Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
1.
Genes Dev ; 32(23-24): 1472-1484, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30463905

ABSTRACT

Modification of mRNA by N6-adenosine methylation (m6A) on internal bases influences gene expression in eukaryotes. How the dynamic genome-wide landscape of m6A-modified mRNAs impacts virus infection and host immune responses remains poorly understood. Here, we show that type I interferon (IFN) production triggered by dsDNA or human cytomegalovirus (HCMV) is controlled by the cellular m6A methyltrasferase subunit METTL14 and ALKBH5 demethylase. While METTL14 depletion reduced virus reproduction and stimulated dsDNA- or HCMV-induced IFNB1 mRNA accumulation, ALKBH5 depletion had the opposite effect. Depleting METTL14 increased both nascent IFNB1 mRNA production and stability in response to dsDNA. In contrast, ALKBH5 depletion reduced nascent IFNB1 mRNA production without detectably influencing IFN1B mRNA decay. Genome-wide transcriptome profiling following ALKBH5 depletion identified differentially expressed genes regulating antiviral immune responses, while METTL14 depletion altered pathways impacting metabolic reprogramming, stress responses, and aging. Finally, we determined that IFNB1 mRNA was m6A-modified within both the coding sequence and the 3' untranslated region (UTR). This establishes that the host m6A modification machinery controls IFNß production triggered by HCMV or dsDNA. Moreover, it demonstrates that responses to nonmicrobial dsDNA in uninfected cells, which shape host immunity and contribute to autoimmune disease, are regulated by enzymes controlling m6A epitranscriptomic changes.


Subject(s)
DNA/immunology , Gene Expression Regulation/genetics , Immune System/enzymology , Immunity, Innate/genetics , Interferon-beta/genetics , Methyltransferases/metabolism , AlkB Homolog 5, RNA Demethylase/genetics , AlkB Homolog 5, RNA Demethylase/metabolism , Animals , Cell Line , Chlorocebus aethiops , Cytomegalovirus/immunology , Gene Expression Profiling , Humans , Interferon-beta/metabolism , RNA Stability/genetics , Vero Cells , Virus Replication/genetics
2.
Immunology ; 164(4): 722-736, 2021 12.
Article in English | MEDLINE | ID: mdl-34534359

ABSTRACT

Bruton's tyrosine kinase (BTK) is a TEC kinase with a multifaceted role in B-cell biology and function, highlighted by its position as a critical component of the B-cell receptor signalling pathway. Due to its role as a therapeutic target in several haematological malignancies including chronic lymphocytic leukaemia, BTK has been gaining tremendous momentum in recent years. Within the immune system, BTK plays a part in numerous pathways and cells beyond B cells (i.e. T cells, macrophages). Not surprisingly, BTK has been elucidated to be a driving factor not only in lymphoproliferative disorders but also in autoimmune diseases and response to infection. To extort this role, BTK inhibitors such as ibrutinib have been developed to target BTK in other diseases. However, due to rising levels of resistance, the urgency to develop new inhibitors with alternative modes of targeting BTK is high. To meet this demand, an expanding list of BTK inhibitors is currently being trialled. In this review, we synopsize recent discoveries regarding BTK and its role within different immune cells and pathways. Additionally, we discuss the broad significance and relevance of BTK for various diseases ranging from haematology and rheumatology to the COVID-19 pandemic. Overall, BTK signalling and its targetable nature have emerged as immensely important for a wide range of clinical applications. The development of novel, more specific and less toxic BTK inhibitors could be revolutionary for a significant number of diseases with yet unmet treatment needs.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , B-Lymphocytes/enzymology , Immune System/enzymology , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/enzymology , Autoimmune Diseases/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , COVID-19/enzymology , COVID-19/immunology , Humans , Immune System/drug effects , Immune System/immunology , Lymphoproliferative Disorders/drug therapy , Lymphoproliferative Disorders/enzymology , Lymphoproliferative Disorders/immunology , Molecular Targeted Therapy , Protein Kinase Inhibitors/therapeutic use , Receptors, Antigen, B-Cell/metabolism , Receptors, Chemokine/metabolism , Signal Transduction , Toll-Like Receptors/metabolism , COVID-19 Drug Treatment
3.
Int J Mol Sci ; 22(24)2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34948077

