Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Cells ; 13(5)2024 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-38474394

RESUMO

Giant cell arteritis (GCA) is an autoimmune disease affecting large vessels in patients over 50 years old. It is an exemplary model of a classic inflammatory disorder with IL-6 playing the leading role. The main comorbidities that may appear acutely or chronically are vascular occlusion leading to blindness and thoracic aorta aneurysm formation, respectively. The tissue inflammatory bulk is expressed as acute or chronic delayed-type hypersensitivity reactions, the latter being apparent by giant cell formation. The activated monocytes/macrophages are associated with pronounced Th1 and Th17 responses. B-cells and neutrophils also participate in the inflammatory lesion. However, the exact order of appearance and mechanistic interactions between cells are hindered by the lack of cellular and molecular information from early disease stages and accurate experimental models. Recently, senescent cells and neutrophil extracellular traps have been described in tissue lesions. These structures can remain in tissues for a prolonged period, potentially favoring inflammatory responses and tissue remodeling. In this review, current advances in GCA pathogenesis are discussed in different inflammatory phases. Through the description of these-often overlapping-phases, cells, molecules, and small lipid mediators with pathogenetic potential are described.


Assuntos
Arterite de Células Gigantes , Humanos , Pessoa de Meia-Idade , Arterite de Células Gigantes/etiologia , Arterite de Células Gigantes/patologia , Inflamação/complicações , Macrófagos/patologia , Neutrófilos/patologia , Linfócitos B/patologia
2.
Eur J Immunol ; 54(5): e2350669, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38339772

RESUMO

The importance of macrophages in adipose tissue (AT) homeostasis and inflammation is well established. However, the potential cues that regulate their function remain incompletely understood. To bridge this important gap, we sought to characterize novel pathways involved using a mouse model of diet-induced obesity. By performing transcriptomics analysis of AT macrophages (ATMs), we found that late-stage ATMs from high-fat diet mice presented with perturbed Notch signaling accompanied by robust proinflammatory and metabolic changes. To explore the hypothesis that the deregulated Notch pathway contributes to the development of AT inflammation and diet-induced obesity, we employed a genetic approach to abrogate myeloid Notch1 and Notch2 receptors. Our results revealed that the combined loss of Notch1 and Notch2 worsened obesity-related metabolic dysregulation. Body and AT weight gain was higher, blood glucose levels increased and metabolic parameters were substantially worsened in deficient mice fed high-fat diet. Moreover, serum insulin and leptin were elevated as were triglycerides. Molecular analysis of ATMs showed that deletion of Notch receptors escalated inflammation through the induction of an M1-like pro-inflammatory phenotype. Our findings thus support a protective role of myeloid Notch signaling in adipose tissue inflammation and metabolic dysregulation.


Assuntos
Tecido Adiposo , Dieta Hiperlipídica , Inflamação , Macrófagos , Obesidade , Receptor Notch1 , Receptor Notch2 , Transdução de Sinais , Animais , Macrófagos/imunologia , Macrófagos/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo/imunologia , Camundongos , Dieta Hiperlipídica/efeitos adversos , Inflamação/imunologia , Inflamação/metabolismo , Transdução de Sinais/imunologia , Obesidade/metabolismo , Obesidade/imunologia , Receptor Notch1/metabolismo , Receptor Notch1/genética , Receptor Notch2/metabolismo , Receptor Notch2/genética , Camundongos Knockout , Camundongos Endogâmicos C57BL , Masculino
3.
Semin Immunol ; 72: 101863, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38271892

RESUMO

Type I and type III interferons (IFNs) constitute a key antiviral defense systems of the body, inducing viral resistance to cells and mediating diverse innate and adaptive immune functions. Defective type I and type III IFN responses have recently emerged as the 'Achilles heel' in COVID-19, with such patients developing severe disease and exhibiting a high risk for critical pneumonia and death. Here, we review the biology of type I and type III IFNs, their similarities and important functional differences, and their roles in SARS-CoV-2 infection. We also appraise the various mechanisms proposed to drive defective IFN responses in COVID-19 with particular emphasis to the ability of SARS-CoV-2 to suppress IFN production and activities, the genetic factors involved and the presence of autoantibodies neutralizing IFNs and accounting for a large proportion of individuals with severe COVID-19. Finally, we discuss the long history of the type I IFN therapeutics for the treatment of viral diseases, cancer and multiple sclerosis, the various efforts to use them in respiratory infections, and the newly emerging type III IFN therapeutics, with emphasis to the more recent studies on COVID-19 and their potential use as broad spectrum antivirals for future epidemics or pandemics.


