Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Cells ; 12(14)2023 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-37508538

RESUMO

CD45 is a transmembrane glycoprotein that is located on the surface of all leukocytes and modulates both innate and adaptive immune system functions. In a recent study, inflammation modulated the CD45 expression in leukocytes, but the effect on the expression of CD45 subtypes is unknown. In the present study, we therefore investigated the effect of inflammatory conditions in humans (surgery, sepsis) and ex vivo incubation with lipopolysaccharides (LPS) on the expression of the subtypes CD45RA and CD45RO in granulocytes, lymphocytes, and monocytes. Whole blood samples were obtained from healthy volunteers, postoperative patients, and patients with sepsis at day 1 of diagnosis, respectively. Samples were incubated with fluorescent antibodies directed against CD45, CD45RA and CD45RO in the absence and presence of lipopolysaccharide and subjected to flow cytometry. In comparison to volunteers, CD45RA surface expression in postoperative and septic patients was reduced by 89% exclusively on granulocytes, but not on lymphocytes or monocytes. In contrast, CD45RO was exclusively reduced on lymphocytes, by 82%, but not on other cell types. Receiver operating characteristic curve analyses demonstrated that CD45RA (on granulocytes) and CD45RO (on lymphocytes) allow a good differentiation of volunteers and patients with sepsis (AUC = 0.9; p = 0.0001). The addition of LPS to the whole blood samples obtained from volunteers, postoperative patients, and septic patients markedly increased the CD45RO expression in granulocytes, lymphocytes, and monocytes. In contrast, LPS reduced CD45RA exclusively on monocytes. In conclusion, the surface expression of CD45RA and CD45RO is regulated in inflammation in a cell-type- and stimulus-specific manner. Considering that CD45 subtypes are critically involved in immune system signaling, the pathophysiologic and diagnostic implications warrant further investigation.


Assuntos
Antígenos Comuns de Leucócito , Sepse , Humanos , Inflamação , Leucócitos , Lipopolissacarídeos/farmacologia , Linfócitos
2.
J Clin Med ; 11(14)2022 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-35887979

RESUMO

CD45 is a transmembrane glycoprotein and protein tyrosine phosphatase expressed on the surface of all nucleated hematopoietic cells. While there is increasing evidence demonstrating the involvement of CD45 in immune system regulation, no information on CD45 expression in inflammation and sepsis is currently available. Therefore, we determined the CD45 surface expression on granulocytes, lymphocytes, and monocytes in patients with COVID-19 and healthy volunteers in both absence and presence of lipopolysaccharide (LPS). Following approval by the local ethics committee, whole blood samples were obtained from patients with COVID-19 infection on day 1 of hospital admission and healthy volunteers. Samples were incubated in absence and presence of LPS and CD45 was measured in granulocytes, lymphocytes, and monocytes using flow cytometry. In comparison with healthy individuals, COVID-19 patients showed an increased CD45 expression on the surface of granulocytes (+35%, p < 0.02) and lymphocytes (+39%, p < 0.0001), but a reduced CD45 expression on monocytes (−35%, p < 0.0001). LPS incubation of whole blood from healthy individuals increased the CD45 expression on granulocytes (+430%, p < 0.0001), lymphocytes (+32%, p = 0.0012), and monocytes (+36%, p = 0.0005), respectively. LPS incubation of whole blood samples from COVID-19 patients increased the CD45 expression on granulocytes and monocytes, and decreased the CD45 expression on lymphocytes. In conclusion, CD45 expression on leucocytes is altered: (1) in COVID-19 patients, and (2) in in vitro endotoxemia in a complex cell-specific way, thus representing a new immunoregulatory mechanism.

3.
Front Immunol ; 12: 711876, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34659202

RESUMO

Cerebral malaria is a potentially lethal disease, which is caused by excessive inflammatory responses to Plasmodium parasites. Here we use a newly developed transgenic Plasmodium berghei ANKA (PbAAma1OVA) parasite that can be used to study parasite-specific T cell responses. Our present study demonstrates that Ifnar1-/- mice, which lack type I interferon receptor-dependent signaling, are protected from experimental cerebral malaria (ECM) when infected with this novel parasite. Although CD8+ T cell responses generated in the spleen are essential for the development of ECM, we measured comparable parasite-specific cytotoxic T cell responses in ECM-protected Ifnar1-/- mice and wild type mice suffering from ECM. Importantly, CD8+ T cells were increased in the spleens of ECM-protected Ifnar1-/- mice and the blood-brain-barrier remained intact. This was associated with elevated splenic levels of CCL5, a T cell and eosinophil chemotactic chemokine, which was mainly produced by eosinophils, and an increase in eosinophil numbers. Depletion of eosinophils enhanced CD8+ T cell infiltration into the brain and increased ECM induction in PbAAma1OVA-infected Ifnar1-/- mice. However, eosinophil-depletion did not reduce the CD8+ T cell population in the spleen or reduce splenic CCL5 concentrations. Our study demonstrates that eosinophils impact CD8+ T cell migration and proliferation during PbAAma1OVA-infection in Ifnar1-/- mice and thereby are contributing to the protection from ECM.


Assuntos
Encéfalo/imunologia , Eosinófilos/fisiologia , Malária Cerebral/imunologia , Parasitemia/imunologia , Plasmodium berghei , Linfócitos T/imunologia , Animais , Animais não Endogâmicos , Anopheles/parasitologia , Antígenos de Protozoários/imunologia , Movimento Celular , Quimiocina CCL5/análise , Quimiocina CCL5/fisiologia , Citotoxicidade Imunológica , Feminino , Contagem de Leucócitos , Malária Cerebral/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mosquitos Vetores/parasitologia , Organismos Geneticamente Modificados , Ovalbumina , Parasitemia/parasitologia , Fragmentos de Peptídeos , Plasmodium berghei/genética , Receptor de Interferon alfa e beta/deficiência , Receptor de Interferon alfa e beta/genética , Receptores CCR5/fisiologia , Baço/química , Baço/imunologia
4.
PLoS One ; 13(2): e0192717, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29438386

RESUMO

Malaria ranks among the most important infectious diseases worldwide and affects mostly people living in tropical countries. Mechanisms involved in disease progression are still not fully understood and specific treatments that might interfere with cerebral malaria (CM) are limited. Here we show that administration of doxycycline (DOX) prevented experimental CM (ECM) in Plasmodium berghei ANKA (PbA)-infected C57BL/6 wildtype (WT) mice in an IL-10-independent manner. DOX-treated mice showed an intact blood-brain barrier (BBB) and attenuated brain inflammation. Importantly, if WT mice were infected with a 20-fold increased parasite load, they could be still protected from ECM if they received DOX from day 4-6 post infection, despite similar parasitemia compared to control-infected mice that did not receive DOX and developed ECM. Infiltration of T cells and cytotoxic responses were reduced in brains of DOX-treated mice. Analysis of brain tissue by RNA-array revealed reduced expression of chemokines and tumour necrosis factor (TNF) in brains of DOX-treated mice. Furthermore, DOX-administration resulted in brains of the mice in reduced expression of matrix metalloproteinase 2 (MMP2) and granzyme B, which are both factors associated with ECM pathology. Systemic interferon gamma production was reduced and activated peripheral T cells accumulated in the spleen in DOX-treated mice. Our results suggest that DOX targeted inflammatory processes in the central nervous system (CNS) and prevented ECM by impaired brain access of effector T cells in addition to its anti-parasitic effect, thereby expanding the understanding of molecular events that underlie DOX-mediated therapeutic interventions.


Assuntos
Antimaláricos/farmacologia , Doxiciclina/farmacologia , Malária Cerebral/prevenção & controle , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/imunologia , Quimiocinas/metabolismo , Modelos Animais de Doenças , Feminino , Inflamação/imunologia , Inflamação/prevenção & controle , Mediadores da Inflamação/metabolismo , Interleucina-10/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Malária Cerebral/imunologia , Malária Cerebral/parasitologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Parasitemia/imunologia , Parasitemia/prevenção & controle , Plasmodium berghei/imunologia , Plasmodium berghei/patogenicidade , Baço/efeitos dos fármacos , Baço/imunologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia
5.
Z Orthop Unfall ; 155(3): 273-280, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28683495

RESUMO

Background Stimulating bone regeneration is a central aim in orthopaedic and trauma surgery. Although the replacement of bone with artificial materials like cement or apatite helps to keep up bone stability, new bone often cannot be regenerated. Increasing research efforts have led to the clinical application of growth factors stimulating bone growth (e.g. bone morphogenic protein, BMP) and inhibitors preventing bone consumption (e.g. RANKL blocking antibodies). These factors mostly concentrate on stimulating osteoblast or preventing osteoclast activity. Current Situation It is widely accepted that osteoblasts and osteoclasts are central players in bone regeneration. This concept assumes that osteoblasts are responsible for bone growth while osteoclasts cause bone consumption by secreting matrix-degrading enzymes such as cathepsin K and matrix metalloproteinases (MMP). However, according to new research results, bone growth or consumption are not regulated by single cell types. It is rather the interaction of various cell types that regulates bone metabolism. While factors secreted by osteoblasts are essential for osteoclast differentiation and activation, factors secreted by activated osteoclasts are essential for osteoblast activity. In addition, recent research results imply that the influence of the immune system on bone metabolism has long been neglected. Factors secreted by macrophages or T cells strongly influence bone growth or degradation, depending on the bone microenvironment. Infections, sterile inflammation or tumour metastases not only affect bone cells directly, but also influence immune cells such as T cells indirectly. Furthermore, immune cells and bone are mechanistically regulated by similar factors such as cytokines, chemokines and transcription factors, suggesting that the definition of bone and immune cells has to be thought over. Outlook Bone and the immune system are regulated by similar mechanisms. These newly identified similarities between bone and the immune system imply that medication developed for tumour and autoimmune patients could also be applied in bone diseases.


Assuntos
Regeneração Óssea/imunologia , Reabsorção Óssea/imunologia , Osso e Ossos/imunologia , Sistema Imunitário/fisiologia , Osteoblastos/imunologia , Osteoclastos/imunologia , Proteínas Morfogenéticas Ósseas/fisiologia , Quimiocinas/fisiologia , Citocinas/fisiologia , Humanos , Macrófagos/imunologia , Ligante RANK/fisiologia , Linfócitos T/imunologia , Fatores de Transcrição/fisiologia
6.
Nanomedicine ; 13(4): 1587-1593, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28115247

RESUMO

Bacterial biofilms cause a considerable amount of prosthetic joint infections every year, resulting in morbidity and expensive revision surgery. To address this problem, surface modifications of implant materials such as carbon nanotube (CNT) coatings have been investigated in the past years. CNTs are biologically compatible and can be utilized as drug delivery systems. In this study, multi-walled carbon nanotube (MWCNT) coated TiAl6V4 titanium alloy discs were fabricated and impregnated with Rifampicin, and tested for their ability to prevent biofilm formation over a period of ten days. Agar plate-based assays were employed to assess the antimicrobial activity of these surfaces against Staphylococcus epidermidis. It was shown that vertically aligned MWCNTs were more stable against attrition on rough surfaces than on polished TiAl6V4 surfaces. Discs with coated surfaces caused a significant inhibition of biofilm formation for up to five days. Therefore, MWCNT-modified surfaces may be effective against pathogenic biofilm formation on endoprostheses.


Assuntos
Antibacterianos/química , Biofilmes/efeitos dos fármacos , Nanotubos de Carbono/química , Staphylococcus epidermidis/efeitos dos fármacos , Titânio/química , Ligas/química , Materiais Revestidos Biocompatíveis/química , Liberação Controlada de Fármacos , Próteses e Implantes/microbiologia , Infecções Relacionadas à Prótese/tratamento farmacológico , Rifampina/química
7.
Sci Transl Med ; 8(362): 362ra146, 2016 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-27797962

RESUMO

In inflammation-associated progressive neuroinflammatory disorders, such as multiple sclerosis (MS), inflammatory infiltrates containing T helper 1 (TH1) and TH17 cells cause demyelination and neuronal degeneration. Regulatory T cells (Treg) control the activation and infiltration of autoreactive T cells into the central nervous system (CNS). In MS and experimental autoimmune encephalomyelitis (EAE) in mice, Treg function is impaired. We show that a recently approved drug, Nle4-d-Phe7-α-melanocyte-stimulating hormone (NDP-MSH), induced functional Treg, resulting in amelioration of EAE progression in mice. NDP-MSH also prevented immune cell infiltration into the CNS by restoring the integrity of the blood-brain barrier. NDP-MSH exerted long-lasting neuroprotective effects in mice with EAE and prevented excitotoxic death and reestablished action potential firing in mouse and human neurons in vitro. Neuroprotection by NDP-MSH was mediated via signaling through the melanocortin-1 and orphan nuclear 4 receptors in mouse and human neurons. NDP-MSH may be of benefit in treating neuroinflammatory diseases such as relapsing-remitting MS and related disorders.


Assuntos
Encefalomielite Autoimune Experimental/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Receptor Tipo 1 de Melanocortina/metabolismo , alfa-MSH/análogos & derivados , Potenciais de Ação , Animais , Barreira Hematoencefálica , Células da Medula Óssea/metabolismo , Proliferação de Células , Sistema Nervoso Central/imunologia , Progressão da Doença , Citometria de Fluxo , Perfilação da Expressão Gênica , Ácido Glutâmico/química , Hipocampo/metabolismo , Imageamento por Ressonância Magnética , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Ligação Proteica , Receptor Tipo 1 de Melanocortina/genética , Linfócitos T Reguladores/citologia , alfa-MSH/farmacologia
8.
Proc Natl Acad Sci U S A ; 113(38): 10649-54, 2016 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-27601670

RESUMO

The mannose receptor (MR) is an endocytic receptor involved in serum homeostasis and antigen presentation. Here, we identify the MR as a direct regulator of CD8(+) T-cell activity. We demonstrate that MR expression on dendritic cells (DCs) impaired T-cell cytotoxicity in vitro and in vivo. This regulatory effect of the MR was mediated by a direct interaction with CD45 on the T cell, inhibiting its phosphatase activity, which resulted in up-regulation of cytotoxic T-lymphocyte-associated Protein 4 (CTLA-4) and the induction of T-cell tolerance. Inhibition of CD45 prevented expression of B-cell lymphoma 6 (Bcl-6), a transcriptional inhibitor that directly bound the CTLA-4 promoter and regulated its activity. These data demonstrate that endocytic receptors expressed on DCs contribute to the regulation of T-cell functionality.


Assuntos
Antígeno CTLA-4/genética , Lectinas Tipo C/genética , Antígenos Comuns de Leucócito/genética , Ativação Linfocitária/genética , Lectinas de Ligação a Manose/genética , Receptores de Superfície Celular/genética , Animais , Apresentação de Antígeno/genética , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Antígeno CTLA-4/imunologia , Regulação da Expressão Gênica/genética , Humanos , Tolerância Imunológica/genética , Lectinas Tipo C/imunologia , Antígenos Comuns de Leucócito/imunologia , Ativação Linfocitária/imunologia , Receptor de Manose , Lectinas de Ligação a Manose/imunologia , Camundongos , Proteínas Proto-Oncogênicas c-bcl-6/genética , Receptores de Superfície Celular/imunologia , Linfócitos T Citotóxicos/imunologia , Ativação Transcricional/genética
9.
Anesthesiology ; 124(4): 923-33, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26808636

RESUMO

BACKGROUND: Critically ill patients are at high risk to suffer from sepsis, even in the absence of an initial infectious source, but the molecular mechanisms for their increased sepsis susceptibility, including a suppressed immune system, remain unclear. Although microbes and pathogen-associated molecular pattern are accepted inducers of sepsis and septic immunosuppression, the role of endogenous Toll-like receptor (TLR) ligands, such as mitochondrial DNA (mtDNA), in altering the immune response is unknown. METHODS: Mitochondrial DNA serum concentrations of the mitochondrial genes D-Loop and adenosine triphosphatase 6 were determined (quantitative polymerase chain reaction) in 165 septic patients and 50 healthy volunteers. Furthermore, cytotoxic T-cell activity was analyzed in wild-type and TLR9 knockout mice, with/without previous mtDNA administration, followed by injection of an ovalbumin-expressing adenoviral vector. RESULTS: Mitochondrial DNA serum concentrations were increased in septic patients (adenosine triphosphatase 6, 123-fold; D-Loop, 76-fold, P < 0.0001) compared with volunteers. Furthermore, a single mtDNA injection caused profound, TLR9-dependent immunosuppression of adaptive T-cell cytotoxicity in wild-type but not in TLR9 knockout mice and evoked various immunosuppressive mechanisms including the destruction of the splenic microstructure, deletion of cross-presenting dendritic cells, and up-regulation of programmed cell death ligand 1 and indoleamine 2,3-dioxygenase. Several of these findings in mice were mirrored in septic patients, and mtDNA concentrations were associated with an increased 30-day mortality. CONCLUSIONS: The findings of this study imply that mtDNA, an endogenous danger associated molecular pattern, is a hitherto unknown inducer of septic immunoparalysis and one possible link between initial inflammation and subsequent immunosuppression in critically ill patients.


Assuntos
DNA Mitocondrial/sangue , DNA Mitocondrial/imunologia , Inflamação/sangue , Inflamação/imunologia , Sepse/sangue , Sepse/imunologia , Adulto , Idoso , Animais , Estado Terminal , Modelos Animais de Doenças , Feminino , Citometria de Fluxo , Humanos , Imunidade/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Estudos Prospectivos
10.
Sci Rep ; 5: 12940, 2015 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-26260698

RESUMO

A main function of splenic red pulp macrophages is the degradation of damaged or aged erythrocytes. Here we show that these macrophages accumulate ferrimagnetic iron oxides that render them intrinsically superparamagnetic. Consequently, these cells routinely contaminate splenic cell isolates obtained with the use of MCS, a technique that has been widely used in immunological research for decades. These contaminations can profoundly alter experimental results. In mice deficient for the transcription factor SpiC, which lack red pulp macrophages, liver Kupffer cells take over the task of erythrocyte degradation and become superparamagnetic. We describe a simple additional magnetic separation step that avoids this problem and substantially improves purity of magnetic cell isolates from the spleen.


Assuntos
Separação Celular/métodos , Eritrócitos/metabolismo , Macrófagos/citologia , Baço/citologia , Animais , Contagem de Células Sanguíneas , Linhagem da Célula , Óxido Ferroso-Férrico/efeitos adversos , Óxido Ferroso-Férrico/química , Citometria de Fluxo , Humanos , Fígado/citologia , Macrófagos/metabolismo , Camundongos , Fagocitose , Baço/metabolismo
11.
PLoS One ; 9(2): e89045, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24586496

RESUMO

The preoperative differentiation between septic and aseptic loosening after total hip or knee arthroplasty is essential for successful therapy and relies in part on biomarkers. The objective of this study was to assess synovial and serum levels of inflammatory proteins as diagnostic tool for periprosthetic joint infection and compare their accuracy with standard tests. 120 patients presenting with a painful knee or hip endoprosthesis for surgical revision were included in this prospective trial. Blood samples and samples of intraoperatively acquired joint fluid aspirate were collected. White blood cell count, C-reactive protein, procalcitonin and interleukin-6 were determined. The joint aspirate was analyzed for total leukocyte count and IL-6. The definite diagnosis of PJI was determined on the basis of purulent synovial fluid, histopathology and microbiology. IL-6 in serum showed significantly higher values in the PJI group as compared to aseptic loosening and control, with specificity at 58.3% and a sensitivity of 79.5% at a cut-off value of 2.6 pg/ml. With a cut-off >6.6 pg/ml, the specificity increased to 88.3%. IL-6 in joint aspirate had, at a cut-off of >2100 pg/ml, a specificity of 85.7% and sensitivity of 59.4%. At levels >9000 pg/ml, specificity was almost at 100% with sensitivity just below 50%, so PJI could be considered proven with IL-6 levels above this threshold. Our data supports the published results on IL-6 as a biomarker in PJI. In our large prospective cohort of revision arthroplasty patients, the use of IL-6 in synovial fluid appears to be a more accurate marker than either the white blood cell count or the C-reactive protein level in serum for the detection of periprosthetic joint infection. On the basis of the results we recommend the use of the synovial fluid biomarker IL-6 for the diagnosis of periprosthetic joint infection following total hip and knee arthroplasty.


Assuntos
Biomarcadores/sangue , Interleucina-6/sangue , Instabilidade Articular/diagnóstico , Infecções Relacionadas à Prótese/diagnóstico , Líquido Sinovial/metabolismo , Artroplastia de Quadril/efeitos adversos , Artroplastia do Joelho/efeitos adversos , Proteína C-Reativa/metabolismo , Diagnóstico Diferencial , Humanos , Interleucina-6/metabolismo , Instabilidade Articular/metabolismo , Contagem de Leucócitos , Estudos Prospectivos , Infecções Relacionadas à Prótese/metabolismo , Sensibilidade e Especificidade
12.
Cancer Discov ; 4(6): 674-87, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24589924

RESUMO

UNLABELLED: Infiltration of human melanomas with cytotoxic immune cells correlates with spontaneous type I IFN activation and a favorable prognosis. Therapeutic blockade of immune-inhibitory receptors in patients with preexisting lymphocytic infiltrates prolongs survival, but new complementary strategies are needed to activate cellular antitumor immunity in immune cell-poor melanomas. Here, we show that primary melanomas in Hgf-Cdk4(R24C) mice, which imitate human immune cell-poor melanomas with a poor outcome, escape IFN-induced immune surveillance and editing. Peritumoral injections of immunostimulatory RNA initiated a cytotoxic inflammatory response in the tumor microenvironment and significantly impaired tumor growth. This critically required the coordinated induction of type I IFN responses by dendritic, myeloid, natural killer, and T cells. Importantly, antibody-mediated blockade of the IFN-induced immune-inhibitory interaction between PD-L1 and PD-1 receptors further prolonged the survival. These results highlight important interconnections between type I IFNs and immune-inhibitory receptors in melanoma pathogenesis, which serve as targets for combination immunotherapies. SIGNIFICANCE: Using a genetically engineered mouse melanoma model, we demonstrate that targeted activation of the type I IFN system with immunostimulatory RNA in combination with blockade of immune-inhibitory receptors is a rational strategy to expose immune cell-poor tumors to cellular immune surveillance.


Assuntos
Interferon Tipo I/imunologia , Melanoma Experimental/tratamento farmacológico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , 9,10-Dimetil-1,2-benzantraceno , Animais , Antígeno B7-H1/imunologia , Carcinógenos , Imunoglobulina G/administração & dosagem , Fatores Imunológicos/administração & dosagem , Melanoma Experimental/imunologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Poli I-C/administração & dosagem , Receptor de Morte Celular Programada 1/imunologia , RNA/administração & dosagem , Microambiente Tumoral/imunologia
13.
Cell Rep ; 2(3): 478-87, 2012 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-22939982

RESUMO

Viruses can escape cytotoxic T cell (CTL) immunity by avoiding presentation of viral components via endogenous MHC class I antigen presentation in infected cells. Cross-priming of viral antigens circumvents such immune escape by allowing noninfected dendritic cells to activate virus-specific CTLs, but they remain ineffective against infected cells in which immune escape is functional. Here, we show that cross-presentation of antigen released from adenovirus-infected hepatocytes by liver sinusoidal endothelial cells stimulated cross-primed effector CTLs to release tumor necrosis factor (TNF), which killed virus-infected hepatocytes through caspase activation. TNF receptor signaling specifically eliminated infected hepatocytes that showed impaired anti-apoptotic defense. Thus, CTL immune surveillance against infection relies on two similarly important but distinct effector functions that are both MHC restricted, requiring either direct antigen recognition on target cells and canonical CTL effector function or cross-presentation and a noncanonical effector function mediated by TNF.


Assuntos
Infecções por Adenoviridae/imunologia , Adenoviridae/imunologia , Apresentação de Antígeno/fisiologia , Receptores Coestimuladores e Inibidores de Linfócitos T/imunologia , Hepatócitos/imunologia , Linfócitos T Citotóxicos/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Hepatócitos/virologia , Imunidade Celular/fisiologia , Vigilância Imunológica/fisiologia , Camundongos , Receptores do Fator de Necrose Tumoral/imunologia , Transdução de Sinais/imunologia
14.
Proc Natl Acad Sci U S A ; 109(10): 3897-902, 2012 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-22355103

RESUMO

Dendritic cells (DCs) are pivotal for the development of experimental autoimmune encephalomyelitis (EAE). However, the mechanisms by which they control disease remain to be determined. This study demonstrates that expression of CC chemokine receptor 4 (CCR4) by DCs is required for EAE induction. CCR4(-/-) mice presented enhanced resistance to EAE associated with a reduction in IL-23 and GM-CSF expression in the CNS. Restoring CCR4 on myeloid cells in bone marrow chimeras or intracerebral microinjection of CCR4-competent DCs, but not macrophages, restored EAE in CCR4(-/-) mice, indicating that CCR4(+) DCs are cellular mediators of EAE development. Mechanistically, CCR4(-/-) DCs were less efficient in GM-CSF and IL-23 production and also T(H)-17 maintenance. Intraspinal IL-23 reconstitution restored EAE in CCR4(-/-) mice, whereas intracerebral inoculation using IL-23(-/-) DCs or GM-CSF(-/-) DCs failed to induce disease. Thus, CCR4-dependent GM-CSF production in DCs required for IL-23 release in these cells is a major component in the development of EAE. Our study identified a unique role for CCR4 in regulating DC function in EAE, harboring therapeutic potential for the treatment of CNS autoimmunity by targeting CCR4 on this specific cell type.


Assuntos
Células Dendríticas/citologia , Encefalomielite Autoimune Experimental/imunologia , Regulação da Expressão Gênica , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Interleucina-23/metabolismo , Receptores CCR4/fisiologia , Animais , Células da Medula Óssea/citologia , Encefalomielite Autoimune Experimental/metabolismo , Inflamação , Ligantes , Macrófagos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Biológicos , Receptores CCR4/metabolismo
15.
PLoS One ; 7(1): e30422, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22276195

RESUMO

Regulatory T cells (T(reg) cells) are increased in context of malignancies and their expansion can be correlated with higher disease burden and decreased survival. Initially, interleukin 2 (IL-2) has been used as T-cell growth factor in clinical vaccination trials. In murine models, however, a role of IL-2 in development, differentiation, homeostasis, and function of T(reg) cells was established. In IL-2 treated cancer patients a further T(reg)-cell expansion was described, yet, the mechanism of expansion is still elusive. Here we report that functional T(reg) cells of a naïve phenotype--as determined by CCR7 and CD45RA expression--are significantly expanded in colorectal cancer patients. Treatment of 15 UICC stage IV colorectal cancer patients with IL-2 in a phase I/II peptide vaccination trial further enlarges the already increased naïve T(reg)-cell pool. Higher frequencies of T-cell receptor excision circles in naïve T(reg) cells indicate IL-2 dependent thymic generation of naïve T(reg) cells as a mechanism leading to increased frequencies of T(reg) cells post IL-2 treatment in cancer patients. This finding could be confirmed in naïve murine T(reg) cells after IL-2 administration. These results point to a more complex regulation of T(reg) cells in context of IL-2 administration. Future strategies therefore might aim at combining IL-2 therapy with novel strategies to circumvent expansion and differentiation of naïve T(reg) cells.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/imunologia , Fatores de Transcrição Forkhead/metabolismo , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Interleucina-2/uso terapêutico , Linfócitos T Reguladores/efeitos dos fármacos , Adulto , Idoso , Linfócitos T CD4-Positivos/efeitos dos fármacos , Antígeno CTLA-4/metabolismo , Neoplasias Colorretais/metabolismo , Feminino , Citometria de Fluxo , Proteína Relacionada a TNFR Induzida por Glucocorticoide/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Linfócitos T Reguladores/metabolismo
16.
EMBO J ; 31(1): 201-13, 2012 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-22036947

RESUMO

Early during Gram-negative sepsis, excessive release of pro-inflammatory cytokines can cause septic shock that is often followed by a state of immune paralysis characterized by the failure to mount adaptive immunity towards secondary microbial infections. Especially, the early mechanisms responsible for such immune hypo-responsiveness are unclear. Here, we show that TLR4 is the key immune sensing receptor to initiate paralysis of T-cell immunity after bacterial sepsis. Downstream of TLR4, signalling through TRIF but not MyD88 impaired the development of specific T-cell immunity against secondary infections. We identified type I interferon (IFN) released from splenic macrophages as the critical factor causing T-cell immune paralysis. Early during sepsis, type I IFN acted selectively on dendritic cells (DCs) by impairing antigen presentation and secretion of pro-inflammatory cytokines. Our results reveal a novel immune regulatory role for type I IFN in the initiation of septic immune paralysis, which is distinct from its well-known immune stimulatory effects. Moreover, we identify potential molecular targets for therapeutic intervention to overcome impairment of T-cell immunity after sepsis.


Assuntos
Imunidade Adaptativa , Interferon Tipo I/metabolismo , Macrófagos/metabolismo , Sepse/imunologia , Baço/metabolismo , Animais , Células Dendríticas/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/metabolismo , Sepse/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/metabolismo
17.
Anticancer Res ; 31(12): 4185-90, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22199278

RESUMO

Orf virus (ORFV, Parapoxvirus ovis) possesses strong immunomodulating activity including the induction of interferon gamma (IFN-γ) and interleukin-12 (IL-12) expression. Antiviral effects have been described which appeared to be facilitated by an ORFV-induced Type 1 helper T-cell (Th1-type) immune response. In this study, we investigated the potential antitumoral activity of inactivated ORFV in transplantable tumor models. We show that parenteral administration of inactivated ORFV mediates antitumor effects in various models including the murine syngenic B16 F10 melanoma and MDA-MB-231 human breast cancer xenograft. Inhibition of natural killer (NK) and NKT cell activity through administration of an anti-mouse NK-1.1 antibody led to a reduction of ORFV-mediated antitumoral effects. However, residual antitumoral activity was observed. This observation was confirmed in MDA-MB-231 tumor-bearing NOD/LtSz-scid/j mice which not only lack functional T and B lymphocytes but, in addition, have virtually no cells positive for the NK 1.1 cell surface marker. Thus, administration of inactivated ORFV induced inhibitory effects on the growth of transplantable tumors even under conditions of severe immunosuppression.


Assuntos
Antineoplásicos/farmacologia , Vírus do Orf/metabolismo , Animais , Linfócitos B/imunologia , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Feminino , Humanos , Sistema Imunitário , Imunoterapia/métodos , Interleucina-12/metabolismo , Melanoma Experimental , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Transplante de Neoplasias , Vírus do Orf/genética , Linfócitos T/imunologia , Fatores de Tempo
18.
Nat Med ; 16(12): 1407-13, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21113155

RESUMO

Localized abdominal surgery can lead to disruption of motility in the entire gastrointestinal tract (postoperative ileus). Intestinal macrophages produce mediators that paralyze myocytes, but it is unclear how the macrophages are activated, especially those in unmanipulated intestinal areas. Here we show that intestinal surgery activates intestinal CD103(+)CD11b(+) dendritic cells (DCs) to produce interleukin-12 (IL-12). This promotes interferon-γ (IFN-γ) secretion by CCR9(+) memory T helper type 1 (T(H)1) cells which activates the macrophages. IL-12 also caused some T(H)1 cells to migrate from surgically manipulated sites through the bloodstream to unmanipulated intestinal areas where they induced ileus. Preventing T cell migration with the drug FTY720 or inhibition of IL-12, T-bet (T(H)1-specific T box transcription factor) or IFN-γ prevented postoperative ileus. CCR9(+) T(H)1 memory cells were detected in the venous blood of subjects 1 h after abdominal surgery. These findings indicate that postoperative ileus is a T(H)1 immune-mediated disease and identify potential targets for disease monitoring and therapy.


Assuntos
Células Dendríticas/metabolismo , Íleus/imunologia , Íleus/prevenção & controle , Interleucina-12/metabolismo , Complicações Pós-Operatórias/imunologia , Animais , Movimento Celular/efeitos dos fármacos , Cloridrato de Fingolimode , Citometria de Fluxo , Humanos , Íleus/etiologia , Memória Imunológica/imunologia , Imunossupressores/farmacologia , Interferon gama/imunologia , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Propilenoglicóis/farmacologia , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Estatísticas não Paramétricas , Células Th1/imunologia
19.
Gastroenterology ; 138(1): 336-46, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19737567

RESUMO

BACKGROUND & AIMS: Dendritic cell activation through ligation of pattern recognition receptors leading to full functional maturation causes induction of CD8(+) T-cell immunity through increased delivery of costimulatory signals instead of tolerance. Here we investigate whether organ-resident antigen-presenting cells, such as liver sinusoidal endothelial cells (LSECs), also switch from tolerogenic to immunogenic CD8(+) T-cell activation upon such stimulation. METHODS: Murine LSECs were isolated by immunomagnetic separation and analyzed for functional maturation upon triggering pattern recognition receptors or viral infection employing gene expression analysis and T cell coculture assays. In vivo relevance of the findings was confirmed with bone-marrow chimeric animals. RESULTS: LSECs expressed numerous pattern recognition receptors that allowed for sentinel function, but ligand-induced activation of these receptors was not sufficient to overcome tolerance induction of CD8(+) T cells. Importantly, viral infection with murine cytomegalovirus caused functional maturation of antigen-presenting LSECs and was sufficient to promote antigen-specific differentiation into effector CD8(+) T cells in the absence of dendritic cells and independent of CD80/86. CONCLUSIONS: These results shed new light on the generation of organ-specific immunity and may contribute to overcoming tolerance in relevant situations, such as cancer.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/virologia , Infecções por Herpesviridae/imunologia , Tolerância Imunológica/imunologia , Fígado/virologia , Muromegalovirus , Transferência Adotiva , Animais , Medula Óssea , Linfócitos T CD8-Positivos/citologia , Diferenciação Celular/imunologia , Células Cultivadas , Quimera , Células Endoteliais/citologia , Células Endoteliais/virologia , Ligantes , Fígado/citologia , Fígado/imunologia , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Especificidade de Órgãos , Receptores de Reconhecimento de Padrão/genética , Receptores de Reconhecimento de Padrão/imunologia , Receptores de Reconhecimento de Padrão/metabolismo
20.
Proc Natl Acad Sci U S A ; 107(1): 216-21, 2010 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-20018690

RESUMO

The spleen is the lymphoid organ that induces immune responses toward blood-borne pathogens. Specialized macrophages in the splenic marginal zone are strategically positioned to phagocytose pathogens and cell debris, but are not known to play a role in the activation of T-cell responses. Here we demonstrate that splenic marginal metallophilic macrophages (MMM) are essential for cross-presentation of blood-borne antigens by splenic dendritic cells (DCs). Our data demonstrate that antigens targeted to MMM as well as blood-borne adenoviruses are efficiently captured by MMM and exclusively transferred to splenic CD8(+) DCs for cross-presentation and for the activation of cytotoxic T lymphocytes. Depletion of macrophages in the marginal zone prevents cytotoxic T-lymphocyte activation by CD8(+) DCs after antibody targeting or adenovirus infection. Moreover, we show that tumor antigen targeting to MMM is very effective as antitumor immunotherapy. Our studies point to an important role for splenic MMM in the initial steps of CD8(+) T-cell immunity by capturing and concentrating blood-borne antigens and the transfer to cross-presenting DCs which can be used to design vaccination strategies to induce antitumor cytotoxic T-cell immunity.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Macrófagos/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Apresentação de Antígeno/imunologia , Conservadores da Densidade Óssea/farmacologia , Ácido Clodrônico/farmacologia , Ativação Linfocitária/imunologia , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia , Baço/citologia , Baço/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA