Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Clin Oral Investig ; 26(12): 7209-7218, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35986791

ABSTRACT

OBJECTIVES: IL-23p19/Ebi3 (IL-39) was described as a new IL-12 family member. The aim of this study is to evaluate the gingival crevicular fluid (GCF) IL-39 levels in periodontal diseases and health and to correlate them to GCF levels of IL-1ß and periostin. MATERIALS AND METHODS: Sixty-six adult patients were included in the study. The study design was comprised of three groups, each containing 22 individuals: the periodontally healthy (PH), gingivitis (G), and periodontitis (P) groups. The clinical periodontal parameters were recorded and GCF samples were collected from the participants. GCF interleukin (IL)-39, IL-1ß, and periostin levels were examined using the enzyme-linked immunosorbent assay. RESULTS: GCF IL­1ß, periostin, and IL-39 levels were higher in the P and G groups than in the PH group (p < 0.001). Positive correlations were detected between all GCF biochemical parameters and clinical periodontal parameters (p < 0.05). In the multivariate generalized linear regression analysis, the P (ß = 37.6, 95% CI = 22.9-52.4) and G (ß = 28.4, 95% CI = 15.8-41) groups were associated with GCF IL-39 levels (p < 0.001). CONCLUSION: IL-39 levels were elevated in the presence of periodontal disease paralleling the increase in IL­1ß and periostin levels. IL-39 may have a role in the periodontal inflammation process. STATEMENT OF CLINICAL RELEVANCE: IL-39, a new cytokine from the IL-12 family, can be a possible predictor marker of periodontal diseases.


Subject(s)
Chronic Periodontitis , Gingivitis , Adult , Humans , Gingival Crevicular Fluid/chemistry , Interleukin-12/analysis , Interleukin-23 Subunit p19/analysis , Interleukins , Minor Histocompatibility Antigens/analysis
2.
Clin Epigenetics ; 13(1): 232, 2021 12 27.
Article in English | MEDLINE | ID: mdl-34961566

ABSTRACT

BACKGROUND: Circulating tumor DNA (ctDNA) is a promising diagnostic and prognostic marker for many cancers and has been actively investigated in recent years. Previous studies have already demonstrated the potential use of ctDNA methylation markers in the diagnosis and prognostication of colorectal cancer (CRC). This retrospective study validated the value of methylation biomarker MYO1-G (cg10673833) in CRC diagnosis and disease monitoring using digital droplet PCR (ddPCR), a biomarker selected from our previous study due to its highest diagnostic efficiency. METHODS: Blood samples of CRC and control samples from tumor-free individuals at two institutions were collected to quantify the methylation ratio using ddPCR. Area under curve (AUC) was calculated after constructing receiver operating characteristic curve (ROC) for CRC diagnosis. Sensitivity and specificity were estimated and comparisons of methylation ratio in different groups were performed. RESULTS: We collected 673 blood samples from 272 patients diagnosed with stage I-IV CRC and 402 normal control samples. The methylation biomarker discriminated patients with CRC from normal controls with high accuracy (area under curve [AUC] = 0.94) and yielded a sensitivity of 84.3% and specificity of 94.5%. Besides, methylation ratio of MYO1-G was associated with tumor burden and treatment response. The methylation ratio was significantly lower in patients after their radical operation than when compared with those before surgeries (P < 0.001). Methylation ratio was significantly higher in patients with disease progression than those with stable disease (P = 0.002) and those with complete response or partial response (P = 0.009). CONCLUSIONS: Together, our study indicated that this methylation marker can serve as a potential biomarker for diagnosing and monitoring CRC.


Subject(s)
Circulating Tumor DNA/analysis , Colorectal Neoplasms/blood , Minor Histocompatibility Antigens/analysis , Myosins/analysis , Adult , Area Under Curve , Biomarkers, Tumor/analysis , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , China/epidemiology , Circulating Tumor DNA/blood , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/genetics , DNA Methylation/genetics , DNA Methylation/physiology , Female , Humans , Male , Middle Aged , Minor Histocompatibility Antigens/blood , Myosins/blood , ROC Curve
3.
Article in English | MEDLINE | ID: mdl-31881361