Assuntos
COVID-19 , Interferon Tipo I , Humanos , Interferon lambda , SARS-CoV-2 , Interferon Tipo I/uso terapêutico , Biologia
4.
Pediatr Hematol Oncol ; 39(6): 540-548, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35139731

RESUMO

Langerhans cell histiocytosis (LCH) is a rare inflammatory myeloid neoplasia with a broad spectrum of clinical manifestations. The activation of the MAP kinase pathway plays an integral role in its pathogenesis with genetic alterations found in the majority of cases that most frequently involve a somatic mutation of the oncogenic BRAFV600E variant. In this study we investigated the prevalence of the BRAFV600E mutation and its clinical relevance in adult Greek patients with LCH. Among 37 patients studied, the BRAFV600E mutation was identified in 12 out of 31 (38.7%), whereas in six patients (19.3%) the results were in conclusive. The presence of the mutation did not correlate with age at diagnosis, organ involvement, disease extent, response to initial treatment, development of diabetes insipidus and relapse risk. In our series the prevalence of the BRAFV600E mutation is at the lower range of the relative percentage found in children, but in line to that obtained in previous studies of adult patients with LCH that have found an up to 50% prevalence of the BRAFV600E mutation in these patients. Further studies with a larger number of adults are needed to identify the exact prevalence of mutations in the RAS-RAF-MEK-ERK pathway and their role on clinical parameters and disease outcomes.Supplemental data for this article is available online at https://doi.org/10.1080/08880018.2022.2029988 .


Assuntos
Histiocitose de Células de Langerhans , Proteínas Proto-Oncogênicas B-raf , Adulto , Criança , Grécia/epidemiologia , Histiocitose de Células de Langerhans/epidemiologia , Histiocitose de Células de Langerhans/genética , Humanos , Mutação , Prevalência , Proteínas Proto-Oncogênicas B-raf/genética
5.
Nat Commun ; 13(1): 215, 2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-35017526

RESUMO

Macrophages are integral to the pathogenesis of atherosclerosis, but the contribution of distinct macrophage subsets to disease remains poorly defined. Using single cell technologies and conditional ablation via a LysMCre+ Clec4a2flox/DTR mouse strain, we demonstrate that the expression of the C-type lectin receptor CLEC4A2 is a distinguishing feature of vascular resident macrophages endowed with athero-protective properties. Through genetic deletion and competitive bone marrow chimera experiments, we identify CLEC4A2 as an intrinsic regulator of macrophage tissue adaptation by promoting a bias in monocyte-to-macrophage in situ differentiation towards colony stimulating factor 1 (CSF1) in vascular health and disease. During atherogenesis, CLEC4A2 deficiency results in loss of resident vascular macrophages and their homeostatic properties causing dysfunctional cholesterol metabolism and enhanced toll-like receptor triggering, exacerbating disease. Our study demonstrates that CLEC4A2 licenses monocytes to join the vascular resident macrophage pool, and that CLEC4A2-mediated macrophage homeostasis is critical to combat cardiovascular disease.


Assuntos
Apolipoproteínas E/genética , Aterosclerose/genética , Vasos Sanguíneos/metabolismo , Lectinas Tipo C/genética , Macrófagos/metabolismo , Animais , Apolipoproteínas E/deficiência , Aterosclerose/metabolismo , Aterosclerose/patologia , Vasos Sanguíneos/patologia , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Morte Celular/genética , Diferenciação Celular , Linhagem da Célula/genética , Colesterol/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Homeostase/genética , Humanos , Lectinas Tipo C/deficiência , Fator Estimulador de Colônias de Macrófagos/genética , Fator Estimulador de Colônias de Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/metabolismo , Monócitos/patologia , Transdução de Sinais , Análise de Célula Única
6.
Allergy ; 77(4): 1150-1164, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34658046

