Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
PLoS One ; 17(11): e0277827, 2022.
Article in English | MEDLINE | ID: mdl-36409702

ABSTRACT

Studies on the humoral response to homologous BNT162b2 mRNA-vaccination focus mainly on IgG antibody dynamics, while long-term IgA kinetics are understudied. Herein, kinetics of IgG and IgA levels against trimeric-Spike (S) and Receptor-Binding-Domain (RBD) were evaluated by in-house ELISAs in 146 two-dose vaccinated Greek healthcare workers (HCWs) in a 9-month period at six time points (up to 270 days after the first dose). The effect of a homologous booster third dose was also studied and evaluated. The peak of immune response was observed 21 days after the second dose; 100% seroconversion rate for anti-S and anti-RBD IgG, and 99.7% and 96.3% respectively for IgA. IgG antibody levels displayed higher increase compared to IgA. Declining but persistent anti-SARS-CoV-2 antibody levels were detected 9 months after vaccination; IgG and IgA anti-S levels approached those after the first dose, while a more rapid reduction rate for anti-RBD antibodies led to significantly lower levels for both classes, supporting the need for a booster dose. Indeed, a homologous booster third dose resulted in enhanced levels of anti-S of both classes, whereas anti-RBD didn't exceed the peak levels after the second dose. Previous SARS-CoV-2 infection, flu vaccination, BMI<35 and the occurrence of an adverse event upon vaccination, were associated with higher IgG antibody levels over time, which however were negatively affected by age increase and the presence of chronic diseases. Overall, after concurrently using the S and RBD target-antigens in in-house ELISAs, we report in addition to IgG, long-term persistence of IgA antibodies. Regarding antibody levels, homologous mRNA vaccination gives rise to an effective anti-viral protection up to 9 months negatively correlated to age. Considering that COVID-19 is still a matter of public concern, booster vaccine doses remain critical to vulnerable individuals.


Subject(s)
BNT162 Vaccine , COVID-19 , Humans , RNA, Messenger , Greece , COVID-19/prevention & control , SARS-CoV-2 , Vaccination , Immunoglobulin A , Immunoglobulin G , Health Personnel
2.
Cells ; 10(9)2021 08 30.
Article in English | MEDLINE | ID: mdl-34571900

ABSTRACT

Iron is crucial to the regulation of the host innate immune system and the outcome of many infections. Hepatitis C virus (HCV), one of the major viral human pathogens that depends on iron to complete its life cycle, is highly skilled in evading the immune system. This study presents the construction and validation of a physiologically relevant triple-cell co-culture model that was used to investigate the input of iron in HCV infection and the interplay between HCV, iron, and determinants of host innate immunity. We recorded the expression patterns of key proteins of iron homeostasis involved in iron import, export and storage and examined their relation to the iron regulatory hormone hepcidin in hepatocytes, enterocytes and macrophages in the presence and absence of HCV. We then assessed the transcriptional profiles of pro-inflammatory cytokines Interleukin-6 (IL-6) and interleukin-15 (IL-15) and anti-inflammatory interleukin-10 (IL-10) under normal or iron-depleted conditions and determined how these were affected by infection. Our data suggest the presence of a link between iron homeostasis and innate immunity unfolding among liver, intestine, and macrophages, which could participate in the deregulation of innate immune responses observed in early HCV infection. Coupled with iron-assisted enhanced viral propagation, such a mechanism may be important for the establishment of viral persistence and the ensuing chronic liver disease.


Subject(s)
Enterocytes/pathology , Hepatitis C/pathology , Hepatocytes/pathology , Homeostasis , Immunity, Innate , Iron/metabolism , Macrophages/pathology , Coculture Techniques , Cytokines/metabolism , Enterocytes/immunology , Enterocytes/metabolism , Enterocytes/virology , Hepacivirus/immunology , Hepacivirus/metabolism , Hepatitis C/immunology , Hepatitis C/metabolism , Hepatitis C/virology , Hepatocytes/immunology , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology
3.
FEBS Open Bio ; 11(1): 237-250, 2021 01.
Article in English | MEDLINE | ID: mdl-33247551

ABSTRACT

Hepcidin, a 25-amino acid peptide encoded by the HAMP gene and produced mainly by hepatocytes and macrophages, is a mediator of innate immunity and the central iron-regulatory hormone. Circulating hepcidin controls iron efflux by inducing degradation of the cellular iron exporter ferroportin. HCV infection is associated with hepatic iron overload and elevated serum iron, which correlate with poor antiviral responses. The HCV nonstructural NS5A protein is known to function in multiple aspects of the HCV life cycle, probably exerting its activity in concert with cellular factor(s). In this study, we attempted to delineate the effect of HCV NS5A on HAMP gene expression. We observed that transient transfection of hepatoma cell lines with HCV NS5A resulted in down-regulation of HAMP promoter activity. A similar effect was evident after transduction of Huh7 cells with a recombinant baculovirus vector expressing NS5A protein. We proceeded to construct an NS5A-expressing stable cell line, which also exhibited down-regulation of HAMP gene promoter activity and significant reduction of HAMP mRNA and hepcidin protein levels. Concurrent expression of HCV core protein, a well-characterized hepcidin inducer, revealed antagonism between those two proteins for hepcidin regulation. In attempting to identify the pathways involved in NS5A-driven reduction of hepcidin levels, we ruled out any NS5A-induced alterations in the expression of the well-known hepcidin inducers SMAD4 and STAT3. Further analysis linked the abundance of intracellular zinc ions and the deregulation of the MTF-1/MRE/hepcidin axis with the observed phenomenon. This effect could be associated with distinct phases in HCV life cycle.


Subject(s)
Hepacivirus/immunology , Hepatitis C/immunology , Hepcidins/genetics , Viral Core Proteins/metabolism , Viral Nonstructural Proteins/isolation & purification , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Hepacivirus/metabolism , Hepatitis C/genetics , Hepatitis C/virology , Hepatocytes/metabolism , Hepatocytes/virology , Host Microbial Interactions/genetics , Host Microbial Interactions/immunology , Humans , Immunity, Innate/genetics , Iron/metabolism , Macrophages/immunology , Macrophages/metabolism , Promoter Regions, Genetic/genetics , Transcription Factors/metabolism , Transcription Factor MTF-1
4.
Hum Gene Ther ; 29(9): 1056-1070, 2018 09.
Article in English | MEDLINE | ID: mdl-30191743

ABSTRACT

Antiviral DNA vaccines are a novel strategy in the vaccine development field, which basically consists of the administration of expression vectors coding viral antigen sequences into the host's cells. Targeting of conserved viral epitopes by antibody fragments specific to activating cell surface co-receptor molecules on antigen-presenting cells could be an alternative approach for inducing protective immunity. It has been shown that FcγRI on human monocytes enhances antigen presentation in vivo. Various DNA constructs, encoding a Single-chain variable antibodies (scFv) from mouse anti-human FcγRI monoclonal antibody, coupled to a sequence encoding a T- and B-cell epitope-containing influenza A virus hemagglutinin inter-subunit peptide were inserted into the eukaryotic expression vector system pTriEx-3 Neo. The constructed chimeric DNA molecules were expressed by transfected Chinese hamster ovary cells and the ability of the engineered proteins to interact with FcγRI-expressing cells was confirmed by flow cytometry. The fusion protein induced a strong signal transduction on human monocytes via FcγRI. The expression vector pTriEx-3 Neo containing the described construct was used as a naked DNA vaccine and introduced directly to experimental humanized NOD SCID gamma mice with or without boosting with the expressed fusion protein. Immunization with the generated DNA chimeric molecules and prime-boost with the expressed recombinant proteins induced significant serum levels of anti-influenza immunoglobulin G antibodies and strong cytotoxic T lymphocyte activity against influenza virus-infected cells in humanized animals.


Subject(s)
Antibodies, Monoclonal/immunology , Epitopes/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Vaccines, DNA/immunology , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/immunology , Antigen-Presenting Cells/immunology , CHO Cells , Cricetulus , Epitopes/biosynthesis , Flow Cytometry , Gene Expression Regulation , Genetic Engineering , Humans , Immunoglobulin Fragments/genetics , Immunoglobulin Fragments/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Mice , Orthomyxoviridae/immunology , Orthomyxoviridae/pathogenicity , T-Lymphocytes, Cytotoxic/immunology
5.
Virulence ; 7(6): 679-90, 2016 08 17.
Article in English | MEDLINE | ID: mdl-27058404

ABSTRACT

Mechanisms that favor Hepatitis C virus (HCV) persistence over clearance are unclear, but involve defective innate immunity. Chronic infection is characterized by hepatic iron overload, hyperferraemia and hyperferittinaemia. Hepcidin modulates iron egress via ferroportin and its storage in ferritin. Chronic HCV patients have decreased hepcidin, while HCV replication is modified by HAMP silencing. We aimed to investigate interactions between HCV and hepcidin, during acute and chronic disease, and putative alterations in cellular iron homeostasis that enhance HCV propagation and promote viral persistence. Thus, we used HCV JFH-1-infected co-cultures of Huh7.5 hepatoma and THP-1 macrophage cells, HCV patients' sera and Huh7 hepcidin-expressing cells transfected with HCV replicons. Hepcidin levels were elevated in acutely infected patients, but correlated with viral load in chronic patients. HAMP expression was up-regulated early in HCV infection in vitro, with corresponding changes in ferritin and FPN. Hepcidin overexpression enhanced both viral translation and replication. In HCV-infected co-cultures, we observed increased hepcidin, reduced hepatoma ferritin and a concurrent rise in macrophaghic ferritin over time. Altered iron levels complemented amplified replication in hepatoma cells and one replication round in macrophages. Iron-loading of macrophages led to enhancement of hepatic HCV replication through reversed ferritin "flow." Viral transmissibility from infected macrophages to naïve hepatoma cells was induced by iron. We propose that HCV control over iron occurs both by intracellular iron sequestration, through hepcidin, and intercellular iron mobilisation via ferritin, as means toward enhanced replication. Persistence could be achieved through HCV-induced changes in macrophagic iron that enhances viral replication in these cells.


Subject(s)
Hepacivirus/physiology , Hepatitis C/virology , Homeostasis , Iron/metabolism , Macrophages/metabolism , Macrophages/virology , Carcinoma, Hepatocellular , Cell Line , Coculture Techniques , Ferritins/metabolism , Hepacivirus/genetics , Hepatitis C/blood , Hepatitis C/metabolism , Hepatitis C, Chronic/blood , Hepatitis C, Chronic/metabolism , Hepatitis C, Chronic/virology , Hepcidins/blood , Hepcidins/genetics , Hepcidins/metabolism , Humans , Iron Overload , Liver Neoplasms , Macrophages/chemistry , Replicon , Virus Replication
6.
PLoS One ; 10(8): e0135486, 2015.
Article in English | MEDLINE | ID: mdl-26270641

ABSTRACT

Hepcidin, a liver hormone, is important for both innate immunity and iron metabolism regulation. As dysfunction of the hepcidin pathway may contribute to liver pathology, we analysed liver hepcidin mRNA and serum hepcidin in patients with chronic liver diseases. Hepcidin mRNA levels were determined in liver biopsies obtained from 126 patients with HCV (n = 21), HBV (n = 23), autoimmune cholestatic disease (primary biliary cirrhosis and primary sclerosing cholangitis; PBC/PSC; n = 34), autoimmune hepatitis (AIH; n = 16) and non-alcoholic fatty liver disease (NAFLD; n = 32). Sera sampled on the biopsy day from the same patients were investigated for serum hepcidin levels. Hepatic hepcidin mRNA levels correlated positively with ferritin and negatively with serum γ-GT levels. However, no correlation was found between serum hepcidin and either ferritin or liver hepcidin mRNA. Both serum hepcidin and the serum hepcidin/ferritin ratio were significantly lower in AIH and PBC/PSC patients' sera compared to HBV, HCV or NAFLD (P<0.001 for each comparison) and correlated negatively with serum ALP levels. PBC/PSC and AIH patients maintained low serum hepcidin during the course of their two-year long treatment. In summary, parallel determination of liver hepcidin mRNA and serum hepcidin in patients with chronic liver diseases shows that circulating hepcidin and its respective ratio to ferritin are significantly diminished in patients with autoimmune liver diseases. These novel findings, once confirmed by follow-up studies involving bigger size and better-matched disease subgroups, should be taken into consideration during diagnosis and treatment of autoimmune liver diseases.


Subject(s)
Cholangitis, Sclerosing/pathology , Hepatitis, Autoimmune/pathology , Hepcidins/blood , Hepcidins/genetics , Liver Cirrhosis, Biliary/pathology , Adult , Aged , Cholangitis, Sclerosing/blood , Cholangitis, Sclerosing/genetics , Diagnosis, Differential , Down-Regulation , Female , Ferritins/blood , Hepatitis B/blood , Hepatitis B/genetics , Hepatitis B/pathology , Hepatitis C/blood , Hepatitis C/genetics , Hepatitis C/pathology , Hepatitis, Autoimmune/blood , Hepatitis, Autoimmune/genetics , Humans , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Biliary/blood , Liver Cirrhosis, Biliary/genetics , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology
8.
Cell Mol Life Sci ; 71(21): 4243-58, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24718935

ABSTRACT

Hepatitis C virus (HCV) infection is associated with hepatic iron overload and elevated serum iron that correlate to poor antiviral responses. Hepcidin (HAMP), a 25-aa cysteine-rich liver-specific peptide, controls iron homeostasis. Its expression is up-regulated in inflammation and iron excess. HCV-mediated hepcidin regulation remains controversial. Chronic HCV patients possess relatively low hepcidin levels; however, elevated HAMP mRNA has been reported in HCV core transgenic mice and HCV replicon-expressing cells. We investigated the effect of HCV core protein on HAMP gene expression and delineated the complex interplay of molecular mechanisms involved. HCV core protein up-regulated HAMP promoter activity, mRNA, and secreted protein levels. Enhanced promoter activity was abolished by co-transfections of core with HAMP promoter constructs containing mutated/deleted BMP and STAT binding sites. Dominant negative constructs, pharmacological inhibitors, and silencing experiments against STAT3 and SMAD4 confirmed the participation of both pathways in HAMP gene regulation by core protein. STAT3 and SMAD4 expression levels were found increased in the presence of HCV core, which orchestrated SMAD4 translocation into the nucleus and STAT3 phosphorylation. To further understand the mechanisms governing the core effect, the role of the JAK/STAT-activating kinase CK2 was investigated. A CK2-dominant negative construct, a CK2-specific inhibitor, and RNAi interference abrogated the core-induced increase on HAMP promoter activity, mRNA, and protein levels, while CK2 acted in synergy with core to significantly enhance HAMP gene expression. Therefore, HCV core up-regulates HAMP gene transcription via a complex signaling network that requires both SMAD/BMP and STAT3 pathways and CK2 involvement.


Subject(s)
Casein Kinase II/metabolism , Gene Expression Regulation, Enzymologic , Hepacivirus/metabolism , Hepcidins/metabolism , Viral Core Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Gene Expression Regulation, Viral , Gene Silencing , Hep G2 Cells , Homeostasis , Humans , Iron/metabolism , Promoter Regions, Genetic , RNA Interference , STAT3 Transcription Factor/metabolism , Signal Transduction , Smad4 Protein/metabolism , Up-Regulation
9.
Virulence ; 5(4): 465-76, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24626108

ABSTRACT

An estimated 30-40% of patients with chronic hepatitis C have elevated serum iron, transferrin saturation, and ferritin levels. Clinical data suggest that iron is a co-morbidity factor for disease progression following HCV infection. Iron is essential for a number of fundamental metabolic processes in cells and organisms. Mammalian iron homeostasis is tightly regulated and this is maintained through the coordinated action of sensory and regulatory networks that modulate the expression of iron-related proteins at the transcriptional and/or posttranscriptional levels. Disturbances of iron homeostasis have been implicated in infectious disease pathogenesis. Viruses, similarly to other pathogens, can escape recognition by the immune system, but they need iron from their host to grow and spread. Hepcidin is a 25-aa peptide, present in human serum and urine and represents the key peptide hormone, which modulates iron homeostasis in the body. It is synthesized predominantly by hepatocytes and its mature form is released in circulation. In this review, we discuss recent advances in the exciting crosstalk of molecular mechanisms and cell signaling pathways by which iron and hepcidin production influences HCV-induced liver disease.


Subject(s)
Hepacivirus/physiology , Hepatitis C, Chronic/metabolism , Hepcidins/metabolism , Iron/metabolism , Animals , Hepatitis C, Chronic/virology , Humans , Liver/metabolism , Liver/virology
10.
Mol Cell Biochem ; 379(1-2): 107-13, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23716178

ABSTRACT

EMMPRIN is a widely expressed transmembrane glycoprotein that plays important roles in many physiological and pathological processes, such as tumor invasion and metastasis. It stimulates the production of matrix metalloproteinase (MMPs) by tumor-associated fibroblasts. In the present study, our aim was to (a) to investigate if the IgII loop domain of the extracellular domain (ECD) of EMMPRIN contributes to the MMP production by fibroblasts and (b) to evaluate the significance of glycosylation in this process. For this purpose, we expressed the ECD, IgI, or IgII domains of EMMPRIN, in their glycosylated and non-glycosylated forms, in the heterologous expression systems of P. pastoris and E. coli, respectively. Dermal fibroblasts were treated with purified recombinant domains and proteins from cell extracts and supernatants were analyzed by Western blot and zymography assays. Fibroblasts treated with ECD-, IgI-, and IgII-glycosylated domains of EMMPRIN significantly stimulated the gelatinolytic activity of MMP-2, compared to untreated fibroblasts, whereas no significant effect was observed after treatment with the non-glycosylated ECD, IgI, and IgII domains. Western blot analysis from cell extracts and supernatants revealed that only the glycosylated forms were able to stimulate MMP-2 production and secretion, respectively. Quantitative PCR revealed that this effect was not attributed to transcriptional alterations. This study showed that N-glycosylation was a prerequisite for efficient MMP-2 production, with the IgII loop domain contributing significantly to this process. Perturbation of the function of IgII-EMMPRIN loop could have potential therapeutic value in the inhibition of MMP-2-dependent cancer cell invasion and metastasis.


Subject(s)
Basigin/metabolism , Matrix Metalloproteinase 2/genetics , Amino Acid Sequence , Base Sequence , Basigin/chemistry , Cells, Cultured , Enzyme Induction , Escherichia coli , Fibroblasts/enzymology , Glycosylation , Humans , Immunoglobulins/chemistry , Immunoglobulins/metabolism , Matrix Metalloproteinase 2/metabolism , Molecular Sequence Data , Pichia , Protein Processing, Post-Translational , Protein Structure, Tertiary
11.
PLoS One ; 7(4): e35739, 2012.
Article in English | MEDLINE | ID: mdl-22558213

ABSTRACT

Metabolic syndrome (MS) is a constellation of metabolic derangements associated with vascular endothelial dysfunction and oxidative stress and is widely regarded as an inflammatory condition, accompanied by an increased risk for cardiovascular disease. The present study tried to investigate the implications of telomerase activity with inflammation and impaired endothelial function in patients with metabolic syndrome. Telomerase activity in circulating peripheral blood mononuclear cells (PBMC), TNF-α, IL-6 and ADMA were monitored in 39 patients with MS and 20 age and sex-matched healthy volunteers. Telomerase activity in PBMC, TNF-α, IL-6 and ADMA were all significantly elevated in patients with MS compared to healthy volunteers. PBMC telomerase was negatively correlated with HDL and positively correlated with ADMA, while no association between TNF-α and IL-6 was observed. IL-6 was increasing with increasing systolic pressure both in the patients with MS and in the healthy volunteers, while smoking and diabetes were positively correlated with IL-6 only in the patients' group. In conclusion, in patients with MS characterised by a strong dyslipidemic profile and low diabetes prevalence, significant telomerase activity was detected in circulating PBMC, along with elevated markers of inflammation and endothelial dysfunction. These findings suggest a prolonged activity of inflammatory cells in the studied state of this metabolic disorder that could represent a contributory pathway in the pathogenesis of atherosclerosis.


Subject(s)
Biomarkers/blood , Endothelium, Vascular/metabolism , Inflammation/blood , Leukocytes, Mononuclear/metabolism , Metabolic Syndrome/blood , Telomerase/blood , Adult , Aged , Arginine/analogs & derivatives , Arginine/blood , Blood Pressure , Case-Control Studies , Endothelium, Vascular/physiopathology , Female , Greece , Humans , Inflammation/complications , Inflammation/physiopathology , Interleukin-6/blood , Leukocytes, Mononuclear/pathology , Lipoproteins, HDL/blood , Male , Metabolic Syndrome/complications , Metabolic Syndrome/physiopathology , Middle Aged , Oxidative Stress , Tumor Necrosis Factor-alpha/blood
12.
PLoS One ; 6(9): e23906, 2011.
Article in English | MEDLINE | ID: mdl-21912649

ABSTRACT

MAb 4C5 is a cell impermeable, anti-HSP90 murine monoclonal antibody, originally produced using hybridoma technology. We have previously shown that mAb 4C5 specifically recognizes both the α- and to a lesser extent the ß-isoform of HSP90. Additionally, in vitro and in vivo studies revealed that by selectively inhibiting the function of cell-surface HSP90, mAb 4C5 significantly impairs cancer cell invasion and metastasis. Here we describe the reconstitution of mAb 4C5 into a mouse-human chimera. More importantly we report that mAb 4C5 and consequently its chimeric counterpart are completely devoid of heavy chain and consist only of a functional kappa light chain dimer. The chimeric antibody is shown to retain the original antibody's specificity and functional properties. Thus it is capable of inhibiting the function of surface HSP90, leading to reduced cancer cell invasion in vitro. Finally, we present in vivo evidence showing that the chimeric 4C5 significantly inhibits the metastatic deposit formation of MDA-MB-453 cells into the lungs of SCID mice. These data suggest that a chimeric kappa light chain antibody could be potentially used as an anti-cancer agent, thereby introducing a novel type of antibody fragment, with reduced possible adverse immunogenic effects, into cancer therapeutics.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , HSP90 Heat-Shock Proteins/immunology , Immunoglobulin kappa-Chains/pharmacology , Protein Multimerization , Single-Chain Antibodies/pharmacology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Antibody Specificity , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Base Sequence , Cell Line, Tumor , Cloning, Molecular , Female , Humans , Hybridomas/cytology , Immunoglobulin kappa-Chains/chemistry , Immunoglobulin kappa-Chains/genetics , Immunoglobulin kappa-Chains/metabolism , Lung/drug effects , Lung/pathology , Mice , Molecular Sequence Data , Neoplasm Invasiveness , Neoplasm Metastasis , Permeability , Protein Structure, Quaternary , Sequence Analysis, DNA , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/genetics , Single-Chain Antibodies/metabolism
13.
Atherosclerosis ; 216(1): 131-8, 2011 May.
Article in English | MEDLINE | ID: mdl-21316675

ABSTRACT

INTRODUCTION: The aim of this study was to evaluate the iron burden of carotid atherosclerotic plaques removed from patients treated for carotid disease and find any relation with haptoglobin genotype and other common cardiovascular risk factors. METHODS: Consecutive patients undergoing carotid endarterectomy were included in the study. All patients had high-grade carotid stenosis (>70%). The clinical characteristics and serum parameters of the study population were recorded and the haptoglobin genotype was determined. The presence of hemosiderin deposits in the plaques was identified using Perl's stain on adjacent serial sections. RESULTS: 70 specimens were processed for histologic examination: 27 plaques from diabetic patients (16 with the Hp 1-1 or 2-1 genotype and 11 with the Hp 2-2 genotype) and 43 plaques from non diabetic patients (20 with the Hp 1-1 or 2-1 genotype and 23 with the Hp 2-2 genotype). In plaques from diabetic patients the density of Perl's iron stain was significantly higher in the Hp 2-2 group compared with that in the Hp 1-1 or 2-1 group (p = 0.008). The correlation and regression analysis of all possible clinical and laboratory predictors of intraplaque iron deposition showed that four factors were independently associated with intraplaque iron deposition: male gender, serum homocysteine, Hp 2-2 genotype and diabetes mellitus treatment. CONCLUSIONS: Male diabetic patients with increased plasma levels of homocysteine and the Hp 2-2 genotype had higher carotid plaque iron deposition. Current evidence and pathophysiological considerations suggest that the increased intraplaque iron deposition may be associated with increased oxidative stress, affecting the stability of the carotid plaque.


Subject(s)
Cardiovascular Diseases/genetics , Carotid Stenosis/genetics , Haptoglobins/genetics , Iron/analysis , Plaque, Atherosclerotic/genetics , Aged , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Carotid Stenosis/complications , Carotid Stenosis/metabolism , Carotid Stenosis/pathology , Carotid Stenosis/surgery , Diabetes Mellitus/drug therapy , Endarterectomy, Carotid , Female , Genetic Predisposition to Disease , Greece , Hemosiderin/analysis , Homocysteine/blood , Humans , Hyperhomocysteinemia/blood , Hyperhomocysteinemia/complications , Hypoglycemic Agents/therapeutic use , Linear Models , Male , Middle Aged , Oxidative Stress , Phenotype , Plaque, Atherosclerotic/complications , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Plaque, Atherosclerotic/surgery , Risk Assessment , Risk Factors , Severity of Illness Index , Sex Factors , Staining and Labeling
14.
Cancer Immunol Immunother ; 59(5): 715-27, 2010 May.
Article in English | MEDLINE | ID: mdl-19904532

ABSTRACT

Our aim is to develop peptide vaccines that stimulate tumor antigen-specific T-lymphocyte responses against frequently detected cancers. We describe herein a novel HLA-A*0201-restricted epitope, encompassing amino acids 828-836 (residues QIAKGMSYL), which is naturally presented by various HER-2/neu (+) tumor cell lines. HER-2/neu(828-836), [HER-2(9(828))], possesses two anchor residues and stabilized HLA-A*0201 on T2 cells in a concentration-dependent Class I binding assay. This peptide was stable for 3.5 h in an off-kinetic assay. HER-2(9(828)) was found to be immunogenic in HLA-A*0201 transgenic (HHD) mice inducing peptide-specific and functionally potent CTL and long-lasting anti-tumor immunity. Most important, using HLA-A*0201 pentamer analysis we could detect increased ex vivo frequencies of CD8(+) T-lymphocytes specifically recognizing HER-2(9(828)) in 8 out of 20 HLA-A*0201(+) HER-2/neu (+) breast cancer patients. Moreover, HER-2(9(828))-specific human CTL recognized the tumor cell line SKOV3.A2 as well as the primary RS.A2.1.DR1 tumor cell line both expressing HER-2/neu and HLA-A*0201. Finally, therapeutic vaccination with HER-2(9(828)) in HHD mice was proven effective against established transplantable ALC.A2.1.HER tumors, inducing complete tumor regression in 50% of mice. Our data encourage further exploitation of HER-2(9(828)) as a promising candidate for peptide-based cancer vaccines.


Subject(s)
Cancer Vaccines/immunology , Epitopes, T-Lymphocyte/immunology , Immunotherapy/methods , Neoplasms/therapy , Receptor, ErbB-2/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Breast Neoplasms/immunology , Cell Separation , Flow Cytometry , HLA-A Antigens/immunology , HLA-A2 Antigen , Humans , Mice , Neoplasms/immunology , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Peptide Fragments/immunology
15.
Haematologica ; 94(12): 1748-52, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19996119

ABSTRACT

The recently discovered iron regulatory peptide hormone hepcidin holds promise as a novel biomarker in iron metabolism disorders. To date, various mass spectrometry and immunochemical methods have been developed for its quantification in plasma and urine. Differences in methodology and analytical performance hinder the comparability of data. As a first step towards method harmonization, several hepcidin assays were compared. Worldwide eight laboratories participated in a urinary and plasma round robin in which hepcidin was analyzed. For both urine and plasma: (i) the absolute hepcidin concentrations differed widely between methods, (ii) the between-sample variation and the analytical variation of the methods are similar. Importantly, the analytical variation as percentage of the total variance is low for all methods, indicating their suitability to distinguish hepcidin levels of different samples. Spearman correlations between methods were generally high. The round robin results inform the scientific and medical community on the status and agreement of the current hepcidin methods. Ongoing initiatives should facilitate standardization by exchanging calibrators and representative samples.


Subject(s)
Antimicrobial Cationic Peptides/blood , Antimicrobial Cationic Peptides/urine , Clinical Laboratory Techniques/standards , Biomarkers/blood , Biomarkers/urine , Hepcidins , Humans , Immunohistochemistry/methods , Immunohistochemistry/standards , International Cooperation , Iron Metabolism Disorders/blood , Iron Metabolism Disorders/diagnosis , Iron Metabolism Disorders/urine , Mass Spectrometry/methods , Mass Spectrometry/standards , Reference Standards
16.
BMC Cancer ; 9: 386, 2009 Oct 30.
Article in English | MEDLINE | ID: mdl-19878568

ABSTRACT

BACKGROUND: Her2/neu is overexpressed in various human cancers of epithelial origin and is associated with increased metastatic potential and poor prognosis. Several attempts have been made using the extracellular domain of Her2/neu (ECD/Her2) as a prophylactic vaccine in mice with no success in tumor prevention. METHODS: The extracellular domain of Her2/neu (ECD/Her2) was expressed in yeast P. pastoris, in a soluble highly mannosylated form. The immune response of the immunization with this recombinant ECD/Her2 was analyzed using immunoprecipitation and western blot analysis, proliferation and cytotoxicity assays as well as specific tumor growth assays. RESULTS: Mannosylated ECD/Her2 elicited a humoral response with HER2/neu specific antibodies in vaccinated mice, which were able to reduce the proliferation rate of cancer cells in vitro. Moreover, it elicited a cellular response with Her2/neu-specific CTL capable of lysing tumor cells, in vitro. When immunized Balb/c and HHD mice were challenged with Her2/neu-overexpressing cells, tumor growth was inhibited. CONCLUSION: Here we report on the efficacy of the extracellular domain of human Her2/neu produced in yeast P. pastoris, which confers mannosylation of the protein, to act as a potent anti-tumor vaccine against Her2/neu overexpressing tumors. Specific cellular and humoral responses were observed as well as efficacy.


Subject(s)
Neoplasms/immunology , Neoplasms/prevention & control , Pichia/metabolism , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/immunology , Angiogenesis Inducing Agents/blood , Animals , Cancer Vaccines/chemistry , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cancer Vaccines/metabolism , Cell Line, Tumor , Female , Gene Expression , Humans , Immunity , Mice , Mice, Inbred BALB C , Pichia/genetics , Protein Processing, Post-Translational , Protein Structure, Tertiary , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism
17.
PLoS One ; 4(2): e4581, 2009.
Article in English | MEDLINE | ID: mdl-19238200

ABSTRACT

BACKGROUND: Hepcidin is a 25-aminoacid cysteine-rich iron regulating peptide. Increased hepcidin concentrations lead to iron sequestration in macrophages, contributing to the pathogenesis of anaemia of chronic disease whereas decreased hepcidin is observed in iron deficiency and primary iron overload diseases such as hereditary hemochromatosis. Hepcidin quantification in human blood or urine may provide further insights for the pathogenesis of disorders of iron homeostasis and might prove a valuable tool for clinicians for the differential diagnosis of anaemia. This study describes a specific and non-operator demanding immunoassay for hepcidin quantification in human sera. METHODS AND FINDINGS: An ELISA assay was developed for measuring hepcidin serum concentration using a recombinant hepcidin25-His peptide and a polyclonal antibody against this peptide, which was able to identify native hepcidin. The ELISA assay had a detection range of 10-1500 microg/L and a detection limit of 5.4 microg/L. The intra- and interassay coefficients of variance ranged from 8-15% and 5-16%, respectively. Mean linearity and recovery were 101% and 107%, respectively. Mean hepcidin levels were significantly lower in 7 patients with juvenile hemochromatosis (12.8 microg/L) and 10 patients with iron deficiency anemia (15.7 microg/L) and higher in 7 patients with Hodgkin lymphoma (116.7 microg/L) compared to 32 age-matched healthy controls (42.7 microg/L). CONCLUSIONS: We describe a new simple ELISA assay for measuring hepcidin in human serum with sufficient accuracy and reproducibility.


Subject(s)
Anemia/diagnosis , Antimicrobial Cationic Peptides/blood , Enzyme-Linked Immunosorbent Assay/methods , Anemia, Iron-Deficiency/blood , Hemochromatosis/blood , Hepcidins , Hodgkin Disease/blood , Humans , Sensitivity and Specificity
18.
Biochimie ; 90(5): 726-35, 2008 May.
Article in English | MEDLINE | ID: mdl-18295606

ABSTRACT

Hepcidin is a circulating cysteine-rich peptide with antimicrobial properties. It functions as a hormonal regulator of iron homeostasis by controlling iron efflux from target cells via ferroportin (FPN1), which is internalized and degraded upon hepcidin binding. Because of its profound biomedical significance, hepcidin has become the target of intense biochemical studies. The aim of this study was to produce functional recombinant hepcidin in sufficient quantities for advanced research or potential clinical use, as the native hepcidin can be isolated from urine in very low yield. We report the expression, purification and functional characterization of hepcidin variants in yeast P. pastoris. The yield of untagged hepcidin 20- and 25-mer peptides was too low for complete functional characterization. By contrast, Hep20 and Hep25 tagged with either single 6xHis or double Myc-6xHis epitopes were expressed at high quantities (5-7mg/l of culture), yet mostly in oligomeric forms. Purification of monomeric tagged hepcidins was achieved by size exclusion chromatography, with a yield of 0.5-1mg/l of culture. All recombinant hepcidins exhibited bacteriostatic activity and the ability to control cellular iron homeostasis, with Hep25-His being the most potent. Thus, Hep25-His promoted an increase in the levels of the labile iron pool (LIP) in macrophages and consistently bound to ferroportin (FPN1) causing its internalization and the subsequent downregulation of transferrin receptor 1 (TfR1) expression. Analysis by mass-spectrometry suggested that all eight cysteines participated in disulfide bond formation. Our results suggest that only the recombinant Hep25-His monomer was a fully active peptide. As Hep25-His faithfully recapitulates the functional properties of native Hep25, it represents a powerful tool for biochemical studies and potential diagnostic and therapeutic applications.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Iron/metabolism , Pichia/genetics , Amino Acid Sequence , Animals , Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/chemistry , Cell Line , Hepcidins , Humans , Macrophages/drug effects , Macrophages/metabolism , Mass Spectrometry , Mice , Molecular Sequence Data , Plasmids , Recombinant Proteins/chemistry , Recombinant Proteins/pharmacology , Sequence Homology, Amino Acid
19.
Cancer Immunol Immunother ; 55(9): 1091-9, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16311733

ABSTRACT

The Her2/neu oncogene is overexpressed in various human cancers of epithelial origin and is associated with increased metastatic potential and poor prognosis. Blocking the Her2/neu signalling has been the focus of most therapeutic approaches. In this paper, the Her2/neu extracellular domain expressed in soluble form in yeast Pichia pastoris was used in order to isolate a fully human Fab fragment from a combinatorial Fab phage display library, derived from invaded lymph nodes of a breast cancer patient. The isolated fully human Fab63 binds specifically the native Her2/neu receptor and competes with Herceptin for binding to soluble Her2/neu receptor. In Her2/neu overexpressing cancer cells, Fab63 is rapidly internalized and has significant antiproliferative effects, where ligand-independent mechanisms dominate signal induction. Moreover, in the presence of the ligand heregulin, growth inhibition was also detected by Fab63. The human Fab63 is a non-immunogenic agent with unique properties that can be applied in diagnosis and cancer therapy, with great potential for further manipulation towards the generation of an effective anticancer molecule.


Subject(s)
Antibodies, Monoclonal/immunology , Breast Neoplasms/drug therapy , Immunoglobulin Fab Fragments/immunology , Peptide Library , Receptor, ErbB-2/antagonists & inhibitors , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Antibody Specificity , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoglobulin Fab Fragments/isolation & purification , Immunoglobulin Fab Fragments/pharmacology , Microscopy, Confocal , Pichia/chemistry , Pichia/genetics , Receptor, ErbB-2/metabolism , Trastuzumab , Up-Regulation
20.
J Neurochem ; 95(1): 146-59, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16181419

ABSTRACT

BM88 is a neurone-specific protein implicated in cell cycle exit and differentiation of neuronal precursors. It is widely expressed in terminally differentiated neurones but also in neuronal progenitors, albeit in lower levels. Thus BM88 expression shows a tight correlation with the progression of progenitor cells towards neuronal differentiation. Here we report the genomic organization and proximal promoter characterization of the human and mouse BM88 genes. Both promoters lie in a CpG island, are TATA-less and have multiple transcription start sites. Deletion analysis performed on the human BM88 gene revealed an 88 bp minimal promoter fragment that is preferentially active in neural cells. Importantly, this minimal promoter is sufficient to confer specific transcriptional activity in primary neurones, but not in glial cells. Within the promoter region there are four functional Sp1-binding sites. Simultaneous mutations to all four Sp1 sites results in complete loss of promoter activity. Transactivation experiments revealed that Sp1 directly activates the BM88 promoter while activation also occurs in the presence of neurogenin-1. Characterization of the promoter elements that control neurone-specific and developmental expression of BM88 should contribute to the elucidation of the transcriptional networks that regulate the transition from a proliferative neural progenitor to a post-mitotic neurone.


Subject(s)
Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Promoter Regions, Genetic/physiology , Animals , Base Sequence , Cell Line , DNA/metabolism , DNA Mutational Analysis , Genome , Humans , Luciferases/metabolism , Membrane Proteins , Mice , Molecular Sequence Data , Neurons/enzymology , Promoter Regions, Genetic/genetics , Pseudogenes , Sequence Deletion , Transcription Initiation Site
SELECTION OF CITATIONS
SEARCH DETAIL
...