ABSTRACT

BACKGROUND & AIMS: Nonalcoholic steatohepatitis (NASH) occurs in the context of aberrant metabolism. Glutaminolysis is required for metabolic reprograming of hepatic stellate cells (HSCs) and liver fibrogenesis in mice. However, it is unclear how changes in HSC glutamine metabolism contribute to net changes in hepatic glutaminolytic activity during fibrosis progression, or whether this could be used to track fibrogenic activity in NASH. We postulated that increased HSC glutaminolysis marks active scarring in NASH. METHODS: Glutaminolysis was assessed in mouse NASH fibrosis models and in NASH patients. Serum and liver levels of glutamine and glutamate and hepatic expression of glutamine transporter/metabolic enzymes were correlated with each other and with fibrosis severity. Glutaminolysis was disrupted in HSCs to examine if this directly influenced fibrogenesis. 18F-fluoroglutamine positron emission tomography was used to determine how liver glutamine assimilation tracked with hepatic fibrogenic activity in situ. RESULTS: The serum glutamate/glutamine ratio increased and correlated with its hepatic ratio, myofibroblast content, and fibrosis severity. Healthy livers almost exclusively expressed liver-type glutaminase (Gls2); Gls2 protein localized in zone 1 hepatocytes, whereas glutamine synthase was restricted to zone 3 hepatocytes. In fibrotic livers, Gls2 levels reduced and glutamine synthase zonality was lost, but both Slc1a5 (glutamine transporter) and kidney-type Gls1 were up-regulated; Gls1 protein was restricted to stromal cells and accumulated in fibrotic septa. Hepatocytes did not compensate for decreased Gls2 by inducing Gls1. Limiting glutamine or directly inhibiting GLS1 inhibited growth and fibrogenic activity in cultured human HSCs. Compared with healthy livers, fibrotic livers were 18F-fluoroglutamine-avid by positron emission tomography, suggesting that glutamine-addicted myofibroblasts drive increased hepatic utilization of glutamine as fibrosis progresses. CONCLUSIONS: Glutaminolysis is a potential diagnostic marker and therapeutic target during NASH fibrosis progression.


Subject(s)
Cicatrix/diagnosis , Liver Cirrhosis/diagnosis , Liver/pathology , Non-alcoholic Fatty Liver Disease/pathology , Adult , Amino Acid Transport System ASC/analysis , Amino Acid Transport System ASC/metabolism , Animals , Biomarkers/analysis , Biomarkers/metabolism , Cell Line , Cicatrix/pathology , Disease Models, Animal , Disease Progression , Female , Glutaminase/analysis , Glutaminase/metabolism , Glutamine/analysis , Glutamine/metabolism , Hepatic Stellate Cells/pathology , Humans , Liver/cytology , Liver/diagnostic imaging , Liver Cirrhosis/pathology , Male , Metabolomics , Mice , Middle Aged , Minor Histocompatibility Antigens/analysis , Minor Histocompatibility Antigens/metabolism , Myofibroblasts/pathology , Positron-Emission Tomography
4.
Intern Emerg Med ; 14(5): 713-718, 2019 08.
Article in English | MEDLINE | ID: mdl-30820838

ABSTRACT

The Endoplasmic reticulum aminopeptidase protein 1 (ERAP1) trims N-terminal amino acids from epitope precursors for Major Histocompatibility Complex class I presentation. Genome-wide association studies demonstrated that ERAP1 gene single nucleotide polymorphisms (SNPs) are associated with Behçet's syndrome (BS). This study was conducted on the two most consistently BS-associated ERAP1 polymorphisms, rs17482078 (NG_027839.1:g.35983G>A) and rs27044 (NG_027839.1:g.35997C>G) to analyse their distribution in 55 Italian BS patients and 65 ethnically matched controls (healthy controls, HC) and to test their association with BS risk. SNPs were detected by isolation, amplification of genomic DNA and direct sequencing. SNPs functional effects were predicted by bioinformatics software. The odds ratio (OR) with 95% confidence intervals was calculated to assess the strength of BS association for genotypes and alleles, also validated by logistic regression (LR). LR was used to test the association between both SNPs and patients HLA genetic data. Bonferroni correction was also applied. Comparing patients and controls, we found a significant higher frequency of rs17482078 A allele (32.73% BS vs 17.69% HC, p = 0.007) and AA genotype (18.18% BS vs 0% HC; p = 0.0003) and rs27044 G allele (63.64% BS vs 46.92% HC; p = 0.0096) in BS group after Bonferroni correction. No association was found between HLA-B*51 and both ERAP1 SNPs. Although preliminary, our data show a stronger association of rs17482078 with BS compared to rs27044 by means of case-control genetic analysis and bioinformatics prediction of protein structure change. A larger series of patients and controls is required to confirm our preliminary findings.


Subject(s)
Aminopeptidases/analysis , Behcet Syndrome/genetics , Minor Histocompatibility Antigens/analysis , Polymorphism, Genetic/genetics , Adult , Case-Control Studies , Chi-Square Distribution , Computer Simulation , Female , Genome-Wide Association Study/methods , Humans , Italy , Logistic Models , Male , Middle Aged , Odds Ratio , Prospective Studies
5.
Histol Histopathol ; 34(2): 167-174, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30191947

ABSTRACT

PURPOSE: We evaluated the expression of glutaminolysis-related proteins in Hurthle cell neoplasms (HCN) and follicular neoplasms (FN) of the thyroid, and investigated its clinical implication. METHODS: Tissue microarrays were constructed from 264 FNs (112 follicular carcinomas [FCs] and 152 follicular adenomas [FAs]) and 108 HCNs (27 Hurthle cell carcinomas [HCCs] and 81 Hurthle cell adenomas [HCAs]. The immunohistochemical staining result of 3 glutaminolysis-related proteins (Glutaminase 1 [GLS1], glutaminate dehydrogenase [GDH] and alanine- serine, cysteine-preferring transporter 2 [ASCT2]) was analyzed. RESULTS: GLS1 and GDH showed significantly higher expression rates in HCN compared to FN (P<0.001). More HCN cases showed co-positivity of multiple glutaminolysis-related proteins than those of FN cases (P<0.001). In silico analysis, both GLUD1 and GLUD2 showed higher expression rate in HCA compared to FA (P=0.027 and P=0.018, respectively). SLC1A5 expression was highest in HCA, followed by FC and FA (HCA vs FC, P=0.023; FC vs FA, P=0.002). CONCLUSION: FN and HCN exhibit a different expression pattern for glutaminolysis-related proteins, and GLS1 and GDH have higher expression rates in HCN and FN.