RESUMO

BACKGROUND: Although FoxP3+ regulatory T (Treg) cells constitute a highly heterogeneous population, with different regulatory potential depending on the disease context, distinct subsets or phenotypes remain poorly defined. This hampers the development of immunotherapy for allergic and autoimmune disorders. The present study aimed at characterizing distinct FoxP3+ Treg subpopulations involved in the suppression of Th2-mediated allergic inflammation in the lung. METHODS: We used an established mouse model of allergic airway disease based on ovalbumin sensitization and challenge to analyze FoxP3+ Tregs during the induction and resolution of inflammation, and identify markers that distinguish their most suppressive phenotypes. We also developed a new knock-in mouse model (Foxp3cre Cd103dtr ) enabling the specific ablation of CD103+ FoxP3+ Tregs for functional studies. RESULTS: We found that during resolution of allergic airway inflammation in mice >50% of FoxP3+ Treg cells expressed the integrin CD103 which marks FoxP3+ Treg cells of high IL-10 production, increased expression of immunoregulatory molecules such as KLRG1, ICOS and CD127, and enhanced suppressive capacity for Th2-mediated inflammatory responses. CD103+ FoxP3+ Tregs were essential for keeping allergic inflammation under control as their specific depletion in Foxp3cre Cd103dtr mice lead to severe alveocapillary damage, eosinophilic pneumonia, and markedly reduced lifespan of the animals. Conversely, adoptive transfer of CD103+ FoxP3+ Tregs effectively treated disease, attenuating Th2 responses and allergic inflammation in an IL-10-dependent manner. CONCLUSIONS: Our study identifies a novel regulatory T-cell population, defined by CD103 expression, programmed to prevent exuberant type 2 inflammation and keep homeostasis in the respiratory tract under control. This has important therapeutic implications.


Assuntos
Antígenos CD/imunologia , Hipersensibilidade , Cadeias alfa de Integrinas/imunologia , Linfócitos T Reguladores , Animais , Citocinas/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Humanos , Inflamação/metabolismo , Integrinas/metabolismo , Interleucina-10/metabolismo , Pulmão , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
7.
J Gen Virol ; 103(12)2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36748697

RESUMO

Bacteriophages represent the most extensive group of viruses within the human virome and have a significant impact on general health and well-being by regulating bacterial population dynamics. Staphylococcus aureus, found in the anterior nostrils, throat and skin, is an opportunistic pathobiont that can cause a wide range of diseases, from chronic inflammation to severe and acute infections. In this study, we developed a human cell-based homeostasis model between a clinically isolated strain of S. aureus 141 and active phages for this strain (PYOSa141) isolated from the commercial Pyophage cocktail (PYO). The cocktail is produced by Eliava BioPreparations Ltd. (Tbilisi, Georgia) and is used as an add-on therapy for bacterial infections, mainly in Georgia. The triptych interaction model was evaluated by time-dependent analysis of cell death and inflammatory response of the nasal and bronchial epithelial cells. Inflammatory mediators (IL-8, CCL5/RANTES, IL-6 and IL-1ß) in the culture supernatants were measured by enzyme-linked immunosorbent assay and cell viability was determined by crystal violet staining. By measuring trans-epithelial electrical resistance, we assessed the epithelial integrity of nasal cells that had differentiated under air-liquid interface conditions. PYOSa141 was found to have a prophylactic effect on airway epithelial cells exposed to S. aureus 141 by effectively down-regulating bacterial-induced inflammation, cell death and epithelial barrier disruption in a time-dependent manner. Overall, the proposed model represents an advance in the way multi-component biological systems can be simulated in vitro.


Assuntos
Bacteriófagos , Humanos , Sobrevivência Celular , Staphylococcus aureus/fisiologia , Imagem com Lapso de Tempo , Inflamação , Células Epiteliais/metabolismo , Células Cultivadas
8.
Nat Immunol ; 22(1): 32-40, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33277638