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is the most common neurodegenerative disorder of motor neurons in adults, with a median survival of 3-5 years after appearance of symptoms, and with no curative treatment currently available. Frontotemporal dementia (FTD) is also an adult-onset neurodegenerative disease, displaying not only clinical overlap with ALS, but also significant similarities at genetic and pathologic levels. Apart from the progressive loss of neurons and the accumulation of protein inclusions in certain cells and tissues, both disorders are characterized by chronic inflammation mediated by activated microglia and astrocytes, with an early and critical impact of neurodegeneration along the disease course. Despite the progress made in the last two decades in our knowledge around these disorders, the underlying molecular mechanisms of such non-cell autonomous neuronal loss still need to be clarified. In particular, immune signaling kinases are currently thought to have a key role in determining the neuroprotective or neurodegenerative nature of the central and peripheral immune states in health and disease. This review provides a comprehensive and updated view of the proposed mechanisms, therapeutic potential, and ongoing clinical trials of immune-related kinases that have been linked to ALS and/or FTD, by covering the more established TBK1, RIPK1/3, RACK I, and EPHA4 kinases, as well as other emerging players in ALS and FTD immune signaling.


Subject(s)
Amyotrophic Lateral Sclerosis/enzymology , Frontotemporal Dementia/enzymology , Immune System/enzymology , Inflammation , Phosphotransferases/metabolism , Signal Transduction , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/metabolism , Frontotemporal Dementia/drug therapy , Frontotemporal Dementia/immunology , Frontotemporal Dementia/metabolism , Humans , Immune System/metabolism , Phosphotransferases/antagonists & inhibitors
4.
Fish Physiol Biochem ; 46(4): 1409-1420, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32240445

ABSTRACT

A 10-week feeding trial was conducted to investigate the effects of dietary carbohydrate-to-lipid (CHO:L) ratios on glycogen content, hematological indices, liver, and intestinal enzyme activity of sub-adult grouper Epinephelus coioides. Five iso-nitrogenous (496.0 g kg-1 protein) and iso-energetic (21.6 KJ g-1 gross energy) diets with varying CHO: L ratios of 0.65 (D1), 1.31 (D2), 2.33 (D3), 4.24 (D4), and 8.51 (D5), respectively, were fed to triplicate groups of 20 fish (average 275.1 ± 1.86 g). Results showed that the weight gain rate (WGR), specific growth rate (SGR), and protein efficiency ratio (PER) of sub-adult grouper increased and then stable when dietary CHO:L ratios reach D4 (CHO:L = 4.24). The trend of feed conversion ratio (FCR) was opposite to PER. Along with the dietary CHO:L ratios, the liver and muscle glycogen level increased gradually. Plasma triglycerides (TG) and glucose (GLU) were all maximized at D5 (CHO:L = 8.51) group, cholesterol (CHOL) at D4 (CHO:L = 4.24) group. Digestive enzyme activities were significantly affected by dietary CHO:L ratios. Liver hexokinase (HK), alkaline phosphatase (AKP), and glucose-6-phosphate dehydrogenase (G6PDH) activity increased significantly as CHO:L ratios increased. Liver lysozyme (LYZ) and superoxide dismutase (SOD) activity of sub-adult grouper fed the D4 diet was significantly higher than that of the D2 (CHO:L = 1.31) diet. The trend of acid phosphatase (ACP) is opposite to AKP. The regression model analysis showed that the most suitable dietary CHO:L ratio to reach the highest SGR is 6.06.


Subject(s)
Bass/physiology , Dietary Carbohydrates/administration & dosage , Dietary Fats/administration & dosage , Lipids/administration & dosage , Analysis of Variance , Animals , Bass/blood , Bass/growth & development , Bass/immunology , Correlation of Data , Digestion/drug effects , Gastrointestinal Tract/physiology , Glycogen/metabolism , Immune System/enzymology , Liver/metabolism , Regression Analysis
5.
Adv Exp Med Biol ; 1172: 207-226, 2019.
Article in English | MEDLINE | ID: mdl-31628658

ABSTRACT

The NF-κB (Nuclear Factor kappa B) transcription factor plays crucial roles in the regulation of numerous biological processes including development of the immune system, inflammation, and innate and adaptive immune responses. Control over the immune cell functions of NF-κB results from signaling through one of two different routes: the canonical and noncanonical NF-κB signaling pathways. Present at the end of both pathways are the proteins NF-κB, IκB, and the IκB kinase (IKK). These proteins work together to deliver the myriad outcomes that influence context-dependent transcriptional control in immune cells. In the present chapter, we review the structural information available on NF-κB, IκB, and IKK, the critical terminal components of the NF-κB signaling, in relation to their physiological function.