Subject(s)
Adenocarcinoma, Follicular/metabolism , Adenoma, Oxyphilic/metabolism , Thyroid Neoplasms/metabolism , Adult , Amino Acid Transport System ASC/analysis , Amino Acid Transport System ASC/biosynthesis , Female , Glutamate Dehydrogenase/analysis , Glutamate Dehydrogenase/biosynthesis , Glutaminase/analysis , Glutaminase/biosynthesis , Glutamine/metabolism , Humans , Male , Middle Aged , Minor Histocompatibility Antigens/analysis , Minor Histocompatibility Antigens/biosynthesis
6.
Front Immunol ; 9: 1750, 2018.
Article in English | MEDLINE | ID: mdl-30116242

ABSTRACT

The immune system has been traditionally divided into two arms called innate and adaptive immunity. Typically, innate immunity refers to rapid defense mechanisms that set in motion within minutes to hours following an insult. Conversely, the adaptive immune response emerges after several days and relies on the innate immune response for its initiation and subsequent outcome. However, the recent discovery of immune cells displaying merged properties indicates that this distinction is not mutually exclusive. These populations that span the innate-adaptive border of immunity comprise, among others, CD1d-restricted natural killer T cells and MR1-restricted mucosal-associated invariant T cells. These cells have the unique ability to swiftly activate in response to non-peptidic antigens through their T cell receptor and/or to activating cytokines in order to modulate many aspects of the immune response. Despite they recirculate all through the body via the bloodstream, these cells mainly establish residency at barrier sites including lungs. Here, we discuss the current knowledge into the biology of these cells during lung (viral and bacterial) infections including activation mechanisms and functions. We also discuss future strategies targeting these cell types to optimize immune responses against respiratory pathogens.


Subject(s)
Mucosal-Associated Invariant T Cells/immunology , Natural Killer T-Cells/immunology , Pneumonia, Bacterial/immunology , Pneumonia, Viral/immunology , Tuberculosis, Pulmonary/immunology , Adaptive Immunity , Animals , Antigens, CD1d/analysis , Histocompatibility Antigens Class I/analysis , Humans , Immunity, Innate , Immunotherapy, Adoptive , Minor Histocompatibility Antigens/analysis , Vaccines/therapeutic use
7.
Biosci Rep ; 38(4)2018 09 03.
Article in English | MEDLINE | ID: mdl-30054427

ABSTRACT

Autoimmune and autoinflammatory conditions represent a group of disorders characterized by self-directed tissue damage due to aberrant changes in innate and adaptive immune responses. These disorders possess widely varying clinical phenotypes and etiology; however, they share a number of similarities in genetic associations and environmental influences. Whilst the pathogenic mechanisms of disease remain poorly understood, genome wide association studies (GWAS) have implicated a number of genetic loci that are shared between several autoimmune and autoinflammatory conditions. Association of particular HLA alleles with disease susceptibility represents one of the strongest genetic associations. Furthermore, recent GWAS findings reveal strong associations with single nucleotide polymorphisms in the endoplasmic reticulum aminopeptidase 1 (ERAP1) gene and susceptibility to a number of these HLA-associated conditions. ERAP1 plays a major role in regulating the repertoire of peptides presented on HLA class I alleles at the cell surface, with the presence of single nucleotide polymorphisms in ERAP1 having a significant impact on peptide processing function and the repertoire of peptides presented. The impact of this dysfunctional peptide generation on CD8+ T-cell responses has been proposed as a mechanism of pathogenesis diseases where HLA and ERAP1 are associated. More recently, studies have highlighted a role for ERAP1 in innate immune-mediated pathways involved in inflammatory responses. Here, we discuss the role of polymorphic ERAP1 in various immune cell functions, and in the context of autoimmune and autoinflammatory disease pathogenesis.


Subject(s)
Aminopeptidases/genetics , Autoimmune Diseases/genetics , Inflammation/genetics , Minor Histocompatibility Antigens/genetics , Polymorphism, Single Nucleotide , Aminopeptidases/analysis , Aminopeptidases/immunology , Animals , Antigen Presentation , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Autoimmunity , Genome-Wide Association Study , Humans , Immunity, Cellular , Immunity, Innate , Inflammation/immunology , Inflammation/pathology , Minor Histocompatibility Antigens/analysis , Minor Histocompatibility Antigens/immunology
8.
Oncol Res Treat ; 41(3): 111-115, 2018.
Article in English | MEDLINE | ID: mdl-29485413

