Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 382
Filtrar
1.
Br J Dermatol ; 185(1): 164-176, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33400270

RESUMO

BACKGROUND: Hidradenitis suppurativa (HS) is a chronic inflammatory disease, characterized by painful, purulent and destructive skin alterations in intertriginous areas. OBJECTIVES: We investigated the expression and role in HS of granulocyte colony-stimulating factor (G-CSF), the regulator of neutrophil biology, as clinical signs of a neutrophilic granulocyte-driven inflammation are distinctive in the disease. METHODS: Skin and blood samples obtained from different cohorts of patients with HS and control individuals were assessed by RNA sequencing, quantitative polymerase chain reaction on reverse transcribed mRNA, and/or enzyme-linked immunosorbent assay. Mechanistic studies using keratinocytes, dermal fibroblasts, immune cell populations and skin biopsies were performed. RESULTS: G-CSF was abundant in HS skin, particularly in inflamed nodules and abscesses. Its levels even exceeded those found in other inflammatory skin diseases. Interleukin (IL)-1 and IL-17, respectively, induced G-CSF production by fibroblasts and keratinocytes. These effects were enhanced by tumour necrosis factor (TNF)-α and IL-36. Accordingly, fibroblasts separated from HS lesions expressed G-CSF, and IL-1 receptor antagonist reduced G-CSF levels in explanted HS skin. G-CSF blood levels positively correlated with severity of HS. Elevated lesional G-CSF receptor levels were linked to upregulation of molecules that contribute to prolonged activation of neutrophils by components of bacteria and damaged host cells [formyl peptide receptor 1 (FPR1), FPR2 and free fatty acid receptor 2 (FFAR2)], neutrophil survival [TNF receptor superfamily member 10C (TNFRSF10C/TRAIL-R3) and TNF receptor superfamily member 6B], kinases (tyrosine-protein kinase HCK and hexokinase 3), and skin destruction [MMP25 (matrix metalloproteinase 25) and ADAM8 (disintegrin and metalloproteinase domain-containing protein 8)]. G-CSF elevated the expression of FPR1, FFAR2, and TNFRSF10C/TRAIL-R3 in neutrophils and synergized with bacterial components to induce skin-destructive enzymes. CONCLUSIONS: The G-CSF pathway engages both tissue and immune cells, is strongly activated in HS lesions, and offers the opportunity to target the neutrophil-driven inflammation.


Assuntos
Hidradenite Supurativa , Proteínas ADAM , Fator Estimulador de Colônias de Granulócitos , Humanos , Queratinócitos , Proteínas de Membrana , Neutrófilos , Pele , Fator de Necrose Tumoral alfa
2.
Br J Dermatol ; 177(5): 1385-1393, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28256718

RESUMO

BACKGROUND: Acne inversa (AI)/hidradenitis suppurativa is a chronic inflammatory disease characterized by painful axillary, inguinal and perianal skin lesions with deep-seated nodules, abscesses and fistulae. OBJECTIVES: This study aimed to identify and characterize the key players in AI pathogenesis. METHODS: Epidemiological and anamnestic data for patients with AI were collected, and blood and skin samples were also taken. Healthy participants and patients with psoriasis served as controls. Assessment of samples and cultures of primary cells was performed by enzyme-linked immunosorbent assay, quantitative polymerase chain reaction on reverse transcribed mRNA, and immunohistochemistry. RESULTS: Of 35 mediators quantified in the blood of patients with AI, lipocalin-2 (LCN2) appeared as one of the most significantly upregulated parameters compared with healthy participants [85·8 ± 12·2 (n = 18) vs. 41·8 ± 4·2 (n = 15); P < 0·001]. Strongly elevated LCN2 expression was present in AI lesions, with granulocytes and keratinocytes being sources of this expression. In vitro, these cells upregulated LCN2 production in response to tumour necrosis factor (TNF)-α, and a positive relationship between systemic TNF-α and LCN2 levels (rs = 0·55, P = 0·011; n = 20) was evident for AI. LCN2 blood levels correlated with AI disease severity (rs = 0·65, P < 0·001; n = 29), but not with disease duration, age, sex, body mass index or smoking habit. Detailed analyses revealed a link with the number of skin regions containing nodules and fistulae, but not scars. CONCLUSIONS: LCN2 might serve as a blood biomarker for the objective assessment of inflammatory activity in AI. We suggest a self-amplification loop comprising TNF-α, neutrophilic granulocytes and LCN2, which contributes to the recurrent skin neutrophil infiltration in AI, clinically evident as pus.