Subject(s)
I-kappa B Kinase , I-kappa B Proteins , Immune System , NF-kappa B , Signal Transduction , Animals , Humans , I-kappa B Kinase/immunology , I-kappa B Proteins/immunology , Immune System/enzymology , NF-kappa B/immunology , Phosphorylation , Signal Transduction/immunology
6.
Nat Rev Immunol ; 6(7): 532-40, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16799472

ABSTRACT

Signals emanating from many cell-surface receptors and environmental cues converge on mitogen-activated protein kinases (MAPKs), which in turn phosphorylate and activate various transcription factors and other molecular effectors. Members of the p38 MAPK family, which respond to pro-inflammatory cytokines and cellular stresses, are typically activated by serial phosphorylation and activation of upstream kinases (the MAPK cascade). In this Review, I highlight the recent studies that indicate that p38-subfamily members can also be activated by non-canonical mechanisms, at least one of which seems to have an important role in antigen-receptor-activated T cells. These alternative pathways might have particular relevance for cells that participate in immune and inflammatory responses.


Subject(s)
Enzyme Activation/immunology , Immune System/enzymology , Models, Immunological , Signal Transduction/immunology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Humans , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , p38 Mitogen-Activated Protein Kinases/immunology
7.
Nat Rev Immunol ; 6(4): 308-17, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16557262

ABSTRACT

Caspases are responsible for crucial aspects of inflammation and immune-cell death that are disrupted in a number of genetic autoimmune and autoinflammatory diseases. The caspase family of proteases can be divided into pro-apoptotic and pro-inflammatory members based on their substrate specificity and participation in separate signalling cascades. However, as discussed here, evidence has emerged over the past few years that a number of the caspases thought to be involved solely in apoptosis also contribute to specific aspects of immune-cell development, activation and differentiation, and can even protect cells from some forms of cell death.


Subject(s)
Apoptosis/physiology , Caspases/physiology , Immune System/physiology , Animals , Apoptosis/immunology , Caspases/metabolism , Cell Differentiation/immunology , Cell Differentiation/physiology , Cell Proliferation , Cell Survival/immunology , Cell Survival/physiology , Drosophila/immunology , Drosophila/physiology , Humans , Immune System/cytology , Immune System/enzymology , Lymphocyte Activation/physiology , Models, Biological , Models, Immunological , Signal Transduction/immunology , Signal Transduction/physiology
8.
Fish Shellfish Immunol ; 53: 50-7, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26973022

ABSTRACT

The environment in which teleosts exist can experience considerable change. Short-term changes can occur in relation to tidal movements or adverse weather events. Long-term changes can be caused by anthropogenic impacts such as climate change, which can result in changes to temperature, acidity, salinity and oxygen capacity of aquatic environments. These changes can have important impacts on the physiology of an animal, including its immune system. This can have consequences on the well-being of the animal and its ability to protect against pathogens. This review will look at recent investigations of these types of environmental change on the immune response in teleosts.


Subject(s)
Environment , Fishes/immunology , Immune System/immunology , Immunity, Innate , Animals , Fishes/microbiology , Fishes/parasitology , Immune System/enzymology
9.
J Exp Ther Oncol ; 11(1): 51-61, 2015.
Article in English | MEDLINE | ID: mdl-26259390

