Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-34045306

RESUMO

OBJECTIVE: To determine whether blocking the neonatal Fc receptor (FcRn) during gestation with an anti-FcRn monoclonal antibody (mAb) reduces transfer of pathogenic maternal antibodies in utero and decreases the likelihood of maternal antibody-mediated neonatal disease in the offspring. METHODS: Using a previously established maternal-to-fetal transfer mouse model of arthrogryposis multiplex congenita (AMC), we assessed the effect of 4470, an anti-FcRn mAb, on the transfer of total human immunoglobulin G (IgG) and specific acetylcholine receptor (AChR)-antibodies from mother to fetus, as well as its effect on the prevention of neurodevelopmental abnormalities in the offspring. RESULTS: Offspring of pregnant dams treated with 4470 during gestation showed a substantial reduction in total human IgG and AChR antibody levels compared with those treated with the isotype mAb control. Treatment with 4470 was also associated with a significant reduction in AMC-IgG-induced deformities (limb or spinal curve malformations) when compared with mAb control-exposed embryos and a nonsignificant increase in the percentage of fetuses showing spontaneous movements. 4470 exposure during pregnancy was not associated with changes in general parameters of maternal well-being or fetal development; indeed, male neonates showed faster weight gain and shorter time to reach developmental milestones. CONCLUSIONS: FcRn blockade is a promising therapeutic strategy to prevent the occurrence of AMC and other human maternal autoantibody-related diseases in the offspring.


Assuntos
Anticorpos Bloqueadores/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Artrogripose/prevenção & controle , Autoanticorpos/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Troca Materno-Fetal , Receptores Fc/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Feminino , Humanos , Imunoglobulina G , Camundongos , Camundongos Endogâmicos ICR , Gravidez , Receptores Colinérgicos/imunologia
2.
Adv Drug Deliv Rev ; 175: 113778, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33887405

RESUMO

Diabetes mellitus is a chronic disease with an elevated risk of micro- and macrovascular complications, such as fibrosis. To prevent diabetes-associated fibrosis, the symptomatology of diabetes must be controlled, which is commonly done by subcutaneous injection of antidiabetic peptides. To minimize the pain and distress associated with such injections, there is an urgent need for non-invasive oral transmucosal drug delivery strategies. However, orally administered peptide-based drugs are exposed to harsh conditions in the gastrointestinal tract and poorly cross the selective intestinal epithelium. Thus, targeting of drugs to receptors expressed in epithelial cells, such as the neonatal Fc receptor (FcRn), may therefore enhance uptake and transport through mucosal barriers. This review compiles how in-depth studies of FcRn biology and engineering of receptor-binding molecules may pave the way for design of new classes of FcRn-targeted nanosystems. Tailored strategies may open new avenues for oral drug delivery and provide better treatment options for diabetes and, consequently, fibrosis prevention.


Assuntos
Diabetes Mellitus/tratamento farmacológico , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Hipoglicemiantes/administração & dosagem , Sistemas de Liberação de Fármacos por Nanopartículas , Receptores Fc/efeitos dos fármacos , Administração Oral , Animais , Diabetes Mellitus/patologia , Sistemas de Liberação de Medicamentos , Fibrose , Humanos , Hipoglicemiantes/uso terapêutico , Sistemas de Liberação de Fármacos por Nanopartículas/uso terapêutico
3.
Drug Resist Updat ; 56: 100752, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33765484

RESUMO

Immunotherapies such as CAR-T cell transfer and antibody-targeted therapy have produced promising clinical outcomes in patients with advanced and metastatic cancer that are resistant to conventional therapies. However, with increasing use of cancer immunotherapy in clinical treatment, multiple therapy-resistance mechanisms have gradually emerged. The tumor microenvironment (TME), an integral component of cancer, can significantly influence the therapeutic response. Thus, it is worth exploring the potential of TME in modulating therapy resistance, in the hope to devise novel strategies to reinforcing anti-cancer treatments such as immunotherapy. As a crucial recycling process in the complex TME, the role of autophagy in tumor immunity has been increasingly appreciated. Firstly, autophagy in tumor cells can affect their immune response through modulating MHC-I-antigen complexes, thus modulating immunogenic tumor cell death, changing functions of immune cells via secretory autophagy, reducing the NK- and CTL-mediated cell lysis and degradation of immune checkpoint proteins. Secondly, autophagy is critical for the differentiation, maturation and survival of immune cells in the TME and can significantly affect the immune function of these cells, thereby regulating the anti-tumor immune response. Thirdly, alteration of autophagic activity in stromal cells, especially in fibroblasts, can reconstruct the three-dimensional stromal environment and metabolic reprogramming in the TME. A number of studies have demonstrated that optimal induction or inhibition of autophagy may lead to effective therapeutic regimens when combined with immunotherapy. This review discusses the important roles of autophagy in tumor cells, immune cells and stromal cells in the context of tumor immunity, and the potential of combining the autophagy-based therapy with immunotherapy as novel therapeutic approaches against cancer.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Autofagia/fisiologia , Microambiente Tumoral/fisiologia , Autofagia/efeitos dos fármacos , Autofagia/imunologia , Morte Celular/fisiologia , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Humanos , Neoplasias/imunologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
4.
Transfusion ; 61(4): 1222-1234, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33580979

RESUMO

BACKGROUND: Patients can form antibodies to foreign human leukocyte antigen (HLA) Class I antigens after exposure to allogeneic cells. These anti-HLA class I antibodies can bind transfused platelets (PLTs) and mediate their destruction, thus leading to PLT refractoriness. Patients with PLT refractoriness need HLA-matched PLTs, which require expensive HLA typing of donors, antibody analyses of patient sera and/or crossmatching. An alternative approach is to reduce PLT HLA Class I expression using a brief incubation in citric acid on ice at low pH. METHODS AND MATERIALS: Apheresis PLT concentrates were depleted of HLA Class I complexes by 5 minutes incubation in ice-cold citric acid, at pH 3.0. Surface expression of HLA Class I complexes, CD62P, CD63, phosphatidylserine, and complement factor C3c was analyzed by flow cytometry. PLT functionality was tested by thromboelastography (TEG). RESULTS: Acid treatment reduced the expression of HLA Class I complexes by 71% and potential for C3c binding by 11.5-fold compared to untreated PLTs. Acid-treated PLTs were significantly more activated than untreated PLTs, but irrespective of this increase in steady-state activation, CD62P and CD63 were strongly upregulated on both acid-treated and untreated PLTs after stimulation with thrombin receptor agonist peptide. Acid treatment did not induce apoptosis over time. X-ray irradiation did not significantly influence the expression of HLA Class I complexes, CD62P, CD63, and TEG variables on acid treated PLTs. CONCLUSION: The relatively simple acid stripping method can be used with irradiated apheresis PLTs and may prevent transfusion-associated HLA sensitization and overcome PLT refractoriness.


Assuntos
Ácido Cítrico/efeitos adversos , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Transfusão de Plaquetas/métodos , Imunodeficiência Combinada Severa/induzido quimicamente , Anticorpos/imunologia , Tipagem e Reações Cruzadas Sanguíneas/métodos , Plaquetas/efeitos da radiação , Feminino , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Antígenos de Histocompatibilidade Classe I/efeitos da radiação , Teste de Histocompatibilidade/economia , Teste de Histocompatibilidade/métodos , Humanos , Selectina-P/metabolismo , Transfusão de Plaquetas/efeitos adversos , Plaquetoferese/métodos , Tetraspanina 30/metabolismo , Tromboelastografia/métodos , Trombocitopenia/terapia , Regulação para Cima/genética
5.
Clin Transl Oncol ; 23(1): 110-121, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32661823

RESUMO

PURPOSE: Chemotherapy for advanced pancreatic cancer has limited efficacy due to the difficultly of treating established tumours and the evolution of tumour resistance. Chemotherapies for pancreatic cancer are typically studied for their cytotoxic properties rather than for their ability to increase the immunogenicity of pancreatic tumour cells. In this study Gemcitabine in combination with immune modulatory chemotherapies Oxaliplatin, zoledronic acid and pomalidomide was studied to determine how combination therapy alters the immunogenicity of pancreatic tumour cell lines and subsequent T-cell responses. METHODS: Pancreatic tumour cell lines were stimulated with the chemotherapeutic agents and markers of immune recognition were assessed. The effect of chemotherapeutic agents on DC function was measured using uptake of CFSE-stained PANC-1 cells, changes in markers of maturation and their ability to activate CD8+ T-cells. The effect of chemotherapeutic agents on T-cell priming prior to activation using anti-CD3 and anti-CD28 antibodies was determined by measuring IFN-γ expression and Annexin V staining using flow cytometry. RESULTS: These agents demonstrate both additive and inhibitory properties on a range of markers of immunogenicity. Gemcitabine was notable for its ability to induce the upregulation of human leukocyte antigen and checkpoints on pancreatic tumour cell lines whilst inhibiting T-cell activation. Pomalidomide demonstrated immune modulatory properties on dendritic cells and T-cells, even in the presence of gemcitabine. DISCUSSION: These data highlight the complex interactions of different agents in the modulation of tumour immunogenicity and immune cell activation and emphasise the complexity in rationally designing chemo immunogenic combinations for use with immunotherapy.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Desoxicitidina/análogos & derivados , Imunomodulação/efeitos dos fármacos , Neoplasias Pancreáticas/imunologia , Anexina A5/metabolismo , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Desoxicitidina/farmacologia , Interações Medicamentosas/imunologia , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Imunomodulação/imunologia , Interferon gama/metabolismo , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Oxaliplatina/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Talidomida/análogos & derivados , Talidomida/farmacologia , Ácido Zoledrônico/farmacologia , Gencitabina
6.
CPT Pharmacometrics Syst Pharmacol ; 9(11): 649-658, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33021075

RESUMO

Isatuximab, a monoclonal antibody (mAb) of immunoglobulin G (IgG) isotype, specifically targets the cluster of differentiation 38 antigen overexpressed in malignant plasma cells. Isatuximab is used to treat multiple myeloma (MM), characterized by the excessive production of abnormal "myeloma proteins" (M-proteins) that may interact with therapeutic IgG mAb on the neonatal Fc receptor (FcRn)-mediated recycling pathway. The clinical pharmacology profile of isatuximab was investigated by population pharmacokinetics (PKs) modeling in 476 patients with MM who received 1-20 mg/kg isatuximab either as single agent or in combination with pomalidomide-dexamethasone in 4 clinical trials. Isatuximab PKs were characterized by a two-compartment model with parallel time-varying linear clearance (CL) and nonlinear elimination. Due to a mechanism-based drug-disease interaction, patients secreting IgG M-protein exhibited a twofold lower drug exposure compared with patients with non-IgG MM. No dose adjustment was required based on MM immunoglobulin type because efficacy and safety profiles were comparable between IgG and non-IgG MM subpopulations. ß2-microglobulin, body weight, sex, drug material, and race have a limited effect on drug exposure and do not require any dose adjustment. A typical 50% decrease in linear CL from initial treatment to steady-state was predicted, and this decrease correlated with the best overall response rate and was slower for patients with IgG MM. These findings suggest that the time-dependent effect of isatuximab is likely mediated by a combined factor of both disease state evolution and the perturbation of the FcRn-mediated recycling pathway.


Assuntos
Anticorpos Monoclonais Humanizados/farmacocinética , Anticorpos Monoclonais/farmacocinética , Antineoplásicos Imunológicos/farmacocinética , Mieloma Múltiplo/tratamento farmacológico , Idoso , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais Humanizados/administração & dosagem , Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos Hormonais/administração & dosagem , Antineoplásicos Hormonais/uso terapêutico , Antineoplásicos Imunológicos/administração & dosagem , Antineoplásicos Imunológicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Peso Corporal/efeitos dos fármacos , Estudos de Casos e Controles , Ensaios Clínicos como Assunto , Dexametasona/administração & dosagem , Dexametasona/uso terapêutico , Interações Medicamentosas , Feminino , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Masculino , Mieloma Múltiplo/imunologia , Proteínas do Mieloma/efeitos dos fármacos , Recidiva Local de Neoplasia/tratamento farmacológico , Preparações Farmacêuticas , Receptores Fc/efeitos dos fármacos , Receptores Fc/metabolismo , Recidiva , Talidomida/administração & dosagem , Talidomida/análogos & derivados , Talidomida/uso terapêutico , Fatores de Tempo , Microglobulina beta-2/efeitos dos fármacos , Microglobulina beta-2/metabolismo
7.
Sci Rep ; 10(1): 15556, 2020 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-32968163

RESUMO

MICA/B proteins are expressed on the surface of various types of stressed cells, including cancer cells. Cytotoxic lymphocytes expressing natural killer group 2D (NKG2D) receptor recognize MICA/B and eliminate the cells. However, cancer cells evade such immune recognition by inducing proteolytic shedding of MICA/B proteins. Therefore, preventing the shedding of MICA/B proteins could enhance antitumor immunity. Here, by screening a protease inhibitor library, we found that the fatty-acid amide hydrolase (FAAH) inhibitor, URB597, suppresses the shedding of MICA/B. URB597 significantly reduced the soluble MICA level in culture medium and increased the MICA level on the surface of cancer cells. The effect was indirect, being mediated by increased expression of tissue inhibitor of metalloproteinases 3 (TIMP3). Knockdown of TIMP3 expression reversed the effect of URB597, confirming that TIMP3 is required for the MICA shedding inhibition by URB597. In contrast, FAAH overexpression reduced TIMP3 expression and the cell-surface MICA level and increased the soluble MICA level. These results suggest that inhibition of FAAH could prevent human cancer cell evasion of immune-mediated clearance.


Assuntos
Amidoidrolases/genética , Antígenos de Histocompatibilidade Classe I/genética , Inibidor Tecidual de Metaloproteinase-3/genética , Benzamidas/química , Benzamidas/farmacologia , Carbamatos/química , Carbamatos/farmacologia , Linhagem Celular Tumoral , Meios de Cultura/química , Meios de Cultura/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Humanos , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia , Neoplasias/tratamento farmacológico , Neoplasias/genética , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T Citotóxicos/imunologia
8.
Am J Physiol Gastrointest Liver Physiol ; 319(4): G432-G442, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32755306

RESUMO

Alcohol consumption worsens hepatitis B virus (HBV) infection pathogenesis. We have recently reported that acetaldehyde suppressed HBV peptide-major histocompatibility complex I (MHC class I) complex display on hepatocytes, limiting recognition and subsequent removal of the infected hepatocytes by HBV-specific cytotoxic T lymphocytes (CTLs). This suppression was attributed to impaired processing of antigenic peptides by the proteasome. However, in addition to proteasome dysfunction, alcohol may induce endoplasmic reticulum (ER) stress and Golgi fragmentation in HBV-infected liver cells to reduce uploading of viral peptides to MHC class I and/or trafficking of this complex to the hepatocyte surface. Hence, the aim of this study was to elucidate whether alcohol-induced ER stress and Golgi fragmentation affect HBV peptide-MHC class I complex presentation on HBV+ hepatocytes. Here, we demonstrate that, while both acetaldehyde and HBV independently cause ER stress and Golgi fragmentation, the combined exposure provided an additive effect. Thus we observed an activation of the inositol-requiring enzyme 1α-X-box binding protein 1 and activation transcription factor (ATF)6α, but not the phospho PKR-like ER kinase-phospho eukaryotic initiation factor 2α-ATF4-C/EBP homologous protein arms of ER stress in HBV-transfected cells treated with acetaldehyde-generating system (AGS). In addition, Golgi proteins trans-Golgi network 46, GM130, and Giantin revealed punctate distribution, indicating Golgi fragmentation upon AGS exposure. Furthermore, the effects of acetaldehyde were reproduced by treatment with ER stress inducers, thapsigargin and tunicamycin, which also decreased the display of this complex and MHC class I turnover in HepG2.2.15 cells and HBV-infected primary human hepatocytes. Taken together, alcohol-induced ER stress and Golgi fragmentation contribute to the suppression of HBV peptide-MHC class I complex presentation on HBV+ hepatocytes, which may diminish their recognition by CTLs and promote persistence of HBV infection in hepatocytes.NEW & NOTEWORTHY Our current findings show that acetaldehyde accelerates endoplasmic reticulum (ER) stress by activating the unfolded protein response arms inositol-requiring enzyme 1α-X-box binding protein 1 and activation transcription factor (ATF)6α but not phospho PKR-like ER kinase-p eukaryotic initiation factor 2α-ATF4-C/EBP homologous protein in hepatitis B virus (HBV)-transfected HepG2.2.15 cells. It also potentiates Golgi fragmentation, as evident by punctate distribution of Golgi proteins, GM130, trans-Golgi network 46, and Giantin. While concomitantly increasing HBV DNA and HBV surface antigen titers, acetaldehyde-induced ER stress suppresses the presentation of HBV peptide-major histocompatibility complex I complexes on hepatocyte surfaces, thereby promoting the persistence of HBV infection in the liver.


Assuntos
Apresentação de Antígeno/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Complexo de Golgi/efeitos dos fármacos , Vírus da Hepatite B/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Fígado/virologia , Acetaldeído , Estresse do Retículo Endoplasmático/genética , Expressão Gênica/efeitos dos fármacos , Complexo de Golgi/ultraestrutura , Antígeno HLA-A2/análise , Células Hep G2 , Vírus da Hepatite B/genética , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Humanos , Fígado/imunologia , RNA Mensageiro/análise , Transfecção , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Resposta a Proteínas não Dobradas/genética
9.
CPT Pharmacometrics Syst Pharmacol ; 9(9): 534-541, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32697437

RESUMO

Monoclonal antibodies (mAbs) can be engineered to have "extended half-life" and "catch and release" properties to improve target coverage. We have developed a mAb physiologically-based pharmacokinetic model that describes intracellular trafficking, neonatal Fc receptor (FcRn) recycling, and nonspecific clearance of mAbs. We extended this model to capture target binding as a function of target affinity, expression, and turnover. For mAbs engineered to have an extended half-life, the model was able to accurately predict the terminal half-life (82% within 2-fold error of the observed value) in the human FcRn transgenic (Tg32) homozygous mouse and human. The model also accurately captures the trend in pharmacokinetic and target coverage data for a set of mAbs with differing catch and release properties in the Tg32 mouse. The mechanistic nature of this model allows us to explore different engineering techniques early in drug discovery, potentially expanding the number of "druggable" targets.


Assuntos
Anticorpos Monoclonais Humanizados/farmacocinética , Anticorpos Monoclonais/farmacocinética , Antígenos de Histocompatibilidade Classe I/genética , Receptores Fc/genética , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais Humanizados/imunologia , Anticorpos Monoclonais Humanizados/metabolismo , Anticorpos Amplamente Neutralizantes/imunologia , Anticorpos Amplamente Neutralizantes/metabolismo , Simulação por Computador , Desenvolvimento de Medicamentos , Descoberta de Drogas , Anticorpos Anti-HIV/imunologia , Anticorpos Anti-HIV/metabolismo , Meia-Vida , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Homozigoto , Humanos , Camundongos , Camundongos Transgênicos , Modelos Imunológicos , Mutação , Ligação Proteica/imunologia , Engenharia de Proteínas/métodos , Receptores Fc/efeitos dos fármacos
10.
Am J Transplant ; 19(7): 1881-1887, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30903736

RESUMO

IgG and albumin are the most abundant proteins in the circulation and have the longest half-lives. These properties are due to a unique receptor, the neonatal Fc receptor (FcRn). Although FcRn is named for its function of transferring IgG across the placenta from maternal to fetal circulation, FcRn functions throughout life to maintain IgG and albumin concentrations. FcRn protects IgG and albumin from intracellular degradation and recycles them back into the circulation. Clinical trials have confirmed that pathogenic antibodies can be depleted by blocking this homeostatic function of FcRn. Moreover, understanding the molecular interactions between IgG and FcRn has resulted in the design of therapeutic monoclonal antibodies with more efficacious pharmacokinetics. As a result of genetic engineering these monoclonals can be delivered at lower doses and at longer intervals. More recent findings have demonstrated that FcRn enhances phagocytosis by neutrophils, immune complex clearance by podocytes and antigen presentation by dendritic cells, macrophages, and B cells. This minireview highlights the relevance of FcRn to transplantation.


Assuntos
Produtos Biológicos/farmacologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Homeostase , Imunoglobulina G/metabolismo , Receptores Fc/metabolismo , Apresentação de Antígeno , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Humanos , Fagocitose , Receptores Fc/efeitos dos fármacos
11.
Immunogenetics ; 71(3): 263-271, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30220008

RESUMO

The immunoproteasome is expressed in cells of hematopoietic origin and is induced during inflammation by IFN-γ. Targeting the immunoproteasome with selective inhibitors has been shown to be therapeutically effective in pre-clinical models for autoimmune diseases, colitis-associated cancer formation, and transplantation. Immunoproteasome inhibition prevents activation and proliferation of lymphocytes, lowers MHC class I cell surface expression, reduces the expression of cytokines of activated immune cells, and curtails  T helper 1 and 17 cell differentiation. This might explain the in vivo efficacy of immunoproteasome inhibition in different pre-clinical disease models for autoimmunity, cancer, and transplantation. In this review, we summarize the effect of immunoproteasome inhibition in different animal models for transplantation.


Assuntos
Doenças Autoimunes/terapia , Rejeição de Enxerto/etiologia , Antígenos de Histocompatibilidade Classe I/imunologia , Transplante de Órgãos/efeitos adversos , Complexo de Endopeptidases do Proteassoma/imunologia , Inibidores de Proteassoma/uso terapêutico , Animais , Rejeição de Enxerto/tratamento farmacológico , Rejeição de Enxerto/enzimologia , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Transdução de Sinais
12.
Cancer Immunol Res ; 5(9): 778-789, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28724544

RESUMO

Natural killer (NK) cells are large granular lymphocytes that promote the antitumor response via communication with other cell types in the tumor microenvironment. Previously, we have shown that NK cells secrete a profile of immune stimulatory factors (e.g., IFNγ, MIP-1α, and TNFα) in response to dual stimulation with the combination of antibody (Ab)-coated tumor cells and cytokines, such as IL12. We now demonstrate that this response is enhanced in the presence of autologous monocytes. Monocyte enhancement of NK cell activity was dependent on cell-to-cell contact as determined by a Transwell assay. It was hypothesized that NK cell effector functions against Ab-coated tumor cells were enhanced via binding of MICA on monocytes to NK cell NKG2D receptors. Strategies to block MICA-NKG2D interactions resulted in reductions in IFNγ production. Depletion of monocytes in vivo resulted in decreased IFNγ production by murine NK cells upon exposure to Ab-coated tumor cells. In mice receiving trastuzumab and IL12 therapy, monocyte depletion resulted in significantly greater tumor growth in comparison to mock-depleted controls (P < 0.05). These data suggest that NK cell-monocyte interactions enhance NK cell antitumor activity in the setting of monoclonal Ab therapy for cancer. Cancer Immunol Res; 5(9); 778-89. ©2017 AACR.


Assuntos
Neoplasias da Mama/terapia , Antígenos de Histocompatibilidade Classe I/imunologia , Interferon gama/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/imunologia , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Feminino , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Humanos , Interleucina-12/imunologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Camundongos , Monócitos/imunologia , Monócitos/patologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/antagonistas & inibidores , Receptores Fc/administração & dosagem , Receptores Fc/imunologia , Trastuzumab/administração & dosagem , Trastuzumab/imunologia
13.
J Hematol Oncol ; 9(1): 134, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27903272

RESUMO

BACKGROUND: Anti-cancer immune responses may contribute to the control of tumors after conventional chemotherapy, and different observations have indicated that chemotherapeutic agents can induce immune responses resulting in cancer cell death and immune-stimulatory side effects. Increasing experimental and clinical evidence highlight the importance of natural killer (NK) cells in immune responses toward multiple myeloma (MM), and combination therapies able to enhance the activity of NK cells against MM are showing promise in treating this hematologic cancer. The epigenetic readers of acetylated histones bromodomain and extra-terminal (BET) proteins are critical regulators of gene expression. In cancer, they can upregulate transcription of key oncogenes such as cMYC, IRF4, and BCL-2. In addition, the activity of these proteins can regulate the expression of osteoclastogenic cytokines during cancer progression. Here, we investigated the effect of BET bromodomain protein inhibition, on the expression of NK cell-activating ligands in MM cells. METHODS: Five MM cell lines [SKO-007(J3), U266, RPMI-8226, ARP-1, JJN3] and CD138+ MM cells isolated from MM patients were used to investigate the activity of BET bromodomain inhibitors (BETi) (JQ1 and I-BET151) and of the selective BRD4-degrader proteolysis targeting chimera (PROTAC) (ARV-825), on the expression and function of several NK cell-activating ligands (NKG2DLs and DNAM-1Ls), using flow cytometry, real-time PCR, transient transfections, and degranulation assays. RESULTS: Our results indicate that inhibition of BET proteins via small molecule inhibitors or their degradation via a hetero-bifunctional PROTAC probe can enhance the expression of MICA, a ligand of the NKG2D receptor, in human MM cell lines and primary malignant plasma cells, rendering myeloma cells more efficient to activate NK cell degranulation. Noteworthy, similar results were obtained using selective CBP/EP300 bromodomain inhibition. Mechanistically, we found that BETi-mediated inhibition of cMYC correlates with the upregulation of miR-125b-5p and the downregulation of the cMYC/miR-125b-5p target gene IRF4, a transcriptional repressor of MICA. CONCLUSIONS: These findings provide new insights on the immuno-mediated antitumor activities of BETi and further elucidate the molecular mechanisms that regulate NK cell-activating ligand expression in MM.


Assuntos
Citotoxicidade Imunológica , Antígenos de Histocompatibilidade Classe I/metabolismo , Células Matadoras Naturais/imunologia , Mieloma Múltiplo/metabolismo , Proteínas/antagonistas & inibidores , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Humanos , Fatores Reguladores de Interferon/metabolismo , MicroRNAs/metabolismo , Mieloma Múltiplo/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Proteínas/fisiologia , Proteínas Proto-Oncogênicas c-myc/metabolismo
14.
BMC Nephrol ; 16: 60, 2015 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-25899529

RESUMO

BACKGROUND: The objective of this study was to investigate the influence of ß2-microglobulin (ß2-M) on the epithelial-mesenchymal transition (EMT) in renal tubular epithelial cells. METHODS: A human kidney proximal tubular cell line (HK-2) was used as the proximal tubular cell model. HK-2 cells were exposed to different concentrations of ß2-M (5, 10, 25, and 50 µM) for up to 24, 48 and 72 h. The effects of ß2-M on cell morphology were observed by phase contrast microscopy, and the possible associated mechanisms were assessed by immunofluorescence staining, western blot, RNA interference, immunoprecipitation, and induced coupled plasma mass spectroscopy. RESULTS: ß2-M induced marked morphological alterations in the HK-2 cells, accompanied by the increased expression of extracellular matrix components and α-smooth muscle actin (α-SMA), vimentin and fibronectin and the reduced expression of E-cadherin. Our results also revealed that ß2-M could induce the EMT in the HK-2 cells without significant affecting cell viability. Excess ß2-M in the HK-2 cells led to a decrease in iron and an increase in hypoxia inducible factor-1α (HIF-1α), which induced EMT in the HK-2 cells. Additionally, disrupting the function of the ß2-M/hemochromatosis (HFE) complex by HFE knockdown was sufficient to reverse ß2-M-mediated EMT in the HK-2 cells. CONCLUSION: These findings demonstrate that the activity of ß2-M is mediated by the ß2-M/HFE complex, which regulates intracellular iron homeostasis and HIF-1α and ultimately induces EMT in HK2 cells.


Assuntos
Células Epiteliais/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Túbulos Renais Proximais/efeitos dos fármacos , Microglobulina beta-2/farmacologia , Actinas/efeitos dos fármacos , Actinas/metabolismo , Western Blotting , Caderinas/efeitos dos fármacos , Caderinas/metabolismo , Linhagem Celular , Células Epiteliais/metabolismo , Fibronectinas/efeitos dos fármacos , Fibronectinas/metabolismo , Técnicas de Silenciamento de Genes , Proteína da Hemocromatose , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imunoprecipitação , Técnicas In Vitro , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/metabolismo , Espectrometria de Massas , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Microscopia de Contraste de Fase , Interferência de RNA , Vimentina/efeitos dos fármacos , Vimentina/metabolismo
15.
Transplantation ; 98(6): 660-5, 2014 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-24798311

RESUMO

BACKGROUND: Bortezomib has been used to reduce HLA antibody in patients either before transplantation or as treatment for antibody-mediated rejection (AMR). Reports on its efficacy show mixed results. The mechanism of action of this agent is via proteasome inhibition. The primary route of synthesis of HLA class I molecules is dependent on peptide generation by the proteasome, whereas that of class II is not. We observed a differential effect of bortezomib on class I versus class II antibody and hypothesized that this was related to a reduced expression of class I HLA antigens. METHODS: The effect of bortezomib on HLA antibody levels was evaluated in 13 patients who were desensitized for incompatible renal transplantation. We calculated the percent difference in HLA antibody level before and after bortezomib treatment and the impact of bortezomib on HLA expression in lymphocytes of healthy control subjects. RESULTS: On average, the level of HLA class I donor-specific antibody (DSA) decreased by 32%, whereas that of class II DSA increased by 29%. In vitro bortezomib treatment of lymphocytes resulted in a mean decrease of 23% in MHC class I expression on B lymphocytes and no change (+1.08%) in MHC class II expression (P=0.0003). The amount of intracellular class I molecules was reduced by a mean of 29% with bortezomib. CONCLUSION: These data indicate that bortezomib reduces HLA class I antibody more effectively than class II antibody. This difference may be due to the reduced expression of class I molecules resulting from treatment with this proteasome inhibitor.


Assuntos
Ácidos Borônicos/farmacologia , Antígenos de Histocompatibilidade Classe II/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Falência Renal Crônica/imunologia , Inibidores de Proteassoma/farmacologia , Pirazinas/farmacologia , Anticorpos/imunologia , Apoptose , Bortezomib , Membrana Celular/metabolismo , Feminino , Rejeição de Enxerto , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Falência Renal Crônica/cirurgia , Transplante de Rim , Linfócitos/imunologia , Masculino , Estudos Retrospectivos
16.
Nat Commun ; 5: 3633, 2014 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-24736453

RESUMO

Subsets of rodent neurons are reported to express major histocompatibility complex class I (MHC-I), but such expression has not been reported in normal adult human neurons. Here we provide evidence from immunolabel, RNA expression and mass spectrometry analysis of postmortem samples that human catecholaminergic substantia nigra and locus coeruleus neurons express MHC-I, and that this molecule is inducible in human stem cell-derived dopamine (DA) neurons. Catecholamine murine cultured neurons are more responsive to induction of MHC-I by gamma-interferon than other neuronal populations. Neuronal MHC-I is also induced by factors released from microglia activated by neuromelanin or alpha-synuclein, or high cytosolic DA and/or oxidative stress. DA neurons internalize foreign ovalbumin and display antigen derived from this protein by MHC-I, which triggers DA neuronal death in the presence of appropriate cytotoxic T cells. Thus, neuronal MHC-I can trigger antigenic response, and catecholamine neurons may be particularly susceptible to T-cell-mediated cytotoxic attack.


Assuntos
Neurônios Adrenérgicos/metabolismo , Neurônios Dopaminérgicos/metabolismo , Genes MHC Classe I , Antígenos de Histocompatibilidade Classe I/genética , Locus Cerúleo/metabolismo , Microglia/metabolismo , RNA Mensageiro/metabolismo , Substância Negra/metabolismo , Neurônios Adrenérgicos/efeitos dos fármacos , Idoso , Idoso de 80 Anos ou mais , Animais , Sobrevivência Celular , Dopamina/metabolismo , Neurônios Dopaminérgicos/efeitos dos fármacos , Feminino , Perfilação da Expressão Gênica , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe I/metabolismo , Humanos , Interferon gama/farmacologia , Masculino , Melaninas/metabolismo , Camundongos , Pessoa de Meia-Idade , Estresse Oxidativo , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , RNA Mensageiro/efeitos dos fármacos , Linfócitos T Citotóxicos , alfa-Sinucleína/metabolismo
17.
World J Urol ; 31(2): 325-30, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22383129

RESUMO

OBJECTIVE: Phosphodiesterases (PDEs) play a role in controlling cyclic nucleotide action, including cyclic guanosine monophosphate (cGMP). Previous studies have ascribed a protective role of cGMP signaling on hypoxia-mediated cancer progression. Herein, we determine their potential role in hypoxia-mediated chemoresistance and immune escape. MATERIALS AND METHODS: Phosphodiesterase assays were used to measure PDE activity in prostate cancer cell lines (DU145, PC3). Immunoblots were performed to determine the presence of PDEs in human prostate tissue samples. The effect of PDE inhibition on hypoxia-induced chemoresistance (compared to normoxic controls, 20% O2) was determined using clonogenic assays. Flow cytometry was used to determine the effects of PDE inhibition on surface MHC class I-related chain A (MICA), a natural killer (NK) cell-activating ligand. A mouse model was used to evaluate the in vivo effects of PDE inhibition on the growth of human prostate cancer cells. RESULTS: PDE5 and PDE11 were the most prominent PDEs in the cell lines, representing between 86 and 95% of the total cGMP-specific PDE activity. Treatment of DU-145 cells with a PDE inhibitor significantly reduced the hypoxia-associated acquisition of resistance to doxorubicin, with a mean 51% reduction in surviving fraction compared to controls (p < 0.001, ANOVA). As well, PDE inhibition completely reversed (p = 0.02, ANOVA) hypoxia-induced shedding of the immune stimulatory molecule, MICA, and attenuated the growth of human prostate tumor xenografts in an NK cell-competent murine model (p = 0.03, Wilcoxon, Mann-Whitney). CONCLUSIONS: These results suggest a rationale for future studies on the potential therapeutic applications of PDE inhibitors in men with prostate cancer.


Assuntos
Adenocarcinoma/enzimologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe I/metabolismo , Inibidores de Fosfodiesterase/farmacologia , Diester Fosfórico Hidrolases/metabolismo , Neoplasias da Próstata/enzimologia , Evasão Tumoral/efeitos dos fármacos , 3',5'-GMP Cíclico Fosfodiesterases , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/imunologia , Animais , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Ensaio de Unidades Formadoras de Colônias , Nucleotídeo Cíclico Fosfodiesterase do Tipo 1/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5/metabolismo , Modelos Animais de Doenças , Doxorrubicina/uso terapêutico , Ensaios Enzimáticos , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Humanos , Masculino , Camundongos , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/imunologia , Evasão Tumoral/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Burns ; 38(7): 1027-34, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22455798

RESUMO

Organ protection is desirable in severe burn/scald injuries, and damage mechanisms and thus effective therapies following scald injury have not been fully elucidated. Our aim was to examine the beneficial effects of ulinastatin on pulmonary damage associated with scald injury. Lewis rats were subjected to 30% total body surface area (TBSA) scald injury and were randomly divided into a burn control (S group) and an ulinastatin-treated group (U group). Lung malondialdehyde (MDA) and superoxide dismutase (SOD) levels were determined, and the lungs were examined histologically with immunohistochemistry (IHC) as well for the major histocompatibility complex (MHC) class I chain-related antigen A (MICA) and Bcl-2 at 24, 48 and 72 h after the injury. The expression of spleen human leucocyte antigen-DR (HLA-DR) was detected by immunohistochemistry analysis. Selectins and adhesion molecules in lungs and serum were also detected. The lung injury degree was represented as wet/dry (W/D) values and alveolar thickness. Ulinastatin decreased MDA levels and ameliorated the down-regulation of SOD activity. MICA was up-regulated after the scald, and this up-regulation was greatly diminished by ulinastatin. Bcl-2 was up-regulated after the scald, especially in the U group. The spleen HLA-DR expression demonstrated the immunoregulatory effects of ulinastatin, which effectively protected the pulmonary tissues from scald-induced injury. Our results demonstrated that pulmonary damage was associated with autoimmunity and oxidant attack after severe scald. Ulinastatin exhibits significant protective effects on these effects.


Assuntos
Queimaduras/complicações , Glicoproteínas/uso terapêutico , Lesão Pulmonar/prevenção & controle , Inibidores da Tripsina/uso terapêutico , Animais , Queimaduras/metabolismo , Regulação para Baixo , Antígenos HLA-DR/efeitos dos fármacos , Antígenos HLA-DR/metabolismo , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe I/metabolismo , Imuno-Histoquímica , Lesão Pulmonar/complicações , Lesão Pulmonar/metabolismo , Masculino , Malondialdeído/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Endogâmicos Lew , Selectinas/efeitos dos fármacos , Selectinas/metabolismo , Superóxido Dismutase/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Regulação para Cima , Molécula 1 de Adesão de Célula Vascular/efeitos dos fármacos , Molécula 1 de Adesão de Célula Vascular/metabolismo
19.
Immunology ; 135(2): 133-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22044191

RESUMO

Many MHC class I molecules contain unpaired cysteine residues in their cytoplasmic tail domains, the function of which remains relatively uncharacterized. Recently, it has been shown that in the small secretory vesicles known as exosomes, fully folded MHC class I dimers can form through a disulphide bond between the cytoplasmic tail domain cysteines, induced by the low levels of glutathione in these extracellular vesicles. Here we address whether similar MHC class I dimers form in whole cells by alteration of the redox environment. Treatment of the HLA-B27-expressing Epstein-Barr virus-transformed B-cell line Jesthom, and the leukaemic T-cell line CEM transfected with HLA-B27 with the strong oxidant diamide, and the apoptosis-inducing and glutathione-depleting agents hydrogen peroxide and thimerosal, induced MHC class I dimers. Furthermore, induction of apoptosis by cross-linking FasR/CD95 on CEM cells with monoclonal antibody CH-11 also induced MHC class I dimers. As with exosomal MHC class I dimers, the formation of these structures on cells is controlled by the cysteine at position 325 in the cytoplasmic tail domain of HLA-B27. Therefore, the redox environment of cells intimately controls induction of MHC class I dimers, the formation of which may provide novel structures for recognition by the immune system.


Assuntos
Apoptose/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Apoptose/efeitos dos fármacos , Linhagem Celular , Dimerização , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/farmacologia , Oxirredução , Timerosal/farmacologia , Receptor fas/imunologia
20.
Turk J Gastroenterol ; 22(4): 400-7, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21948571

RESUMO

BACKGROUND/AIMS: Hepatic stellate cells play an important role as the major source of fibrillar and non-fibrillar matrix proteins in the process of liver fibrosis. Natural killer cells have an anti-fibrotic effect through the killing of activated hepatic stellate cells. Major histocompatibility complex class I-related molecules, MICA and MICB, function as ligands for the NKG2D receptor and play an important role in hepatic stellate cells susceptibility to natural killer cells during hepatic inflammation. The aim of this study was therefore to investigate the effect of vitamin D2 and free fatty acids on stress ligands and pro-fibrotic activity in LX-2 cells and human primary hepatic stellate cells. METHODS: LX-2 cells and primary human hepatic stellate cells were treated with vitamin D2 (10-6 M) and free fatty acids at different concentrations (0.25 mM, 0.5 mM, and 1 mM) for 24 hours, and expressions of the stress ligands MICA/B as well as of transforming growth factor-ß, α-smooth muscle actin and collagen 1α were assessed by quantitative real time-polymerase chain reaction. RESULTS: Treatment of cells with 0.5 mM and 1 mM free fatty acids induced α-smooth muscle actin and transforming growth factor-ß expression in LX-2 cells. Moreover, 1 mM free fatty acids resulted in increased expression of MICA. Surprisingly, collagen 1α expression was reduced after addition of free fatty acids. MICA/B expression in primary hepatic stellate cells was not affected by free fatty acids treatment. Vitamin D2 treatment significantly downregulated the free fatty acids-induced expression of transforming growth factor-ß and α-smooth muscle actin in LX-2 cells. Further, in hepatic stellate cells, a significant decrease in MICA/B mRNA with vitamin D2, independent of free fatty acids treatment, was detectable. CONCLUSIONS: These results indicate that vitamin D2 may reduce inflammatory and pro-fibrogenic activity of stellate cells in vitro.


Assuntos
Actinas/metabolismo , Colágeno Tipo I/metabolismo , Ergocalciferóis/farmacologia , Ácidos Graxos não Esterificados/farmacologia , Células Estreladas do Fígado/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe I/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Actinas/genética , Linhagem Celular , Colágeno Tipo I/genética , Expressão Gênica , Células Estreladas do Fígado/metabolismo , Antígenos de Histocompatibilidade Classe I/efeitos dos fármacos , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Estresse Oxidativo/efeitos dos fármacos , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fator de Crescimento Transformador beta/genética , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...