RESUMO

A central paradigm of immunity is that interferon (IFN)-mediated antiviral responses precede pro-inflammatory ones, optimizing host protection and minimizing collateral damage1,2. Here, we report that for coronavirus disease 2019 (COVID-19) this paradigm does not apply. By investigating temporal IFN and inflammatory cytokine patterns in 32 moderate-to-severe patients with COVID-19 hospitalized for pneumonia and longitudinally followed for the development of respiratory failure and death, we reveal that IFN-λ and type I IFN production were both diminished and delayed, induced only in a fraction of patients as they became critically ill. On the contrary, pro-inflammatory cytokines such as tumor necrosis factor (TNF), interleukin (IL)-6 and IL-8 were produced before IFNs in all patients and persisted for a prolonged time. This condition was reflected in blood transcriptomes wherein prominent IFN signatures were only seen in critically ill patients who also exhibited augmented inflammation. By comparison, in 16 patients with influenza (flu) hospitalized for pneumonia with similar clinicopathological characteristics to those of COVID-19 and 24 nonhospitalized patients with flu with milder symptoms, IFN-λ and type I IFN were robustly induced earlier, at higher levels and independently of disease severity, whereas pro-inflammatory cytokines were only acutely produced. Notably, higher IFN-λ concentrations in patients with COVID-19 correlated with lower viral load in bronchial aspirates and faster viral clearance and a higher IFN-λ to type I IFN ratio correlated with improved outcome for critically ill patients. Moreover, altered cytokine patterns in patients with COVID-19 correlated with longer hospitalization and higher incidence of critical disease and mortality compared to flu. These data point to an untuned antiviral response in COVID-19, contributing to persistent viral presence, hyperinflammation and respiratory failure.


Assuntos
COVID-19/imunologia , Imunidade/imunologia , Influenza Humana/imunologia , Interferon Tipo I/imunologia , Interferons/imunologia , SARS-CoV-2/imunologia , Antivirais/imunologia , Antivirais/metabolismo , COVID-19/genética , COVID-19/virologia , Citocinas/genética , Citocinas/imunologia , Progressão da Doença , Expressão Gênica/genética , Expressão Gênica/imunologia , Perfilação da Expressão Gênica/métodos , Humanos , Imunidade/genética , Inflamação/genética , Inflamação/imunologia , Influenza Humana/genética , Interferon Tipo I/genética , Interferons/genética , Tempo de Internação , Prognóstico , SARS-CoV-2/fisiologia , Carga Viral/genética , Carga Viral/imunologia , Interferon lambda
9.
Eur J Clin Invest ; 50(12): e13411, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32954520

RESUMO

INTRODUCTION: Asymptomatic carotid artery stenosis (ACAS) may cause future stroke and therefore patients with ACAS require best medical treatment. Patients at high risk for stroke may opt for additional revascularization (either surgery or stenting) but the future stroke risk should outweigh the risk for peri/post-operative stroke/death. Current risk stratification for patients with ACAS is largely based on outdated randomized-controlled trials that lack the integration of improved medical therapies and risk factor control. Furthermore, recent circulating and imaging biomarkers for stroke have never been included in a risk stratification model. The TAXINOMISIS Project aims to develop a new risk stratification model for cerebrovascular complications in patients with ACAS and this will be tested through a prospective observational multicentre clinical trial performed in six major European vascular surgery centres. METHODS AND ANALYSIS: The risk stratification model will compromise clinical, circulating, plaque and imaging biomarkers. The prospective multicentre observational study will include 300 patients with 50%-99% ACAS. The primary endpoint is the three-year incidence of cerebrovascular complications. Biomarkers will be retrieved from plasma samples, brain MRI, carotid MRA and duplex ultrasound. The TAXINOMISIS Project will serve as a platform for the development of new computer tools that assess plaque progression based on radiology images and a lab-on-chip with genetic variants that could predict medication response in individual patients. CONCLUSION: Results from the TAXINOMISIS study could potentially improve future risk stratification in patients with ACAS to assist personalized evidence-based treatment decision-making.


Assuntos
Anticoagulantes/uso terapêutico , Doenças Assintomáticas , Estenose das Carótidas/terapia , Endarterectomia das Carótidas , Hipolipemiantes/uso terapêutico , Inibidores da Agregação Plaquetária/uso terapêutico , Acidente Vascular Cerebral/prevenção & controle , Idoso , Biomarcadores/sangue , Estenose das Carótidas/sangue , Estenose das Carótidas/complicações , Regras de Decisão Clínica , Progressão da Doença , Procedimentos Endovasculares , Feminino , Humanos , Dispositivos Lab-On-A-Chip , Masculino , Pessoa de Meia-Idade , Modelos Teóricos , Testes Farmacogenômicos , Estudos Prospectivos , Medição de Risco , Stents , Acidente Vascular Cerebral/epidemiologia , Acidente Vascular Cerebral/etiologia
10.
Endocrine ; 68(2): 438-447, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32114655

