Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
1.
Article in English | MEDLINE | ID: mdl-34045306

ABSTRACT

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.


Subject(s)
Antibodies, Blocking/administration & dosage , Antibodies, Monoclonal/administration & dosage , Arthrogryposis/prevention & control , Autoantibodies/drug effects , Histocompatibility Antigens Class I/drug effects , Maternal-Fetal Exchange , Receptors, Fc/drug effects , Animals , Animals, Newborn , Disease Models, Animal , Female , Humans , Immunoglobulin G , Mice , Mice, Inbred ICR , Pregnancy , Receptors, Cholinergic/immunology
2.
Adv Drug Deliv Rev ; 175: 113778, 2021 08.
Article in English | MEDLINE | ID: mdl-33887405

ABSTRACT

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.


Subject(s)
Diabetes Mellitus/drug therapy , Histocompatibility Antigens Class I/drug effects , Hypoglycemic Agents/administration & dosage , Nanoparticle Drug Delivery System , Receptors, Fc/drug effects , Administration, Oral , Animals , Diabetes Mellitus/pathology , Drug Delivery Systems , Fibrosis , Humans , Hypoglycemic Agents/therapeutic use , Nanoparticle Drug Delivery System/therapeutic use
3.
Drug Resist Updat ; 56: 100752, 2021 05.
Article in English | MEDLINE | ID: mdl-33765484

ABSTRACT

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.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Autophagy/physiology , Tumor Microenvironment/physiology , Autophagy/drug effects , Autophagy/immunology , Cell Death/physiology , Histocompatibility Antigens Class I/drug effects , Humans , Neoplasms/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
4.
Transfusion ; 61(4): 1222-1234, 2021 04.
Article in English | MEDLINE | ID: mdl-33580979

ABSTRACT

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.


Subject(s)
Citric Acid/adverse effects , Histocompatibility Antigens Class I/drug effects , Platelet Transfusion/methods , Severe Combined Immunodeficiency/chemically induced , Antibodies/immunology , Blood Grouping and Crossmatching/methods , Blood Platelets/radiation effects , Female , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/radiation effects , Histocompatibility Testing/economics , Histocompatibility Testing/methods , Humans , P-Selectin/metabolism , Platelet Transfusion/adverse effects , Plateletpheresis/methods , Tetraspanin 30/metabolism , Thrombelastography/methods , Thrombocytopenia/therapy , Up-Regulation/genetics
5.
Clin Transl Oncol ; 23(1): 110-121, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32661823

ABSTRACT

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.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Deoxycytidine/analogs & derivatives , Immunomodulation/drug effects , Pancreatic Neoplasms/immunology , Annexin A5/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Dendritic Cells/drug effects , Dendritic Cells/immunology , Deoxycytidine/pharmacology , Drug Interactions/immunology , Histocompatibility Antigens Class I/drug effects , Histocompatibility Antigens Class I/metabolism , Humans , Immunomodulation/immunology , Interferon-gamma/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Oxaliplatin/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Zoledronic Acid/pharmacology , Gemcitabine
6.
CPT Pharmacometrics Syst Pharmacol ; 9(11): 649-658, 2020 11.
Article in English | MEDLINE | ID: mdl-33021075

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents, Immunological/pharmacokinetics , Multiple Myeloma/drug therapy , Aged , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/administration & dosage , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/therapeutic use , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Body Weight/drug effects , Case-Control Studies , Clinical Trials as Topic , Dexamethasone/administration & dosage , Dexamethasone/therapeutic use , Drug Interactions , Female , Histocompatibility Antigens Class I/drug effects , Histocompatibility Antigens Class I/metabolism , Humans , Male , Multiple Myeloma/immunology , Myeloma Proteins/drug effects , Neoplasm Recurrence, Local/drug therapy , Pharmaceutical Preparations , Receptors, Fc/drug effects , Receptors, Fc/metabolism , Recurrence , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Time Factors , beta 2-Microglobulin/drug effects , beta 2-Microglobulin/metabolism
7.
Sci Rep ; 10(1): 15556, 2020 09 23.
Article in English | MEDLINE | ID: mdl-32968163

ABSTRACT

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.


Subject(s)
Amidohydrolases/genetics , Histocompatibility Antigens Class I/genetics , Tissue Inhibitor of Metalloproteinase-3/genetics , Benzamides/chemistry , Benzamides/pharmacology , Carbamates/chemistry , Carbamates/pharmacology , Cell Line, Tumor , Culture Media/chemistry , Culture Media/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Histocompatibility Antigens Class I/drug effects , Humans , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Neoplasms/drug therapy , Neoplasms/genetics , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology
8.
Am J Physiol Gastrointest Liver Physiol ; 319(4): G432-G442, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32755306

ABSTRACT

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.


