Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
1.
Front Immunol ; 15: 1347901, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38571963

RESUMO

Most host-microbiota interactions occur within the intestinal barrier, which is essential for separating the intestinal epithelium from toxins, microorganisms, and antigens in the gut lumen. Gut inflammation allows pathogenic bacteria to enter the blood stream, forming immune complexes which may deposit on organs. Despite increased circulating immune complexes (CICs) in patients with inflammatory bowel disease (IBD) and discussions among IBD experts regarding their potential pathogenic role in extra-intestinal manifestations, this phenomenon is overlooked because definitive evidence demonstrating CIC-induced extra-intestinal manifestations in IBD animal models is lacking. However, clinical observations of elevated CICs in newly diagnosed, untreated patients with IBD have reignited research into their potential pathogenic implications. Musculoskeletal symptoms are the most prevalent extra-intestinal IBD manifestations. CICs are pivotal in various arthritis forms, including reactive, rheumatoid, and Lyme arthritis and systemic lupus erythematosus. Research indicates that intestinal barrier restoration during the pre-phase of arthritis could inhibit arthritis development. In the absence of animal models supporting extra-intestinal IBD manifestations, this paper aims to comprehensively explore the relationship between CICs and arthritis onset via a multifaceted analysis to offer a fresh perspective for further investigation and provide novel insights into the interplay between CICs and arthritis development in IBD.


Assuntos
Artrite , Doenças Inflamatórias Intestinais , Animais , Humanos , Complexo Antígeno-Anticorpo/uso terapêutico , Artrite/etiologia , Inflamação , Artralgia/etiologia
2.
Inn Med (Heidelb) ; 65(2): 114-121, 2024 Feb.
Artigo em Alemão | MEDLINE | ID: mdl-38236411

RESUMO

The immune-mediated small vessel vasculitis is known as Schoenlein-Henoch purpura predominantly from pediatrics and in these cases occurs more frequently after infections of the upper airways. In adults, immunoglobulin A (IgA) vasculitis often proceeds more severely und recurrently with the classical tetrad of skin manifestations in the sense of leukocytoclastic vasculitis, joint affection, gastrointestinal involvement and IgA nephritis, in contrast to the mostly mild and self-limiting course in children. The background of this systemic vasculitis with formation of IgA immune complexes is considered to be an altered glycosylation of IgA, as this causes the exposure of binding sites for autoantibodies so that an immune complex reaction can be elicited. This ultimately leads to perivascular deposition of IgA and a further activation of neutrophils. Groundbreaking in the diagnostics is the histological detection of leukocytoclastic vasculitis and in cases of renal manifestations a kidney biopsy with characteristic deposits of immune complexes, which cannot be clearly differentiated from IgA nephropathy. The treatment is aimed at the respective manifestation and is mostly based on consensus recommendations due to the lack of randomized studies. In addition to immunosuppressive medication, in the presence of a chronic kidney disease general nephroprotection is becoming increasingly more important also by inhibition of sodium-glucose transporter 2 (SGLT2). The type and extent of kidney involvement and also rare cardiac manifestations are the main determinants of the prognosis. Continuous medical accompaniment of those affected is necessary due to the possible progression of the disease and the risk of recurrence.


Assuntos
Vasculite por IgA , Poliarterite Nodosa , Vasculite Leucocitoclástica Cutânea , Vasculite , Adulto , Humanos , Criança , Vasculite por IgA/diagnóstico , Complexo Antígeno-Anticorpo/uso terapêutico , Imunoglobulina A , Vasculite/diagnóstico
3.
Naunyn Schmiedebergs Arch Pharmacol ; 396(11): 3269-3283, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37243760

RESUMO

The primary objectives of this research were to investigate the protective effects of liriodendrin against IgG immune complex (IgG-IC)-induced acute lung injury (ALI) and to elucidate the underlying mechanisms. This study employed a mouse and cell model of IgG-IC-induced acute lung injury. Lung tissue was stained with hematoxylin-eosin to observe pathological alterations and arterial blood gas analysis was tested. Inflammatory cytokines, including interleukin-6 (IL-6), interleukin-1ß (IL-1ß), and tumor necrosis factor-alpha (TNF-α), were measured using ELISA. The mRNA expression of inflammatory cytokines was assessed via RT-qPCR. Molecular docking and enrichment analysis were combined to identify the most potential signaling pathways modulated by liriodendrin, which were then verified using western blot analysis in IgG-IC-induced ALI models. We identified 253 shared targets between liriodendrin and IgG-IC-induced acute lung injury from the database. Through network pharmacology, enrichment analysis, and molecular docking, SRC was determined to be the most closely associated target of liriodendrin in IgG-IC-induced ALI. Pretreatment with liriodendrin notably reduced the increased cytokine secretion of IL-1ß, IL-6, and TNF-α. Histopathological analysis of lung tissue demonstrated a protective effect of liriodendrin on IgG-IC-induced acute lung injury in mice. Arterial blood gas analysis showed liriodendrin ameliorated acidosis and hypoxemia efficiently. Further studies revealed that liriodendrin pretreatment substantially attenuated the elevated phosphorylation levels of SRC's downstream components (JNK, P38, and STAT3), suggesting that liriodendrin may protect against IgG-IC-induced ALI via the SRC/STAT3/MAPK pathway. Our findings indicate that liriodendrin protects against IgG-IC-induced acute lung injury by inhibiting the SRC/STAT3/MAPK signaling pathway, suggesting that liriodendrin may serve as a potential treatment for acute lung injury caused by IgG-IC.