ABSTRACT

The aim of the study is to develop a technology for cost effective immunomodulator from natural products to combat adverse effects during cancer chemotherapy. In the present study, the immunomodulatory efficacy of Vivartana, a poly herbal formulation in immunosuppressed animal model induced by cyclophosphamide (CTX) and its comparison with standard herbal immunostimulators Chyawanprash and Brahma Rasayana was investigated. The effect of Vivartana (500 mg/kg x bw) (p.o.), Chyawanprash (20 mg/kg.bw) (p.o.) and Brahma Rasayana (20 mg/kg x bw) (p.o.) on hematological parameters, relative organ weight, Bone marrow cellularity and α-esterase activity were determined in Swiss albino mice by using the standard methods. Among the herbal formulations Vivartana showed the maximum number of leukocytes (13150 cells/mm3) on the 15th day. The leukocyte count in Vivartana treated CTX induced group shows significant increase (5375 cells/mm3) when compared with CTX alone induced group (3358 cells/mm3) on the same day. The Vivartana treated CTX induced group shows increase in the hemoglobin level compared with the CTX induced group. Moreover, Vivartana treatment prevented the loss of organ weight in the CTX induced group by the enhancement of spleenocytes on the 7th day and thymocytes on the 11th day. Similarly the lowered bone marrow cellularity and number of α-esterase positive cells in CTX induced group were restored in the Vivartana treatment. Treatment with vivartana also exhibits hepatoprotective activity by regulating the SGOT and SGPT levels in CTX induced group. The study indicates that Vivartana has the considerable potential as an immunostimulant and chemoprotectant against CTX induced immunosuppression in Swiss albino mice.


Subject(s)
Antineoplastic Agents, Alkylating/toxicity , Cyclophosphamide/toxicity , Immune System/drug effects , Immunologic Factors/pharmacology , Plant Preparations/pharmacology , Protective Agents/pharmacology , Animals , Body Weight/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/enzymology , Bone Marrow Cells/immunology , Cytoprotection , Female , Hemoglobins/metabolism , Immune System/enzymology , Immune System/immunology , Immunocompromised Host , Liver/drug effects , Liver/enzymology , Male , Mice , Organ Size/drug effects , Phytotherapy , Plants, Medicinal , Time Factors
10.
Pharmacol Rev ; 64(4): 1027-54, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23023033

ABSTRACT

The phosphoinositide 3-kinase/mammalian target of rapamycin/protein kinase B (PI3K/mTOR/Akt) signaling pathway is central to a plethora of cellular mechanisms in a wide variety of cells including leukocytes. Perturbation of this signaling cascade is implicated in inflammatory and autoimmune disorders as well as hematological malignancies. Proteins within the PI3K/mTOR/Akt pathway therefore represent attractive targets for therapeutic intervention. There has been a remarkable evolution of PI3K inhibitors in the past 20 years from the early chemical tool compounds to drugs that are showing promise as anticancer agents in clinical trials. The use of animal models and pharmacological tools has expanded our knowledge about the contribution of individual class I PI3K isoforms to immune cell function. In addition, class II and III PI3K isoforms are emerging as nonredundant regulators of immune cell signaling revealing potentially novel targets for disease treatment. Further complexity is added to the PI3K/mTOR/Akt pathway by a number of novel signaling inputs and feedback mechanisms. These can present either caveats or opportunities for novel drug targets. Here, we consider recent advances in 1) our understanding of the contribution of individual PI3K isoforms to immune cell function and their relevance to inflammatory/autoimmune diseases as well as lymphoma and 2) development of small molecules with which to inhibit the PI3K pathway. We also consider whether manipulating other proximal elements of the PI3K signaling cascade (such as class II and III PI3Ks or lipid phosphatases) are likely to be successful in fighting off different immune diseases.


Subject(s)
Autoimmune Diseases/enzymology , Hematologic Neoplasms/enzymology , Phosphatidylinositol 3-Kinases/immunology , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Enzyme Inhibitors/pharmacology , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/immunology , Humans , Immune System/enzymology , Inflammation/drug therapy , Inflammation/enzymology , Inflammation/immunology , Phosphatidylinositol 3-Kinases/chemistry , Phosphoinositide-3 Kinase Inhibitors , Protein Structure, Tertiary , Signal Transduction
11.
Postepy Biochem ; 61(2): 147-58, 2015.
Article in Polish | MEDLINE | ID: mdl-26689007

ABSTRACT

Heme oxygenase-1 (HO-1) is an enzyme degrading heme to three products - ferrous ions, carbon monoxide and biliverdin. Its function extends, however, far beyond removal of pro-oxidative heme from microenvironment. During the last few decades it was proven that apart from cytoprotective and antioxidative properties HO-1 regulates also a variety of cellular processes. It exerts an impact on both innate and adaptive immune response. HO-1 accelerates development of new blood vessels in a process called angiogenesis. Moreover, it controls cell cycle and depending on a cell type increases or decreases the rate of cell division. Finally, the most recent data indicate, that HO-1 regulates also differentiation of various stem and progenitor cells. Interestingly, that aspect of HO-1 function seems also to depend on cell type. In this review, both effects and mechanisms of above-mentioned processes in different cell types are discussed.


Subject(s)
Cell Differentiation , Heme Oxygenase-1/physiology , Immune System/enzymology , Neovascularization, Physiologic , Cell Cycle , Cytoprotection , Gene Expression Regulation , Heme Oxygenase-1/genetics , Heme Oxygenase-1/immunology , Humans
12.
Endocr J ; 61(6): 529-38, 2014.
Article in English | MEDLINE | ID: mdl-24614797

ABSTRACT

Obesity causes type 2 diabetes, atherosclerosis and cardiovascular diseases by inducing systemic insulin resistance. It is now recognized that obesity is related to chronic low-grade inflammation in adipose tissue. Specifically, activated immune cells infiltrate adipose tissue and cause inflammation. There is increasing evidence that activated macrophages accumulate in the hypertrophied adipose tissue of rodents and humans and induce systemic insulin resistance by secreting inflammatory cytokines. Accordingly, a better understanding of the molecular mechanisms underlying macrophage activation in adipose tissue will facilitate the development of new therapeutic strategies. Currently, little is known about the regulation of macrophage activation, although E3 ubiquitin ligase Casitas B-lineage lymphoma (Cbl)-b was identified recently as a novel negative regulator of macrophage activation in adipose tissue. Cbl-b, which is a suppressor of T- and B-cell activation, inhibits intracellular signal transduction by targeting some tyrosine kinases. Notably, preventing Cbl-b-mediated macrophage activation improves obesity-induced insulin resistance in mice. c-Cbl is another member of the Cbl family that is associated with insulin resistance in obesity. These reports suggest that Cbl-b and c-Cbl are potential therapeutic targets for treating obesity-induced insulin resistance. In this review, we focus on the importance of Cbl-b in macrophage activation in aging-induced and high-fat diet-induced obesity.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Insulin Resistance/genetics , Obesity/metabolism , Proto-Oncogene Proteins c-cbl/physiology , Aging/physiology , Animals , Diet, High-Fat , Humans , Immune System/enzymology , Immune System/metabolism , Macrophage Activation/genetics , Mice , Obesity/complications , Obesity/genetics
13.
Biochim Biophys Acta ; 1824(1): 105-12, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21664991

ABSTRACT

Cathepsin E is an intracellular aspartic proteinase of the pepsin superfamily, which is predominantly expressed in certain cell types, including the immune system cells and rapidly regenerating gastric mucosal and epidermal keratinocytes. The intracellular localization of this protein varies with different cell types. The endosomal localization is primarily found in antigen-presenting cells and gastric cells. The membrane association is observed with certain cell types such as erythrocytes, osteoclasts, gastric parietal cells and renal proximal tubule cells. This enzyme is also found in the endoplasmic reticulum, Golgi complex and cytosolic compartments in various cell types. In addition to its intracellular localization, cathepsin E occurs in the culture medium of activated phagocytes and cancer cells as the catalytically active enzyme. Its strategic expression and localization thus suggests the association of this enzyme with specific biological functions of the individual cell types. Recent genetic and pharmacological studies have particularly suggested that cathepsin E plays an important role in host defense against cancer cells and invading microorganisms. This review focuses emerging roles of cathepsin E in immune system cells and skin keratinocytes, and in host defense against cancer cells. This article is part of a Special Issue entitled: Proteolysis 50 years after the discovery of lysosome.


Subject(s)
Cathepsin E/physiology , Immune System/enzymology , Immune System/physiology , Animals , Antigen-Presenting Cells/enzymology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/physiology , Cathepsin E/genetics , Cathepsin E/metabolism , Humans , Immune System/metabolism , Models, Biological , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/metabolism , Skin/enzymology , Skin/immunology , Skin/metabolism , Skin Physiological Phenomena/genetics , Skin Physiological Phenomena/immunology
14.
Biochim Biophys Acta ; 1824(1): 186-94, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21801859

ABSTRACT

Influenza A virus (IAV) is one of the most common infectious pathogens in humans. Since the IVA genome does not have the processing protease for the viral hemagglutinin (HA) envelope glycoprotein precursors, entry of this virus into cells and infectious organ tropism of IAV are primarily determined by host cellular trypsin-type HA processing proteases. Several secretion-type HA processing proteases for seasonal IAV in the airway, and ubiquitously expressed furin and pro-protein convertases for highly pathogenic avian influenza (HPAI) virus, have been reported. Recently, other HA-processing proteases for seasonal IAV and HPAI have been identified in the membrane fraction. These proteases proteolytically activate viral multiplication at the time of viral entry and budding. In addition to the role of host cellular proteases in IAV pathogenicity, IAV infection results in marked upregulation of cellular trypsins and matrix metalloproteinase-9 in various organs and cells, particularly endothelial cells, through induced pro-inflammatory cytokines. These host cellular factors interact with each other as the influenza virus-cytokine-protease cycle, which is the major mechanism that induces vascular hyperpermeability and multiorgan failure in severe influenza. This mini-review discusses the roles of cellular proteases in the pathogenesis of IAV and highlights the molecular mechanisms of upregulation of trypsins as effective targets for the control of IAV infection. This article is part of a Special Issue entitled: Proteolysis 50 years after the discovery of lysosome.


Subject(s)
Host-Pathogen Interactions/immunology , Influenza, Human/complications , Influenza, Human/etiology , Multiple Organ Failure/etiology , Peptide Hydrolases/physiology , Animals , Antigen Presentation/physiology , Birds , Capillary Permeability/immunology , Capillary Permeability/physiology , Humans , Immune System/enzymology , Immune System/metabolism , Influenza A virus/immunology , Influenza A virus/metabolism , Influenza A virus/pathogenicity , Influenza A virus/physiology , Influenza in Birds/virology , Influenza, Human/enzymology , Models, Biological , Multiple Organ Failure/genetics , Multiple Organ Failure/immunology , Multiple Organ Failure/metabolism , Peptide Hydrolases/metabolism
15.
Biochem Biophys Res Commun ; 431(2): 152-7, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23313512

ABSTRACT

Xenopus tadpoles have high regenerative ability of amputated tails except during the 'refractory period', when the ability is transiently lost. We previously demonstrated that distinct immune responses occur in tail stumps between the refractory and pre/post-refractory regeneration periods. Furthermore, treatment with an immunosuppressant, FK506, restores the tail regenerative ability during the refractory period. Based on these findings, we previously proposed that autoreactive immune cells infiltrate the tail stumps to attack blastema cells as 'non-self' during the refractory period, resulting in the impaired regenerative ability. The immune cells that attack the blastema cells, however, remained unclear. Here we screened for genes whose expression in the tail stumps was altered by FK506 treatment during the refractory period and identified a Xenopus homolog of phytanoyl-CoA dioxygenase (PhyH)-like. XPhyH-like expression transiently increased in tail stumps after amputation during the refractory period, and was reduced by FK506 treatment. XPhyH-like expression in the whole tadpole body specifically increased during the refractory period and was enriched in the blood cell fraction. These findings suggest that XPhyH-like is expressed in autoreactive immune cells that are distributed in the whole body during the refractory period and transiently infiltrate the tail stumps to attack the blastema cells as 'non-self'.


Subject(s)
Dioxygenases/biosynthesis , Immune System/enzymology , Regeneration/immunology , Tail/physiology , Xenopus Proteins/biosynthesis , Xenopus laevis/growth & development , Animals , Dioxygenases/genetics , Gene Expression , Immunosuppressive Agents/pharmacology , Larva/enzymology , Larva/genetics , Larva/physiology , Regeneration/drug effects , Tacrolimus/pharmacology , Tail/enzymology , Xenopus Proteins/genetics , Xenopus laevis/genetics , Xenopus laevis/metabolism
16.
FASEB J ; 26(8): 3550-62, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22593545

ABSTRACT

Nicotinamide nucleotide transhydrogenase (NNT) is a mitochondrial redox-driven proton pump that couples the production of NADPH to the mitochondrial metabolic rate. In this study, we demonstrated for the first time that NNT has a significant effect in the modulation of the immune response and host defense against pathogens. We found that NNT mRNA is enriched in immune system-related tissues and regulated during macrophage activation. Overexpression of NNT in a macrophage cell-line resulted in decreased levels of reactive oxygen species (ROS) and nitric oxide upon induction of the macrophage inflammatory responses. These cells failed to fully activate MAPK signaling pathways, resulting in defective secretion of proinflammatory cytokines in response to LPS, and were inefficient in clearance of intracellular bacteria. We have shown that C57BL/6J mice, which have a deletion in the Nnt gene, exhibited greater resistance to acute pulmonary infection with Streptococcus pneumoniae. Macrophages from these mice generated more ROS and established a stronger inflammatory response to this pathogen. Our results demonstrate a novel role for NNT as a regulator of macrophage-mediated inflammatory responses.


Subject(s)
Inflammation/physiopathology , Macrophages/immunology , NADP Transhydrogenases/physiology , Animals , Cell Line , Immune System/enzymology , Lung/pathology , Macrophages/enzymology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phagocytosis/physiology , Pneumococcal Infections/pathology , Pneumococcal Infections/physiopathology , Pneumonia, Bacterial/etiology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/pharmacology
17.
Bioorg Med Chem ; 21(24): 7595-603, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24262887

ABSTRACT

Screening of a fragment library identified 2-hydrazinobenzothiazole as a potent inhibitor of indoleamine 2,3-dioxygenase 1 (IDO1), an enzyme expressed by tumours that suppresses the immune system. Spectroscopic studies indicated that 2-hydrazinobenzothiazole interacted with the IDO1 haem and in silico docking predicted that the interaction was through hydrazine. Subsequent studies of hydrazine derivatives identified phenylhydrazine (IC50=0.25 ± 0.07 µM) to be 32-fold more potent than 2-hydrazinobenzothiazole (IC50=8.0 ± 2.3 µM) in inhibiting rhIDO1 and that it inhibited cellular IDO1 at concentrations that were noncytotoxic to cells. Here, phenylhydrazine is shown to inhibit IDO1 through binding to haem.


Subject(s)
Drug Discovery , Enzyme Inhibitors/pharmacology , Hydrazines/pharmacology , Immune System/enzymology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Humans , Hydrazines/chemistry , Immune System/drug effects , Immune System/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Mice , Models, Molecular , Molecular Structure , Recombinant Proteins/metabolism , Structure-Activity Relationship
18.
Clin Exp Pharmacol Physiol ; 40(12): 885-94, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23662678

ABSTRACT

Identified over a decade ago, the with-no-lysine[K] kinases (WNKs) have been the subsequent focus of intense research into the renal handling of Na(+) , Cl(-) and K(+) and several rare monogenetic diseases. However, the potential extrarenal roles for WNKs have been less well explored. Thiazides and Gordon syndrome are known to have effects on bone mineral density, Ca(2+) and PO4 (3-) homeostasis, which were originally assumed to be an indirect effect through the kidney. However, current data suggest a complex and direct role for WNKs in the physiology of bone. The WNKs also modulate systemic blood pressure at several levels, including the vascular resistance vessels, where they cause vasoconstriction by altering the abundance of the transient receptor potential canonical channel 3 and/or phosphorylation of the Na(+) -K(+) -2Cl(-) cotransporter 1 in vascular smooth muscle cells. The WNKs and many of the cation-coupled Cl(-) cotransporters they regulate are highly expressed in the central nervous system and recent work suggests that WNK dysfunction may have a role in the development of autism, schizophrenia and hereditary sensory and autonomic neuropathy Type 2. Finally, the WNK-sterile 20 kinase signalling axis represents an evolutionarily ancient mechanism for maintaining osmotic homeostasis, but a rapidly expanding body of evidence also shows a role in immunity and cellular regulation.


Subject(s)
Bone and Bones/enzymology , Cardiovascular System/enzymology , Central Nervous System/enzymology , Immune System/enzymology , Intracellular Signaling Peptides and Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Autistic Disorder/enzymology , Homeostasis/physiology , Humans , Hypertension/enzymology , Intracellular Signaling Peptides and Proteins/genetics , Minor Histocompatibility Antigens , Protein Serine-Threonine Kinases/genetics , WNK Lysine-Deficient Protein Kinase 1
19.
Immunol Cell Biol ; 90(1): 95-100, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22124370

ABSTRACT

Several histone deacetylases (HDACs) are involved in the regulation of forkhead box protein P3 (FOXP3) expression and function by affecting features of FOXP3 protein stability. FOXP3, a forkhead family transcription factor specially expressed in regulatory T (Treg) cells, controls the expression of many key immune-regulatory genes. Treg cells are a population of T lymphocytes that have critical roles in the immune system homeostasis and tolerance to self and foreign antigens, the body's response to cancer and infectious agents. FOXP3 forms oligomeric complexes with other proteins, the components of which are believed to be regulated dynamically. In addition, HDAC activities influence FOXP3 interactions with other partners to form transcriptional regulatory complexes. By understanding the details of the biochemical and structural basis of the regulation of FOXP3 acetylation, therapeutic strategies for diseases related to Treg cells may emerge.


Subject(s)
Forkhead Transcription Factors/metabolism , Histone Deacetylases/metabolism , Immune System/immunology , T-Lymphocytes, Regulatory/immunology , Acetylation , Animals , Forkhead Transcription Factors/chemistry , Histone Acetyltransferases/metabolism , Humans , Immune System/enzymology , Immune System/metabolism , Protein Multimerization , T-Lymphocytes, Regulatory/metabolism
20.
J Transl Med ; 10: 247, 2012 Dec 11.
Article in English | MEDLINE | ID: mdl-23232072

ABSTRACT

BACKGROUND: Multiple myeloma (MM) is a plasma cell malignancy with a multifaceted immune dysfunction. Indoleamine 2,3-dioxygenase 1 (IDO1) degrades tryptophan into kynurenine (KYN), which inhibits effector T cells and promote regulatory T-cell (Treg) differentiation. It is presently unknown whether MM cells express IDO1 and whether IDO1 activity correlates with immune system impairment. METHODS: We investigated IDO1 expression in 25 consecutive patients with symptomatic MM and in 7 patients with either monoclonal gammopathy of unknown significance (MGUS; n=3) or smoldering MM (SMM; n=4). IDO1-driven tryptophan breakdown was correlated with the release of hepatocyte growth factor (HGF) and with the frequency of Treg cells and NY-ESO-1-specific CD8(+) T cells. RESULTS: KYN was increased in 75% of patients with symptomatic MM and correlated with the expansion of CD4(+)CD25(+)FoxP3(+) Treg cells and the contraction of NY-ESO-1-specific CD8(+) T cells. In vitro, primary MM cells promoted the differentiation of allogeneic CD4(+) T cells into bona fide CD4(+)CD25(hi)FoxP3(hi) Treg cells and suppressed IFN-γ/IL-2 secretion, while preserving IL-4 and IL-10 production. Both Treg expansion and inhibition of Th1 differentiation by MM cells were reverted, at least in part, by D,L-1-methyl-tryptophan, a chemical inhibitor of IDO. Notably, HGF levels were higher within the BM microenvironment of patients with IDO(+) myeloma disease compared with patients having IDO(-) MM. Mechanistically, the antagonism of MET receptor for HGF with SU11274, a MET inhibitor, prevented HGF-induced AKT phosphorylation in MM cells and translated into reduced IDO protein levels and functional activity. CONCLUSIONS: These data suggest that IDO1 expression may contribute to immune suppression in patients with MM and possibly other HGF-producing cancers.


Subject(s)
Immune System/abnormalities , Immune System/enzymology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Multiple Myeloma/enzymology , Multiple Myeloma/immunology , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation/immunology , Cell Line, Tumor , Cell Proliferation , Hepatocyte Growth Factor/metabolism , Humans , Interleukin-10/metabolism , Membrane Proteins/metabolism , Plasma Cells/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Transforming Growth Factor beta/metabolism , Tumor Burden/immunology
SELECTION OF CITATIONS
SEARCH DETAIL