Subject(s)
Antigen Presentation/drug effects , Endoplasmic Reticulum Stress/drug effects , Golgi Apparatus/drug effects , Hepatitis B virus/immunology , Histocompatibility Antigens Class I/immunology , Liver/virology , Acetaldehyde , Endoplasmic Reticulum Stress/genetics , Gene Expression/drug effects , Golgi Apparatus/ultrastructure , HLA-A2 Antigen/analysis , Hep G2 Cells , Hepatitis B virus/genetics , Histocompatibility Antigens Class I/drug effects , Humans , Liver/immunology , RNA, Messenger/analysis , Transfection , Unfolded Protein Response/drug effects , Unfolded Protein Response/genetics
9.
CPT Pharmacometrics Syst Pharmacol ; 9(9): 534-541, 2020 09.
Article in English | MEDLINE | ID: mdl-32697437

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacokinetics , Antibodies, Monoclonal/pharmacokinetics , Histocompatibility Antigens Class I/genetics , Receptors, Fc/genetics , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/metabolism , Broadly Neutralizing Antibodies/immunology , Broadly Neutralizing Antibodies/metabolism , Computer Simulation , Drug Development , Drug Discovery , HIV Antibodies/immunology , HIV Antibodies/metabolism , Half-Life , Histocompatibility Antigens Class I/drug effects , Homozygote , Humans , Mice , Mice, Transgenic , Models, Immunological , Mutation , Protein Binding/immunology , Protein Engineering/methods , Receptors, Fc/drug effects
10.
Am J Transplant ; 19(7): 1881-1887, 2019 07.
Article in English | MEDLINE | ID: mdl-30903736

ABSTRACT

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.


Subject(s)
Biological Products/pharmacology , Histocompatibility Antigens Class I/metabolism , Homeostasis , Immunoglobulin G/metabolism , Receptors, Fc/metabolism , Antigen Presentation , Histocompatibility Antigens Class I/drug effects , Humans , Phagocytosis , Receptors, Fc/drug effects
11.
Immunogenetics ; 71(3): 263-271, 2019 03.
Article in English | MEDLINE | ID: mdl-30220008

ABSTRACT

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.


Subject(s)
Autoimmune Diseases/therapy , Graft Rejection/etiology , Histocompatibility Antigens Class I/immunology , Organ Transplantation/adverse effects , Proteasome Endopeptidase Complex/immunology , Proteasome Inhibitors/therapeutic use , Animals , Graft Rejection/drug therapy , Graft Rejection/enzymology , Histocompatibility Antigens Class I/drug effects , Histocompatibility Antigens Class I/metabolism , Humans , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , Signal Transduction
12.
Cancer Immunol Res ; 5(9): 778-789, 2017 09.
Article in English | MEDLINE | ID: mdl-28724544

ABSTRACT

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.


Subject(s)
Breast Neoplasms/therapy , Histocompatibility Antigens Class I/immunology , Interferon-gamma/immunology , NK Cell Lectin-Like Receptor Subfamily K/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Histocompatibility Antigens Class I/drug effects , Humans , Interleukin-12/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Mice , Monocytes/immunology , Monocytes/pathology , NK Cell Lectin-Like Receptor Subfamily K/antagonists & inhibitors , Receptors, Fc/administration & dosage , Receptors, Fc/immunology , Trastuzumab/administration & dosage , Trastuzumab/immunology
13.
J Hematol Oncol ; 9(1): 134, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27903272

ABSTRACT

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.


Subject(s)
Cytotoxicity, Immunologic , Histocompatibility Antigens Class I/metabolism , Killer Cells, Natural/immunology , Multiple Myeloma/metabolism , Proteins/antagonists & inhibitors , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class I/drug effects , Humans , Interferon Regulatory Factors/metabolism , MicroRNAs/metabolism , Multiple Myeloma/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Proteins/physiology , Proto-Oncogene Proteins c-myc/metabolism
14.
BMC Nephrol ; 16: 60, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25899529

ABSTRACT

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.


Subject(s)
Epithelial Cells/drug effects , Epithelial-Mesenchymal Transition/drug effects , Kidney Tubules, Proximal/drug effects , beta 2-Microglobulin/pharmacology , Actins/drug effects , Actins/metabolism , Blotting, Western , Cadherins/drug effects , Cadherins/metabolism , Cell Line , Epithelial Cells/metabolism , Fibronectins/drug effects , Fibronectins/metabolism , Gene Knockdown Techniques , Hemochromatosis Protein , Histocompatibility Antigens Class I/drug effects , Histocompatibility Antigens Class I/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunoprecipitation , In Vitro Techniques , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/metabolism , Mass Spectrometry , Membrane Proteins/drug effects , Membrane Proteins/metabolism , Microscopy, Phase-Contrast , RNA Interference , Vimentin/drug effects , Vimentin/metabolism
15.
Transplantation ; 98(6): 660-5, 2014 Sep 27.
Article in English | MEDLINE | ID: mdl-24798311

ABSTRACT

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.


Subject(s)
Boronic Acids/pharmacology , Histocompatibility Antigens Class II/drug effects , Histocompatibility Antigens Class I/drug effects , Kidney Failure, Chronic/immunology , Proteasome Inhibitors/pharmacology , Pyrazines/pharmacology , Antibodies/immunology , Apoptosis , Bortezomib , Cell Membrane/metabolism , Female , Graft Rejection , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Humans , Kidney Failure, Chronic/surgery , Kidney Transplantation , Lymphocytes/immunology , Male , Retrospective Studies
16.
Nat Commun ; 5: 3633, 2014 Apr 16.
Article in English | MEDLINE | ID: mdl-24736453

ABSTRACT

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.


Subject(s)
Adrenergic Neurons/metabolism , Dopaminergic Neurons/metabolism , Genes, MHC Class I , Histocompatibility Antigens Class I/genetics , Locus Coeruleus/metabolism , Microglia/metabolism , RNA, Messenger/metabolism , Substantia Nigra/metabolism , Adrenergic Neurons/drug effects , Aged , Aged, 80 and over , Animals , Cell Survival , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Female , Gene Expression Profiling , Histocompatibility Antigens Class I/drug effects , Histocompatibility Antigens Class I/metabolism , Humans , Interferon-gamma/pharmacology , Male , Melanins/metabolism , Mice , Middle Aged , Oxidative Stress , Parkinson Disease/genetics , Parkinson Disease/metabolism , RNA, Messenger/drug effects , T-Lymphocytes, Cytotoxic , alpha-Synuclein/metabolism
17.
World J Urol ; 31(2): 325-30, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22383129

ABSTRACT

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.


Subject(s)
Adenocarcinoma/enzymology , Drug Resistance, Neoplasm/drug effects , Histocompatibility Antigens Class I/metabolism , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Prostatic Neoplasms/enzymology , Tumor Escape/drug effects , 3',5'-Cyclic-GMP Phosphodiesterases , Adenocarcinoma/drug therapy , Adenocarcinoma/immunology , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Colony-Forming Units Assay , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Disease Models, Animal , Doxorubicin/therapeutic use , Enzyme Assays , Histocompatibility Antigens Class I/drug effects , Humans , Male , Mice , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/immunology , Tumor Escape/physiology , Xenograft Model Antitumor Assays
18.
Burns ; 38(7): 1027-34, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22455798

ABSTRACT

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.


Subject(s)
Burns/complications , Glycoproteins/therapeutic use , Lung Injury/prevention & control , Trypsin Inhibitors/therapeutic use , Animals , Burns/metabolism , Down-Regulation , HLA-DR Antigens/drug effects , HLA-DR Antigens/metabolism , Histocompatibility Antigens Class I/drug effects , Histocompatibility Antigens Class I/metabolism , Immunohistochemistry , Lung Injury/complications , Lung Injury/metabolism , Male , Malondialdehyde/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Inbred Lew , Selectins/drug effects , Selectins/metabolism , Superoxide Dismutase/drug effects , Superoxide Dismutase/metabolism , Up-Regulation , Vascular Cell Adhesion Molecule-1/drug effects , Vascular Cell Adhesion Molecule-1/metabolism
19.
Immunology ; 135(2): 133-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22044191

ABSTRACT

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.


Subject(s)
Apoptosis/immunology , Histocompatibility Antigens Class I/immunology , Apoptosis/drug effects , Cell Line , Dimerization , Histocompatibility Antigens Class I/drug effects , Humans , Hydrogen Peroxide/pharmacology , Oxidation-Reduction , Thimerosal/pharmacology , fas Receptor/immunology
20.
Turk J Gastroenterol ; 22(4): 400-7, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21948571

ABSTRACT

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.


Subject(s)
Actins/metabolism , Collagen Type I/metabolism , Ergocalciferols/pharmacology , Fatty Acids, Nonesterified/pharmacology , Hepatic Stellate Cells/drug effects , Histocompatibility Antigens Class I/metabolism , Transforming Growth Factor beta/metabolism , Actins/genetics , Cell Line , Collagen Type I/genetics , Gene Expression , Hepatic Stellate Cells/metabolism , Histocompatibility Antigens Class I/drug effects , Histocompatibility Antigens Class I/genetics , Humans , Oxidative Stress/drug effects , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Transforming Growth Factor beta/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...