Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 466
Filter
1.
Bioengineered ; 13(2): 2412-2429, 2022 02.
Article in English | MEDLINE | ID: mdl-35094638

ABSTRACT

This study investigated the role of ADAM metallopeptidase domain 12 (ADAM12) in clear cell renal cell carcinoma (ccRCC). The mRNA expression of ADAM12 was analyzed using The Cancer Genome Atlas (TCGA) database and the Gene Expression Omnibus (GEO) database, and the protein expression level of ADAM12 in renal clear cell carcinoma cell lines was detected by Western blot analysis. The Wilcoxon rank-sum test, logistic regression analysis, Cox regression analysis, and Kaplan-Meier analysis were used to assess the relationship between the clinicopathological characteristics and the prognosis of ccRCC patients and ADAM12 expression. The miRNAs and lncRNAs associated with ADAM12 were predicted, and a ceRNA network was constructed using the Starbase database. Gene Set Enrichment Analysis (GSEA) and Gene Ontology (GO) analysis were used to identify relevant pathways. The relationship between ADAM12 and immune infiltration and checkpoints was analyzed using the TIMER and Gene Expression Profiling Interactive Analysis (GEPIA) databases. The results showed that ADAM12 expression was increased in ccRCC tissues and cells and significantly correlated with patient gender, Tumor stage, Metastasis stage, Node stage, and clinical grade. Survival analysis showed that ccRCC patients with high ADAM12 expression had a low overall survival rate. Univariate and multivariate Cox regression analyses showed that ADAM12 was an independent prognostic factor. Enrichment analysis showed that ADAM12 expression was associated with immune-related pathways. Immune infiltration analysis showed that ADAM12 expression was related to immune cell infiltration, PD-1, PD-L1, and CTLA4. These results suggest that ADAM12 may be a potential diagnostic and prognostic biomarker for ccRCC.


Subject(s)
ADAM12 Protein/immunology , Carcinoma, Renal Cell/immunology , Gene Expression Regulation, Enzymologic/immunology , Gene Expression Regulation, Neoplastic/immunology , Kidney Neoplasms/immunology , Neoplasm Proteins/immunology , Carcinoma, Renal Cell/diagnosis , Cell Line, Tumor , Humans , Kidney Neoplasms/diagnosis , Prognosis
2.
Eur J Immunol ; 52(2): 338-351, 2022 02.
Article in English | MEDLINE | ID: mdl-34755333

ABSTRACT

PSMA3, a member of the proteasome subunit, has been shown to play a major player in protein degradation. Reportedly, PSMA3 functions as a negative regulator in various cancers including colon, pancreatic and gastric cancers. However, the contributions of PSMA3 to the progression of esophageal squamous cell carcinoma (ESCC) and the underlying mechanism remain unclear. Therefore, in this study, we investigated whether PSMA3 is involved in ESCC progression and the potential underlying mechanism. The results revealed that PSMA3 was highly expressed in the ESCC tumor tissues and functioned as a negative indicator according to the data from The Cancer Genome Atlas (TCGA)/Gene Expression Omnibus (GEO) datasets and clinical patients' samples. Pathway enrichment analysis showed that PSMA3 was closely correlated with ESCC cancer stemness and the inflammatory response; however, this correlation was absent after knockdown of PSMA3 in vitro. We further demonstrated that PSMA3 suppressed CD8+ T-cells infiltration depending on the C-C motif chemokine ligand 3 (CCL3)/C-C motif chemokine receptor 5 (CCR5) axis. Collectively, these results demonstrate the role of PSMA3 in ESCC cancer stemness and the negative regulation of CD8 T-cells infiltration mediated by PSMA3. The results of this study may provide a potential target for the immuno-oncology effect of PSMA3 in ESCC therapy.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Gene Expression Regulation, Enzymologic/immunology , Gene Expression Regulation, Neoplastic/immunology , Neoplasm Proteins , Cell Line, Tumor , Databases, Nucleic Acid , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/immunology , Esophageal Squamous Cell Carcinoma/enzymology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/immunology , Humans , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Proteasome Endopeptidase Complex/biosynthesis , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/immunology
3.
Proc Natl Acad Sci U S A ; 118(31)2021 08 03.
Article in English | MEDLINE | ID: mdl-34312224

ABSTRACT

Regulatory T cells (Tregs) play fundamental roles in maintaining peripheral tolerance to prevent autoimmunity and limit legitimate immune responses, a feature hijacked in tumor microenvironments in which the recruitment of Tregs often extinguishes immune surveillance through suppression of T-effector cell signaling and tumor cell killing. The pharmacological tuning of Treg activity without impacting on T conventional (Tconv) cell activity would likely be beneficial in the treatment of various human pathologies. PIP4K2A, 2B, and 2C constitute a family of lipid kinases that phosphorylate PtdIns5P to PtdIns(4,5)P2 They are involved in stress signaling, act as synthetic lethal targets in p53-null tumors, and in mice, the loss of PIP4K2C leads to late onset hyperinflammation. Accordingly, a human single nucleotide polymorphism (SNP) near the PIP4K2C gene is linked with susceptibility to autoimmune diseases. How PIP4Ks impact on human T cell signaling is not known. Using ex vivo human primary T cells, we found that PIP4K activity is required for Treg cell signaling and immunosuppressive activity. Genetic and pharmacological inhibition of PIP4K in Tregs reduces signaling through the PI3K, mTORC1/S6, and MAPK pathways, impairs cell proliferation, and increases activation-induced cell death while sparing Tconv. PIP4K and PI3K signaling regulate the expression of the Treg master transcriptional activator FOXP3 and the epigenetic signaling protein Ubiquitin-like containing PHD and RING finger domains 1 (UHRF1). Our studies suggest that the pharmacological inhibition of PIP4K can reprogram human Treg identity while leaving Tconv cell signaling and T-helper differentiation to largely intact potentially enhancing overall immunological activity.


Subject(s)
CCAAT-Enhancer-Binding Proteins/metabolism , Forkhead Transcription Factors/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , T-Lymphocytes, Regulatory/physiology , Ubiquitin-Protein Ligases/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , Cell Proliferation , Cell Survival , Cloning, Molecular , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/immunology , Gene Expression Regulation, Enzymologic/physiology , Humans , Immunosuppression Therapy , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Quinazolines/pharmacology , Signal Transduction , Thiophenes/pharmacology , Ubiquitin-Protein Ligases/genetics
4.
Proc Natl Acad Sci U S A ; 118(22)2021 06 01.
Article in English | MEDLINE | ID: mdl-34035164

ABSTRACT

Inducible regulatory T (iTreg) cells play a crucial role in immune suppression and are important for the maintenance of immune homeostasis. Mounting evidence has demonstrated connections between iTreg differentiation and metabolic reprogramming, especially rewiring in fatty acid oxidation (FAO). Previous work showed that butyrate, a specific type of short-chain fatty acid (SCFA) readily produced from fiber-rich diets through microbial fermentation, was critical for the maintenance of intestinal homeostasis and capable of promoting iTreg generation by up-regulating histone acetylation for gene expression as an HDAC inhibitor. Here, we revealed that butyrate could also accelerate FAO to facilitate iTreg differentiation. Moreover, butyrate was converted, by acyl-CoA synthetase short-chain family member 2 (ACSS2), into butyryl-CoA (BCoA), which up-regulated CPT1A activity through antagonizing the association of malonyl-CoA (MCoA), the best known metabolic intermediate inhibiting CPT1A, to promote FAO and thereby iTreg differentiation. Mutation of CPT1A at Arg243, a reported amino acid required for MCoA association, impaired both MCoA and BCoA binding, indicating that Arg243 is probably the responsible site for MCoA and BCoA association. Furthermore, blocking BCoA formation by ACSS2 inhibitor compromised butyrate-mediated iTreg generation and mitigation of mouse colitis. Together, we unveil a previously unappreciated role for butyrate in iTreg differentiation and illustrate butyrate-BCoA-CPT1A axis for the regulation of immune homeostasis.


Subject(s)
Butyrates/immunology , Carnitine O-Palmitoyltransferase/immunology , Cell Differentiation/immunology , Fatty Acids/immunology , Gastrointestinal Microbiome/immunology , T-Lymphocytes, Regulatory/immunology , Acetate-CoA Ligase/immunology , Animals , Gene Expression Regulation, Enzymologic/immunology , Mice , Oxidation-Reduction , Up-Regulation/immunology
5.
Fish Shellfish Immunol ; 114: 199-206, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33940173

ABSTRACT

Co-infection with parasites and bacteria is of frequent occurrence in aquaculture, leads to growth impedance otherwise mortality in fish depending on the varying degree of a load of primary pathogen either parasite or bacteria. The mechanistic regulation of immune response during co-infection in fish has merely documented. The aim of this study was to determine the impact of co-infection with Aeromonas hydrophila at three exposure doses of Argulus sp. on the innate immune responses and antioxidative stress enzymes of goldfish (Carassius auratus). The experimental fish were randomly distributed into eight treatment groups viz. T1 (control group without Argulus and A. hydrophila infection), T2 (fish exposed to a sub-lethal dose of A. hydrophila), T3 (low Argulus-infested fish), T4 (T3 + sub-lethal dose of A. hydrophila), T5 (moderate Argulus-infested fish), T6 (T5 + sub-lethal dose of A. hydrophila), T7 (high Argulus-infested fish) and T8 (T7+ sub-lethal dose of A. hydrophila) in duplicates. After distributing experimental fish into their respective treatment group, A. hydrophila was injected to T2, T4, T6 and T8. After the bacterial challenge, four fish from each experimental group were randomly sampled on 24, 72, and 168 h and subjected to the hematological, innate immune parameters and enzymatic analysis. In the co-infection group T8, a high degree of enhanced pathogenicity of A. hydrophila was noticed with increased mortalities (84.2%) in comparison to other groups. The current study shows a declining pattern in RBC, PCV and Hb values with the degree of parasite infestation without co-infection groups. Moreover, in the T8 group, exposure of a sub-lethal dose of bacteria resulted in a drastic reduction of the recorded parameters. Furthermore, a decreased value for WBC, monocyte and neutrophil was found in higher parasite group co-infected with a sub-lethal dose of bacteria relative to other co-infected groups during the experimental period. Also, a decrease in innate immune parameters and antioxidative stress enzymes were observed in the T8 group compared to T7 and T2 groups throughout the trial period. These findings indicate that a rise in the dose of Argulus infection improves A. hydrophila colonization in goldfish and contributes to suppression of the innate immune system and increased mortality.


Subject(s)
Aeromonas hydrophila , Arguloida , Goldfish , Gram-Negative Bacterial Infections/veterinary , Immunity, Innate/physiology , Parasitic Diseases, Animal/parasitology , Animals , Antioxidants , Catalase/genetics , Catalase/metabolism , Gene Expression Regulation, Enzymologic/immunology , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Gram-Negative Bacterial Infections/complications , Gram-Negative Bacterial Infections/immunology , Parasitic Diseases, Animal/complications , Parasitic Diseases, Animal/immunology , Stress, Physiological , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism
6.
Fish Shellfish Immunol ; 114: 36-48, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33864947

ABSTRACT

By using immunohistochemistry detection, yellow head virus (YHV) was found to replicate in granule-containing hemocytes including semi-granular hemocytes (SGC) and granular hemocytes (GC) during the early phase (24 h post injection) of YHV-infected shrimp. Higher signal of YHV infection was found in GC more than in SGC. Comparative phosphoproteomic profiles between YHV-infected and non-infected GC reveal a number of phosphoproteins with different expression levels. The phosphoprotein spot with later on identified as caspase-3 in YHV-infected GC is most interesting. Blocking caspase-3 function using a specific inhibitor (Ac-DEVD-CMK) demonstrated high replication of YHV and consequently, high shrimp mortality. The immunohistochemistry results confirmed the high viral load in shrimp that caspase-3 activity was blocked. Caspase-3 is regulated through a variety of posttranslational modifications, including phosphorylation. Analysis of phosphorylation sites of shrimp caspase-3 revealed phosphorylation sites at serine residue. Taken together, caspase-3 is a hemocytic protein isolated from shrimp granular hemocytes with a role in anti-YHV response and regulated through the phosphorylation process.


Subject(s)
Caspase 3/metabolism , Hemocytes/enzymology , Penaeidae/virology , Roniviridae , Animals , Caspase 3/genetics , Gene Expression Regulation, Enzymologic/immunology , Host-Pathogen Interactions/immunology , Host-Pathogen Interactions/physiology
7.
Front Immunol ; 12: 655528, 2021.
Article in English | MEDLINE | ID: mdl-33841439

ABSTRACT

The pandemic distribution of SARS-CoV-2 together with its particular feature of inactivating the interferon-based endogenous response and accordingly, impairing the innate immunity, has become a challenge for the international scientific and medical community. Fortunately, recombinant interferons as therapeutic products have accumulated a long history of beneficial therapeutic results in the treatment of chronic and acute viral diseases and also in the therapy of some types of cancer. One of the first antiviral treatments during the onset of COVID-19 in China was based on the use of recombinant interferon alfa 2b, so many clinicians began to use it, not only as therapy but also as a prophylactic approach, mainly in medical personnel. At the same time, basic research on interferons provided new insights that have contributed to a much better understanding of how treatment with interferons, initially considered as antivirals, actually has a much broader pharmacological scope. In this review, we briefly describe interferons, how they are induced in the event of a viral infection, and how they elicit signaling after contact with their specific receptor on target cells. Additionally, some of the genes stimulated by type I interferons are described, as well as the way interferon-mediated signaling is torpedoed by coronaviruses and in particular by SARS-CoV-2. Angiotensin converting enzyme 2 (ACE2) gene is one of the interferon response genes. Although for many scientists this fact could result in an adverse effect of interferon treatment in COVID-19 patients, ACE2 expression contributes to the balance of the renin-angiotensin system, which is greatly affected by SARS-CoV-2 in its internalization into the cell. This manuscript also includes the relationship between type I interferons and neutrophils, NETosis, and interleukin 17. Finally, under the subtitle of "take-home messages", we discuss the rationale behind a timely treatment with interferons in the context of COVID-19 is emphasized.


Subject(s)
COVID-19 Drug Treatment , COVID-19 , Interferon Type I/therapeutic use , SARS-CoV-2/immunology , Angiotensin-Converting Enzyme 2/immunology , COVID-19/immunology , COVID-19/pathology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/immunology , Humans , Interferon Type I/immunology
8.
Sci Rep ; 11(1): 7251, 2021 03 31.
Article in English | MEDLINE | ID: mdl-33790364

ABSTRACT

The hormone renin plays a crucial role in the regulation of blood pressure and fluid-electrolyte homeostasis. Normally, renin is synthesized by juxtaglomerular (JG) cells, a specialized group of myoepithelial cells located near the entrance to the kidney glomeruli. In response to low blood pressure and/or a decrease in extracellular fluid volume (as it occurs during dehydration, hypotension, or septic shock) JG cells respond by releasing renin to the circulation to reestablish homeostasis. Interestingly, renin-expressing cells also exist outside of the kidney, where their function has remained a mystery. We discovered a unique type of renin-expressing B-1 lymphocyte that may have unrecognized roles in defending the organism against infections. These cells synthesize renin, entrap and phagocyte bacteria and control bacterial growth. The ability of renin-bearing lymphocytes to control infections-which is enhanced by the presence of renin-adds a novel, previously unsuspected dimension to the defense role of renin-expressing cells, linking the endocrine control of circulatory homeostasis with the immune control of infections to ensure survival.


Subject(s)
Bacteria/immunology , Bacterial Infections/immunology , Cell Differentiation/immunology , Gene Expression Regulation, Enzymologic/immunology , Lymphocytes/immunology , Renin/immunology , Animals , Mice , Mice, Transgenic , Renin/genetics
9.
J Fish Dis ; 44(7): 961-977, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33645734

ABSTRACT

The bacterium Aeromonas salmonicida is the pathogen responsible for furunculosis, which is a serious disease of salmonids. This disease has a significant economic impact on the economic benefits of the global salmon farming industry. However, the pathogenesis of this disease in fish is still unknown. Members of the aldehyde dehydrogenase gene (ALDH) superfamily play a key role in the enzyme detoxification of endogenous and exogenous aldehydes. In this study, we obtained a recombinant aldehyde dehydrogenase 7A1 (ALDH7A1) protein to find its functions on Atlantic salmon infected by A. salmonicida. The transcriptional response in the liver of Atlantic salmon (Salmo salar) with differing levels of A. salmonicida infection was analysed and compared in order to reveal mechanisms by which ALDH7A1 may confer infection resistance. With the addition of ALDH7A1 protein, it was found that a total of 13,369 genes were annotated with one or more KEGG and localized to 360 KEGG pathways in the high concentration infection group. The differential expression genes were more enriched in immune signalling pathways such as the Toll-like receptor signalling pathway, NF-kappa B signalling pathway and TNF signalling pathway. On the other hand, at low concentrations of infection, KEGG enriched a smaller number of differential expression genes. However, these differential genes were more concentrated in immune signalling pathways such as the PI3K-Akt signalling pathway, JAK-STAT signalling pathway and complement and coagulation cascades. In addition, several known immune-related genes including HSP90α, HSP70, DNA damage-inducible transcript 4, integrin alpha 5 and microtubule-associated protein 2 were among the differentially expressed transcripts. These data provide the first insights into the host-ALDH7A1 vaccine interactome. The results of this study contribute to identifying the potential resistance mechanisms of Atlantic salmon to A. salmonicida infection and determining future treatment strategies.


Subject(s)
Aeromonas salmonicida , Aldehyde Dehydrogenase/metabolism , Gene Expression Regulation, Enzymologic/immunology , Gram-Negative Bacterial Infections/veterinary , Salmo salar/microbiology , Aldehyde Dehydrogenase/genetics , Animals , Fish Diseases/metabolism , Fish Diseases/microbiology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/metabolism , Reproducibility of Results , Transcriptome
10.
Front Immunol ; 12: 756606, 2021.
Article in English | MEDLINE | ID: mdl-35116021

ABSTRACT

Gliomas are malignant tumors that originate from the central nervous system. The aldehyde dehydrogenase family has been documented to affect cancer progression; however, its role in gliomas remains largely unexplored. Bulk RNA-seq analysis and single-cell RNA-Seq analysis were performed to explore the role of the aldehyde dehydrogenases family in gliomas. Training cohort contained The Cancer Genome Atlas data, while data from Chinese Glioma Genome Atlas and Gene Expression Omnibus were set as validation cohorts. Our scoring system based on the aldehyde dehydrogenases family suggested that high-scoring samples were associated with worse survival outcomes. The enrichment score of pathways were calculated by AUCell to substantiate the biofunction prediction results that the aldehyde dehydrogenases family affected glioma progression by modulating tumor cell proliferation, migration, and immune landscape. Tumor immune landscape was mapped from high-scoring samples. Moreover, ALDH3B1 and ALDH16A1, two main contributors of the scoring system, could affect glioblastoma cell proliferation and migration by inducing cell-cycle arrest and the epithelial-mesenchymal transition. Taken together, the aldehyde dehydrogenases family could play a significant role in the tumor immune landscape and could be used to predict patient prognosis. ALDH3B1 and ALDH16A1 could influence tumor cell proliferation and migration.


Subject(s)
Aldehyde Dehydrogenase/immunology , Biomarkers, Tumor/immunology , Brain Neoplasms/immunology , Epithelial-Mesenchymal Transition/immunology , Gene Expression Regulation, Enzymologic/immunology , Gene Expression Regulation, Neoplastic/immunology , Glioma/immunology , Neoplasm Proteins/immunology , Aldehyde Dehydrogenase/genetics , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Epithelial-Mesenchymal Transition/genetics , Glioma/genetics , Humans , Neoplasm Proteins/genetics
11.
Front Immunol ; 11: 531491, 2020.
Article in English | MEDLINE | ID: mdl-33072086

ABSTRACT

Indoleamine 2,3-dioxygenase 1 (IDO1) is a cytosolic haem-containing enzyme involved in the degradation of tryptophan to kynurenine. Although initially thought to be solely implicated in the modulation of innate immune responses during infection, subsequent discoveries demonstrated IDO1 as a mechanism of acquired immune tolerance. In cancer, IDO1 expression/activity has been observed in tumor cells as well as in the tumor-surrounding stroma, which is composed of endothelial cells, immune cells, fibroblasts, and mesenchymal cells. IDO1 expression/activity has also been reported in the peripheral blood. This manuscript reviews available data on IDO1 expression, mechanisms of its induction, and its function in cancer for each of these compartments. In-depth study of the biological function of IDO1 according to the expressing (tumor) cell can help to understand if and when IDO1 inhibition can play a role in cancer therapy.


Subject(s)
Gene Expression Regulation, Enzymologic/immunology , Gene Expression Regulation, Neoplastic/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Neoplasm Proteins/immunology , Neoplasms/immunology , Tumor Microenvironment/immunology , Animals , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis , Neoplasm Proteins/biosynthesis , Neoplasms/enzymology , Neoplasms/pathology , Neoplasms/therapy
12.
Mech Ageing Dev ; 192: 111363, 2020 12.
Article in English | MEDLINE | ID: mdl-32987038

ABSTRACT

Neprilysin (NEP) is an integral membrane-bound metallopeptidase with a wide spectrum of substrates and physiological functions. It plays an important role in proteolytic processes in the kidney, cardiovascular regulation, immune response, cell proliferation, foetal development etc. It is an important neuropeptidase and amyloid-degrading enzyme which makes NEP a therapeutic target in Alzheimer's disease (AD). Moreover, it plays a preventive role in development of cancer, obesity and type-2 diabetes. Recently a role of NEP in COVID-19 pathogenesis has also been suggested. Despite intensive research into NEP structure and functions in different organisms, changes in its expression and regulation during brain development and ageing, especially in age-related pathologies, is still not fully understood. This prevents development of pharmacological treatments from various diseases in which NEP is implicated although recently a dual-acting drug sacubitril-valsartan (LCZ696) combining a NEP inhibitor and angiotensin receptor blocker has been approved for treatment of heart failure. Also, various natural compounds capable of upregulating NEP expression, including green tea (EGCG), have been proposed as a preventive medicine in prostate cancer and AD. This review summarizes the existing literature and our own research on the expression and activity of NEP in normal brain development, ageing and under pathological conditions.


Subject(s)
Aging/immunology , Alzheimer Disease/immunology , COVID-19/immunology , Diabetes Mellitus, Type 2/immunology , Gene Expression Regulation, Enzymologic/immunology , Gene Expression Regulation, Neoplastic/immunology , Neoplasm Proteins/immunology , Neoplasms/immunology , Neprilysin/immunology , SARS-CoV-2/immunology , Aging/pathology , Alzheimer Disease/pathology , Animals , COVID-19/pathology , Diabetes Mellitus, Type 2/pathology , Humans , Neoplasms/pathology
13.
J Immunol ; 205(8): 2243-2254, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32917786

ABSTRACT

In IgA nephropathy (IgAN), IgA immune complexes are deposited in the mesangium and drive inflammation and extracellular matrix (ECM) remodelling. The functional links between IgA deposition, inflammation, and matrix remodelling are not well characterized. We recently performed urine liquid chromatography-tandem mass spectrometry proteomics and identified multiple ECM glycoproteins whose expression and function in IgAN is unclear. None of the urine glycoproteins was regulated in IgAN transcriptomics, indicating that tissue remodelling rather than increased expression might contribute to their presence in urine. To investigate this, we examined the IgAN expression profile of metalloproteinases, enzymes involved in the remodelling of ECM proteins, and noted that the proteoglycanase ADAMTS5 was upregulated in IgAN kidneys. ADAMTS5 accumulated in areas of inflammation, and ADAMTS5+ cells were seen in the tubulointerstitium and glomeruli. The enzyme was expressed by CD64+ cells and its expression was increased by IL-1 and LPS. Analysis of myeloid cell transcriptomics revealed that ADAMTS5 is enriched in human classical monocytes. ADAMTS5+ cells were present in areas of matrix remodelling and associated with ECM proteins lumican, versican, and collagen-4. Liquid chromatography-tandem mass spectrometry proteomics of kidney explants digested with ADAMTS5, identified multiple kidney proteins affected by ADAMTS5 and revealed specific proteolysis of complement C3 and fibronectin associated with IgA on immune complexes. ADAMTS5 processing of immune complex proteins reduced binding to cultured mesangial cells. ADAMTS5 is associated with interstitial inflammatory cells in IgAN and other kidney lesions and fragments relevant extracellular proteins. The proteolytic enzyme might be a new translational target relevant to inflammation and scarring in kidney disease.


Subject(s)
ADAMTS5 Protein/immunology , Extracellular Matrix/immunology , Gene Expression Regulation, Enzymologic/immunology , Glomerulonephritis, IGA/immunology , Kidney Glomerulus/immunology , Monocytes/immunology , Adult , Aged , Extracellular Matrix/pathology , Female , Glomerulonephritis, IGA/pathology , Humans , Inflammation/immunology , Inflammation/pathology , Kidney Glomerulus/pathology , Male , Middle Aged , Monocytes/pathology
14.
J Leukoc Biol ; 108(6): 1727-1733, 2020 12.
Article in English | MEDLINE | ID: mdl-32640496

ABSTRACT

This study tested the hypothesis that heparanase (HPSE) is related to tumor metastasis and curcumin (CCM) inhibits tumor metastasis by down-regulating HPSE expression. MTT, Transwell assays, and RT-PCR were used to study the effects of CCM on the migration and invasion of CT26 cells and the expression of HPSE. CT26 cells were transfected with lentivirus to establish HPSE-overexpressing cells (OE) and corresponding negative control cells (NC). Signal pathways involved in down-regulating the expression of HPSE and inhibiting the migration and invasion of CT26 cells by CCM were screened by the liquid crystal chip. HPSE promoted CT26 cells migration and invasion, and CCM inhibited the proliferation and metastasis of CT26 cells. The results of RT-PCR indicated that CCM down-regulated HPSE expression. Liquid phase microarray showed that CCM inhibited the phosphorylation of P38 and STAT5 in CT26 cells and NC cells. In contrast, the inhibitory function of CCM was markedly enhanced when HPSE was overexpressed (P < 0.05). In short, HPSE is closely related to metastasis of colon cancer cells. CCM inhibits colon cancer cell migration and invasion by inhibiting HPSE expression, which may be related to P38 MAPK and JAK/STAT5 signal pathways.


Subject(s)
Colonic Neoplasms/immunology , Curcumin/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Glucuronidase/immunology , Neoplasm Proteins/immunology , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Gene Expression Regulation, Enzymologic/immunology , Gene Expression Regulation, Neoplastic/immunology , Humans , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/immunology , Neoplasm Metastasis
15.
Genes Cells ; 25(7): 443-449, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32394600

ABSTRACT

Histamine is a bioactive monoamine that is synthesized by the enzymatic activity of histidine decarboxylase (HDC) in basophils, mast cells, gastric enterochromaffin-like (ECL) cells and histaminergic neuronal cells. Upon a series of cellular stimuli, these cells release stored histamine, which elicits allergies, inflammation, and gastric acid secretion and regulates neuronal activity. Recent studies have shown that certain other types of myeloid lineage cells also produce histamine with HDC induction under various pathogenic stimuli. Histamine has been shown to play a series of pathophysiological roles by modulating immune and inflammatory responses in a number of disease conditions, whereas the mechanistic aspects underlying induced HDC expression remain elusive. In the present review, we summarize the current understanding of the regulatory mechanism of Hdc gene expression and the roles played by histamine in physiological contexts as well as pathogenic processes. We also introduce a newly developed histaminergic cell-monitoring transgenic mouse line (Hdc-BAC-GFP) that serves as a valuable experimental tool to identify the source of histamine and dissect upstream regulatory signals.


Subject(s)
Histamine/metabolism , Histidine Decarboxylase/metabolism , Receptors, Histamine/metabolism , Sepsis/immunology , Animals , Chromosomes, Artificial, Bacterial , Gene Expression Regulation, Enzymologic/immunology , Histamine/physiology , Histidine Decarboxylase/genetics , Histones/metabolism , Methylation , Mice , Mice, Transgenic , Myeloid Cells/metabolism , Sepsis/metabolism
16.
J Clin Invest ; 130(7): 3422-3436, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32452837

ABSTRACT

Vaccination is a mainstay in preventive medicine, reducing morbidity and mortality from infection, largely by generating pathogen-specific neutralizing antibodies. However, standard immunization strategies are insufficient with increasing age due to immunological impediments, including defects in T follicular helper (Tfh) cells. Here, we found that Tfh generation is inversely linked to the expression of the ecto-NTPDase CD39 that modifies purinergic signaling. The lineage-determining transcription factor BCL6 inhibited CD39 expression, while increased Tfh frequencies were found in individuals with a germline polymorphism preventing transcription of ENTPD1, encoding CD39. In in vitro human and in vivo mouse studies, Tfh generation and germinal center responses were enhanced by reducing CD39 expression through the inhibition of the cAMP/PKA/p-CREB pathway, or by blocking adenosine signaling downstream of CD39 using the selective adenosine A2a receptor antagonist istradefylline. Thus, purinergic signaling in differentiating T cells can be targeted to improve vaccine responses, in particular in older individuals who have increased CD39 expression.


Subject(s)
Apyrase/immunology , Cell Differentiation/immunology , Gene Expression Regulation, Enzymologic/immunology , Germinal Center/immunology , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Apyrase/genetics , Germinal Center/cytology , Humans , Mice , Mice, Transgenic , T-Lymphocytes, Helper-Inducer/cytology
17.
Fish Shellfish Immunol ; 99: 386-391, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32081808

ABSTRACT

DEAD-box helicase 41 (DDX41) is a key cytosolic DNA sensor playing critical roles in the regulation of type I IFN responses, and their functions have been well-characterized in mammals. However, little information is available regarding the function of fish DDX41. In this study, a DDX41 gene, named On-DDX41, was identified in Nile tilapia, Oreochromis niloticus. The predicted protein of On-DDX41 contains several structural features known in DDX41, including conserved DEADc and HELICc domains, and a conserved sequence "Asp-Glu-Ala-Asp (D-E-A-D)". On-DDX41 gene was constitutively expressed in all tissues examined, with the highest expression level observed in liver and muscle, and was inducible after poly(I:C) stimulation. Moreover, the overexpression of On-DDX41 can elicit a strong activation of both zebrafish IFN1 and IFN3 promoter in fish cells treated with poly(dA:dT). The present study thus contributes to a better understanding of the functional properties of DDX41 in fish.


Subject(s)
DEAD-box RNA Helicases/metabolism , Fish Proteins/metabolism , Gene Expression Regulation, Enzymologic/immunology , Interferons/metabolism , Tilapia/metabolism , Amino Acid Sequence , Animals , Cloning, Molecular , DEAD-box RNA Helicases/genetics , Fish Proteins/genetics , Interferons/genetics , Phylogeny
18.
Front Immunol ; 11: 594620, 2020.
Article in English | MEDLINE | ID: mdl-33537030

ABSTRACT

Interleukin-15 (IL-15) is a cytokine that has been shown to expand CD8 T cell and natural killer (NK) cell populations, and therefore has potential for potentiating adoptive immune cell therapy for cancer. Previously, IL-15 has been shown to induce proliferation of CD8 memory T cells through activation of telomerase. Here, we investigated whether telomerase is also activated during the IL-15 mediated proliferation of NK and NKT-like (CD56+CD3+) cells. We also examined the extent that each of the three signaling pathways known to be stimulated by IL-2/IL-15 (JAK-STAT, PI3K-AKT Ras-RAF/MAPK) were activated and involved in the telomerase expression in the three cell types NK, NKT, or CD8 T cells. To assess cell proliferation and doubling, peripheral blood mononuclear cells (PBMCs) or isolated NK, NKT-like or CD8 T cells were incubated with varying concentrations of IL-15 or IL-2 for 7 days. CD8 T, NK, and NKT cell expansion was determined by fluorophore-conjugated antibody staining and flow cytometry. Cell doubling was investigated using carboxyfluorescein-succinimidyl-ester (CFSE). Telomerase expression was investigated by staining cells with anti-telomerase reverse transcriptase (anti-TERT). Telomerase activity in CD56+ and CD8 T cells was also measured via Telomerase Repeat Amplification Protocol (TRAP). Analysis of cellular expansion, proliferation and TERT expression concluded that IL-15 increased cellular growth of NK, NKT, and CD8 T cells more effectively than IL-2 using low or high doses. IL-15, increased TERT expression in NK and NKT cells by up to 2.5 fold, the same increase seen in CD8 T cells. IL-2 had effects on TERT expression only at high doses (100-1000 ng/ml). Proteome profiling identified that IL-15 activated selected signaling proteins in the three pathways (JAK-STAT, PI3K-AKT, Ras-MAPK) known to mediate IL-2/IL-15 signaling, more strongly than IL-2. Evaluation by signaling pathway inhibitors revealed that JAK/STAT and PI3K/AKT pathways are important in IL-15's ability to upregulate TERT expression in NK and NKT cells, whereas all three pathways were involved in CD8 T cell TERT expression. In conclusion, this study shows that IL-15 potently stimulates TERT upregulation in NK and NKT cells in addition to CD8 T cells and is therefore a valuable tool for adoptive cell therapies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Gene Expression Regulation, Enzymologic/immunology , Interleukin-15/immunology , Killer Cells, Natural/immunology , Natural Killer T-Cells/immunology , Telomerase/immunology , Up-Regulation/immunology , Humans
19.
J Fish Biol ; 96(3): 580-589, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31886519

ABSTRACT

Mitogen-activated protein kinases (MAPKs) are crucial Ser/Thr protein kinases that play important roles in innate immunity by converting extracellular stimuli into a wide range of cellular responses, including the production of cytokines. In this study, two MAPK genes, jnk1 and erk1, were cloned and characterized in rohu (Labeo rohita), a commercially important freshwater fish species in the Indian subcontinent. In healthy rohu, both jnk1 and erk1 gene expressions were highest in the spleen as compared to gill, liver, blood and kidney tissues. In vitro stimulation of the L. rohita gill (LRG) cell line with γ-D-glutamyl-meso-diaminopimelic acid, muramyl dipeptide and polyinosinic: polycytidylic acid (poly I:C) resulted in significantly enhanced expressions of jnk1 and erk1 genes. In the in vivo experiments, jnk1 and erk1 gene expressions were also enhanced in lipopolysaccharides and poly I:C-treatment. Infection of rohu fingerlings with Aeromonas hydrophila and Bacillus subtilis revealed significantly enhanced expressions of the jnk1 and erk1 genes in all of the tested organs/tissues. Together these results imply the important role of jnk1 and erk1 genes in fish during pathogenic invasion and diseases.


Subject(s)
Cyprinidae , Fish Diseases/immunology , Gene Expression Regulation, Enzymologic/immunology , Gram-Negative Bacterial Infections/immunology , JNK Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinase 3/genetics , Signal Transduction/genetics , Adjuvants, Immunologic/pharmacology , Aeromonas hydrophila/physiology , Animals , Cyprinidae/genetics , Cyprinidae/metabolism , Cyprinidae/microbiology , Fish Diseases/enzymology , Fish Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation, Enzymologic/drug effects , Gram-Negative Bacterial Infections/enzymology , JNK Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Pathogen-Associated Molecular Pattern Molecules/immunology , Poly I-C/pharmacology
20.
J Infect Dis ; 221(6): 973-982, 2020 03 02.
Article in English | MEDLINE | ID: mdl-31748808

ABSTRACT

BACKGROUND: Skin lesions from patients infected with Leishmania braziliensis has been associated with inflammation induced by cytotoxic CD8+ T cells. In addition, CD8+ T cell-mediated cytotoxicity has not been linked to parasite killing. Meanwhile, the cytotoxic role played by natural killer (NK) cells in cutaneous leishmaniasis (CL) remains poorly understood. METHODS: In this study, we observed higher frequencies of NK cells in the peripheral blood of CL patients compared with healthy subjects, and that NK cells expressed more interferon-γ, tumor necrosis factor (TNF), granzyme B, and perforin than CD8+ T cells. RESULTS: We also found that most of the cytotoxic activity in CL lesions was triggered by NK cells, and that the high levels of granzyme B produced in CL lesions was associated with larger lesion size. Furthermore, an in vitro blockade of granzyme B was observed to decrease TNF production. CONCCLUSIONS: Our data, taken together, suggest an important role by NK cells in inducing inflammation in CL, thereby contributing to disease immunopathology.


Subject(s)
Gene Expression Regulation, Enzymologic/immunology , Granzymes/metabolism , Inflammation/metabolism , Killer Cells, Natural/enzymology , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/pathology , CD4-Positive T-Lymphocytes , Case-Control Studies , Granzymes/genetics , Humans , Interferon-gamma/genetics , Interferon-gamma/metabolism , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Perforin/genetics , Perforin/metabolism , T-Lymphocytes, Cytotoxic , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...