ABSTRACT

BACKGROUND AND AIM: So far, the understanding of the role of Epstein-Barr virus-induced gene 3 (EBI3) in breast cancer has been limited. This study uncovers the functional role and clinical significance of EBI3 in breast cancer patients. PATIENTS AND METHODS: The expression levels of EBI3, IL-27p28, and IL-12p35 were measured by quantitative real-time reverse transcription (RT)-polymerase chain reaction (PCR). Correlations of EBI3 expression with IL-27p28 and IL-12p35 expression were analyzed using Pearson's correlation assay. The prognostic performance of EBI3 was assessed via Kaplan-Meier survival assay and Cox regression analysis. RESULTS: EBI3 expression was increased in cancerous tissues compared with the controls (P < 0.05). This overexpression of EBI3 was correlated with lymph node metastasis and clinical stage (both P < 0.05). Besides, elevated expression of EBI3 was usually found in patients with positive lymph node metastasis (P < 0.05), and similar results were obtained in advanced clinical-stage breast cancer cases (P < 0.05). Increases in both IL-27p28 and IL-12p35 expression were identified in breast cancer tissues (all P < 0.05), and IL-12p35 expression was found to be associated with EBI3 expression (R = 0.888, P < 0.001). Survival curves revealed that high EBI3 expression was correlated with poor overall survival (log-rank P < 0.05). The Cox analysis indicated that EBI3 was an independent prognostic factor in breast cancer. CONCLUSION: Taken together, overexpression of EBI3 was associated with poor prognosis and might be involved in the progression of breast cancer.


Subject(s)
Breast Neoplasms/pathology , Interleukins/physiology , Minor Histocompatibility Antigens/physiology , Adult , Aged , Breast Neoplasms/mortality , Disease Progression , Female , Humans , Interleukins/analysis , Middle Aged , Minor Histocompatibility Antigens/analysis , Proportional Hazards Models , Up-Regulation
9.
Mol Cell Probes ; 37: 39-47, 2018 02.
Article in English | MEDLINE | ID: mdl-29179987

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is a common cancer with high morbidity and mortality. However, its molecular mechanism is not clear, nor the genes related to CRC stages. METHODS: Gene expression data in CRC and healthy colorectal tissues were obtained from gene expression omnibus. Limma package was used to identify the differentially expressed genes (DEGs) between control and CRC (stage I, II, III, and IV), obtaining 4 DEG sets. VennPlex was utilized to find all DEGs and intersection DEGs. Functional interactions between all DEGs and protein-protein interactions (PPIs) between intersection DEGs were analyzed using ReactomeFIViz and STRING, respectively, and networks were visualized. Known CRC-related genes were down-loaded from Comparative Toxicogenomics Database and mapped to PPI network. RESULTS: Totally, 851, 760, 729, and 878 DEGs were found between control and CRC stage I, II, III, and IV, respectively. Taken together, 1235 DEGs were found, as well as 128 up-regulated intersection DEGs, 365 down-regulated intersection DEGs, and 0 contra-regulated DEG. A functional interaction network of all DEGs and a PPI network of intersection DEGs were constructed, in which CDC20, PTTG1, and MAD2L1 interacted with BUB1B; UGT2B17 interacted with ADH1B; MCM7 interacted with MCM2. BUB1B, ADH1B, and MCM2 were known CRC-related genes. Gradually upregulated expressions of CDC20, PTTG1, MAD2L1, UGT2B17, and MCM7 in stage I, II, III, and IV CRC were confirmed by using quantitative PCR. Besides, up-regulated intersection DEGs enriched in pathways about Cell cycle, DNA replication, and p53 signaling. CONCLUSION: CDC20, PTTG1, MAD2L1, UGT2B17, and MCM7 might be CRC stage-related genes.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Profiling , Adaptor Proteins, Signal Transducing/analysis , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cdc20 Proteins/analysis , Cdc20 Proteins/genetics , Cdc20 Proteins/metabolism , Cell Cycle Proteins/analysis , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Databases, Genetic , Gene Expression Regulation, Neoplastic , Glucuronosyltransferase/analysis , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , Minichromosome Maintenance Complex Component 7/analysis , Minichromosome Maintenance Complex Component 7/genetics , Minichromosome Maintenance Complex Component 7/metabolism , Minor Histocompatibility Antigens/analysis , Minor Histocompatibility Antigens/genetics , Minor Histocompatibility Antigens/metabolism , Neoplasm Staging , Nuclear Proteins/analysis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Interaction Maps , Securin/analysis , Securin/genetics , Securin/metabolism
10.
Nat Immunol ; 18(12): 1321-1331, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28991267