Assuntos
Granulócitos/metabolismo , Hidradenite Supurativa/etiologia , Queratinócitos/metabolismo , Lipocalina-2/metabolismo , Adulto , Biomarcadores/metabolismo , Células Cultivadas , Feminino , Hidradenite Supurativa/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Infiltração de Neutrófilos/fisiologia , Pele/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Regulação para Cima/fisiologia
3.
Stem Cells ; 33(10): 3087-99, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26184374

RESUMO

Although the immunomodulatory potency of mesenchymal stromal cells (MSC) is well established, the mechanisms behind are still not clear. The crosstalk between myeloid dendritic cells (mDC) and natural killer (NK) cells and especially NK cell-derived interferon-gamma (IFN-γ) play a pivotal role in the development of type 1 helper (Th1) cell immune responses. While many studies explored the isolated impact of MSC on either in vitro generated DC, NK, or T cells, there are only few data available on the complex interplay between these cells. Here, we investigated the impact of MSC on the functionality of human mDC and the consequences for NK cell and Th1 priming in vitro and in vivo. In critical limb ischemia patients, who have been treated with allogeneic placenta-derived mesenchymal-like stromal cells (PLX-PAD), no in vivo priming of Th1 responses toward the major histocompatibility complex (MHC) mismatches could be detected. Further in vitro studies revealed that mDC reprogramming could play a central role for these effects. Following crosstalk with MSC, activated mDC acquired a tolerogenic phenotype characterized by reduced migration toward CCR7 ligand and impaired ability to stimulate NK cell-derived IFN-γ production. These effects, which were strongly related to an altered interleukin (IL)-12/IL-10 production by mDC, were accompanied by an effective prevention of Th1 priming in vivo. Our findings provide novel evidence for the regulation of Th1 priming by MSC via modulation of mDC and NK cell crosstalk and show that off-the-shelf produced MHC-mismatched PLX-PAD can be used in patients without any sign of immunogenicity.


Assuntos
Células Dendríticas/imunologia , Imunidade Celular , Células Matadoras Naturais/imunologia , Células-Tronco Mesenquimais/imunologia , Células Th1/imunologia , Comunicação Celular/imunologia , Diferenciação Celular/imunologia , Técnicas de Cocultura , Células Dendríticas/metabolismo , Feminino , Humanos , Imunomodulação , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-10/imunologia , Interleucina-10/metabolismo , Células Matadoras Naturais/metabolismo , Linfonodos/imunologia , Linfonodos/metabolismo , Ativação Linfocitária/imunologia , Células-Tronco Mesenquimais/metabolismo , Células Mieloides/imunologia , Células Mieloides/metabolismo , Placenta/citologia , Placenta/metabolismo , Gravidez , Células Th1/metabolismo
4.
Genes Immun ; 16(1): 8-14, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25472783

RESUMO

Interleukin (IL)-10 is an important immunoregulatory cytokine that mediates its effects via a transmembrane receptor complex consisting of two different chains, IL-10R1 and IL-10R2. While IL-10R2 is ubiquitously expressed and does not bind IL-10 primarily, the expression of IL-10R1 determines cellular responsiveness. However, the current knowledge about the expression and regulation of IL-10R1 is still limited. Here we analyzed the expression of IL-10R1 on monocytic cells and demonstrated that human blood monocytes carried about 720 IL-10-binding sites on their surface. Compared with lymphocytes and various tissue cells and tissues, blood monocytes expressed the highest IL-10R1 levels. The in vitro differentiation of these cells into macrophages provoked a further increase of IL-10R1 surface expression. In contrast, their differentiation into myeloid dendritic cells (mDCs) resulted in reduced surface IL-10R1 levels. The different IL-10R1 levels expressed by monocyte-derived antigen-presenting cell populations were reflected in their different responsiveness toward IL-10. Importantly, also in vivo developed immature macrophages and mDCs showed different IL-10 sensitivity. These data suggest that, compared with monocytes and macrophages, mDCs partially escape from IL-10's inhibitory mechanisms by downregulating IL-10R1.


Assuntos
Subunidade alfa de Receptor de Interleucina-10/imunologia , Interleucina-10/imunologia , Células Dendríticas/imunologia , Fibroblastos/metabolismo , Expressão Gênica , Humanos , Subunidade alfa de Receptor de Interleucina-10/genética , Queratinócitos/metabolismo , Leucócitos Mononucleares/imunologia
5.
Am J Transplant ; 15(10): 2625-35, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25988290

RESUMO

Adoptive immunotherapy with regulatory T cells (Treg) is a new option to promote immune tolerance following solid organ transplantation (SOT). However, Treg from elderly patients awaiting transplantation are dominated by the CD45RA(-) CD62L(+) central memory type Treg subset (TregCM), and the yield of well-characterized and stable naïve Treg (TregN) is low. It is, therefore, important to determine whether these TregCM are derived from the thymus and express high stability, suppressive capacity and a broad antigen repertoire like TregN. In this study, we showed that TregCM use a different T cell receptor (TCR) repertoire from conventional T cells (Tconv), using next-generation sequencing of all 24 Vß families, with an average depth of 534 677 sequences. This showed almost no contamination with induced Treg. Furthermore, TregCM showed enhanced suppressive activity on Tconv at early checkpoints of immune activation controlling activation markers expression and cytokine secretion, but comparable inhibition of proliferation. Following in vitro expansion under mTOR inhibition, TregCM expanded equally as well as TregN without losing their function. Despite relatively limited TCR repertoire, TregCM also showed specific alloresponse, although slightly reduced compared to TregN. These results support the therapeutic usefulness of manufacturing Treg products from CD45RA(-) CD62L(+) Treg-enriched starting material to be applied for adoptive Treg therapy.


Assuntos
Linfócitos T Reguladores/imunologia , Adulto , Fatores Etários , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/metabolismo , Citocinas/metabolismo , Citometria de Fluxo , Fatores de Transcrição Forkhead/metabolismo , Voluntários Saudáveis , Humanos , Transplante de Rim , Antígenos Comuns de Leucócito/metabolismo , Pessoa de Meia-Idade , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T Reguladores/metabolismo , Timo/citologia , Timo/imunologia
6.
Am J Transplant ; 14(3): 594-606, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24467477

RESUMO

The adoptive transfer of natural regulatory T cells (nTreg) is a new option to reshape undesired immune reactivity in autoimmunity and transplantation toward "tolerance." The first clinical trials using adoptive transfer of polyclonal nTreg demonstrated safety and hints of efficacy. However, the low frequencies of antigen-specific cells among the pool of polyclonal nTreg and their broad antigen nonspecific suppression are limitations of this approach regarding efficacy and safety. Recently, the isolation and expansion of (allo)antigen-specific nTreg have successfully been achieved by using Treg-specific activation markers but the yield is relatively low. Here, we describe a novel good manufacturing practice (GMP)-compatible expansion protocol of alloantigen-specific nTreg based on the stimulation of nTreg by allogeneic activated B cells. Their functionality and specificity are superior compared to polyclonal nTreg both in vitro and in vivo. Employing an allogeneic B cell bank, designed to cover the majority of HLA types, allows fast GMP-compliant manufacturing for donor-specific nTreg for clinical application in organ and stem cell transplantation. TCR repertoire analyses by next generation sequencing revealed impressive expansion by several log-steps of even very low-abundance alloantigen-specific nTreg clones. This novel method offers a simple approach for expanding antigen-specific nTreg and is characterized by high replicability and easy transferability to full GMP standards.


Assuntos
Linfócitos B/imunologia , Protocolos Clínicos/normas , Rejeição de Enxerto/imunologia , Tolerância Imunológica/imunologia , Isoantígenos/imunologia , Transplante de Pele , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Linfócitos B/citologia , Células Cultivadas , Proteínas de Ligação a DNA/fisiologia , Teste de Histocompatibilidade , Humanos , Terapia de Imunossupressão , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T Reguladores/citologia
7.
Am J Transplant ; 13(11): 2842-54, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24020931

RESUMO

Clonotype analysis is essential for complete characterization of antigen-specific T cells. Moreover, knowledge on clonal identity allows tracking of antigen-specific T cells in whole blood and tissue infiltrates and can provide information on antigenic specificity. Here, we developed a next generation sequencing (NGS)-based platform for the highly quantitative clonotype characterization of T cells and determined requirements for the unbiased characterization of the input material (DNA, RNA, ex vivo derived or cell culture expanded T cells). Thereafter we performed T cell receptor (TCR) repertoire analysis of various specimens in clinical settings including cytomegalovirus (CMV), polyomavirus BK (BKV) reactivation and acute cellular allograft rejection. Our results revealed dynamic nature of virus-specific T cell clonotypes; CMV reactivation was linked to appearance of new highly abundant antigen-specific clonalities. Moreover, analysis of clonotype overlap between BKV-, alloantigen-specific T cell-, kidney allograft- and urine-derived lymphocytes provided hints for the differential diagnosis of allograft dysfunction and enabled appropriate therapy adjustment. We believe that the established approach will provide insights into the regulation of virus-specific/anti-tumor immunity and has high diagnostic potential in the clinical routine.


Assuntos
Infecções por Citomegalovirus/diagnóstico , Citomegalovirus/imunologia , Rejeição de Enxerto/genética , Sequenciamento de Nucleotídeos em Larga Escala , Infecções por Polyomavirus/diagnóstico , Receptores de Antígenos de Linfócitos T/genética , Linfócitos T/patologia , Infecções Tumorais por Vírus/diagnóstico , Vírus BK/genética , Citomegalovirus/patogenicidade , Infecções por Citomegalovirus/genética , Infecções por Citomegalovirus/virologia , Diagnóstico Diferencial , Humanos , Transplante de Rim/efeitos adversos , Infecções por Polyomavirus/genética , Infecções por Polyomavirus/virologia , Estudos Retrospectivos , Linfócitos T/imunologia , Linfócitos T/virologia , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/virologia , Ativação Viral
8.
Am J Transplant ; 13(7): 1880-90, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23763435

RESUMO

Assessment of donor-specific alloreactive memory/effector T cell responses using an IFN-γ Elispot assay has been suggested to be a novel immune-monitoring tool for evaluating the cellular immune risk in renal transplantation. Here, we report the cross-validation data of the IFN-γ Elispot assay performed within different European laboratories taking part of the EU RISET consortium. For this purpose, development of a standard operating procedure (SOP), comparisons of lectures of IFN-γ plates assessing intra- and interlaboratory assay variability of allogeneic or peptide stimuli in both healthy and kidney transplant individuals have been the main objectives. We show that the use of a same SOP and count-settings of the Elispot bioreader allow low coefficient variation between laboratories. Frozen and shipped samples display slightly lower detectable IFN-γ frequencies than fresh samples. Importantly, a close correlation between different laboratories is obtained when measuring high frequencies of antigen-specific primed/memory T cell alloresponses. Interestingly, significant high donor-specific alloreactive T cell responses can be similarly detected among different laboratories in kidney transplant patients displaying histological patterns of acute T cell mediated rejection. In conclusion, assessment of circulating alloreactive memory/effector T cells using an INF-γ Elispot assay can be accurately achieved using the same SOP, Elispot bioreader and experienced technicians in kidney transplantation.


Assuntos
ELISPOT/métodos , Rejeição de Enxerto/imunologia , Imunidade Celular/imunologia , Memória Imunológica , Interferon gama/imunologia , Transplante de Rim/imunologia , Ensaio de Imunoadsorção Enzimática/métodos , Humanos , Linfócitos T/imunologia
9.
Am J Transplant ; 13(9): 2308-21, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23855618

RESUMO

To ensure safety tolerance induction protocols are accompanied by conventional immunosuppressive drugs (IS). But IS such as calcineurin inhibitors (CNI), for example, cyclosporin A (CsA), can interfere with tolerance induction. We investigated the effect of an additional transient CsA treatment on anti-CD4mAb-induced tolerance induction upon rat kidney transplantation. Additional CsA treatment induced deteriorated graft function, resulting in chronic rejection characterized by glomerulosclerosis, interstitial fibrosis, tubular atrophy and vascular changes. Microarray analysis revealed enhanced intragraft expression of the B cell attracting chemokine CXCL13 early during CsA treatment. Increase in CXCL13 expression is accompanied by enhanced B cell infiltration with local and systemic IgG production and C3d deposition as early as 5 days upon CsA withdrawal. Adding different CNIs to cultures of primary mesangial cells isolated from glomeruli resulted in a concentration-dependent increase in CXCL13 transcription. CsA in synergy with TNF-α can enhance the B cell attracting and activating potential of mesangial cells. Transient B cell depletion or transfer of splenocytes from tolerant recipients 3 weeks after transplantation could rescue tolerance induction and did inhibit intragraft B cell accumulation, alloantibody production and ameliorate chronic rejection.


Assuntos
Anticorpos Monoclonais/farmacologia , Linfócitos T CD4-Positivos/imunologia , Inibidores de Calcineurina , Tolerância Imunológica/imunologia , Imunossupressores/farmacologia , Transplante de Rim , Animais , Linfócitos B/imunologia , Calcineurina/farmacologia , Quimiocina CXCL13/biossíntese , Ciclosporina/farmacologia , Rejeição de Enxerto/tratamento farmacológico , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Humanos , Tolerância Imunológica/efeitos dos fármacos , Rim/metabolismo , Ativação Linfocitária , Masculino , Ratos , Ratos Endogâmicos Lew
10.
Eur J Clin Microbiol Infect Dis ; 32(4): 451-60, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23179251

RESUMO

Viruses can manipulate the immune response against them by various strategies to influence immune cells, i.e. by over-activation leading to functional inactivation, bypassing antigen presentation or even suppression of effector functions. Little is known, however, about how these features of immune regulation and modulation could be used for therapeutic purposes. Reasons for this include the complexity of immune regulatory mechanisms under certain disease conditions and the risks that infections with viruses pose to human beings. The orf virus (ORFV), a member of the Parapoxvirus genus of the poxvirus family, is known as a common pathogen in sheep and goats worldwide. The inactivated ORFV, however, has been used as a preventative as well as therapeutic immunomodulator in veterinary medicine in different species. Here, we review the key results obtained in pre-clinical studies or clinical studies in veterinary medicine to characterise the therapeutic potential of inactivated ORFV. Inactivated ORFV has strong effects on cytokine secretion in mice and human immune cells, leading to an auto-regulated loop of initial up-regulation of inflammatory and Th1-related cytokines, followed by Th2-related cytokines that attenuate immunopathology. The therapeutic potential of inactivated ORFV has been recognised in several difficult-to-treat disease areas, such as chronic viral diseases, liver fibrosis or various forms of cancer. Further research will be required in order to evaluate the full beneficial potential of inactivated ORFV for therapeutic immunomodulation.


Assuntos
Fatores Imunológicos/administração & dosagem , Imunomodulação , Imunoterapia/métodos , Vírus do Orf/imunologia , Medicina Veterinária/métodos , Animais , Ensaios Clínicos como Assunto , Avaliação Pré-Clínica de Medicamentos , Cabras , Camundongos , Ovinos
11.
Am J Transplant ; 12(6): 1469-78, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22390272

RESUMO

Quantification of the humoral alloimmune response is generally achieved by measuring serum HLA antibodies, which provides no information about the cells involved in the humoral immune response. Therefore, we have developed an HLA-specific B-cell ELISPOT assay allowing for quantification of B cells producing HLA antibodies. We used recombinant HLA monomers as target in the ELISPOT assay. Validation was performed with human B-cell hybridomas producing HLA antibodies. Subsequently, we quantified B cells producing HLA antibodies in HLA-immunized individuals, non-HLA-immunized individuals and transplant patients with serum HLA antibodies. B-cell hybridomas exclusively formed spots against HLA molecules of corresponding specificity with the sensitivity similar to that found in total IgG ELISPOT assays. HLA-immunized healthy individuals showed up to 182 HLA-specific B cells per million total B cells while nonimmunized individuals had none. Patients who were immunized by an HLA-A2-mismatched graft had up to 143 HLA-A2-specific B cells per million total B cells. In conclusion, we have developed and validated a highly specific and sensitive HLA-specific B-cell ELISPOT assay, which needs further validation in a larger series of transplant patients. This technique constitutes a new tool for quantifying humoral immune responses.


Assuntos
Linfócitos B/imunologia , Ensaio de Imunoadsorção Enzimática/métodos , Antígenos HLA/imunologia , Formação de Anticorpos , Humanos , Limite de Detecção
12.
Am J Transplant ; 12(3): 669-81, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22081907

RESUMO

Cytomegalovirus (CMV) infections have a major impact on morbidity and mortality of transplant patients. Among the complex antiviral T-cell response, CMV-IE-1 antigen-specific CD8+ cells are crucial for preventing CMV disease but do not protect from recurring/lasting CMV reactivation. Recently, we confirmed that adoptive transfer of autologous IE-1/pp65-specific T-cell lines was able to combat severe CMV disease; however, the control of CMV infection was only temporary. We hypothesized that CMV-induced regulatory T cells (iTreg) might be related to recurring/lasting CMV infection. In fact, kidney transplant patients with recurring CMV infections expressed enhanced suppression on CMV response. Analysis of in vitro expanded CD4+ epitope-specific cells revealed that CMV-specific CD4+CD25(high) Treg cells functionally suppress CD25(low) effector T cells (Teff) upon epitope-specific reactivation. Their phenotype is similar to iTreg - CD39(high) /Helios-/IL-2(low) /IFNγ(high) /IL-10±/TGFß-LAP±/FOXP3+ and methylated foxp3 locus. Remarkably, in vitro expanded CD4+CD25(high) iTreg share the same dominant TCR-Vß-CDR3 clones with functionally distinct CD4+CD25(low) Teff. Moreover, the same clones were present in freshly isolated CD4+CD25(high) and CD4+CD25(low) T cells suggesting their in vivo generation. These findings directly demonstrate that Teff and iTreg can differentiate from one "mother" clone with specificity to the same viral epitope and indicate that peripheral iTreg generation is related to frequent antigen appearance.


Assuntos
Antígenos Virais/imunologia , Infecções por Citomegalovirus/imunologia , Citomegalovirus/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T/imunologia , Proliferação de Células , Citocinas/metabolismo , Infecções por Citomegalovirus/microbiologia , Citometria de Fluxo , Humanos , Receptores de Antígenos de Linfócitos T/imunologia , Recidiva
13.
Am J Transplant ; 12(11): 2909-19, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22882762

RESUMO

Despite remarkable progress in organ transplantation through the development of a wealth of immunosuppressive drugs highly effective at controlling acute rejection, two major problems still remain, the loss of transplants due to chronic rejection and the growing number of sensitized recipients due to previous transplants, transfusions or pregnancies. Induction of immune tolerance appears to be the only way to curb this complex situation. Here we describe that a therapy, already successfully used to restore immune tolerance to self-antigens in overt autoimmunity, is effective at promoting transplant tolerance. We demonstrate that a short low-dose course with CD3 antibodies started after transplantation, at the time of effector T cell priming to alloantigens, induces permanent acceptance of fully mismatched islet allografts. Mechanistic studies revealed that antigen-specific regulatory and effector T cells are differentially affected by the treatment. CD3 antibody treatment preferentially induces apoptosis of activated alloreactive T cells which is mandatory for tolerance induction. In contrast, regulatory T cells are relatively spared from CD3 antibody-induced depletion and can transfer antigen-specific tolerance thus arguing for their prominent role in sustaining long-term graft survival.


Assuntos
Anticorpos Monoclonais/farmacologia , Complexo CD3/farmacologia , Tolerância Imunológica/imunologia , Ilhotas Pancreáticas/imunologia , Tolerância ao Transplante/imunologia , Animais , Anticorpos Monoclonais/imunologia , Complexo CD3/imunologia , Transplante de Células/métodos , Modelos Animais de Doenças , Rejeição de Enxerto , Sobrevivência de Enxerto , Tolerância Imunológica/efeitos dos fármacos , Isoantígenos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Distribuição Aleatória , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Risco , Sensibilidade e Especificidade , Fatores de Tempo , Imunologia de Transplantes/fisiologia , Tolerância ao Transplante/fisiologia
14.
Am J Transplant ; 12(9): 2384-94, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22702307

RESUMO

Recent data suggest that donor-specific memory T cells (T(mem)) are an independent risk factor for rejection and poor graft function in patients and a major challenge for immunosuppression minimizing strategies. Many tolerance induction protocols successfully proven in small animal models e.g. costimulatory blockade, T cell depletion failed in patients. Consequently, there is a need for more predictive transplant models to evaluate novel promising strategies, such as adoptive transfer of regulatory T cells (Treg). We established a clinically more relevant, life-supporting rat kidney transplant model using a high responder (DA to LEW) recipients that received donor-specific CD4(+)/ 8(+) GFP(+) T(mem) before transplantation to achieve similar pre-transplant frequencies of donor-specific T(mem) as seen in many patients. T cell depletion alone induced long-term graft survival in naïve recipients but could not prevent acute rejection in T(mem)(+) rats, like in patients. Only if T cell depletion was combined with permanent CNI-treatment, the intragraft inflammation, and acute/chronic allograft rejection could be controlled long-term. Remarkably, combining 10 days CNI treatment and adoptive transfer of Tregs (day 3) but not Treg alone also induced long-term graft survival and an intragraft tolerance profile (e.g. high TOAG-1) in T(mem)(+) rats. Our model allows evaluation of novel therapies under clinically relevant conditions.


Assuntos
Inibidores de Calcineurina , Rejeição de Enxerto , Imunossupressores/farmacologia , Transplante de Rim , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Citometria de Fluxo , Memória Imunológica , Depleção Linfocítica , Ratos , Ratos Endogâmicos Lew , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Nat Cell Biol ; 1(2): 94-7, 1999 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10559880

RESUMO

MAPKAP kinase 2 (MK2) is one of several kinases that are regulated through direct phosphorylation by p38 MAP kinase. By introducing a targeted mutation into the mouse MK2 gene, we have determined the physiological function of MK2 in vivo. Mice that lack MK2 show increased stress resistance and survive LPS-induced endotoxic shock. This is due to a reduction of approximately 90% in the production of tumor necrosis factor-alpha (TNF-alpha) and not to a change in signalling from the TNF receptor. The level and stability of TNF-alpha mRNA is not reduced and TNF-alpha secretion is not affected. We conclude that MK2 is an essential component in the inflammatory response which regulates biosynthesis of TNF-alpha at a post-transcriptional level.


Assuntos
Citocinas/genética , Regulação da Expressão Gênica/fisiologia , Lipopolissacarídeos/farmacologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Fator de Necrose Tumoral alfa/genética , Animais , Citocinas/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamação/genética , Inflamação/imunologia , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutagênese , Fosforilação , Proteínas Serina-Treonina Quinases/deficiência , RNA Mensageiro/metabolismo , Mapeamento por Restrição , Salmonella typhi , Baço/imunologia , Transcrição Gênica , Fator de Necrose Tumoral alfa/biossíntese , Proteínas Quinases p38 Ativadas por Mitógeno
16.
Nat Med ; 3(6): 678-81, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9176497

RESUMO

Neutralization of proinflammatory cytokines, such as tumor necrosis factor-alpha (TNF-alpha) or interleukin-1 (IL-1), decreases mortality in several animal models of sepsis. However, recent clinical trials did not show an unequivocal improvement in survival. In contrast to animals, which succumb to shock during the first 72 hours, we found that many patients die much later with signs of opportunistic infections accompanied by downregulation of their monocytic HLA-DR expression and reduced ability to produce lipopolysaccharide (LPS)-induced TNF-alpha in vitro. This phenomenon of monocyte deactivation in septic patients with fatal outcome shows similarities to experimental monocytic refractoriness induced by LPS desensitization or by pretreatment with its endogenous mediators IL-10 and transforming growth factor-beta (TGF-beta). In order to strengthen their antimicrobial defense, here we tested whether interferon-gamma (IFN-gamma) can improve monocytic functions in these patients and in experimental monocytic deactivation. The considerably lowered in vitro levels of LPS-induced TNF-alpha in these situations were significantly enhanced by IFN-gamma, but did not reach the extremely high levels of IFN-gamma primed naive cells from healthy donors. Moreover, IFN-gamma applied to septic patients with low monocytic HLA-DR expression restored the deficient HLA-DR expression and in vitro LPS-induced TNF-alpha secretion. Recovery of monocyte function resulted in clearance of sepsis in eight of nine patients. These data suggest that IFN-gamma treatment in carefully selected septic patients is a novel therapeutic strategy worth pursuing.


Assuntos
Interferon gama/uso terapêutico , Monócitos/imunologia , Sepse/tratamento farmacológico , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Relação Dose-Resposta a Droga , Antígenos HLA-DR/sangue , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Lipopolissacarídeos/farmacologia , Pessoa de Meia-Idade , Monócitos/efeitos dos fármacos , Sepse/imunologia , Fator de Necrose Tumoral alfa/metabolismo
17.
Nat Med ; 4(7): 808-13, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9662372

RESUMO

The mechanism of immunodepression after brain injury is not yet clear. Here we demonstrate rapid systemic release of the immunoinhibitory cytokine interleukin-10, monocytic deactivation and a high incidence of infection in patients with 'sympathetic storm' due to acute accidental or iatrogenic brain trauma. In vitro studies showed that within minutes catecholamines trigger the secretion of interleukin-10 from unstimulated monocytes through a beta-adrenoreceptor-mediated, cAMP/protein kinase A-dependent pathway. We found that in a rat model of acute brain injury, the beta-receptor antagonist propranolol prevented the increase of interleukin-10 plasma levels. Rapid monocytic interleukin-10 release after sympathetic activation may represent a common pathway for immunodepression induced by stress and injury.


Assuntos
Lesões Encefálicas/sangue , Tolerância Imunológica , Interleucina-10/sangue , Sistema Nervoso Simpático/fisiopatologia , Antagonistas Adrenérgicos beta/farmacologia , Adulto , Idoso , Animais , Encéfalo/cirurgia , Lesões Encefálicas/complicações , Lesões Encefálicas/fisiopatologia , Neoplasias Encefálicas/sangue , Neoplasias Encefálicas/cirurgia , Tronco Encefálico/fisiopatologia , Catecolaminas/farmacologia , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias de Tecido Nervoso/sangue , Neoplasias de Tecido Nervoso/cirurgia , Propranolol/farmacologia , Ratos , Ratos Sprague-Dawley , Sistema Nervoso Simpático/efeitos dos fármacos , Simpatolíticos/farmacologia , Simpatomiméticos/farmacologia
19.
J Exp Med ; 181(5): 1887-92, 1995 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-7722463

RESUMO

Tolerance of monocytes/macrophages to endotoxin (lipopolysaccharide [LPS]) can be induced both in vivo and in vitro by LPS itself. Exposure to LPS, even at a very low dose, induces a downregulation of cytokine response to a second high dose LPS challenge. To learn more about the unknown mechanisms of this phenomenon, we studied the role of antiinflammatory cytokines in this process. Preculture of human peripheral blood monocytes for 24 hours with low concentrations of LPS induced hyporesponsiveness to high-dose LPS rechallenge with respect to tumor necrosis factor (TNF) alpha and interleukin (IL) 10 but not IL-1RA production. These results suggest that LPS tolerance reflects a functional switch of monocytes rather than a general LPS hyporesponsiveness. IL-10 and transforming growth factor (TGF) beta 1 showed additive effects in replacing LPS for induction of LPS hyporesponsiveness in vitro. Additionally, neutralizing anti-IL-10 and anti-TGF-beta monoclonal antibodies prevented induction of LPS tolerance. In vitro induced LPS tolerance looks like the ex vivo LPS hyporesponsiveness of monocytes from septic patients with fatal outcome: downregulation of LPS-induced TNF-alpha and IL-10 production but not of IL-1RA secretion. LPS hyporesponsiveness in septic patients was preceded by expression of IL-10 at both the mRNA and protein level. In summary, our data suggests that IL-10 and TGF-beta mediate the phenomenon of LPS tolerance in vitro and perhaps in vivo (septic patients), too.


Assuntos
Interleucina-10/fisiologia , Lipopolissacarídeos/toxicidade , Fator de Crescimento Transformador beta/fisiologia , Células Cultivadas , Antígenos HLA-DR/análise , Humanos , Proteína Antagonista do Receptor de Interleucina 1 , Sialoglicoproteínas/biossíntese , Fator de Necrose Tumoral alfa/biossíntese
20.
J Exp Med ; 173(5): 1047-52, 1991 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-2022919

RESUMO

The tumor necrosis factor alpha (TNF-alpha) gene was introduced by retroviral gene transfer into the TNF-alpha-insensitive tumor cell line J558L. Production of 40 pg/ml TNF-alpha by clone J2T12 consistently did not change the growth rate in vitro, but drastically suppressed tumor growth when injected into syngeneic BALB/c mice. Within 2 wk, 90% of the mice inoculated with J558L cells developed a tumor, but none of the mice injected with J2T12 did so. Within the observation period (greater than 3 mo), 60% of the mice inoculated with J2T12 did not develop a tumor. In the other 40% of the mice, tumor manifestation was significantly delayed. Mice injected simultaneously with J2T12 cells and an anti-TNF-alpha monoclonal antibody developed tumors similar to parental J558L cells. Similarly, the tumor-suppressive effects of TNF-alpha were abolished, e.g., by injection of an anti-type 3 complement receptor (CR3) monoclonal antibody that is known to prevent migration of inflammatory cells. These results and the observation of tumor-infiltrating macrophages suggest that lack of tumorigenicity of J2T12 cells is due to the TNF-alpha secretion by the tumor cells and that TNF-alpha acts indirectly by a mechanism that involves chemotactic recruitment and activation of cells, predominantly of macrophages. In contrast, the tumor growth was not affected when, instead of TNF-alpha, interleukin 6 was expressed by J558L cells. Together, our results support the concept of tumor cell-targeted cytokine gene transfer as a tool for cancer treatment, and particularly demonstrate that extremely low doses of TNF-alpha produced by tumor cells are sufficient to inhibit tumor growth without detectable side effects.


Assuntos
Mieloma Múltiplo/patologia , Plasmocitoma/patologia , Transfecção/genética , Fator de Necrose Tumoral alfa/genética , Fosfatase Alcalina/metabolismo , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/fisiologia , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Genes Supressores de Tumor/genética , Genes Supressores de Tumor/fisiologia , Imuno-Histoquímica , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-6/fisiologia , Antígeno de Macrófago 1/imunologia , Macrófagos/efeitos dos fármacos , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Fenótipo , Plasmocitoma/genética , Plasmocitoma/metabolismo , Retroviridae/genética , Células Tumorais Cultivadas/efeitos dos fármacos , Células Tumorais Cultivadas/metabolismo , Células Tumorais Cultivadas/patologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA