Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Vaccines (Basel) ; 12(3)2024 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-38543863

RESUMO

BACKGROUND: COVID-19, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has become a recurrent endemic disease affecting the whole world. Since November 2021, Omicron and its subvariants have dominated in the spread of the disease. In order to prevent severe courses of disease, vaccines are needed to boost and maintain antibody levels capable of neutralizing Omicron. Recently, we produced and characterized a SARS-CoV-2 vaccine based on a recombinant fusion protein consisting of hepatitis B virus (HBV)-derived PreS and two SARS-CoV-2 wild-type RBDs. OBJECTIVES: To develop a PreS-RBD vaccine which induces high levels of Omicron-specific neutralizing antibodies. METHODS: We designed, produced, characterized and compared strain-specific (wild-type: W-PreS-W; Omicron: O-PreS-O), bivalent (mix of W-PreS-W and O-PreS-O) and chimeric (i.e., W-PreS-O) SARS-CoV-2 protein subunit vaccines. Immunogens were characterized in vitro using protein chemical methods, mass spectrometry, and circular dichroism in combination with thermal denaturation and immunological methods. In addition, BALB/c mice were immunized with aluminum-hydroxide-adsorbed proteins and aluminum hydroxide alone (i.e., placebo) to study the specific antibody and cytokine responses, safety and Omicron neutralization. RESULTS: Defined and pure immunogens could be produced in significant quantities as secreted and folded proteins in mammalian cells. The antibodies induced after vaccination with different doses of strain-specific, bivalent and chimeric PreS-RBD fusion proteins reacted with wild-type and Omicron RBD in a dose-dependent manner and resulted in a mixed Th1/Th2 immune response. Interestingly, the RBD-specific IgG levels induced with the different vaccines were comparable, but the W-PreS-O-induced virus neutralization titers against Omicron (median VNT50: 5000) were seven- and twofold higher than the W-PreS-W- and O-PreS-O-specific ones, respectively, and they were six-fold higher than those of the bivalent vaccine. CONCLUSION: Among the tested immunogens, the chimeric PreS-RBD subunit vaccine, W-PreS-O, induced the highest neutralizing antibody titers against Omicron. Thus, W-PreS-O seems to be a highly promising COVID-19 vaccine candidate for further preclinical and clinical evaluation.

2.
Pharmaceutics ; 15(4)2023 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-37111542

RESUMO

Lactoferrin is an iron-binding glycoprotein present in most human exocrine fluids, particularly breast milk. Lactoferrin is also released from neutrophil granules, and its concentration increases rapidly at the site of inflammation. Immune cells of both the innate and the adaptive immune system express receptors for lactoferrin to modulate their functions in response to it. On the basis of these interactions, lactoferrin plays many roles in host defense, ranging from augmenting or calming inflammatory pathways to direct killing of pathogens. Complex biological activities of lactoferrin are determined by its ability to sequester iron and by its highly basic N-terminus, via which lactoferrin binds to a plethora of negatively charged surfaces of microorganisms and viruses, as well as to mammalian cells, both normal and cancerous. Proteolytic cleavage of lactoferrin in the digestive tract generates smaller peptides, such as N-terminally derived lactoferricin. Lactoferricin shares some of the properties of lactoferrin, but also exhibits unique characteristics and functions. In this review, we discuss the structure, functions, and potential therapeutic uses of lactoferrin, lactoferricin, and other lactoferrin-derived bioactive peptides in treating various infections and inflammatory conditions. Furthermore, we summarize clinical trials examining the effect of lactoferrin supplementation in disease treatment, with a special focus on its potential use in treating COVID-19.

3.
Int J Mol Sci ; 24(6)2023 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-36982424

RESUMO

More than three years ago, the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) caused the unforeseen COVID-19 pandemic with millions of deaths. In the meantime, SARS-CoV-2 has become endemic and is now part of the repertoire of viruses causing seasonal severe respiratory infections. Due to several factors, among them the development of SARS-CoV-2 immunity through natural infection, vaccination and the current dominance of seemingly less pathogenic strains belonging to the omicron lineage, the COVID-19 situation has stabilized. However, several challenges remain and the possible new occurrence of highly pathogenic variants remains a threat. Here we review the development, features and importance of assays measuring SARS-CoV-2 neutralizing antibodies (NAbs). In particular we focus on in vitro infection assays and molecular interaction assays studying the binding of the receptor binding domain (RBD) with its cognate cellular receptor ACE2. These assays, but not the measurement of SARS-CoV-2-specific antibodies per se, can inform us of whether antibodies produced by convalescent or vaccinated subjects may protect against the infection and thus have the potential to predict the risk of becoming newly infected. This information is extremely important given the fact that a considerable number of subjects, in particular vulnerable persons, respond poorly to the vaccination with the production of neutralizing antibodies. Furthermore, these assays allow to determine and evaluate the virus-neutralizing capacity of antibodies induced by vaccines and administration of plasma-, immunoglobulin preparations, monoclonal antibodies, ACE2 variants or synthetic compounds to be used for therapy of COVID-19 and assist in the preclinical evaluation of vaccines. Both types of assays can be relatively quickly adapted to newly emerging virus variants to inform us about the magnitude of cross-neutralization, which may even allow us to estimate the risk of becoming infected by newly appearing virus variants. Given the paramount importance of the infection and interaction assays we discuss their specific features, possible advantages and disadvantages, technical aspects and not yet fully resolved issues, such as cut-off levels predicting the degree of in vivo protection.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , Enzima de Conversão de Angiotensina 2 , Pandemias , Anticorpos Antivirais , Anticorpos Neutralizantes
4.
Immunol Lett ; 253: 30-40, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36608905

RESUMO

Interstitial lung disease comprises numerous clinical entities posing significant challenges towards a prompt and accurate diagnosis. Amongst the contributing factors are intricate pathophysiological mechanisms, an overlap between conditions, and interobserver disagreement. We developed a model for patient clustering offering an additional approach to such complex clinical cases. The model is based on surface phenotyping of over 40 markers on immune cells isolated from bronchoalveolar lavage in combination with clinical data. Based on the marker expression pattern we constructed an individual immune cell profile, then merged these to create a global profile encompassing various pathologies. The contribution of each participant to the global profile was assessed through dimensionality reduction tools and the ensuing similarity between samples was calculated. Our model enables two approaches. First, assessing the immune cell population landscape similarity between patients within a diagnostic group allows rapid identification of divergent profiles, which is particularly helpful for cases with uncertain diagnoses. Second, sample clustering is based exclusively on the calculated similarity of the immune cell profiles, thereby removing physician bias and relying on cellular nearest neighbors.


Assuntos
Doenças Pulmonares Intersticiais , Humanos , Líquido da Lavagem Broncoalveolar , Doenças Pulmonares Intersticiais/diagnóstico , Lavagem Broncoalveolar
5.
Front Immunol ; 13: 958581, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36081512

RESUMO

In addition to vaccines, there is an urgent need for supplemental antiviral therapeutics to dampen the persistent COVID-19 pandemic caused by the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). The transmembrane protease serine 2 (TMPRSS2), that is responsible for proteolytic priming of the SARS-CoV-2 spike protein, appears as a rational therapeutic target. Accordingly, selective inhibitors of TMPRSS2 represent potential tools for prevention and treatment of COVID-19. Previously, we identified the human milk glycoprotein lactoferrin as a natural inhibitor of plasminogen conversion to plasmin, a serine protease homologous to TMPRSS2. Here, we tested whether lactoferrin and lactoferricin, a biologically active natural peptide produced by pepsin-mediated digestion of lactoferrin, together with synthetic peptides derived from lactoferrin, were able to block TMPRSS2 and SARS-CoV-2 infection. Particularly, we revealed that both lactoferricin and the N-terminal synthetic peptide pLF1 significantly inhibited: i) proteolytic activity of TMPRSS2 and plasmin, ii) proteolytic processing of the SARS-CoV-2 spike protein, and iii) SARS-CoV-2 infection of SARS-CoV-2-permissive cells. Thus, natural and synthetic peptides derived from lactoferrin represent feasible candidates for supporting prevention and treatment of COVID-19.


Assuntos
Tratamento Farmacológico da COVID-19 , Lactoferrina , SARS-CoV-2 , Serina Endopeptidases , Inibidores de Serina Proteinase , Fibrinolisina , Humanos , Lactoferrina/farmacologia , Pandemias , Inibidores de Serina Proteinase/química , Inibidores de Serina Proteinase/farmacologia , Glicoproteína da Espícula de Coronavírus
6.
Immunol Lett ; 250: 1-6, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-36108774

RESUMO

Antibody testing after COVID-19 vaccination is generally not recommended. Here, we present the results of a retrospective study, in which we analyzed antibody levels before and after the first dose of the ChAdOx1 vector vaccine. We identified 5% non-responders (43.6 ± 10.6 years; females: 41%) and 3.4% low-responders (44.2 ± 10.1 years; females: 64%) after the first dose. Of these, 61 individuals received a timely second dose either with a homologous (ChAdOx1/ChAdOx1) or heterologous (ChAdOx1/mRNA-1273) schedule. All vaccinees achieved positive S1-specific IgG titers to the ancestral SARS-CoV-2 strain after the second dose, but antibody levels as well as neutralization titers against the ancestral SARS-CoV-2 strain were higher after the heterologous schedule. However, Omicron-specific neutralizing antibodies were not detectable after two doses in either group, indicating that a third vaccine dose is needed to enhance cross-reactive antibodies against currently circulating and emerging variants of concern.


Assuntos
Vacinas contra COVID-19 , COVID-19 , Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19/prevenção & controle , Feminino , Humanos , Imunoglobulina G , Estudos Retrospectivos , SARS-CoV-2 , Soroconversão , Vacinação
7.
Front Immunol ; 13: 889138, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35634285

RESUMO

Background: Individuals with secondary immunodeficiencies belong to the most vulnerable groups to succumb to COVID-19 and thus are prioritized for SARS-CoV-2 vaccination. However, knowledge about the persistence and anamnestic responses following SARS-CoV-2-mRNA vaccinations is limited in these patients. Methods: In a prospective, open-label, phase four trial we analyzed S1-specific IgG, neutralizing antibodies and cytokine responses in previously non-infected patients with cancer or autoimmune disease during primary mRNA vaccination and up to one month after booster. Results: 263 patients with solid tumors (SOT, n=63), multiple myeloma (MM, n=70), inflammatory bowel diseases (IBD, n=130) and 66 controls were analyzed. One month after the two-dose primary vaccination the highest non-responder rate was associated with lower CD19+ B-cell counts and was found in MM patients (17%). S1-specific IgG levels correlated with IL-2 and IFN-γ responses in controls and IBD patients, but not in cancer patients. Six months after the second dose, 18% of patients with MM, 10% with SOT and 4% with IBD became seronegative; no one from the control group became negative. However, in IBD patients treated with TNF-α inhibitors, antibody levels declined more rapidly than in controls. Overall, vaccination with mRNA-1273 led to higher antibody levels than with BNT162b2. Importantly, booster vaccination increased antibody levels >8-fold in seroresponders and induced anamnestic responses even in those with undetectable pre-booster antibody levels. Nevertheless, in IBD patients with TNF-α inhibitors even after booster vaccination, antibody levels were lower than in untreated IBD patients and controls. Conclusion: Immunomonitoring of vaccine-specific antibody and cellular responses seems advisable to identify vaccination failures and consequently establishing personalized vaccination schedules, including shorter booster intervals, and helps to improve vaccine effectiveness in all patients with secondary immunodeficiencies. Trial registration: EudraCT Number: 2021-000291-11.


Assuntos
COVID-19 , Doenças Inflamatórias Intestinais , Mieloma Múltiplo , Vacina BNT162 , COVID-19/prevenção & controle , Vacinas contra COVID-19 , Humanos , Imunização Secundária , Hospedeiro Imunocomprometido , Imunoglobulina G , Memória Imunológica , Mieloma Múltiplo/terapia , Estudos Prospectivos , RNA Mensageiro , SARS-CoV-2 , Fator de Necrose Tumoral alfa , Vacinação
8.
Allergy ; 77(8): 2431-2445, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35357709

RESUMO

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the ongoing global COVID-19 pandemic. One possibility to control the pandemic is to induce sterilizing immunity through the induction and maintenance of neutralizing antibodies preventing SARS-CoV-2 from entering human cells to replicate in. METHODS: We report the construction and in vitro and in vivo characterization of a SARS-CoV-2 subunit vaccine (PreS-RBD) based on a structurally folded recombinant fusion protein consisting of two SARS-CoV-2 Spike protein receptor-binding domains (RBD) fused to the N- and C-terminus of hepatitis B virus (HBV) surface antigen PreS to enable the two unrelated proteins serving as immunologic carriers for each other. RESULTS: PreS-RBD, but not RBD alone, induced a robust and uniform RBD-specific IgG response in rabbits. Currently available genetic SARS-CoV-2 vaccines induce mainly transient IgG1 responses in vaccinated subjects whereas the PreS-RBD vaccine induced RBD-specific IgG antibodies consisting of an early IgG1 and sustained IgG4 antibody response in a SARS-CoV-2 naive subject. PreS-RBD-specific IgG antibodies were detected in serum and mucosal secretions, reacted with SARS-CoV-2 variants, including the omicron variant of concern and the HBV receptor-binding sites on PreS of currently known HBV genotypes. PreS-RBD-specific antibodies of the immunized subject more potently inhibited the interaction of RBD with its human receptor ACE2 and their virus-neutralizing titers (VNTs) were higher than median VNTs in a random sample of healthy subjects fully immunized with registered SARS-CoV-2 vaccines or in COVID-19 convalescent subjects. CONCLUSION: The PreS-RBD vaccine has the potential to serve as a combination vaccine for inducing sterilizing immunity against SARS-CoV-2 and HBV by stopping viral replication through the inhibition of cellular virus entry.


Assuntos
Vacinas contra COVID-19 , COVID-19 , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , COVID-19/prevenção & controle , Vacinas contra COVID-19/imunologia , Humanos , Imunoglobulina G , Pandemias/prevenção & controle , Coelhos , Glicoproteína da Espícula de Coronavírus/imunologia
9.
Front Med (Lausanne) ; 9: 822316, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35242786

RESUMO

In a SARS-CoV-2 seroprevalence study conducted with 1,655 working adults in spring of 2020, 12 of the subjects presented with positive neutralization test (NT) titers (>1:10). They were here followed up for 1 year to assess their Ab persistence. We report that 7/12 individuals (58%) had NT_50 titers ≥1:50 and S1-specific IgG ≥50 BAU/ml 1 year after mild COVID-19 infection. S1-specific IgG were retained until a year when these levels were at least >60 BAU/ml at 3 months post-infection. For both the initial fast and subsequent slow decline phase of Abs, we observed a significant correlation between NT_50 titers and S1-specific IgG and thus propose S1-IgG of 60 BAU/ml 3 months post-infection as a potential threshold to predict neutralizing Ab persistence for 1 year. NT_50 titers and S1-specific IgG also correlated with circulating S1-specific memory B-cells. SARS-CoV-2-specific Ab levels after primary mRNA vaccination in healthy controls were higher (Geometric Mean Concentration [GMC] 3158 BAU/ml [CI 2592 to 3848]) than after mild COVID-19 infection (GMC 82 BAU/ml [CI 48 to 139]), but showed a stronger fold-decline within 5-6 months (0.20-fold, to GMC 619 BAU/ml [CI 479 to 801] vs. 0.56-fold, to GMC 46 BAU/ml [CI 26 to 82]). Of particular interest, the decline of both infection- and vaccine-induced Abs correlated with body mass index. Our data contribute to describe decline and persistence of SARS-CoV-2-specific Abs after infection and vaccination, yet the relevance of the maintained Ab levels for protection against infection and/or disease depends on the so far undefined correlate of protection.

10.
Elife ; 112022 01 13.
Artigo em Inglês | MEDLINE | ID: mdl-35023830

RESUMO

Despite tremendous progress in the understanding of COVID-19, mechanistic insight into immunological, disease-driving factors remains limited. We generated maVie16, a mouse-adapted SARS-CoV-2, by serial passaging of a human isolate. In silico modeling revealed how only three Spike mutations of maVie16 enhanced interaction with murine ACE2. maVie16 induced profound pathology in BALB/c and C57BL/6 mice, and the resulting mouse COVID-19 (mCOVID-19) replicated critical aspects of human disease, including early lymphopenia, pulmonary immune cell infiltration, pneumonia, and specific adaptive immunity. Inhibition of the proinflammatory cytokines IFNγ and TNF substantially reduced immunopathology. Importantly, genetic ACE2-deficiency completely prevented mCOVID-19 development. Finally, inhalation therapy with recombinant ACE2 fully protected mice from mCOVID-19, revealing a novel and efficient treatment. Thus, we here present maVie16 as a new tool to model COVID-19 for the discovery of new therapies and show that disease severity is determined by cytokine-driven immunopathology and critically dependent on ACE2 in vivo.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19/virologia , Interferon gama/farmacologia , SARS-CoV-2/patogenicidade , Imunidade Adaptativa/imunologia , Animais , Modelos Animais de Doenças , Interferon gama/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Peptidil Dipeptidase A/genética , Glicoproteína da Espícula de Coronavírus/genética
11.
J Hepatol ; 75(5): 1164-1176, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34242699

RESUMO

BACKGROUND & AIMS: 24-Norursodeoxycholic acid (NorUDCA) is a novel therapeutic bile acid used to treat immune-mediated cholestatic liver diseases, such as primary sclerosing cholangitis (PSC), where dysregulated T cells including CD8+ T cells contribute to hepatobiliary immunopathology. We hypothesized that NorUDCA may directly modulate CD8+ T cell function thus contributing to its therapeutic efficacy. METHODS: NorUDCA's immunomodulatory effects were first studied in Mdr2-/- mice, as a cholestatic model of PSC. To differentiate NorUDCA's immunomodulatory effects on CD8+ T cell function from its anticholestatic actions, we also used a non-cholestatic model of hepatic injury induced by an excessive CD8+ T cell immune response upon acute non-cytolytic lymphocytic choriomeningitis virus (LCMV) infection. Studies included molecular and biochemical approaches, flow cytometry and metabolic assays in murine CD8+ T cells in vitro. Mass spectrometry was used to identify potential CD8+ T cell targets modulated by NorUDCA. The signaling effects of NorUDCA observed in murine cells were validated in circulating T cells from patients with PSC. RESULTS: NorUDCA demonstrated immunomodulatory effects by reducing hepatic innate and adaptive immune cells, including CD8+ T cells in the Mdr2-/- model. In the non-cholestatic model of CD8+ T cell-driven immunopathology induced by acute LCMV infection, NorUDCA ameliorated hepatic injury and systemic inflammation. Mechanistically, NorUDCA demonstrated strong immunomodulatory efficacy in CD8+ T cells affecting lymphoblastogenesis, expansion, glycolysis and mTORC1 signaling. Mass spectrometry identified that NorUDCA regulates CD8+ T cells by targeting mTORC1. NorUDCA's impact on mTORC1 signaling was further confirmed in circulating PSC CD8+ T cells. CONCLUSIONS: NorUDCA has a direct modulatory impact on CD8+ T cells and attenuates excessive CD8+ T cell-driven hepatic immunopathology. These findings are relevant for treatment of immune-mediated liver diseases such as PSC. LAY SUMMARY: Elucidating the mechanisms by which 24-norursodeoxycholic acid (NorUDCA) works for the treatment of immune-mediated liver diseases, such as primary sclerosing cholangitis, is of considerable clinical interest. Herein, we uncovered an unrecognized property of NorUDCA in the immunometabolic regulation of CD8+ T cells, which has therapeutic relevance for immune-mediated liver diseases, including PSC.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Inflamação/tratamento farmacológico , Fígado/efeitos dos fármacos , Ácido Ursodesoxicólico/análogos & derivados , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Modelos Animais de Doenças , Inflamação/fisiopatologia , Fígado/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Ácido Ursodesoxicólico/farmacologia , Ácido Ursodesoxicólico/uso terapêutico
13.
Anticancer Agents Med Chem ; 19(13): 1609-1617, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31038078

RESUMO

BACKGROUND: Based on recent studies, new therapeutic strategies have been developed for cancer treatment using microRNAs (miRNAs). With this view, miRNAs manipulating techniques can be considered as novel therapeutic prospects for cancer treatment. In this study, we evaluated the expression of miR-4301 in human lung cancer cell lines and investigated its potential role in cell proliferation and tumor suppression on Non-Small Cell Lung Cancer (NSCLC) cells. METHODS: We used quantitative Real-Time Polymerase Chain Reaction (qRT-PCR) to examine the level of miR- 4301 expression in human lung cancer cell lines (A549, QU-DB) and non-malignant lung epithelial cells (HFLF-PI5). Then, we investigated the effect of miR-4301 by transfecting it into these cell lines and probing for cancer cell viability and apoptosis using the MTT assay, flow cytometry and immunofluorescence staining. RESULTS: Our results showed that the expression level of miR-4301 was significantly reduced in human lung cancer cell lines (P<0.001). When miR-4301 was transfected in lung cancer cells, their cell proliferation was suppressed and apoptosis induced. This decline in cell survival was confirmed by the MTT assay. Transfection of miR-4301 caused an increase in early and late apoptotic cells in all lung cancer cell lines tested. CONCLUSIONS: Our findings show that miR-4301 may act as a lung cancer suppressor through targeting of proteins involved in cell proliferation and survival. For this reason, targeting miR-4301 may provide a new strategy for the diagnosis and treatment of patients with this deadly disease. This article is protected by copyright. All rights reserved.


Assuntos
Apoptose/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/patologia , Transfecção , Linhagem Celular Tumoral , Proliferação de Células/genética , Sobrevivência Celular/genética , Regulação para Baixo , Humanos , Reação em Cadeia da Polimerase em Tempo Real
14.
J Leukoc Biol ; 105(3): 519-530, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30657605

RESUMO

The plasminogen system is harnessed in a wide variety of physiological processes, such as fibrinolysis, cell migration, or efferocytosis; and accordingly, it is essential upon inflammation, tissue remodeling, wound healing, and for homeostatic maintenance in general. Previously, we identified a plasminogen receptor in the mannose 6-phosphate/insulin-like growth factor 2 receptor (M6P/IGF2R, CD222). Here, we demonstrate by means of genetic knockdown, knockout, and rescue approaches combined with functional studies that M6P/IGF2R is up-regulated on the surface of macrophages, recognizes plasminogen exposed on the surface of apoptotic cells, and mediates plasminogen-induced efferocytosis. The level of uptake of plasminogen-coated apoptotic cells inversely correlates with the TNF-α production by phagocytes indicating tissue clearance without inflammation by this mechanism. Our results reveal an up-to-now undetermined function of M6P/IGF2R in clearance of apoptotic cells, which is crucial for tissue homeostasis.


Assuntos
Fagocitose/efeitos dos fármacos , Plasminogênio/farmacologia , Receptor IGF Tipo 2/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Técnicas de Inativação de Genes , Humanos , Células Jurkat , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Células THP-1 , Fator de Necrose Tumoral alfa/metabolismo
15.
Allergy ; 74(3): 483-494, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30338531

RESUMO

BACKGROUND: Macrophages can be converted in vitro into immunoregulatory M2b macrophages in the presence of immune complexes (ICs), but the role of the specific subclasses IgG1 or IgG4 in this phenotypic and functional change is not known. OBJECTIVE: We aimed to refine the original method by applying precisely defined ICs of the subclasses IgG4 or IgG1 constructed by two independent methods. METHODS: Monocyte-derived macrophages (MDMs) were treated with M-CSF, followed by IL-4/IL-13 to induce the M2a allergic phenotype. To mimic unspecific or allergen-specific ICs, plates were coated with myeloma IgG1 or IgG4, or with grass pollen allergen Phl p 5 followed by recombinant human Phl p 5-specific IgG1 or IgG4. M2a polarized macrophages were then added, cultured, and examined for cellular markers and cytokines by flow cytometry, ELISA, and rtPCR. Alternatively, immune complexes with IgG1 or IgG4 were formed using protein L. RESULTS: IgG4 ICs down regulated CD163 and CD206 on M2a cells, and significantly increased IL-10, IL-6, TNFα, and CCL1 secretion, indicating a shift to an M2b-like phenotype. Treatment with IgG4 ICs resulted in expression of FcγRII and down modulation of FcγRII compared with IgG1 treated cells (P = 0.0335) or untreated cells (P < 0.00001). CONCLUSION: Immune complexes with subclasses IgG1 and IgG4 can in vitro be generated by plate absorption, and in fluid form by protein L. Cross-linking of FcγRIIb by the IgG4 subclass redirects pro-allergic M2a macrophages to an M2b-like immunosuppressive phenotype. This suggests an interplay of macrophages with IgG4 in immune tolerance, likely relevant in allergen immunotherapy.


Assuntos
Tolerância Imunológica , Imunoglobulina G/imunologia , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Fenótipo , Alérgenos/imunologia , Complexo Antígeno-Anticorpo/imunologia , Biomarcadores , Citocinas/metabolismo , Perfilação da Expressão Gênica , Humanos , Imunofenotipagem , Macrófagos/metabolismo , Monócitos/imunologia , Monócitos/metabolismo , Receptores de IgG/metabolismo
16.
Brain ; 141(8): 2329-2342, 2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29860501

RESUMO

X-linked adrenoleukodystrophy is caused by ATP-binding cassette transporter D1 (ABCD1) mutations and manifests by default as slowly progressive spinal cord axonopathy with associated demyelination (adrenomyloneuropathy). In 60% of male cases, however, X-linked adrenoleukodystrophy converts to devastating cerebral inflammation and demyelination (cerebral adrenoleukodystrophy) with infiltrating blood-derived monocytes and macrophages and cytotoxic T cells that can only be stopped by allogeneic haematopoietic stem cell transplantation or gene therapy at an early stage of the disease. Recently, we identified monocytes/macrophages but not T cells to be severely affected metabolically by ABCD1 deficiency. Here we found by whole transcriptome analysis that, although monocytes of patients with X-linked adrenoleukodystrophy have normal capacity for macrophage differentiation and phagocytosis, they are pro-inflammatory skewed also in patients with adrenomyloneuropathy in the absence of cerebral inflammation. Following lipopolysaccharide activation, the ingestion of myelin debris, normally triggering anti-inflammatory polarization, did not fully reverse the pro-inflammatory status of X-linked adrenoleukodystrophy macrophages. Immunohistochemistry on post-mortem cerebral adrenoleukodystrophy lesions reflected the activation pattern by prominent presence of enlarged lipid-laden macrophages strongly positive for the pro-inflammatory marker co-stimulatory molecule CD86. Comparative analyses of lesions with matching macrophage density in cases of cerebral adrenoleukodystrophy and acute multiple sclerosis showed a similar extent of pro-inflammatory activation but a striking reduction of anti-inflammatory mannose receptor (CD206) and haemoglobin-haptoglobin receptor (CD163) expression on cerebral adrenoleukodystrophy macrophages. Accordingly, ABCD1-deficiency leads to an impaired plasticity of macrophages that is reflected in incomplete establishment of anti-inflammatory responses, thus possibly contributing to the devastating rapidly progressive demyelination in cerebral adrenoleukodystrophy that only in rare cases arrests spontaneously. These findings emphasize monocytes/macrophages as crucial therapeutic targets for preventing or stopping myelin destruction in patients with X-linked adrenoleukodystrophy.


Assuntos
Membro 1 da Subfamília D de Transportadores de Cassetes de Ligação de ATP/genética , Adrenoleucodistrofia/imunologia , Macrófagos/metabolismo , Membro 1 da Subfamília D de Transportadores de Cassetes de Ligação de ATP/metabolismo , Membro 1 da Subfamília D de Transportadores de Cassetes de Ligação de ATP/fisiologia , Transportadores de Cassetes de Ligação de ATP/genética , Adrenoleucodistrofia/genética , Adrenoleucodistrofia/fisiopatologia , Adulto , Plasticidade Celular/genética , Plasticidade Celular/fisiologia , Doenças Desmielinizantes/metabolismo , Humanos , Macrófagos/fisiologia , Masculino , Pessoa de Meia-Idade , Monócitos/metabolismo , Monócitos/fisiologia , Bainha de Mielina/metabolismo , População Branca , Sequenciamento do Exoma/métodos
17.
Front Immunol ; 9: 852, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29780382

RESUMO

If misregulated, macrophage (Mϕ)-T cell interactions can drive chronic inflammation thereby causing diseases, such as rheumatoid arthritis (RA). We report that in a proinflammatory environment, granulocyte-Mϕ (GM-CSF)- and Mϕ colony-stimulating factor (M-CSF)-dependent Mϕs have dichotomous effects on T cell activity. While GM-CSF-dependent Mϕs show a highly stimulatory activity typical for M1 Mϕs, M-CSF-dependent Mϕs, marked by folate receptor ß (FRß), adopt an immunosuppressive M2 phenotype. We find the latter to be caused by the purinergic pathway that directs release of extracellular ATP and its conversion to immunosuppressive adenosine by co-expressed CD39 and CD73. Since we observed a misbalance between immunosuppressive and immunostimulatory Mϕs in human and murine arthritic joints, we devised a new strategy for RA treatment based on targeted delivery of a novel methotrexate (MTX) formulation to the immunosuppressive FRß+CD39+CD73+ Mϕs, which boosts adenosine production and curtails the dominance of proinflammatory Mϕs. In contrast to untargeted MTX, this approach leads to potent alleviation of inflammation in the murine arthritis model. In conclusion, we define the Mϕ extracellular purine metabolism as a novel checkpoint in Mϕ cell fate decision-making and an attractive target to control pathological Mϕs in immune-mediated diseases.


Assuntos
Artrite Reumatoide/imunologia , Diferenciação Celular , Macrófagos/imunologia , Macrófagos/metabolismo , Purinas/metabolismo , Adenosina/imunologia , Animais , Artrite Reumatoide/tratamento farmacológico , Proliferação de Células , Modelos Animais de Doenças , Fator Estimulador de Colônias de Granulócitos e Macrófagos/farmacologia , Humanos , Imunossupressores/administração & dosagem , Imunossupressores/uso terapêutico , Inflamação/tratamento farmacológico , Fator Estimulador de Colônias de Macrófagos/farmacologia , Masculino , Metotrexato/administração & dosagem , Metotrexato/uso terapêutico , Camundongos , Monócitos/efeitos dos fármacos , Líquido Sinovial/citologia , Líquido Sinovial/imunologia
18.
J Biol Chem ; 293(22): 8600-8613, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29669808

RESUMO

The plasminogen system is essential for dissolution of fibrin clots, and in addition, it is involved in a wide variety of other physiological processes, including proteolytic activation of growth factors, cell migration, and removal of protein aggregates. On the other hand, uncontrolled plasminogen activation contributes to many pathological processes (e.g. tumor cells' invasion in cancer progression). Moreover, some virulent bacterial species (e.g. Streptococci or Borrelia) bind human plasminogen and hijack the host's plasminogen system to penetrate tissue barriers. Thus, the conversion of plasminogen to the active serine protease plasmin must be tightly regulated. Here, we show that human lactoferrin, an iron-binding milk glycoprotein, blocks plasminogen activation on the cell surface by direct binding to human plasminogen. We mapped the mutual binding sites to the N-terminal region of lactoferrin, encompassed also in the bioactive peptide lactoferricin, and kringle 5 of plasminogen. Finally, lactoferrin blocked tumor cell invasion in vitro and also plasminogen activation driven by Borrelia Our results explain many diverse biological properties of lactoferrin and also suggest that lactoferrin may be useful as a potential tool for therapeutic interventions to prevent both invasive malignant cells and virulent bacteria from penetrating host tissues.


Assuntos
Borrelia/metabolismo , Fibrinolisina/metabolismo , Fibrinólise , Lactoferrina/metabolismo , Plasminogênio/antagonistas & inibidores , Streptococcus/metabolismo , Movimento Celular , Células Cultivadas , Cristalografia por Raios X , Humanos , Lactoferrina/química , Lactoferrina/genética , Plasminogênio/metabolismo , Conformação Proteica
19.
Water Res ; 141: 428-438, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29409685

RESUMO

Legionella infections are among the most important waterborne infections with constantly increasing numbers of cases in industrialized countries, as a result of aging populations, rising numbers of immunocompromised individuals and increased need for conditioned water due to climate change. Surveillance of water systems is based on microbiological culture-based techniques; however, it has been shown that high percentages of the Legionella populations in water systems are not culturable. In the past two decades, the relevance of such viable but non-culturable (VBNC) legionellae has been controversially discussed, and whether VBNC legionellae can directly infect human macrophages, the primary targets of Legionella infections, remains unclear. In this study, it was demonstrated for the first time that several starved VBNC Legionella strains (four L. pneumophila serogroup 1 strains, a serogroup 6 strain and a L. micdadei strain) can directly infect different types of human macrophages and amoebae even after one year of starvation in ultrapure water. However, under these conditions, the strains caused infection with reduced efficacy, as represented by the lower percentages of infected cells, prolonged time in co-culture and higher multiplicities of infection required. Interestingly, the VBNC cells remained mostly non-culturable even after multiplication within the host cells. Amoebal infection by starved VBNC Legionella, which likely occurs in oligotrophic biofilms, would result in an increase in the bacterial concentration in drinking-water systems. If cells remain in the VBNC state, the real number of active legionellae will be underestimated by the use of culture-based standard techniques. Thus, further quantitative research is needed in order to determine, whether and how many starved VBNC Legionella cells are able to cause disease in humans.


Assuntos
Amoeba/microbiologia , Legionella/patogenicidade , Macrófagos/microbiologia , Técnicas de Cocultura , Humanos , Legionelose , Virulência , Poluentes da Água
20.
J Cell Biochem ; 119(8): 6408-6417, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29236292

RESUMO

In several cancers, microRNA (miRNAs) play vital roles in tumor initiation, drug resistance, and metastasis. The aim of this study was to examine the expression levels of miR-4301 in human breast cancer and investigate whether its potential roles involved targeting Dopamine receptor D2 (DRD2). Real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR) was also used to examine the expression levels of miR-4301 in human breast cancer cell lines MDA-MB-231, MCF-7, and SKBR3. In these cell lines, MTT assay, immunofluorescence staining, caspase assay, proliferation assay, and flow cytometry were conducted to explore the potential functions of miR-4301. The effects of modulating miR-4301 on transcription levels of DRD2 were subsequently confirmed via qRT-PCR. miR-4301 expression levels were significantly decreased in human breast cancer specimens and cell lines (P < 0.05). Transfection of miR-4301 in breast cancer cells suppressed cell proliferation and induced apoptosis. Expression analysis indicated that miR-4301 was inversely correlated with DRD2 expression in breast cancer specimens. qRT-PCR showed that miR-4301 negatively regulated DRD2 expression. Downregulation of DRD2 expression in MDA-MB-231, MCF-7, and SKBR3 cells suppressed cell proliferation and promoted apoptosis.


Assuntos
Apoptose , Neoplasias da Mama/metabolismo , Regulação Neoplásica da Expressão Gênica , MicroRNAs/biossíntese , Proteínas de Neoplasias/biossíntese , RNA Neoplásico/biossíntese , Receptores de Dopamina D2/biossíntese , Adulto , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Feminino , Humanos , Células MCF-7 , MicroRNAs/genética , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , RNA Neoplásico/genética , Receptores de Dopamina D2/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...