ABSTRACT

Type 1 diabetes (T1D) is an autoimmune disease that results from the destruction of pancreatic ß-cells by the immune system that involves innate and adaptive immune cells. Mucosal-associated invariant T cells (MAIT cells) are innate-like T-cells that recognize derivatives of precursors of bacterial riboflavin presented by the major histocompatibility complex (MHC) class I-related molecule MR1. Since T1D is associated with modification of the gut microbiota, we investigated MAIT cells in this pathology. In patients with T1D and mice of the non-obese diabetic (NOD) strain, we detected alterations in MAIT cells, including increased production of granzyme B, which occurred before the onset of diabetes. Analysis of NOD mice that were deficient in MR1, and therefore lacked MAIT cells, revealed a loss of gut integrity and increased anti-islet responses associated with exacerbated diabetes. Together our data highlight the role of MAIT cells in the maintenance of gut integrity and the control of anti-islet autoimmune responses. Monitoring of MAIT cells might represent a new biomarker of T1D, while manipulation of these cells might open new therapeutic strategies.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Histocompatibility Antigens Class I/analysis , Intestinal Mucosa/immunology , Minor Histocompatibility Antigens/analysis , Mucosal-Associated Invariant T Cells/immunology , Pancreas/immunology , Animals , Cells, Cultured , Gastrointestinal Microbiome/immunology , Granzymes/biosynthesis , Humans , Insulin-Secreting Cells/immunology , Intestinal Mucosa/cytology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Pancreas/cytology
11.
J Am Soc Mass Spectrom ; 28(9): 1796-1804, 2017 09.
Article in English | MEDLINE | ID: mdl-28681360

ABSTRACT

Structural technologies are an essential component in the design of precision therapeutics. Precision medicine entails the development of therapeutics directed toward a designated target protein, with the goal to deliver the right drug to the right patient at the right time. In the field of oncology, protein structural variants are often associated with oncogenic potential. In a previous proteogenomic screen of patient-derived glioblastoma (GBM) tumor materials, we identified a sequence variant of human mitochondrial branched-chain amino acid aminotransferase 2 as a putative factor of resistance of GBM to standard-of-care-treatments. The enzyme generates glutamate, which is neurotoxic. To elucidate structural coordinates that may confer altered substrate binding or activity of the variant BCAT2 T186R, a ~45 kDa protein, we applied combined ETD and CID top-down mass spectrometry in a LC-FT-ICR MS at 21 T, and X-Ray crystallography in the study of both the variant and non-variant intact proteins. The combined ETD/CID fragmentation pattern allowed for not only extensive sequence coverage but also confident localization of the amino acid variant to its position in the sequence. The crystallographic experiments confirmed the hypothesis generated by in silico structural homology modeling, that the Lys59 side-chain of BCAT2 may repulse the Arg186 in the variant protein (PDB code: 5MPR), leading to destabilization of the protein dimer and altered enzyme kinetics. Taken together, the MS and novel 3D structural data give us reason to further pursue BCAT2 T186R as a precision drug target in GBM. Graphical Abstract ᅟ.


Subject(s)
Crystallography, X-Ray/methods , Mass Spectrometry/methods , Minor Histocompatibility Antigens/chemistry , Models, Molecular , Pregnancy Proteins/chemistry , Transaminases/chemistry , Amino Acid Sequence , Humans , Minor Histocompatibility Antigens/analysis , Minor Histocompatibility Antigens/genetics , Mutation , Precision Medicine , Pregnancy Proteins/analysis , Pregnancy Proteins/genetics , Transaminases/analysis , Transaminases/genetics
12.
Anal Chem ; 89(14): 7641-7648, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28613839

ABSTRACT

Endoplasmic reticulum aminopeptidase 1 (ERAP1), a metallopeptidase belonging to the M1 peptidase family, plays an important role in antigen processing in vivo. Additionally, many diseases are caused by ERAP1 perturbation. Thus, an efficient method for monitoring its content is extremely important for disease diagnosis and treatment. However, few fluorescent probes have been reported for efficiently monitoring ERAP1 in living cells and tissues. In this work, a two-photon fluorescent probe (SNCL) containing 1,8-naphthalimide (two-photon fluorophore), l-leucine (trigger moiety), and a methyl sulfonamide moiety (endoplasmic reticulum-targeting group) for imaging ERAP1 activity in living cells is reported for the first time. The optimized probe exhibited high sensitivity toward ERAP1, with about a 95-fold fluorescence enhancement at 550 nm. Herein, we monitored ERAP1 with SNCL by introducing interferon-γ to induce ERAP1 activity in living cells. The content of ERAP1 was dependent on the redox state of the endoplasmic reticulum, which was demonstrated by using SNCL to monitor the enzymatic activity of ERAP1 under different redox conditions. Excitingly, SNCL was also successfully applied for monitoring ERAP1 in tumor tissue with an imaging depth of 50-120 µm. In conclusion, SNCL not only can be used for the sensitive detection of endogenous ERAP1 in living cells and tumor tissues but also can serve as a potentially useful tool to reveal ERAP1-related diseases.


Subject(s)
Aminopeptidases/analysis , Endoplasmic Reticulum/enzymology , Fluorescent Dyes/chemistry , Minor Histocompatibility Antigens/analysis , Photons , Aminopeptidases/metabolism , Animals , Fluorescent Dyes/chemical synthesis , HeLa Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Microscopy, Fluorescence , Minor Histocompatibility Antigens/metabolism , Molecular Structure , Optical Imaging , Oxidation-Reduction
13.
Am J Hum Genet ; 99(3): 711-719, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27545680

ABSTRACT

The overall understanding of the molecular etiologies of intellectual disability (ID) and developmental delay (DD) is increasing as next-generation sequencing technologies identify genetic variants in individuals with such disorders. However, detailed analyses conclusively confirming these variants, as well as the underlying molecular mechanisms explaining the diseases, are often lacking. Here, we report on an ID syndrome caused by de novo heterozygous loss-of-function (LoF) mutations in SON. The syndrome is characterized by ID and/or DD, malformations of the cerebral cortex, epilepsy, vision problems, musculoskeletal abnormalities, and congenital malformations. Knockdown of son in zebrafish resulted in severe malformation of the spine, brain, and eyes. Importantly, analyses of RNA from affected individuals revealed that genes critical for neuronal migration and cortex organization (TUBG1, FLNA, PNKP, WDR62, PSMD3, and HDAC6) and metabolism (PCK2, PFKL, IDH2, ACY1, and ADA) are significantly downregulated because of the accumulation of mis-spliced transcripts resulting from erroneous SON-mediated RNA splicing. Our data highlight SON as a master regulator governing neurodevelopment and demonstrate the importance of SON-mediated RNA splicing in human development.


Subject(s)
Brain/embryology , Brain/metabolism , DNA-Binding Proteins/genetics , Genes, Essential/genetics , Intellectual Disability/genetics , Minor Histocompatibility Antigens/genetics , Mutation/genetics , RNA Splicing/genetics , Animals , Brain/abnormalities , Brain/pathology , DNA-Binding Proteins/analysis , DNA-Binding Proteins/metabolism , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Developmental Disabilities/physiopathology , Eye Abnormalities/genetics , Female , Haploinsufficiency/genetics , Head/abnormalities , Heterozygote , Humans , Intellectual Disability/pathology , Intellectual Disability/physiopathology , Male , Metabolic Diseases/genetics , Metabolic Diseases/metabolism , Minor Histocompatibility Antigens/analysis , Minor Histocompatibility Antigens/metabolism , Pedigree , RNA, Messenger/analysis , Spine/abnormalities , Syndrome , Zebrafish/abnormalities , Zebrafish/embryology , Zebrafish/genetics
14.
Pancreatology ; 16(5): 905-16, 2016.
Article in English | MEDLINE | ID: mdl-27453460

ABSTRACT

BACKGROUND/OBJECTIVES: The aim of this study was to investigate the role of the guanine nucleotide exchange factor Vav3 in the motility and invasiveness of pancreatic ductal adenocarcinoma (PDAC) cells. METHODS: Immunohistochemistry was used to determine whether high Vav3 expression in human PDAC tissues is correlated with poor prognosis. Immunocytochemistry was used to determine the association and intracellular distribution of Vav3, Rac1 and Akt in PDAC cells. Phosphoprotein array analysis was performed to determine the Vav3-associated intracellular signaling pathways. Immunocytochemistry and Matrigel invasion assays were used to examine the effects of Vav3 on the formation of cell protrusions and PDAC cell invasion. RESULTS: Expression of Vav3 in PDAC tissue was significantly correlated with overall survival. Vav3 was localized in cell protrusions of migrating PDAC cells. Knockdown of Vav3 inhibited the motility and invasiveness of PDAC cells through a decrease in cell protrusions. The levels of active Rac1 or active Akt were not associated with the concentration of Vav3 in cell protrusions. The Vav3-dependent promotion of motility and invasiveness was not modulated by Rac1 or Akt. Additionally, knockdown of Vav3 increased phosphorylated WNK1 in PDAC cells, and knockdown of WNK1 inhibited the motility and invasiveness. This study suggests that Vav3 can be a useful marker for predicting the outcome of patients with PDAC and that Vav3 can promote PDAC cell motility and invasion through association with dephosphorylation of WNK1. CONCLUSIONS: Vav3 was accumulated in cell protrusions, contributed to the formation of membrane protrusions, and thereby increased the motility and invasiveness of PDAC cells.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins c-vav/analysis , Proto-Oncogene Proteins c-vav/genetics , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Carcinoma, Pancreatic Ductal/surgery , Cell Movement/genetics , Female , Gene Knockdown Techniques , Humans , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/analysis , Intracellular Signaling Peptides and Proteins/genetics , Male , Middle Aged , Minor Histocompatibility Antigens/analysis , Minor Histocompatibility Antigens/genetics , Neoplasm Invasiveness/genetics , Pancreatic Neoplasms/surgery , Phosphorylation , Prognosis , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/blood , Survival Analysis , WNK Lysine-Deficient Protein Kinase 1 , rac1 GTP-Binding Protein/blood
15.
Rev Med Chil ; 140(5): 555-60, 2012 May.
Article in Spanish | MEDLINE | ID: mdl-23096659

ABSTRACT

BACKGROUND: Minor histocompatibility antigens (mHAgs) play a critical role in the immune responses associated with allogeneic stem cell transplantation, such as graft versus host disease (GVHD) and graft-versus-tumor (GVT). AIM: To determine the gene frequencies of the mHAgs HA-1, HA-2 and HA-8 in Chilean Blood Bank donors. MATERIAL AND METHODS: Blood from 192 blood donors was analyzed. The presence of haplotype HLA-A*02 was determined by flow cytometry. The frequency of mHAgs was determined by allele specific polymerase chain reaction in genomic DNA. RESULTS: Sixty one participants were carriers of the haplotype HLA-A*02. The relative allele frequency HA-1H was 45%, HA-Ir 55%, HA-2V 80.6%, HA-2M 19.4%, HA-8R 49.8% and HA-8P was 50.2%. Based on mHAgs disparity between HA-1, HA-2 or HA-8, the probability to generate a GVT response in HLA-A*02 individuals was 40%. CONCLUSIONS: The mHAgs frequency in Chilean population is under Hardy-Weinberg equilibrium and they are similar to those of other ethnic populations in the world.


Subject(s)
Blood Donors , Gene Frequency/genetics , Graft vs Host Disease , HLA Antigens/genetics , Minor Histocompatibility Antigens/genetics , Chile , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Tumor Effect/genetics , Histocompatibility Testing , Humans , Minor Histocompatibility Antigens/analysis , Minor Histocompatibility Antigens/immunology , Polymerase Chain Reaction , Stem Cell Transplantation , Transplantation, Homologous
16.
Rev. méd. Chile ; 140(5): 555-560, mayo 2012. ilus, tab
Article in Spanish | LILACS | ID: lil-648580

ABSTRACT

Background: Minor histocompatibility antigens (mHAgs) play a critical role in the immune responses associated with allogeneic stem cell transplantation, such as graft versus host disease (GVHD) and graft-versus-tumor (GVT). Aim: To determine the gene frequencies of the mHAgs HA-1, HA-2 and HA-8 in Chilean Blood Bank donors. Material and Methods: Blood from 192 blood donors was analyzed. The presence of haplotype HLA-A*02 was determined by flow cytometry. The frequency of mHAgs was determined by allele specific polymerase chain reaction in genomic DNA. Results: Sixty one participants were carriers of the haplotype HLA-A*02. The relative allele frequency HA-1H was 45%, HA-Ir 55%, HA-2V 80.6%, HA-2M 19.4%, HA-8R 49.8% and HA-8P was 50.2%. Based on mHAgs disparity between HA-1, HA-2 or HA-8, the probability to generate a GVT response in HLA-A*02 individuals was 40%. Conclusions: The mHAgs frequency in Chilean population is under Hardy-Weinberg equilibrium and they are similar to those of other ethnic populations in the world.


Subject(s)
Humans , Blood Donors , Gene Frequency/genetics , Graft vs Host Disease , HLA Antigens/genetics , Minor Histocompatibility Antigens/genetics , Chile , Graft vs Host Disease/genetics , Graft vs Host Disease/immunology , Graft vs Tumor Effect/genetics , Histocompatibility Testing , Minor Histocompatibility Antigens/analysis , Minor Histocompatibility Antigens/immunology , Polymerase Chain Reaction , Stem Cell Transplantation , Transplantation, Homologous
17.
Clin Cancer Res ; 15(23): 7137-43, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19934307

ABSTRACT

PURPOSE: Identification of minor histocompatibility antigens (mHag) with classic methods often requires sophisticated technologies, determination, and patience. We here describe and validate a nonlaborious and convenient genetic approach, based on genome-wide correlations of mHag zygosities with HapMap single-nucleotide polymorphism genotypes, to identify clinical relevant mHags within a reasonable time frame. EXPERIMENTAL DESIGN: Using this approach, we sought for the mHag recognized by a HLA-DRB1*1501-restricted T-cell clone, isolated from a multiple myeloma patient during a strong graft-versus-tumor effect associated with acute graft-versus-host disease grade 3. RESULTS: In a period of 3 months, we determined the mHag phenotype of 54 HapMap individuals, deduced the zygosity of 20 individuals, defined the mHag locus by zygosity-genotype correlation analyses, tested the putative mHag peptides from this locus, and finally showed that the mHag is encoded by the arginine (R) allele of a nonsynonymous single-nucleotide polymorphism in the SLC19A1 gene. CONCLUSIONS: We conclude that this powerful and convenient strategy offers a broadly accessible platform toward rapid identification of mHags associated with graft-versus-tumor effect and graft-versus-host disease.


Subject(s)
Minor Histocompatibility Antigens/analysis , Multiple Myeloma/metabolism , Alleles , Arginine/chemistry , CD4-Positive T-Lymphocytes/metabolism , Genome , Genotype , Graft vs Host Disease , Graft vs Tumor Effect , Humans , Membrane Transport Proteins/genetics , Models, Genetic , Peptides/chemistry , Phenotype , Polymorphism, Single Nucleotide , Reduced Folate Carrier Protein
18.
Leukemia ; 22(9): 1721-7, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18548092

ABSTRACT

The cure of chronic myeloid leukemia (CML) patients following allogeneic stem cell transplantation (SCT) is attributed to graft-versus-leukemia (GVL) effects targeting alloantigens and/or leukemia-associated antigens (LAA) on leukemia cells. To assess the potential of LAA-peptide vaccines in eliminating leukemia in CML patients, we measured WT1, PR3, ELA2 and PRAME expression in CD34+ progenitor subpopulations in CML patients and compared them with minor histocompatibility antigens (mHAgs) HA1 and SMCY. All CD34+ subpopulations expressed similar levels of mHAgs irrespective of disease phase, suggesting that in the SCT setting, mHAgs are the best target for GVL. Furthermore, WT1 was consistently overexpressed in advanced phase (AdP) CML in all CD34+ subpopulations, and mature progenitors of chronic phase (CP) CML compared to healthy individuals. PRAME overexpression was limited to more mature AdP-CML progenitors only. Conversely, only CP-CML progenitors had PR3 overexpression, suggesting that PR1-peptide vaccines are only appropriate in CP-CML. Surface expression of WT1 protein in the most primitive hematopoietic stem cells in AdP-CML suggest that they could be targets for WT1 peptide-based vaccines, which in combination with PRAME, could additionally improve targeting differentiated progeny, and benefit patients responding suboptimally to tyrosine kinase inhibitors, or enhance GVL effects in SCT patients.


Subject(s)
Antigens, Neoplasm/analysis , Hematopoietic Stem Cells/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Neoplastic Stem Cells/immunology , Antigens, CD34 , Cancer Vaccines/chemistry , Case-Control Studies , Graft vs Leukemia Effect/immunology , Humans , Immunotherapy/methods , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Minor Histocompatibility Antigens/analysis , Neoplasm Proteins/analysis
19.
Blood ; 111(9): 4588-95, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18195091

ABSTRACT

Endothelial cells (ECs) presenting minor histocompatibility antigen (mhAg) are major target cells for alloreactive effector CD8(+) T cells during chronic transplant rejection and graft-versus-host disease (GVHD). The contribution of ECs to T-cell activation, however, is still a controversial issue. In this study, we have assessed the antigen-presenting capacity of ECs in vivo using a transgenic mouse model with beta-galactosidase (beta-gal) expression confined to the vascular endothelium (Tie2-LacZ mice). In a GVHD-like setting with adoptive transfer of beta-gal-specific T-cell receptor-transgenic T cells, beta-gal expression by ECs was not sufficient to either activate or tolerize CD8(+) T cells. Likewise, transplantation of fully vascularized heart or liver grafts from Tie2-LacZ mice into nontransgenic recipients did not suffice to activate beta-gal-specific CD8(+) T cells, indicating that CD8(+) T-cell responses against mhAg cannot be initiated by ECs. Moreover, we could show that spontaneous activation of beta-gal-specific CD8(+) T cells in Tie2-LacZ mice was exclusively dependent on CD11c(+) dendritic cells (DCs), demonstrating that mhAgs presented by ECs remain immunologically ignored unless presentation by DCs is granted.


Subject(s)
Endothelial Cells/immunology , Minor Histocompatibility Antigens/analysis , Transplantation Immunology , Animals , Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Endothelium, Vascular/cytology , Mice , Mice, Transgenic , Minor Histocompatibility Antigens/immunology , Organ Transplantation/adverse effects
20.
J Immunol Methods ; 329(1-2): 125-37, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17996247

ABSTRACT

Minor histocompatibility antigens (mHAgs) constitute the target antigens of the T cell-mediated graft-versus-leukemia response after HLA-identical allogeneic stem cell transplantation (SCT). Several human mHAgs have been identified, but only a few are selectively expressed by hematopoietic cells representing potential targets for specific immunotherapy. Molecular approaches including cDNA library screening and genetic linkage analysis have been successfully applied to identify T cell-defined mHAgs, but each approach has its drawbacks which may lead to mis-identification of the mHAg of interest. We improved both molecular strategies to facilitate more robust identification of hematopoietic-restricted mHAgs. First, we adapted cDNA library cloning by using 293T cells with stable expression of the relevant MHC class I allele, CD80 and CD54. We demonstrated that cDNA library screening using this 293T expression system results in strong activation of cytotoxic T lymphocytes, which significantly contributes to improvement of the assay sensitivity. Second, we refined genetic linkage analysis using single nucleotide polymorphism (SNP) genotyping to narrow down the defined genetic region that holds the mHAg-encoding gene. We showed that SNP marker analysis provides additional information about the genetic position of the antigen-encoding gene. Application of these optimized molecular approaches will lead to more rapid and reliable molecular identification of hematopoietic-restricted mHAgs.


Subject(s)
Genetic Linkage , Lymphocyte Activation/genetics , Minor Histocompatibility Antigens/genetics , Polymorphism, Single Nucleotide , T-Lymphocytes, Cytotoxic/immunology , Animals , B7-1 Antigen/genetics , COS Cells , Chlorocebus aethiops , Cloning, Molecular , Female , Gene Frequency , Gene Library , HLA-B Antigens/genetics , HLA-B44 Antigen , HLA-B7 Antigen , Humans , Intercellular Adhesion Molecule-1/genetics , Interferon-gamma/biosynthesis , K562 Cells , Male , Minor Histocompatibility Antigens/analysis , Pedigree , Reproducibility of Results , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...