RESUMO

BACKGROUND/AIMS: We assessed the levels of autophagy and mitophagy, that are linked to cancer development and drug resistance, in well differentiated pancreatic neuroendocrine neoplasms (PanNENs) and correlated them with clinico-pathological parameters. METHODS: Fluorescent immunostaining for the autophagy markers LC3Β and p62/or LAMP1 was performed on 22 PanNENs and 11 controls of normal pancreatic tissues and validated through Western blotting. Autophagy quantitative scoring was generated for LC3B-positive puncta and analysed in relation to clinico-pathological parameters. TOMM20/LC3B qualitative assessment of mitophagy levels was undertaken by fluorescent immunostaining. The presence of autophagy/mitophagy was validated by transmission electron microscopy. RESULTS: Autophagy levels (LC3B-positive puncta/cell) were discriminative for normal vs. NEN pancreatic tissue (p = 0.007). A significant association was observed between autophagy levels and tumour grade (Ki67 < 3% vs. Ki67 ≥ 3%; p = 0.021), but not functionality (p = 0.266) size (cut-off of 20 mm; p = 0.808), local invasion (p = 0.481), lymph node- (p = 0.849) and distant metastases (p = 0.699). Qualitative assessment of TOMM20/LC3B demonstrated strong mitophagy levels in PanNENs by fluorescent immunostaining as compared with normal tissue. Transmission electron microscopy revealed enhanced autophagy and mitophagy in PanNEN tissue. Response to molecular targeted therapies in metastatic cases (n = 4) did not reveal any patterns of association to autophagy levels. CONCLUSIONS: Increased autophagy levels are present in primary tumours of patients with PanNENs and are partially attributed to upregulated mitophagy. Grade was the only clinico-pathological parameter associated with autophagy scores.


Assuntos
Tumores Neuroendócrinos , Neoplasias Pancreáticas , Autofagia , Humanos , Mitofagia , Pâncreas
12.
J Autoimmun ; 104: 102319, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31481283

RESUMO

Type I IFNs are well known players in immunity and autoimmunity. They induce potent innate and adaptive immune responses essential for mediating host defenses against viral and bacterial infections but also driving inflammation during chronic inflammatory and autoimmune diseases. Lambda interferons (IFNλs) or type III IFNs, on the other hand, comprise a relatively new family of cytokines sharing homology and functional resemblance with type I IFNs but whose spectrum of activities remains poorly understood. Although IFNλs induce antiviral responses similar to type I IFNs, their restricted pattern of expression suggested that may have more specialized functions at specific body sites such as barrier surfaces. However, recent developments in the field have revealed broader roles of IFNλs in immunity against a diverse range of pathogens including viral, bacterial and fungal infections, and have highlighted unique non-redundant functions of IFNλs that cannot be compensated by type I IFNs. They have also positioned IFNλs as a non-inflammatory or immunoregulatory form of IFNs that possesses the antimicrobial functions of type I IFNs but lacks their pro-inflammatory effects, playing a crucial role in the fine tuning of immune defenses for optimal host protection and minimal host damage. Beyond infections, IFNλs are also emerging as important players in immunity against cancer and autoimmunity, with several studies now demonstrating up-regulation of these molecules at disease sites, and functional involvement in experimental animal models. Here, we critically assess recent advances in our understanding of the IFNλ biology, with emphasis to their emerging roles in cancer and autoimmune diseases, and discuss their potential therapeutic implications.


Assuntos
Doenças Autoimunes/imunologia , Autoimunidade , Regulação da Expressão Gênica/imunologia , Infecções/imunologia , Interferon Tipo I/imunologia , Proteínas de Neoplasias/imunologia , Neoplasias/imunologia , Animais , Doenças Autoimunes/patologia , Modelos Animais de Doenças , Humanos , Neoplasias/patologia
13.
Biochem Pharmacol ; 165: 152-169, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30910693