Assuntos
Lesão Pulmonar Aguda , Complexo Antígeno-Anticorpo , Camundongos , Animais , Complexo Antígeno-Anticorpo/farmacologia , Complexo Antígeno-Anticorpo/uso terapêutico , Interleucina-6 , Fator de Necrose Tumoral alfa/farmacologia , Simulação de Acoplamento Molecular , Farmacologia em Rede , Transdução de Sinais , Lesão Pulmonar Aguda/tratamento farmacológico , Lesão Pulmonar Aguda/prevenção & controle , Lesão Pulmonar Aguda/etiologia , Pulmão/patologia , Citocinas/metabolismo , Imunoglobulina G/farmacologia , Imunoglobulina G/uso terapêutico , Lipopolissacarídeos/farmacologia
4.
Mikrobiyol Bul ; 57(2): 293-300, 2023 Apr.
Artigo em Turco | MEDLINE | ID: mdl-37067213

RESUMO

Q fever is a zoonosis caused by the intracellular gram-negative bacterium Coxiella burnetii. Infection can be asymptomatic, acute or can cause chronic disease. Chronic disease often presents with infective endocarditis (IE). Diagnosis of IE is difficult because the agent does not grow easily in standard blood cultures and valve vegetations are difficult to detect. Glomerular involvement in patients with Q fever endocarditis is limited to the case reports. In addition, a total of three cases of Q fever endocarditis from Türkiye have been published so far. In this case report, a fourth case of Q fever endocarditis from Türkiye accompanied by immune complex-mediated glomerulonephritis was presented. A 35-year-old male patient with a history of mitral and aortic heart valve replacement was admitted with complaints of fever, night sweats and involuntary weight loss. Cervical lymphadenopathy and hepatosplenomegaly were found during the examination. Laboratory investigations revealed anemia inflammation, acute kidney injury (AKI), hematuria and proteinuria. While no causative agent was detected in blood and urine cultures, no diagnosis could be made as a result of bone marrow and cervical lymph node biopsies.Transesophageal echocardiography was performed for the etiology of fever and revealed 7 mm vegetation on the prosthetic mitral valve. C.burnetii phase 1 IgG tested with indirect immunofluorescent antibody method was reported positive at 1/16384 titer and doxycycline and hydroxychloroquine treatments were initiated. Kidney biopsy for the etiology of AKI revealed focal segmental endocapillary proliferative glomerulonephritis with C3, C1q and IgM immunocomplex deposition. After the addition of methylprednisolone to the treatment, the patient's symptoms improved and creatinine and proteinuria levels decreased dramatically. Although Q fever is endemic in our country, it is detected in fewer numbers than expected. In addition to the difficulties in microbiological and clinical diagnosis, the low awareness of physicians about the disease is one of the important reasons for this situation. When the disease comes to mind, the diagnosis can be easily reached by serological methods. Therefore, Q fever should be investigated in the presence of lymphoproliferative disease-like findings fever of unknown origin and culture-negative endocarditis.


Assuntos
Injúria Renal Aguda , Coxiella burnetii , Endocardite Bacteriana , Endocardite , Glomerulonefrite , Febre Q , Masculino , Humanos , Adulto , Febre Q/complicações , Febre Q/diagnóstico , Febre Q/microbiologia , Complexo Antígeno-Anticorpo/uso terapêutico , Endocardite Bacteriana/complicações , Endocardite Bacteriana/diagnóstico , Endocardite Bacteriana/microbiologia , Endocardite/microbiologia , Glomerulonefrite/complicações , Injúria Renal Aguda/complicações , Proteinúria/complicações , Doença Crônica
5.
Front Immunol ; 13: 938306, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36311755

RESUMO

Epidermolysis bullosa acquisita (EBA) is a subepidermal autoimmune bullous disease caused by autoantibodies targeting type VII collagen (COL7). It is characterized by inflammation and subepidermal blistering mainly through immune complex (IC)-mediated activation of neutrophils. In experimental EBA, binding of neutrophils to ICs in the skin and induction of clinical disease depends on the expression of the Fc gamma receptor (FcγR) IV. As activating FcγR mediate both neutrophil extravasation and activation, we used multiphoton imaging to obtain further insights into the mechanistic contribution of FcγRIV in the pathogenesis of EBA. First, we demonstrated that blocking FcγRIV function completely protects LysM-eGFP mice against induction of antibody transfer-induced EBA. To visualize the interactions of anti-COL7 IgG and neutrophils in vivo, fluorescently labeled anti-COL7 IgG was injected into LysM-eGFP mice. Multiphoton microscopy was sequentially performed over a period of 8 days. At all time points, we observed a significantly higher extravasation of neutrophils into the skin of mice treated with anti-FcγRIV antibody compared to controls. However, the percentage of detected neutrophils localized to the target antigen along the dermal-epidermal junction was comparable between both groups. Additionally, reactive oxygen release and migration in vitro assay data demonstrate that FcγRIV antibody treatment inhibits the activation, but not the migration, of neutrophils. Our findings underscore the importance of advanced in vivo imaging techniques to understand the complexity of IC-mediated neutrophil-dependent inflammation, and indicate that the therapeutic utility of FcγRIV blockade is achieved through impairment of IC-mediated neutrophil activation.


Assuntos
Epidermólise Bolhosa Adquirida , Camundongos , Animais , Ativação de Neutrófilo , Complexo Antígeno-Anticorpo/uso terapêutico , Modelos Animais de Doenças , Receptores de IgG/genética , Inflamação , Imunoglobulina G
6.
Front Immunol ; 13: 865241, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36248903

RESUMO

Chronic blistering at the skin and/or mucous membranes, accompanied by a varying degree of inflammation, is the clinical hallmark of pemphigoid diseases that impose a major medical burden. Pemphigoid diseases are caused by autoantibodies targeting structural proteins of the epithelial basement membrane. One major pathogenic pathway of skin blistering and inflammation is activation of myeloid cells following Fc gamma receptor-dependent binding to the skin-bound immune complexes. This process requires activation of specific kinases, such as PI3Kδ, which have emerged as potential targets for the treatment of pemphigoid diseases. Yet, it is unknown if global cutaneous kinase activity present in lesional pemphigoid disease correlates with therapeutic effects following treatment with a given target-selective kinase inhibitor. To address this, we here first determined the kinase activity in three different mouse models of pemphigoid diseases: Antibody transfer-induced mucous membrane pemphigoid (MMP), antibody transfer-induced epidermolysis bullosa acquisita (EBA) and immunization-induced EBA. Interestingly, the kinome signatures were different among the three models. More specifically, PI3Kδ was within the kinome activation network of antibody transfer-induced MMP and immunization-induced EBA, but not in antibody transfer-induced EBA. Next, the therapeutic impact of the PI3Kδ-selective inhibitor parsaclisib was evaluated in the three model systems. In line with the kinome signatures, parsaclisib had therapeutic effects in antibody transfer-induced MMP and immunization-induced EBA, but not in autoantibody-induced EBA. In conclusion, kinase activation signatures of inflamed skin, herein exemplified by pemphigoid diseases, correlate with the therapeutic outcomes following kinase inhibition, demonstrated here by the PI3Kδ inhibitor parsaclisib.


Assuntos
Epidermólise Bolhosa Adquirida , Penfigoide Mucomembranoso Benigno , Penfigoide Bolhoso , Animais , Complexo Antígeno-Anticorpo/uso terapêutico , Autoanticorpos , Inflamação/patologia , Camundongos , Penfigoide Mucomembranoso Benigno/patologia , Fosfatidilinositol 3-Quinases , Receptores de IgG , Pele , Resultado do Tratamento
7.
Adv Chronic Kidney Dis ; 29(3): 231-242, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-36084970

RESUMO

Atypical hemolytic uremic syndrome and C3 glomerulopathy/immune complex membranoproliferative glomerulonephritis are ultra-rare chronic, complement-mediated diseases with childhood manifestation in a majority of cases. Transition of clinical care of patients from pediatric to adult nephrologists-typically with controlled disease in native or transplant kidneys in case of atypical hemolytic uremic syndrome and often with chronic progressive disease despite treatment efforts in case of C3 glomerulopathy/immune complex membranoproliferative glomerulonephritis-identifies a challenging juncture in the journey of these patients. Raising awareness for the vulnerability of this patient cohort; providing education on disease pathophysiology and management including the use of new, high-precision complement antagonists; and establishing an ongoing dialog of patients, families, and all members of the health care team involved on either side of the age divide will be inevitable to ensure optimal patient outcomes and a safe transition of these patients to adulthood.


Assuntos
Síndrome Hemolítico-Urêmica Atípica , Glomerulonefrite Membranoproliferativa , Nefropatias , Adolescente , Adulto , Complexo Antígeno-Anticorpo/uso terapêutico , Síndrome Hemolítico-Urêmica Atípica/diagnóstico , Síndrome Hemolítico-Urêmica Atípica/terapia , Criança , Proteínas do Sistema Complemento , Glomerulonefrite Membranoproliferativa/terapia , Humanos , Nefrologistas , Adulto Jovem
8.
Curr Med Sci ; 42(5): 991-999, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-36107305

RESUMO

OBJECTIVE: The main pathological feature of immunoglobulin A nephropathy (IgAN), an autoimmune kidney disease, is the deposition of IgA immune complexes, accompanied by mesangial cell proliferation and elevated urine protein. The Guben Tongluo formula (GTF) is a traditional Chinese medicine prescription, which has predominant protective effects on IgAN. However, the therapeutic mechanism of the GTF in IgAN remains elusive. The present study aimed to determine the effects of GTF in treating IgAN via regulating the TLR4/MyD88/NF-κB pathway. METHODS: In the present study, lamina propria B lymphocytes were treated with different concentrations of lipopolysaccharide (LPS) (0, 1, 5, 10 and 20 ng/mL). Flow cytometry was used to define positive CD86+CD19+ cells. CCK-8 assay was used to examine cell proliferation. RNAi was used to induce TLR4 silencing. qRT-PCR and Western blotting were used to determine gene expression. RESULTS: It was found that the LPS dose-dependently increased the content of IgA and galactose-deficient IgA1 (Gd-IgA), the levels of TLR4, Cosmc, MyD88 and phosphorylated (p)-NF-κB, and the ratio of CD86+CD19+ and IgA-producing B cells. However, the TLR4 knockdown reversed the role of LPS. This suggests that TLR4 mediates the effects of LPS on lamina propria B lymphocytes. Furthermore, the GTF could dose-dependently counteract the effects of LPS and TLR4 overexpression on lamina propria B lymphocytes through the TLR4/MyD88/NF-κB pathway. CONCLUSION: Collectively, these results demonstrate that the GTF can regulate the TLR4/MyD88/NF-κB pathway to treat IgAN model lamina propria B lymphocytes stimulated by LPS.


Assuntos
Glomerulonefrite por IGA , NF-kappa B , Humanos , NF-kappa B/metabolismo , Lipopolissacarídeos/efeitos adversos , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Complexo Antígeno-Anticorpo/metabolismo , Complexo Antígeno-Anticorpo/farmacologia , Complexo Antígeno-Anticorpo/uso terapêutico , Galactose/farmacologia , Galactose/uso terapêutico , Transdução de Sinais , Linfócitos B/metabolismo , Imunoglobulina A/metabolismo , Mucosa/metabolismo
9.
BMC Nephrol ; 23(1): 181, 2022 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-35549887

RESUMO

BACKGROUND: Bartonella endocarditis is often a diagnostic challenge due to its variable clinical manifestations, especially when it is first presented with involvement of organs other than skin and lymph nodes, such as the kidney. CASE PRESENTATION: This was a 13-year-old girl presenting with fever, chest and abdominal pain, acute kidney injury, nephrotic-range proteinuria and low complement levels. Her kidney biopsy showed diffuse crescentic proliferative glomerulonephritis with a full-house pattern of immune complex deposition shown by immunofluorescence, which was initially considered consistent with systemic lupus erythematous-associated glomerulonephritis (lupus nephritis). After extensive workup, Bartonella endocarditis was diagnosed. Antibiotic treatment and valvular replacement surgery were undertaken with subsequent return of kidney function to normal range. CONCLUSION: This case demonstrates the importance of considering the full clinical picture when interpreting clinical, laboratory and biopsy findings, because the treatment strategy for infective endocarditis versus lupus nephritis is drastically different.


Assuntos
Bartonella , Endocardite , Glomerulonefrite Membranoproliferativa , Glomerulonefrite , Nefrite Lúpica , Adolescente , Complexo Antígeno-Anticorpo/uso terapêutico , Endocardite/tratamento farmacológico , Feminino , Glomerulonefrite/complicações , Glomerulonefrite/diagnóstico , Glomerulonefrite/tratamento farmacológico , Glomerulonefrite Membranoproliferativa/complicações , Humanos , Nefrite Lúpica/complicações , Masculino
10.
Leuk Lymphoma ; 61(9): 2050-2058, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32336174

RESUMO

A complex interplay between chronic lymphocytic leukemia (CLL) cells and different constituents of the immune system generally results in immune tolerance. As targeted therapies are gaining a critical role in the therapeutic landscape of this disease, their impact on the already perturbed immune milieu needs to be considered. This review addresses the issues of basic immune dysfunction in CLL which is further complicated by the effects of a number of novel targeted therapies used for this malignancy. These new approaches may simultaneously facilitate both anti- and pro-cancer activity, potentially compromising the depth of response to therapy. Current evidence suggests that exploiting combination therapy could potentially overcome at least part of these deleterious effects, thereby prolonging response to treatment and helping to restore immune activity.


Assuntos
Leucemia Linfocítica Crônica de Células B , Complexo Antígeno-Anticorpo/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica , Terapia Combinada , Humanos , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico
11.
Cancer Immunol Res ; 7(3): 443-457, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30651291

RESUMO

High-dose IL2 immunotherapy can induce long-lasting cancer regression but is toxic and insufficiently efficacious. Improvements are obtained with IL2/anti-IL2 complexes (IL2Cx), which redirect IL2 action to CD8+ T and natural killer (NK) cells. Here, we evaluated the efficacy of combining IL2Cx with blockade of inhibitory immune pathways. In an autochthonous lung adenocarcinoma model, we show that the IL2Cx/anti-PD-1 combination increases CD8+ T-cell infiltration of the lung and controls tumor growth. In the B16-OVA model, which is resistant to checkpoint inhibition, combination of IL2Cx with PD-1 or CTLA-4 pathway blockade reverses that resistance. Both combinations work by reinvigorating exhausted intratumoral CD8+ T cells and by increasing the breadth of tumor-specific T-cell responses. However, only the IL2Cx/anti-CTLA-4 combination is able to rescue NK cell antitumor function by modulating intratumoral regulatory T cells. Overall, association of IL2Cx with PD-1 or CTLA-4 pathway blockade acts by different cellular mechanisms, paving the way for the rational design of combinatorial antitumor therapies.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Complexo Antígeno-Anticorpo/uso terapêutico , Antígeno CTLA-4/imunologia , Interleucina-2/imunologia , Células Matadoras Naturais/imunologia , Linfócitos T Reguladores/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Imunoterapia , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Receptor de Morte Celular Programada 1/imunologia , Linfócitos T Citotóxicos/imunologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
12.
Front Immunol ; 9: 656, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29670626

RESUMO

Murine chronic graft-versus-host-disease (cGvHD) induced by injection of parental lymphocytes into F1 hybrids results in a disease similar to systemic lupus erythematosus. Here, we have used DBA/2 T cell injection into (C57BL/6 × DBA/2)F1 (BDF1) mice as a model system to test the prophylactic and therapeutic effects of interleukin-2 (IL-2)/anti-IL-2 immune complexes on the course of cGvHD. Our findings demonstrate that pretreatment with Treg inducing JES6/IL-2 complexes render BDF1 mice largely resistant to induction of cGvHD, whereas pretreatment with CD8+ T cell/NK cell inducing S4B6/IL-2 complexes results in a more severe cGvHD. In contrast, treatment with JES6/IL-2 complexes 4 weeks after induction had no beneficial effect on disease symptoms. However, similar treatment with S4B6/IL-2 complexes led to a significant amelioration of the disease. This therapeutic effect seems to be mediated by donor CD8+ T cells. The fact that a much stronger cGvHD is induced in BDF1 mice depleted of donor CD8+ T cells strongly supports this conclusion. The contrasting effects of the two different IL-2 complexes are likely due to different mechanisms.


Assuntos
Complexo Antígeno-Anticorpo/uso terapêutico , Linfócitos T CD8-Positivos/imunologia , Doença Enxerto-Hospedeiro/terapia , Imunoterapia/métodos , Interleucina-2/metabolismo , Lúpus Eritematoso Sistêmico/terapia , Linfócitos T Reguladores/imunologia , Animais , Anticorpos/metabolismo , Células Cultivadas , Modelos Animais de Doenças , Doença Enxerto-Hospedeiro/imunologia , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Transfusão de Linfócitos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Transplante Homólogo
13.
Cell Physiol Biochem ; 44(5): 1810-1827, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29224017

RESUMO

BACKGROUND/AIMS: Regulatory T cells (Tregs) can suppress immunologic damage in myocardial ischaemia/reperfusion injury (MIRI), however, the isolation and ex vivo expansion of these cells for clinical application remains challenging. Here, we investigated whether the IL-2/anti-IL-2 complex (IL-2C), a mediator of Treg expansion, can attenuate MIRI in mice. METHODS: Myocardial I/R was surgically induced in male C57BL/6 mice, aged 8-10 weeks, that were randomly assigned to 1) sham group (Sham), 2) Phosphate Buffered Saline (PBS), 3) IL-2-anti-IL-2 Ab complex (IL-2C), or 4) sham group, 5) PBS, 6) IL-2C after MIRI, or 7) IL-2C, 8) IL-2C+anti-CD25 mAbs, or 9) IL-2C; 10) IL-2C+anti-TGF-ß1 mAbs, 11) IL-2C+anti-IL-10 mAbs. The following parameters were measured at different time points: infarct area, myocardial apoptosis, splenocytes, the inhibitory function of Tregs, and presence of inflammatory factors. In addition, immunohistochemistry analysis was performed. RESULTS: We observed that Tregs were activated in response to MIRI. IL-2C administered before MIRI induced Treg expansion in both spleen and heart, attenuated Th1 and Th17 cell numbers, improved myocardial function, and attenuated both infiltration of inflammatory cells and apoptosis after MIRI. Furthermore, IL-2C administration reduced expression of inflammatory cytokines in the heart and attenuated proliferation of splenic cells. Depletion of Tregs with anti-CD25 mAb abrogated the beneficial effects of IL-2C. However, IL-2C-mediated myocardial protection was not dependent on either IL-10 or TGF-ß. In addition, IL-2C administration after MIRI did not reduce infarct area, but did improve myocardial function slightly and reduced myocardial fibrosis. CONCLUSION: Our results demonstrate that IL-2C-induced Treg expansion attenuates MIRI and improves myocardial recovery in vivo, suggesting that IL-2C is a promising therapeutic target for myocardial IRI.


Assuntos
Complexo Antígeno-Anticorpo/farmacologia , Interleucina-2/imunologia , Traumatismo por Reperfusão Miocárdica/patologia , Linfócitos T Reguladores/efeitos dos fármacos , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Complexo Antígeno-Anticorpo/uso terapêutico , Apoptose/efeitos dos fármacos , Citocinas/metabolismo , Modelos Animais de Doenças , Hemodinâmica/efeitos dos fármacos , Interleucina-10/imunologia , Subunidade alfa de Receptor de Interleucina-2/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miocárdio/citologia , Miocárdio/imunologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Baço/citologia , Baço/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Células Th1/citologia , Células Th1/imunologia , Células Th17/citologia , Células Th17/imunologia , Fator de Crescimento Transformador beta1/imunologia
14.
Clin Exp Rheumatol ; 33(4 Suppl 92): S54-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26457438

RESUMO

Interleukin-2 (IL-2) is a cytokine centrally involved in the regulation of immune tolerance and activation by its effects on CD4+ T regulatory (Treg) cells and cytotoxic effector lymphocytes, respectively. Due to these properties IL-2 immunotherapy has been used, as low-dose IL-2, in the treatment of autoimmune and chronic-inflammatory disorders; conversely, at high doses, IL-2 has shown efficacy in a subset of patients with metastatic cancer. Recent advances have highlighted the possibility of using improved IL-2-based therapies, such IL-2-antibody complexes (IL-2 complexes), able to selectively and potently stimulate either Treg cells or cytotoxic effector cells. This article discusses the properties and clinical implications of IL-2 and IL-2 complexes.


Assuntos
Complexo Antígeno-Anticorpo/uso terapêutico , Fatores Imunológicos/uso terapêutico , Interleucina-2/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Subpopulações de Linfócitos T/efeitos dos fármacos , Animais , Complexo Antígeno-Anticorpo/efeitos adversos , Complexo Antígeno-Anticorpo/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Citotoxicidade Imunológica/efeitos dos fármacos , Humanos , Fatores Imunológicos/efeitos adversos , Fatores Imunológicos/imunologia , Interleucina-2/efeitos adversos , Interleucina-2/imunologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia
15.
J Immunol ; 195(11): 5203-14, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26482409

RESUMO

Regulatory T cells (Treg) play a crucial role in the maintenance of self-tolerance. In this study, we sought to expand Ag-specific Tregs in vivo and investigate whether the expanded Tregs can prevent or delay the development of type 1 diabetes (T1D) in the NOD mouse model. NOD mice were treated with a combination of IL-2/anti-IL-2 Ab complex, islet Ag peptide, and rapamycin. After the combined treatment, CD4(+)CD25(+)Foxp3(+) Tregs were significantly expanded in vivo, they expressed classical Treg markers, exerted enhanced suppressive functions in vitro, and protected against spontaneous development of T1D in NOD mice. Moreover, treated mice were almost completely protected from the adoptively transferred, aggressive form of T1D caused by in vitro-activated cytotoxic islet Ag-specific CD8 T cells. Protection from T1D was transferrable by Tregs and could be attributed to reduced islet infiltration of immune cells as well as the skewing of the immune response toward a Th2 cytokine profile. This new method of peripheral immune regulation could potentially contribute to development of novel immunotherapeutic strategies to prevent the development of T1D or to promote tolerance to islet transplants without using immunosuppressive drugs for long terms.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Diabetes Mellitus Tipo 1/prevenção & controle , Interleucina-2/imunologia , Interleucina-2/uso terapêutico , Sirolimo/uso terapêutico , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Complexo Antígeno-Anticorpo/imunologia , Complexo Antígeno-Anticorpo/uso terapêutico , Autoantígenos/imunologia , Proliferação de Células , Quimioterapia Combinada , Feminino , Ilhotas Pancreáticas/imunologia , Ativação Linfocitária , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Peptídeos/imunologia , Linfócitos T Citotóxicos/imunologia , Células Th2/imunologia
17.
J Immunol ; 193(6): 2961-70, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25098291

RESUMO

Leishmania (Viannia) parasites are etiological agents of cutaneous leishmaniasis in the New World. Infection is characterized by a mixed Th1/Th2 inflammatory response, which contributes to disease pathology. However, the role of regulatory T cells (Tregs) in Leishmania (Viannia) disease pathogenesis is unclear. Using the mouse model of chronic L. (V.) panamensis infection, we examined the hypothesis that Treg functionality contributes to control of pathogenesis. Upon infection, Tregs (CD4(+)Foxp3(+)) presented with a dysregulated phenotype, in that they produced IFN-γ, expressed Tbet, and had a reduced ability to suppress T cell proliferation in vitro. Targeted ablation of Tregs resulted in enlarged lesions, increased parasite load, and enhanced production of IL-17 and IFN-γ, with no change in IL-10 and IL-13 levels. This indicated that an increased inflammatory response was commensurate with disease exacerbation and that the remaining impaired Tregs were important in regulation of disease pathology. Conversely, adoptive transfer of Tregs from naive mice halted disease progression, lowered parasite burden, and reduced cytokine production (IL-10, IL-13, IL-17, IFN-γ). Because Tregs appeared to be important for controlling infection, we hypothesized that their expansion could be used as an immunotherapeutic treatment approach. As a proof of principle, chronically infected mice were treated with rIL-2/anti-IL-2 Ab complex to expand Tregs. Treatment transitorily increased the numbers and percentage of Tregs (draining lymph node, spleen), which resulted in reduced cytokine responses, ameliorated lesions, and reduced parasite load (10(5)-fold). Thus, immunotherapy targeting Tregs could provide an alternate treatment strategy for leishmaniasis caused by Leishmania (Viannia) parasites.


Assuntos
Imunoterapia Adotiva , Leishmania guyanensis/imunologia , Leishmaniose Mucocutânea/imunologia , Leishmaniose Mucocutânea/terapia , Linfócitos T Reguladores/imunologia , Animais , Anticorpos/imunologia , Anticorpos/uso terapêutico , Complexo Antígeno-Anticorpo/uso terapêutico , Proliferação de Células , Feminino , Indolamina-Pirrol 2,3,-Dioxigenase/antagonistas & inibidores , Inflamação/imunologia , Interferon gama/biossíntese , Interleucina-10/biossíntese , Interleucina-13/biossíntese , Interleucina-17/biossíntese , Interleucina-2/imunologia , Interleucina-2/uso terapêutico , Leishmaniose Mucocutânea/parasitologia , Contagem de Linfócitos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Carga Parasitária , Linfócitos T Reguladores/transplante , Fator de Crescimento Transformador beta/biossíntese
18.
J Leukoc Biol ; 96(6): 951-5, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25009233

RESUMO

RSV is the most significant cause of serious lower respiratory tract infection in infants and young children worldwide. There is currently no vaccine for the virus, and antiviral therapy (e.g., ribavirin) has shown no efficacy against the disease. We reported that alternatively activated macrophages (AAMs) mediate resolution of RSV-induced pathology. AAM differentiation requires macrophage-derived IL-4 and -13, autocrine/paracrine signaling through the type I IL-4 receptor, and STAT6 activation. Based on these findings, we reasoned that it would be possible to intervene therapeutically in RSV disease by increasing AAM differentiation, thereby decreasing lung pathology. Mice treated with the IL-4/anti-IL-4 immune complexes, shown previously to sustain levels of circulating IL-4, increased the RSV-induced AAM markers arginase-1 and mannose receptor and decreased the lung pathology. Induction of PPARγ, shown to play a role in AAM development, by the PPARγ agonist rosiglitazone or treatment of mice with the macrolide antibiotic AZM, also reported to skew macrophage differentiation to an AAM phenotype, increased the AAM markers and mitigated RSV-induced lung pathology. Collectively, our data suggest that therapeutic manipulation of macrophage differentiation to enhance the AAM phenotype is a viable approach for ameliorating RSV-induced disease.


Assuntos
Complexo Antígeno-Anticorpo/uso terapêutico , Interleucina-4/uso terapêutico , Pulmão/patologia , Macrófagos/efeitos dos fármacos , Infecções por Vírus Respiratório Sincicial/tratamento farmacológico , Animais , Araquidonato 5-Lipoxigenase/fisiologia , Arginase/biossíntese , Arginase/genética , Azitromicina/farmacologia , Azitromicina/uso terapêutico , Diferenciação Celular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Regulação da Expressão Gênica/efeitos dos fármacos , Interleucina-4/imunologia , Interleucina-4/farmacologia , Interleucina-4/fisiologia , Lectinas Tipo C/biossíntese , Lectinas Tipo C/genética , Pulmão/efeitos dos fármacos , Pulmão/virologia , Receptor de Manose , Lectinas de Ligação a Manose/biossíntese , Lectinas de Ligação a Manose/genética , Camundongos , Camundongos Endogâmicos BALB C , PPAR gama/agonistas , PPAR gama/fisiologia , RNA Mensageiro/biossíntese , Receptores de Superfície Celular/biossíntese , Receptores de Superfície Celular/genética , Proteínas Recombinantes/uso terapêutico , Infecções por Vírus Respiratório Sincicial/patologia , Rosiglitazona , Fator de Transcrição STAT6/fisiologia , Sigmodontinae , Transdução de Sinais , Tiazolidinedionas/farmacologia , Tiazolidinedionas/uso terapêutico
19.
J Hepatol ; 59(3): 450-6, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23669281

RESUMO

BACKGROUND & AIMS: Even though various experimental therapeutic approaches for chronic hepatitis B infection have been reported, few of them have been verified by clinical trials. We have developed an antigen-antibody (HBsAg-HBIG) immunogenic complex therapeutic vaccine candidate with alum as adjuvant (YIC), aimed at breaking immune tolerance to HBV by modulating viral antigen processing and presentation. A double-blind, placebo-controlled, phase II B clinical trial of YIC has been reported previously, and herein we present the results of the phase III clinical trial of 450 patients. METHODS: Twelve doses of either YIC or alum alone as placebo were administered randomly to 450 CHB patients and they were followed for 24weeks after the completion of immunization. The primary end point was HBeAg seroconversion, and the secondary end points were decrease in viral load, improvement of liver function, and histology. RESULTS: In contrast to the previous phase II B trial using six doses of YIC and alum as placebo, six more injections of YIC or alum resulted in a decrease of the HBeAg seroconversion rate from 21.8% to 14.0% in the YIC group, but an increase from 9% to 21.9% in the alum group. Decrease in serum HBV DNA and normalization of liver function were similar in both groups (p>0.05). CONCLUSIONS: Overstimulation with YIC did not increase but decreased its efficacy due to immune fatigue in hosts. An appropriate immunization protocol should be explored and is crucial for therapeutic vaccination. Multiple injections of alum alone could have stimulated potent inflammatory and innate immune responses contributing to its therapeutic efficacy, and needs further investigation.


Assuntos
Antígenos de Superfície da Hepatite B/uso terapêutico , Hepatite B Crônica/terapia , Imunoglobulinas/uso terapêutico , Vacinas Virais/uso terapêutico , Adjuvantes Imunológicos/administração & dosagem , Adulto , Compostos de Alúmen/administração & dosagem , Complexo Antígeno-Anticorpo/administração & dosagem , Complexo Antígeno-Anticorpo/uso terapêutico , Citocinas/sangue , Método Duplo-Cego , Feminino , Genótipo , Antígenos de Superfície da Hepatite B/administração & dosagem , Antígenos E da Hepatite B/sangue , Vírus da Hepatite B/classificação , Vírus da Hepatite B/genética , Vírus da Hepatite B/imunologia , Hepatite B Crônica/imunologia , Hepatite B Crônica/patologia , Humanos , Imunoglobulinas/administração & dosagem , Masculino , Vacinas Virais/efeitos adversos , Adulto Jovem
20.
Immunology ; 137(4): 305-16, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23167249

RESUMO

In this study, we investigated the effects of administration of interleukin-2 (IL-2)/JES6-1 (anti-IL-2 monoclonal antibody) immune complexes on the expansion and activation of regulatory T (Treg) cells, the down-regulation of T helper type 17 (Th17) cells, and the control of the severity of collagen-induced arthritis (CIA). Wild-type and CIA-induced wild-type mice were injected intraperitoneally (i.p.) with IL-2 or IL-2/JES6-1 complex three times at 2-day intervals. Treg cell surface markers were analysed by flow cytometry. After injecting IL-2 or IL-2/JES6-1, the time kinetics of IL-2 signalling molecules was examined by FACS and Western blotting. Concentrations of IL-17 and IL-10 were measured by ELISA. Injection of IL-2/JES6-1 increased the proportion of Foxp3+ Treg cells among splenic CD4+ T cells, which reached the highest level on day 4 after injection. Up-regulation of CTLA4, GITR and glycoprotein-A repetitions predominant (GARP) was observed. Activation of p-signal transducer and activator of transcription 5 (STAT5) was apparent within 3 hr after injection of IL-2/JES6-1 complexes. Expression of IL-2 signalling molecules, including p-AKT and p-p38/mitogen-activated protein kinase, was also higher in splenocytes treated with IL-2/JES6-1 complexes. Injection of IL-2/JES6-1 complexes suppressed the induction of CIA and the production of IL-17 and inflammatory responses while increasing the level of IL-10 in the spleen. The expansion of Treg cells (via STAT5) and the concomitant increase in IL-2 signalling pathways by IL-2/JES6-1 complexes suggests their potential use as a novel therapeutic agent for the treatment of autoimmune arthritis.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Complexo Antígeno-Anticorpo/uso terapêutico , Artrite Experimental/tratamento farmacológico , Interleucina-2/imunologia , Fator de Transcrição STAT5/fisiologia , Transdução de Sinais/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Complexo Antígeno-Anticorpo/imunologia , Complexo Antígeno-Anticorpo/farmacologia , Interleucina-2/uso terapêutico , Masculino , Camundongos , Camundongos Endogâmicos DBA , Transdução de Sinais/efeitos dos fármacos , Linfócitos T Reguladores/fisiologia , Células Th17/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...