19.
J Clin Invest ; 130(5): 2301-2318, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-31971917

RESUMO

Increased microvascular permeability to plasma proteins and neutrophil emigration are hallmarks of innate immunity and key features of numerous inflammatory disorders. Although neutrophils can promote microvascular leakage, the impact of vascular permeability on neutrophil trafficking is unknown. Here, through the application of confocal intravital microscopy, we report that vascular permeability-enhancing stimuli caused a significant frequency of neutrophil reverse transendothelial cell migration (rTEM). Furthermore, mice with a selective defect in microvascular permeability enhancement (VEC-Y685F-ki) showed reduced incidence of neutrophil rTEM. Mechanistically, elevated vascular leakage promoted movement of interstitial chemokines into the bloodstream, a response that supported abluminal-to-luminal neutrophil TEM. Through development of an in vivo cell labeling method we provide direct evidence for the systemic dissemination of rTEM neutrophils, and showed them to exhibit an activated phenotype and be capable of trafficking to the lungs where their presence was aligned with regions of vascular injury. Collectively, we demonstrate that increased microvascular leakage reverses the localization of directional cues across venular walls, thus causing neutrophils engaged in diapedesis to reenter the systemic circulation. This cascade of events offers a mechanism to explain how local tissue inflammation and vascular permeability can induce downstream pathological effects in remote organs, most notably in the lungs.


Assuntos
Permeabilidade Capilar/imunologia , Microvasos/imunologia , Ativação de Neutrófilo , Neutrófilos/imunologia , Migração Transendotelial e Transepitelial/imunologia , Animais , Permeabilidade Capilar/genética , Masculino , Camundongos , Camundongos Transgênicos , Microvasos/patologia , Neutrófilos/patologia , Migração Transendotelial e Transepitelial/genética
20.
Int Immunopharmacol ; 78: 106079, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31841756

RESUMO

Hereditary angioedema is a disabling, life-threatening condition caused by deficiency (type I) or dysfunction (type II) of the C1 inhibitor protein (C1-INH-HAE) leading to bradykinin accumulation and recurrent episodes of edema attack. Vascular leakage is a complex process sustained by the coordinated production of several permeabilizing factors including vascular endothelial growth factors (VEGFs), angiopoietins (ANGPTs) and phospholipase A2 enzymes (PLA2). We previously reported that patients with C1-INH-HAE in remission have increased plasma levels of VEGFs, ANGPTs and secreted PLA2. In this study, we sought to analyze plasma levels of these mediators in 15 patients with C1-INH-HAE during the acute attack compared to remission. Plasma concentrations of VEGF-A, VEGF-C and VEGF-D were not altered during attack compared to remission. Moreover, VEGF-D concentrations were not altered also in remission phase compared to controls. Concentrations of ANGPT1, a vascular stabilizer, were increased during attacks compared to symptoms-free periods, whereas ANGPT2 levels were not altered. The ANGPT2/ANGPT1 ratio was decreased during angioedema attacks. Platelet activating factor acetylhydrolase activity was increased in patients with C1-INH-HAE in remission compared to controls and was decreased during angioedema attacks. Our results emphasize the complexity by which several vasoactive mediators are involved not only in the pathophysiology of C1-INH-HAE, but also during angioedema attacks and its resolution.


Assuntos
1-Alquil-2-acetilglicerofosfocolina Esterase/sangue , Angiopoietina-1/sangue , Proteína Inibidora do Complemento C1/metabolismo , Angioedema Hereditário Tipos I e II/imunologia , 1-Alquil-2-acetilglicerofosfocolina Esterase/imunologia , 1-Alquil-2-acetilglicerofosfocolina Esterase/metabolismo , Adolescente , Adulto , Angiopoietina-1/imunologia , Angiopoietina-1/metabolismo , Angiopoietina-2/sangue , Angiopoietina-2/imunologia , Angiopoietina-2/metabolismo , Bradicinina/imunologia , Bradicinina/metabolismo , Permeabilidade Capilar/imunologia , Estudos de Casos e Controles , Proteína Inibidora do Complemento C1/genética , Feminino , Voluntários Saudáveis , Angioedema Hereditário Tipos I e II/sangue , Angioedema Hereditário Tipos I e II/genética , Humanos , Masculino , Pessoa de Meia-Idade , Exacerbação dos Sintomas , Fator A de Crescimento do Endotélio Vascular/sangue , Fator A de Crescimento do Endotélio Vascular/imunologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator C de Crescimento do Endotélio Vascular/sangue , Fator C de Crescimento do Endotélio Vascular/imunologia , Fator C de Crescimento do Endotélio Vascular/metabolismo , Fator D de Crescimento do Endotélio Vascular/sangue , Fator D de Crescimento do Endotélio Vascular/imunologia , Fator D de Crescimento do Endotélio Vascular/metabolismo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...