RESUMO

Macrophages are central to the pathophysiology of rheumatoid arthritis (RA). They constitute the main source of pro-inflammatory cytokines and chemokines such as TNF and IL-1ß, they activate a wide range of immune and non-immune cells, and they secrete diverse tissue degrading enzymes driving chronic pro-inflammatory, tissue destructive and pain responses in RA. However, they can also produce anti-inflammatory cytokines such as IL-10, secrete inhibitors of tissue degrading enzymes and promote immunoregulatory and protective responses, suggesting the existence of macrophages with distinct and diverse functional activities. Although the underlying basis of this phenomenon has remained obscure for years, emerging evidence has now provided insight into the mechanisms and molecular processes involved. Here, we review current knowledge on the biology of macrophages in RA, and highlight recent literature on the heterogeneity, origins and ontogeny of macrophages as part of the mononuclear phagocyte system. We also discuss their plasticity in the context of the M1/M2 paradigm, and the emerging theme of metabolic rewiring as a major mechanism for programming macrophage functions and pro-inflammatory activities. This sheds light into the many facets of macrophages in RA, their molecular regulation and their translational potential for developing novel protective and therapeutic strategies in the clinic.


Assuntos
Artrite Reumatoide/imunologia , Imunidade Celular/imunologia , Mediadores da Inflamação/imunologia , Macrófagos/imunologia , Animais , Anti-Inflamatórios/administração & dosagem , Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/metabolismo , Humanos , Imunidade Celular/efeitos dos fármacos , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Fagócitos/imunologia , Fagócitos/metabolismo
14.
Nat Commun ; 9(1): 5015, 2018 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-30467313

RESUMO

The original version of this Article contained an error in the spelling of the author Emilien Etienne, which was incorrectly given as Emilien Ettiene. These errors have now been corrected in both the PDF and HTML versions of the Article.

15.
Nat Commun ; 9(1): 3333, 2018 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-30127354

RESUMO

Mucormycosis is a life-threatening respiratory fungal infection predominantly caused by Rhizopus species. Mucormycosis has incompletely understood pathogenesis, particularly how abnormalities in iron metabolism compromise immune responses. Here we show how, as opposed to other filamentous fungi, Rhizopus spp. establish intracellular persistence inside alveolar macrophages (AMs). Mechanistically, lack of intracellular swelling of Rhizopus conidia results in surface retention of melanin, which induces phagosome maturation arrest through inhibition of LC3-associated phagocytosis. Intracellular inhibition of Rhizopus is an important effector mechanism, as infection of immunocompetent mice with swollen conidia, which evade phagocytosis, results in acute lethality. Concordantly, AM depletion markedly increases susceptibility to mucormycosis. Host and pathogen transcriptomics, iron supplementation studies, and genetic manipulation of iron assimilation of fungal pathways demonstrate that iron restriction inside macrophages regulates immunity against Rhizopus. Our findings shed light on the pathogenetic mechanisms of mucormycosis and reveal the role of macrophage-mediated nutritional immunity against filamentous fungi.


Assuntos
Interações Hospedeiro-Patógeno , Ferro/metabolismo , Pulmão/microbiologia , Macrófagos Alveolares/metabolismo , Rhizopus/fisiologia , Animais , Parede Celular/metabolismo , Regulação da Expressão Gênica , Macrófagos Alveolares/ultraestrutura , Melaninas/metabolismo , Camundongos Endogâmicos C57BL , Viabilidade Microbiana , Modelos Biológicos , Mucormicose/genética , Mucormicose/microbiologia , Mucormicose/patologia , Fagossomos/metabolismo , Fagossomos/ultraestrutura , Rhizopus/crescimento & desenvolvimento , Esporos Fúngicos/fisiologia
16.
J Pathol ; 243(1): 111-122, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28678391

RESUMO

Neutrophils and neutrophil-released meshwork structures termed neutrophil extracellular traps (NETs) are major mediators of thromboinflammation and emerging targets for therapy, yet the mechanisms and pathways that control the role of neutrophils in thromboinflammation remain poorly understood. Here, we explored the role of IFN-λ1/IL-29, a major antiviral cytokine recently shown to suppress the neutrophil migratory capacity, in prothrombotic and proNETotic functions of neutrophils. In an ex vivo human experimental setting of acute ST-segment elevation myocardial infarction (STEMI), we show that IFN-λ1/IL-29 hinders NET release and diminishes the amount of cytoplasmic TF in neutrophils. Since platelet-neutrophil interaction plays a major role in NET-induced thromboinflammation, we further studied how IFN-λ1/IL-29 may interrupt this interaction. In this context, we identified inorganic polyphosphate (polyP) as a platelet-derived NET inducer in STEMI. In arterial STEMI thrombi, polyP was present in platelets and in close proximity to NET remnants. PolyP release from activated platelets was dependent on thrombin present in infarcted artery plasma, resulting in NET formation by promoting mTOR inhibition and autophagy induction. The effect of polyP on mTOR inhibition was counteracted by IFN-λ1/IL-29 treatment, leading to inhibition of NET formation. Consistently, we show in an in vivo model of FeCl3 -induced arterial thrombosis that IFN-λ2/IL-28A exerts strong antithrombotic potential. Taken together, these findings reveal a novel function of IFN-λ1/IL-29 in the suppression of thromboinflammation. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Coagulação Sanguínea , Plaquetas/metabolismo , Inflamação/sangue , Interleucinas/sangue , Neutrófilos/metabolismo , Polifosfatos/sangue , Infarto do Miocárdio com Supradesnível do Segmento ST/sangue , Trombose/sangue , Animais , Autofagia , Estudos de Casos e Controles , Cloretos , Modelos Animais de Doenças , Armadilhas Extracelulares/metabolismo , Compostos Férricos , Humanos , Inflamação/induzido quimicamente , Inflamação/prevenção & controle , Interferons , Interleucinas/administração & dosagem , Masculino , Camundongos Endogâmicos C57BL , Ativação Plaquetária , Infarto do Miocárdio com Supradesnível do Segmento ST/diagnóstico por imagem , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Trombina/metabolismo , Trombose/induzido quimicamente , Trombose/prevenção & controle
17.
Adv Exp Med Biol ; 850: 1-15, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26324342

RESUMO

Lambda interferons (IFN-λs), type III interferons or interleukins 28 and 29 are the latest addition to the class II cytokine family. They share low homology with the interferon (IFN) and IL-10 cytokine families, yet they exhibit common and unique activities, the full spectrum of which still remains incompletely understood. Although initially described for their antiviral functions, it is now appreciated that IFN-λs also mediate diverse antitumor and immune-modulatory effects, and are key determinants of innate immunity at mucosal sites such as the gastrointestinal and respiratory tracks. Here, we are reviewing the biological functions of IFN-λs, the mechanisms controlling their expression, their downstream effects and their role in the maintenance of homeostasis and disease. We are also exploring the potential application of IFN-λs as novel therapeutics.


Assuntos
Imunidade nas Mucosas/efeitos dos fármacos , Interleucinas/imunologia , Receptores de Interferon/imunologia , Viroses/tratamento farmacológico , Animais , Cromossomos Humanos Par 19 , Éxons , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/virologia , Regulação da Expressão Gênica , Humanos , Imunidade Inata , Interleucinas/genética , Interleucinas/farmacologia , Camundongos , Fases de Leitura Aberta , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Isoformas de Proteínas/farmacologia , Receptores de Interferon/genética , Sistema Respiratório/efeitos dos fármacos , Sistema Respiratório/imunologia , Sistema Respiratório/virologia , Transdução de Sinais , Viroses/imunologia , Viroses/virologia , Receptor de Interferon gama
18.
Autoimmunity ; 48(7): 453-9, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26287441

RESUMO

Antibodies against aquaporin-4 (AQP4) are specific and pathogenetic for Neuromyelitis Optica (NMO). In a previous study, three linear intracellular AQP4 B-cell epitopes were uncovered in NMO patients. A particular epitope showed high-sequence similarity with a segment of the human TAX1BP1 protein, which is necessary for the replication of HTLV-1 virus. The aim of the present study was to investigate whether immunization of mice with the TAX1BP1 peptide could produce specific antibodies against AQP4 epitopes or induce symptoms. Eight C57Bl/6 mice were immunized with TAX1BP1pep in Complete Freund's Adjuvant and eight with adjuvant only. Animals received three subcutaneous injections and sera were obtained before each immunization and at sacrifice. All sera were evaluated by ELISA for antibodies against the TAX1BP1peptide, the homologous AQP4 peptide and all linear AQP4 epitopes. Homologous and cross-inhibition assays were performed to ensure binding specificity, and reactivity against conformational AQP4 epitopes was evaluated by a cell-based assay. Sera from immunized animals showed high reactivity against the immunization peptide, and the homologous AQP4 epitope. Inhibition assays confirmed binding specificity. No antibodies were produced against any other epitopes, either linear or conformational. No clinical or brain inflammatory signs were observed in the animals. The induction of antibodies to an AQP4 epitope in mice immunized with the TAX1BP1-derived peptide suggests that a latent HTLV-1 infection could lead to TAX1BP1 antigen presentation and the production of anti-AQP4 antibodies, probably through T cell-mediated mechanisms. Further studies are needed for exploring triggering factors for NMO especially in HTLV-1-endemic regions.


Assuntos
Anticorpos/química , Aquaporina 4/genética , Epitopos de Linfócito B/genética , Infecções por HTLV-I/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Neoplasias/genética , Neuromielite Óptica/imunologia , Receptores Virais/genética , Animais , Anticorpos/sangue , Especificidade de Anticorpos , Apresentação de Antígeno , Aquaporina 4/química , Aquaporina 4/imunologia , Linfócitos B/imunologia , Linfócitos B/patologia , Linfócitos B/virologia , Reações Cruzadas , Epitopos de Linfócito B/química , Epitopos de Linfócito B/imunologia , Feminino , Adjuvante de Freund/administração & dosagem , Expressão Gênica , Infecções por HTLV-I/genética , Infecções por HTLV-I/patologia , Infecções por HTLV-I/virologia , Vírus Linfotrópico T Tipo 1 Humano/imunologia , Vírus Linfotrópico T Tipo 1 Humano/patogenicidade , Humanos , Imunização , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Neoplasias/química , Proteínas de Neoplasias/imunologia , Neuromielite Óptica/genética , Neuromielite Óptica/patologia , Neuromielite Óptica/virologia , Peptídeos/administração & dosagem , Peptídeos/síntese química , Receptores Virais/química , Receptores Virais/imunologia , Homologia de Sequência de Aminoácidos , Replicação Viral
19.
J Clin Endocrinol Metab ; 100(2): E282-6, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25375981

RESUMO

CONTEXT: Langerhans Cell Histiocytosis (LCH) is a rare disease exhibiting both neoplastic and inflammatory features including abundant cytokine secretion both at the lesional and systemic level. OBJECTIVE: Evaluation of RANKL expression within LCH lesions in various tissues. DESIGN: Cross-sectional study involving paraffin blocks from the diagnostic biopsies of adults with LCH. SETTING: The study was conducted among patients followed in an adult outpatient clinic. SUBJECTS: Eleven patients with active LCH, who were 41.27 ± 3.44 years old, and five patients who were 46.8 ± 7.19 years old with non-LCH diagnosis serving as controls. INTERVENTIONS: RANKL, p65 and CD1a immunostaining of deparaffinized sections from LCH lesions and control tissues. MAIN OUTCOME MEASURE: Comparison of RANKL and p65 expression between LCH lesions, as indicated from concomitant CD1a immunostaining, and control tissue counterparts. RESULTS: A focal positive granular cytoplasmic RANKL staining was found at all lesional sites in a number of cells of the pathological infiltrate, mostly with morphologic features of pathological Langerhans cells (LCs). Compared to control tissues, RANKL positivity in LCH cases showed an excess of staining, both in intensity of staining and the number of stained cells, especially in areas of pathologic infiltration. RANKL staining also coincided with strong p65 NFκB nuclear positivity, especially in lesional infiltrates. CONCLUSIONS: RANKL is highly expressed in active LCH at the lesional level, concomitant to p65 NFκB activation. The use of RANKL inhibition as a rational therapeutic approach of the disease now needs further clinical evaluation.


Assuntos
Osso e Ossos/metabolismo , Histiocitose de Células de Langerhans/metabolismo , Células de Langerhans/metabolismo , Pulmão/metabolismo , Ligante RANK/metabolismo , Adulto , Idoso , Antígenos CD1/metabolismo , Osso e Ossos/patologia , Estudos Transversais , Feminino , Histiocitose de Células de Langerhans/tratamento farmacológico , Histiocitose de Células de Langerhans/patologia , Humanos , Imuno-Histoquímica , Células de Langerhans/patologia , Pulmão/patologia , Masculino , Pessoa de Meia-Idade , Ligante RANK/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA