Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Gynecol Endocrinol ; 38(1): 22-27, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34402730

ABSTRACT

OBJECTIVE: The present meta-analysis was conducted to investigate the association between circulating chemerin levels and polycystic ovary syndrome (PCOS) in women. METHODS: Relevant studies published up to May 2020 were searched from PubMed, Ovid, the Cochrane Library, and Clinical Trial Database. A random effects model was used to measure the strength of association between PCOS and chemerin by using the standardized mean difference (SMD) and 95% confidence interval (CI). All data were analyzed using Stata 12.0 (version 12; Stata-Corp, College Station, TX). RESULTS: The final meta-analysis included eight studies with 15 results including a total of 897 participants (524 patients with PCOS and 373 controls). The circulating chemerin levels were higher in patients with PCOS (random effects SMD = 1.07; 95% CI: 0.55-1.59; p < .001) than in controls. However, considerable heterogeneity across studies was not eliminated in subgroup analyses. The meta-regression analysis further suggested that region is the main source of heterogeneity (p = .001). CONCLUSIONS: Our meta-analysis indicated that women with PCOS have significantly higher circulating chemerin levels than in healthy women, indicating that chemerin may be involved in the pathogenesis of PCOS.


Subject(s)
Chimerin Proteins/blood , Polycystic Ovary Syndrome/blood , Female , Humans , Regression Analysis
2.
J Biol Chem ; 295(12): 3932-3944, 2020 03 20.
Article in English | MEDLINE | ID: mdl-32060095

ABSTRACT

Eph receptors are a family of receptor tyrosine kinases that control directional cell movement during various biological processes, including embryogenesis, neuronal pathfinding, and tumor formation. The biochemical pathways of Eph receptors are context-dependent in part because of the varied composition of a heterotypic, oligomeric, active Eph receptor complex. Downstream of the Eph receptors, little is known about the essential phosphorylation events that define the context and instruct cell movement. Here, we define a pathway that is required for Eph receptor B2 (EphB2)-mediated cell sorting and is conserved among multiple Eph receptors. Utilizing a HEK293 model of EphB2+/ephrinB1+ cell segregation, we found that the scaffold adaptor protein SH2 domain-containing adaptor protein B (Shb) is essential for EphB2 functionality. Further characterization revealed that Shb interacts with known modulators of cytoskeletal rearrangement and cell mobility, including Nck adaptor protein (Nck), p120-Ras GTPase-activating protein (RasGAP), and the α- and ß-Chimaerin Rac GAPs. We noted that phosphorylation of Tyr297, Tyr246, and Tyr336 of Shb is required for EphB2-ephrinB1 boundary formation, as well as binding of Nck, RasGAP, and the chimaerins, respectively. Similar complexes were formed in the context of EphA4, EphA8, EphB2, and EphB4 receptor activation. These results indicate that phosphotyrosine-mediated signaling through Shb is essential in EphB2-mediated heterotypic cell segregation and suggest a conserved function for Shb downstream of multiple Eph receptors.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Chimerin Proteins/metabolism , Oncogene Proteins/metabolism , Proto-Oncogene Proteins/metabolism , RNA-Binding Proteins/metabolism , Receptor, EphB2/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Cell Separation , Chimerin Proteins/chemistry , Ephrin-B1/genetics , Ephrin-B1/metabolism , HEK293 Cells , Humans , Mass Spectrometry , Oncogene Proteins/chemistry , Phosphorylation , Protein Binding , Protein Subunits/chemistry , Protein Subunits/metabolism , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , RNA-Binding Proteins/chemistry , Receptor, EphB2/chemistry , Receptor, EphB2/genetics , Signal Transduction , src Homology Domains
3.
J Biol Chem ; 295(5): 1300-1314, 2020 01 31.
Article in English | MEDLINE | ID: mdl-31871052

ABSTRACT

ß1-chimaerin belongs to the chimaerin family of GTPase-activating proteins (GAPs) and is encoded by the CHN2 gene, which also encodes the ß2- and ß3-chimaerin isoforms. All chimaerin isoforms have a C1 domain that binds diacylglycerol as well as tumor-promoting phorbol esters and a catalytic GAP domain that inactivates the small GTPase Rac. Nuclear Rac has emerged as a key regulator of various cell functions, including cell division, and has a pathological role by promoting tumorigenesis and metastasis. However, how nuclear Rac is regulated has not been fully addressed. Here, using several approaches, including siRNA-mediated gene silencing, confocal microscopy, and subcellular fractionation, we identified a nuclear variant of ß1-chimaerin, ß1-Δ7p-chimaerin, that participates in the regulation of nuclear Rac1. We show that ß1-Δ7p-chimaerin is a truncated variant generated by alternative splicing at a cryptic splice site in exon 7. We found that, unlike other chimaerin isoforms, ß1-Δ7p-chimaerin lacks a functional C1 domain and is not regulated by diacylglycerol. We found that ß1-Δ7p-chimaerin localizes to the nucleus via a nuclear localization signal in its N terminus. We also identified a key nuclear export signal in ß1-chimaerin that is absent in ß1-Δ7p-chimaerin, causing nuclear retention of this truncated variant. Functionally analyses revealed that ß1-Δ7p-chimaerin inactivates nuclear Rac and negatively regulates the cell cycle. Our results provide important insights into the diversity of chimaerin Rac-GAP regulation and function and highlight a potential mechanism of nuclear Rac inactivation that may play significant roles in pathologies such as cancer.


Subject(s)
Cell Nucleus/metabolism , Chimerin Proteins/genetics , Chimerin Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Alternative Splicing , Amino Acid Motifs/genetics , Animals , COS Cells , Cell Cycle/genetics , Cell Line, Tumor , Chlorocebus aethiops , Diglycerides/metabolism , Exons/genetics , Gene Silencing , Humans , Protein Domains/genetics , Protein Isoforms/metabolism , RNA, Small Interfering , Sequence Deletion , rac1 GTP-Binding Protein/genetics
4.
Transl Psychiatry ; 9(1): 254, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31594917

ABSTRACT

Major depressive disorder (MDD) is primarily treated with antidepressants, yet many patients fail to respond adequately, and identifying antidepressant response biomarkers is thus of clinical significance. Some hypothesis-driven investigations of epigenetic markers for treatment response have been previously made, but genome-wide approaches remain unexplored. Healthy participants (n = 112) and MDD patients (n = 211) between 18-60 years old were recruited for an 8-week trial of escitalopram treatment. Responders and non-responders were identified using differential Montgomery-Åsberg Depression Rating Scale scores before and after treatment. Genome-wide DNA methylation and gene expression analyses were assessed using the Infinium MethylationEPIC Beadchip and HumanHT-12 v4 Expression Beadchip, respectively, on pre-treatment peripheral blood DNA and RNA samples. Differentially methylated positions (DMPs) located in regions of differentially expressed genes between responders (n = 82) and non-responders (n = 95) were identified, and technically validated using a targeted sequencing approach. Three DMPs located in the genes CHN2 (cg23687322, p = 0.00043 and cg06926818, p = 0.0014) and JAK2 (cg08339825, p = 0.00021) were the most significantly associated with mRNA expression changes and subsequently validated. Replication was then conducted with non-responders (n = 76) and responders (n = 71) in an external cohort that underwent a similar antidepressant trial. One CHN2 site (cg06926818; p = 0.03) was successfully replicated. Our findings indicate that differential methylation at CpG sites upstream of the CHN2 and JAK2 TSS regions are possible peripheral predictors of antidepressant treatment response. Future studies can provide further insight on robustness of our candidate biomarkers, and greater characterization of functional components.


Subject(s)
Antidepressive Agents/therapeutic use , Citalopram/therapeutic use , DNA Methylation , Depressive Disorder, Major/drug therapy , Depressive Disorder, Major/genetics , Adolescent , Adult , Canada , Case-Control Studies , Chimerin Proteins/genetics , CpG Islands , Female , Genome-Wide Association Study , Humans , Janus Kinase 2/genetics , Linear Models , Male , Middle Aged , Polymorphism, Single Nucleotide , Psychiatric Status Rating Scales , ROC Curve , Young Adult
5.
Proc Natl Acad Sci U S A ; 116(27): 13651-13660, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31209021

ABSTRACT

Adult hippocampal neurogenesis involves the lifelong generation of neurons. The process depends on the homeostasis of the production of neurons and maintenance of the adult neural stem cell (NSC) pool. Here, we report that α2-chimaerin, a Rho GTPase-activating protein, is essential for NSC homeostasis in adult hippocampal neurogenesis. Conditional deletion of α2-chimaerin in adult NSCs resulted in the premature differentiation of NSCs into intermediate progenitor cells (IPCs), which ultimately depleted the NSC pool and impaired neuron generation. Single-cell RNA sequencing and pseudotime analyses revealed that α2-chimaerin-conditional knockout (α2-CKO) mice lacked a unique NSC subpopulation, termed Klotho-expressing NSCs, during the transition of NSCs to IPCs. Furthermore, α2-CKO led to defects in hippocampal synaptic plasticity and anxiety/depression-like behaviors in mice. Our findings collectively demonstrate that α2-chimaerin plays an essential role in adult hippocampal NSC homeostasis to maintain proper brain function.


Subject(s)
Chimerin Proteins/physiology , GTP Phosphohydrolase Activators/metabolism , Neural Stem Cells/metabolism , Neurogenesis/physiology , Animals , Cell Differentiation , Gene Knockdown Techniques , Hippocampus/physiology , Homeostasis , Mice , Mice, Knockout , Neural Stem Cells/physiology , Stem Cells/physiology
6.
Semin Nephrol ; 38(4): 317-324, 2018 07.
Article in English | MEDLINE | ID: mdl-30082052

ABSTRACT

Recent advances in genetics of renal disease have deepened our understanding of progressive kidney disease. Here, we review genetic variants that are of particular importance to progressive glomerular disease that result in end-stage kidney disease (ESKD). Some of the most striking findings relate to APOL1 genetic variants, seen exclusively in individuals of sub-Saharan African descent, that create a predisposition to particular renal disorders, including focal segmental glomerulosclerosis and arterionephrosclerosis. We also review the genetics of cardiovascular disease in ESKD and note that little work has been published on the genetics of other ESKD complications, including anemia, bone disease, and infections. Deeper understanding of the genetics of ESKD and its complications may lead to new therapies that are tailored to an individual patient's genetic profile or are discovered based on genetic approaches that identify novel pathways of renal cell injury and repair.


Subject(s)
Apolipoprotein L1/genetics , Genomics , Kidney Failure, Chronic/genetics , Precision Medicine , Anemia/complications , Anemia/genetics , Arteriosclerosis/genetics , Bone Diseases, Metabolic/complications , Bone Diseases, Metabolic/genetics , Cardiovascular Diseases/complications , Cardiovascular Diseases/genetics , Chimerin Proteins/genetics , Complement Factor H/genetics , Diabetic Nephropathies/genetics , Dipeptidases/genetics , Genetic Predisposition to Disease , Genetic Variation , Genotype , Glomerulosclerosis, Focal Segmental/genetics , Humans , Infections/complications , Infections/genetics , Kidney Failure, Chronic/complications , Lim Kinases/genetics , Molecular Motor Proteins/genetics , Myosin Heavy Chains/genetics , Nephrosclerosis/genetics , Nerve Tissue Proteins/genetics , Oxidative Stress/genetics , Polymorphism, Single Nucleotide , Receptor, Angiotensin, Type 1/genetics , Sickle Cell Trait/genetics
7.
Antiviral Res ; 158: 78-87, 2018 10.
Article in English | MEDLINE | ID: mdl-30071204

ABSTRACT

Human respiratory syncytial virus (RSV) and parainfluenza virus type 3 (PIV3) are major causes of serious lower respiratory tract disease in infants. Currently there is no licensed vaccine against RSV or PIV3. To make an effective bivalent subunit vaccine, a chimeric truncated FRHN protein containing the N-terminal ectodomain of the RSV fusion (F) protein linked to the C-terminal ectodomain of the PIV3 haemagglutinin-neuraminidase (HN) protein was produced in HEK293T cells. Mice, cotton rats and hamsters were immunized intramuscularly (IM) with both RSV F and PIV3 HN (FR+HN) or FRHN, formulated with TriAdj, which consists of poly(I:C), innate defense regulator peptide and poly[di(sodium carboxylatoethylphenoxy)]-phosphazene. Both formulations were immunogenic and elicited full protection from RSV; however, animals vaccinated with FRHN/TriAdj were significantly better protected from PIV3 than animals vaccinated with FR+HN/TriAdj. To develop a potentially more effective subunit vaccine, a chimeric glycoprotein (FRipScHN), encoding the RSV F ectodomain stabilized in the pre-fusion form linked to PIV3 HN was generated. Intramuscular vaccination with FRipScHN/TriAdj induced virus neutralizing antibodies followed by complete protection from RSV and PIV3 replication in the lungs of challenged cotton rats. Furthermore, intranasal vaccination with FRipScHN/TriAdj significantly reduced both RSV and PIV3 replication in cotton rats. Mucosal immunization with FRipScHN/TriAdj also elicited strong antigen-specific mucosal and systemic immune responses in a lamb model. In conclusion, the chimeric FRipScHN protein combined with TriAdj has potential for development of a safe, effective, bivalent vaccine against both RSV and PIV3.


Subject(s)
Adjuvants, Immunologic/pharmacology , Glycoproteins/pharmacology , Parainfluenza Virus 3, Human/immunology , Protective Agents/pharmacology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus, Human/immunology , Respirovirus Infections/prevention & control , Animals , Chimerin Proteins/immunology , Chlorocebus aethiops , Cricetinae , Disease Models, Animal , HEK293 Cells , Humans , Immunity, Innate , Immunity, Mucosal , Immunization , Mice , Mice, Inbred BALB C , Poly I-C , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Vaccines/immunology , Respirovirus Infections/immunology , Sheep , Sigmodontinae , Vaccination , Vaccines, Subunit , Viral Fusion Proteins/immunology , Virus Replication/drug effects
8.
Palliat Support Care ; 15(4): 499-503, 2017 08.
Article in English | MEDLINE | ID: mdl-28049548

ABSTRACT

Chimeric antigen receptor T cells are used in the treatment of B-cell leukemias. Common chimeric antigen receptor T-cell toxicities can range from mild flu-like symptoms, such as fever and myalgia, to a more striking neuropsychiatric toxicity that can present as discrete neurological symptoms and delirium. We report here two cases of chimeric antigen receptor T-cell neuropsychiatric toxicity, one who presented as a mild delirium and aphasia that resolved without intervention, and one who presented with delirium, seizures, and respiratory insufficiency requiring intensive treatment. The current literature on the treatment and proposed mechanisms of this clinically challenging chimeric antigen receptor T-cell complication is also presented.


Subject(s)
Chimerin Proteins/toxicity , Neurotoxicity Syndromes/etiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Receptors, Antigen, T-Cell/metabolism , Adult , Female , Humans , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Receptors, Antigen, T-Cell/therapeutic use
9.
Cytotherapy ; 19(2): 200-210, 2017 02.
Article in English | MEDLINE | ID: mdl-27939374

ABSTRACT

BACKGROUND: Mesenchymal stromal cells (MSCs) are multipotent cells characterized by broad immunomodulatory properties exploited for the treatment of inflammatory disorders. However, the efficacy of MSC-based therapy is highly variable and tightly linked to MSC culture conditions and treatment schedule. Thus, the identification of novel key molecules regulating MSC immunomodulatory activities in vivo might constitute a crucial step toward the optimization of currently available clinical protocols. In this regard, herein, we sought to determine whether the newly identified chemotactic protein, chemerin, plays a role in MSC-mediated regulation of inflammation. METHODS: Chemerin production by human MSCs was investigated under different culture conditions using enzyme-linked immunosorbent assay (ELISA). After purification, MSC-secreted chemerin was identified using mass spectrometry analysis and the biological activity of secreted isoforms was evaluated using migration assay. RESULTS: Bone marrow-derived MSCs secrete chemerin and express its receptors ChemR23 and CCRL2. Chemerin production is dependent on culture conditions and increases upon stimulation with inflammatory cytokines. In particular, platelet lysate (PL)-MSCs produce higher levels of chemerin compared with fetal bovine serum (FBS)-MSCs. Furthermore, chemerin is secreted by MSCs as an inactive precursor, which can be converted into its active form by exogenous chemerin-activating serine and cysteine proteases. DISCUSSION: Our data indicate that, in response to various inflammatory stimuli, MSCs secrete high amounts of inactive chemerin, which can then be activated by inflammation-induced tissue proteases. In light of these initial findings, we propose that further analysis of chemerin functions in vivo might constitute a crucial step toward optimizing MSC-based therapy for inflammatory diseases.


Subject(s)
Chemotaxis/drug effects , Chimerin Proteins/pharmacology , Immunomodulation/drug effects , Mesenchymal Stem Cells/metabolism , Receptors, Chemokine/metabolism , Blood Platelets/chemistry , Cell Culture Techniques , Cell Extracts/chemistry , Cell Extracts/pharmacology , Cells, Cultured , Chemotaxis/genetics , Chimerin Proteins/genetics , Chimerin Proteins/metabolism , Culture Media/metabolism , Culture Media/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Immunomodulation/genetics , Inflammation/metabolism , Inflammation/therapy , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/cytology , Receptors, Chemokine/genetics
10.
J Am Heart Assoc ; 5(10)2016 10 14.
Article in English | MEDLINE | ID: mdl-27742615

ABSTRACT

BACKGROUND: Circulating levels of chemerin are significantly higher in hypertensive patients and positively correlate with blood pressure. Chemerin activates chemokine-like receptor 1 (CMKLR1 or ChemR23) and is proposed to activate the "orphan" G-protein-coupled receptor 1 (GPR1), which has been linked with hypertension. Our aim was to localize chemerin, CMKLR1, and GPR1 in the human vasculature and determine whether 1 or both of these receptors mediate vasoconstriction. METHODS AND RESULTS: Using immunohistochemistry and molecular biology in conduit arteries and veins and resistance vessels, we localized chemerin to endothelium, smooth muscle, and adventitia and found that CMKLR1 and GPR1 were widely expressed in smooth muscle. C9 (chemerin149-157) contracted human saphenous vein (pD2=7.30±0.31) and resistance arteries (pD2=7.05±0.54) and increased blood pressure in rats by 9.1±1.0 mm Hg at 200 nmol. Crucially, these in vitro and in vivo vascular actions were blocked by CCX832, which we confirmed to be highly selective for CMKLR1 over GPR1. C9 inhibited cAMP accumulation in human aortic smooth muscle cells and preconstricted rat aorta, consistent with the observed vasoconstrictor action. Downstream signaling was explored further and, compared to chemerin, C9 showed a bias factor=≈5000 for the Gi protein pathway, suggesting that CMKLR1 exhibits biased agonism. CONCLUSIONS: Our data suggest that chemerin acts at CMKLR1, but not GPR1, to increase blood pressure. Chemerin has an established detrimental role in metabolic syndrome, and these direct vascular actions may contribute to hypertension, an additional risk factor for cardiovascular disease. This study provides proof of principle for the therapeutic potential of selective CMKLR1 antagonists.


Subject(s)
Adventitia/metabolism , Chemokines/metabolism , Endothelium, Vascular/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Receptors, Chemokine/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Arteries/metabolism , Blood Pressure/drug effects , Chimerin Proteins/pharmacology , Humans , Immunohistochemistry , Male , Peptide Fragments/pharmacology , Rats , Rats, Sprague-Dawley , Saphenous Vein/drug effects , Vasoconstriction/drug effects , Veins/metabolism
11.
Emerg Microbes Infect ; 5: e51, 2016 May 25.
Article in English | MEDLINE | ID: mdl-27222326

ABSTRACT

The ectodomain of the influenza A virus (IAV) hemagglutinin (HA) stem is highly conserved across strains and has shown promise as a universal influenza vaccine in a mouse model. In this study, potential B-cell epitopes were found through sequence alignment and epitope prediction in a stem fragment, HA2:90-105, which is highly conserved among virus subtypes H1, H3 and B. A norovirus (NoV) P particle platform was used to express the HA2:90-105 sequences from subtypes H1, H3 and B in loops 1, 2 and 3 of the protrusion (P) domain, respectively. Through mouse immunization and microneutralization assays, the immunogenicity and protective efficacy of the chimeric NoV P particle (trivalent HA2-PP) were tested against infection with three subtypes (H1N1, H3N2 and B) of IAV in Madin-Darby canine kidney cells. The protective efficacy of the trivalent HA2-PP was also evaluated preliminarily in vivo by virus challenge in the mouse model. The trivalent HA2-PP immunogen induced significant IgG antibody responses, which could be enhanced by a virus booster vaccination. Moreover, the trivalent HA2-PP immunogen also demonstrated in vitro neutralization of the H3 and B viruses, and in vivo protection against the H3 virus. Our results support the notion that a broadly protective vaccine approach using an HA2-based NoV P particle platform can provide cross-protection against challenge viruses of different IAV subtypes. The efficacy of the immunogen should be further enhanced for practicality, and a better understanding of the protective immune mechanism will be critical for the development of HA2-based multivalent vaccines.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunogenicity, Vaccine , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza B virus/genetics , Influenza Vaccines/immunology , Norovirus/genetics , A549 Cells , Animals , Antibodies, Viral/blood , Chimerin Proteins/administration & dosage , Chimerin Proteins/genetics , Chimerin Proteins/immunology , Cross Protection , Dogs , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/immunology , Female , Humans , Immunization , Immunoglobulin G/blood , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza B virus/immunology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control
12.
Support Care Cancer ; 24(7): 2927-35, 2016 07.
Article in English | MEDLINE | ID: mdl-26847448

ABSTRACT

PURPOSE: Since circulating level of insulin is associated with colorectal cancer prognosis, it is important to identify factors contributing to fasting insulin level in colorectal cancer patients. The purpose of the current study is to investigate the association of physical fitness, adiponectin, and chemerin levels with circulating level of insulin in colorectal cancer patients. METHODS: A total of 123 stage II-III colorectal cancer patients who completed standard cancer treatment were recruited. Anthropometric characteristics, fitness measurements, fasting insulin level, homeostasis model assessment of insulin resistance, lipid profiles, and adiponectin and chemerin levels were analyzed. RESULT: Cardiopulmonary fitness level inversely associated with fasting insulin levels (the least fit (1st tertile): 8.11 ± 0.64, moderately fit (2nd tertile): 6.02 ± 0.63, and highly fit (3rd tertile): 5.58 ± 0.66 µU/ml, unfit vs. moderately fit, p < 0.01; unfit vs. highly fit, p < 0.05) after adjustment for gender, age, stage, and BMI. In addition, fasting adiponectin and chemerin levels were associated with fasting insulin levels after adjustment for gender, age, stage, and BMI. In our combined analyses, participants with high adiponectin and low chemerin levels showed significantly lower fasting insulin levels (4.92 ± 0.75 vs. 8.07 ± 0.80 µU/ml, p < 0.01) compared with participants with low adiponectin and high chemerin levels. Multiple linear regression analysis confirmed that cardiopulmonary fitness and adiponectin levels (ß = -0.299, p = 0.002; ß = -0.201, p = 0.033) were independently associated with fasting insulin level. CONCLUSION: Our results suggest that physical fitness and adiponectin and chemerin levels may contribute to circulating levels of insulin. These results suggest that exercise may influence the prognosis of colorectal cancer patients by influencing physical fitness level, circulating levels of adiponectin and chemerin.


Subject(s)
Adiponectin/metabolism , Chimerin Proteins/metabolism , Exercise/physiology , Insulin/metabolism , Colorectal Neoplasms , Female , Humans , Male , Middle Aged
13.
Oncotarget ; 6(25): 20863-74, 2015 Aug 28.
Article in English | MEDLINE | ID: mdl-26315110

ABSTRACT

Small bowel accounts for only 0.5% of cancer cases in the US but incidence rates have been rising at 2.4% per year over the past decade. One-third of these are adenocarcinomas but little is known about their molecular pathology and no molecular markers are available for clinical use. Using a retrospective 28 patient matched normal-tumor cohort, next-generation sequencing, gene expression arrays and CpG methylation arrays were used for molecular profiling. Next-generation sequencing identified novel mutations in IDH1, CDH1, KIT, FGFR2, FLT3, NPM1, PTEN, MET, AKT1, RET, NOTCH1 and ERBB4. Array data revealed 17% of CpGs and 5% of RNA transcripts assayed to be differentially methylated and expressed respectively (p < 0.01). Merging gene expression and DNA methylation data revealed CHN2 as consistently hypermethylated and downregulated in this disease (Spearman -0.71, p < 0.001). Mutations in TP53 which were found in more than half of the cohort (15/28) and Kazald1 hypomethylation were both were indicative of poor survival (p = 0.03, HR = 3.2 and p = 0.01, HR = 4.9 respectively). By integrating high-throughput mutational, gene expression and DNA methylation data, this study reveals for the first time the distinct molecular profile of small bowel adenocarcinoma and highlights potential clinically exploitable markers.


Subject(s)
Adenocarcinoma/pathology , Adenocarcinoma/therapy , Intestinal Neoplasms/pathology , Intestinal Neoplasms/therapy , Intestine, Small/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor , Chimerin Proteins/genetics , CpG Islands , DNA Methylation , DNA Mutational Analysis , Epigenesis, Genetic , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, p53 , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Mutation , Nucleophosmin , Oligonucleotide Array Sequence Analysis , Pathology, Molecular , Retrospective Studies , Treatment Outcome
14.
PLoS One ; 9(7): e102977, 2014.
Article in English | MEDLINE | ID: mdl-25057852

ABSTRACT

Hepatosplenic T-cell lymphoma (HSTL) is an aggressive lymphoma cytogenetically characterized by isochromosome 7q [i(7)(q10)], of which the molecular consequences remain unknown. We report here results of an integrative genomic and transcriptomic (expression microarray and RNA-sequencing) study of six i(7)(q10)-positive HSTL cases, including HSTL-derived cell line (DERL-2), and three cases with ring 7 [r(7)], the recently identified rare variant aberration. Using high resolution array CGH, we profiled all cases and mapped the common deleted region (CDR) at 7p22.1p14.1 (34.88 Mb; 3506316-38406226 bp) and the common gained region (CGR) at 7q22.11q31.1 (38.77 Mb; 86259620-124892276 bp). Interestingly, CDR spans a smaller region of 13 Mb (86259620-99271246 bp) constantly amplified in cases with r(7). In addition, we found that TCRG (7p14.1) and TCRB (7q32) are involved in formation of r(7), which seems to be a byproduct of illegitimate somatic rearrangement of both loci. Further transcriptomic analysis has not identified any CDR-related candidate tumor suppressor gene. Instead, loss of 7p22.1p14.1 correlated with an enhanced expression of CHN2 (7p14.1) and the encoded ß2-chimerin. Gain and amplification of 7q22.11q31.1 are associated with an increased expression of several genes postulated to be implicated in cancer, including RUNDC3B, PPP1R9A and ABCB1, a known multidrug resistance gene. RNA-sequencing did not identify any disease-defining mutation or gene fusion. Thus, chromosome 7 imbalances remain the only driver events detected in this tumor. We hypothesize that the Δ7p22.1p14.1-associated enhanced expression of CHN2/ß2-chimerin leads to downmodulation of the NFAT pathway and a proliferative response, while upregulation of the CGR-related genes provides growth advantage for neoplastic δγT-cells and underlies their intrinsic chemoresistance. Finally, our study confirms the previously described gene expression profile of HSTL and identifies a set of 24 genes, including three located on chromosome 7 (CHN2, ABCB1 and PPP1R9A), distinguishing HSTL from other malignancies.


Subject(s)
Chimerin Proteins/genetics , Chromosome Aberrations , Chromosomes, Human, Pair 7 , Liver Neoplasms/genetics , Lymphoma, T-Cell/genetics , Microfilament Proteins/genetics , Neoplasm Proteins/genetics , Nerve Tissue Proteins/genetics , Splenic Neoplasms/genetics , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Adolescent , Adult , Child , Chimerin Proteins/metabolism , Chromosome Mapping , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genetic Loci , Genome, Human , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Lymphoma, T-Cell/metabolism , Lymphoma, T-Cell/pathology , Male , Microfilament Proteins/metabolism , Middle Aged , Neoplasm Proteins/metabolism , Nerve Tissue Proteins/metabolism , Splenic Neoplasms/metabolism , Splenic Neoplasms/pathology , Transcriptome
15.
Braz. j. infect. dis ; 18(3): 308-314, May-June/2014. tab, graf
Article in English | LILACS | ID: lil-712955

ABSTRACT

OBJECTIVES: Enterotoxigenic Escherichia coli (ETEC), a major cause of diarrhea in children under 5, is an important agent for traveler's diarrhea. Heat-labile enterotoxin (LT) and colonization factors (CFs) are two main virulence mechanisms in ETEC. CS6 is one of the most prevalent CFs consisting of two structural subunits viz., CssA, CssB, necessary for attachment to the intestinal cells. METHODS: In the present research, a chimeric trivalent protein composed of CssB, CssA and LTB was constructed. The chimeric gene was synthesized with codon bias of E. coli for enhanced expression of the protein. Recombinant proteins were expressed and purified. Mice were immunized with the recombinant protein. The antibody titer and specificity of the immune sera were analyzed by ELISA and Western blotting. Efficiency of the immune sera against ETEC was evaluated. RESULTS: Antibody induction was followed by immunization of mice with the chimeric protein. Pretreatment of the ETEC cells with immunized animal antisera remarkably decreased their adhesion to Caco-2 cells. DISCUSSION: The results indicate efficacy of the recombinant chimeric protein as an effective immunogen, which induces strong humoral response as well as protection against ETEC adherence and toxicity. .


Subject(s)
Animals , Female , Mice , Antigens, Bacterial/genetics , Chimerin Proteins/immunology , Enterotoxigenic Escherichia coli/genetics , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Blotting, Western , Chimerin Proteins/chemistry , Enterotoxigenic Escherichia coli/immunology , Escherichia coli Infections/immunology , Mice, Inbred BALB C
16.
J Diabetes Complications ; 28(4): 460-3, 2014.
Article in English | MEDLINE | ID: mdl-24854763

ABSTRACT

AIM: To investigate whether chimerin 2 (CHN2) genetic polymorphisms were associated with the susceptibility to diabetic retinopathy (DR) in Taiwanese individuals with type 2 diabetes. METHODS: This case-control study comprised of 171 individuals with DR and 548 without DR. Four rs39059, rs2023908, rs1002630 and rs1362363 polymorphism of CHN2 were genotyped for each subjects. All subjects underwent a complete ophthalmologic examination, and basic information (age, gender, age at diagnosis of diabetes, and ocular history of the patient) was record. Several clinical parameters (systolic and diastolic blood pressure, waist and hip circumferences, body mass index levels, fasting glucose and HbA1c) were measured. RESULTS: Logistic regressions were used to analyze odds ratios between SNPs and DR after controlling for gender, systolic blood pressure, waist and hip ratio, duration of diabetes, serum HbA1c levels and nephropathy classification. A protective effect of rs1002630 (GA+AA) and rs1362363 (AG+GG) [odds ratio (OR) (95% confidence interval)=0.45 (0.22-0.88), 0.66 (0.44-0.99), respectively) was observed. Furthermore, the protective effect of rs1002630 was observed when compared subjects with non-proliferative DR with subjects without DR [OR=0.25 (95%C.I. = 0.09-0.73)]. CONCLUSIONS: This study showed that the rs1002630 of CHN2 were associated with DR risk and non-proliferative DR risk in Taiwanese individuals with type 2 diabetes. Variations at this locus may contribute to the pathogenesis of DR.


Subject(s)
Chimerin Proteins/genetics , Diabetes Mellitus, Type 2/complications , Diabetic Retinopathy/genetics , Polymorphism, Single Nucleotide , Aged , Alleles , Asian People , Case-Control Studies , Chimerin Proteins/metabolism , China/ethnology , Cross-Sectional Studies , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Diabetic Retinopathy/physiopathology , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Logistic Models , Male , Middle Aged , Retina/pathology , Severity of Illness Index , Taiwan
17.
J Cell Mol Med ; 18(7): 1313-20, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24702860

ABSTRACT

The prevalence of obesity has been increasing worldwide. Chemerin is a recently discovered adipokine secreted by the enlarged adipose tissue with diverse biological effects that are not well detailed yet. This study aimed to elucidate the potential role of chemerin in oxidative stress and inflammation that are characteristics for excess weight and may eventually lead to insulin resistance and atherosclerotic complications. We also analysed the associations between chemerin and classical adipokines, namely leptin and adiponectin. Therefore, we investigated non-diabetic obese patients without manifest cardiovascular disease and compared their data to healthy lean individuals. Chemerin correlated positively with markers of oxidative stress and inflammation, while it showed a negative correlation with the measure of antioxidant status, characterized by the HDL-linked paraoxonase-1 enzyme. Chemerin also correlated positively with leptin and negatively with adiponectin respectively. In our study population, oxidized low-density lipoprotein and high-sensitivity C-reactive protein were found to be the strongest predictors of chemerin level. We conclude that chemerin may contribute to chronic inflammation and increased oxidative stress in obese individuals, even in the absence of manifest insulin resistance.


Subject(s)
Adipokines/metabolism , Biomarkers/metabolism , Chimerin Proteins/metabolism , Inflammation/pathology , Obesity/pathology , Oxidative Stress , Adiponectin/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Aryldialkylphosphatase/metabolism , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Humans , Inflammation/complications , Insulin Resistance , Leptin/metabolism , Lipoproteins, LDL/metabolism , Obesity/etiology
18.
Am J Reprod Immunol ; 71(3): 265-77, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24506805

ABSTRACT

PROBLEM: Chemerin is a novel chemo-attractant and adipokine involved in leukocyte recruitment, inflammation, adipogenesis, lipid/carbohydrate metabolism, and reproduction. Based on the bioinformatic search for putative small peptides in the conserved region of pre-pro-chemerin, an evolutionary conserved region flanked by potential convertase cleavage sites was identified and we named it as C-20. The binding capacity of C-20 to chemerin receptors and its potential bioactivities were investigated in this study. METHOD OF STUDY: Radioligand binding assay, receptor internalization assay, and early response gene C-FOS simulation, cAMP assay were carried out in chemokine-like receptor 1 (CMKLR1)/HEK293 transfectants and G protein-coupled receptor 1 (GPR1)/HEK293 transfectants. In vitro transwell chemotaxis assay in CMKLR1/L1.2 transfectants, primary Leydig cell culture, and antral follicle culture was explored to investigate the bioactivity of C-20. RESULTS: C-20 bound to chemerin receptors CMKLR1 and GPR1 with high affinity triggered CMKLR1 internalization and stimulated subsequent signal C-FOS expression and cAMP production. C-20, such as chemerin, showed CMKLR1-dependent chemotactic property. Furthermore, in primary Leydig cells and antral follicles, C-20 showed similar but less potent suppressive effect on human chorionic gonadotropin-stimulated testosterone production and progesterone production, compared with chemerin. CONCLUSION: The novel chemerin-derived C-20 peptide binds to chemerin receptors CMKLR1 and GPR1 and showed similar but less potent bioactivity in chemotaxis and the suppression of gonadal steroidogenesis, suggesting that after optimization, C-20 is possible to be a useful experimental tool for the understanding of the biological functions of chemerin/CMKLR1 and chemerin/GPR1 signaling.


Subject(s)
Chimerin Proteins/metabolism , Peptide Fragments/metabolism , Progesterone/biosynthesis , Testis/physiology , Testosterone/biosynthesis , Chemotaxis , Chimerin Proteins/genetics , Computational Biology , Cyclic AMP/metabolism , HEK293 Cells , Humans , Leydig Cells , Male , Peptide Fragments/genetics , Protein Binding , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
19.
Mol Biol Rep ; 41(4): 2067-76, 2014.
Article in English | MEDLINE | ID: mdl-24430297

ABSTRACT

Chimaerins are a family of diacylglycerol- and phorbol ester-regulated GTPase activating proteins (GAPs) for the small G-protein Rac. Extensive evidence indicates that these proteins play important roles in development, axon guidance, metabolism, cell motility, and T cell activation. Four isoforms have been reported to-date, which are products of CHN1 (α1- and α2-chimaerins) and CHN2 (ß1- and ß2-chimaerins) genes. Although these gene products are assumed to be generated by alternative splicing, bioinformatics analysis of the CHN2 gene revealed that ß1- and ß2-chimaerins are the products of alternative transcription start sites (TSSs) in different promoter regions. Furthermore, we found an additional TSS in CHN2 gene that leads to a novel product, which we named ß3-chimaerin. Expression profile analysis revealed predominantly low levels for the ß3-chimaerin transcript, with higher expression levels in epididymis, plasma blood leucocytes, spleen, thymus, as well as various areas of the brain. In addition to the prototypical SH2, C1, and Rac-GAP domains, ß3-chimaerin has a unique N-terminal domain. Studies in cells established that ß3-chimaerin has Rac-GAP activity and is responsive to phorbol esters. The enhanced responsiveness of ß3-chimaerin for phorbol ester-induced translocation relative to ß2-chimaerin suggests differential ligand accessibility to the C1 domain.


Subject(s)
Chimerin Proteins/genetics , Chimerin Proteins/metabolism , Protein Isoforms , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Cell Line , Chimerin Proteins/chemistry , Chlorocebus aethiops , Enzyme Activation , Gene Expression , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Order , Humans , Molecular Sequence Data , Organ Specificity/genetics , Promoter Regions, Genetic , Protein Binding , Protein Interaction Domains and Motifs , Tetradecanoylphorbol Acetate/pharmacology , rac GTP-Binding Proteins/metabolism
20.
Braz J Infect Dis ; 18(3): 308-14, 2014.
Article in English | MEDLINE | ID: mdl-24389278

ABSTRACT

OBJECTIVES: Enterotoxigenic Escherichia coli (ETEC), a major cause of diarrhea in children under 5, is an important agent for traveler's diarrhea. Heat-labile enterotoxin (LT) and colonization factors (CFs) are two main virulence mechanisms in ETEC. CS6 is one of the most prevalent CFs consisting of two structural subunits viz., CssA, CssB, necessary for attachment to the intestinal cells. METHODS: In the present research, a chimeric trivalent protein composed of CssB, CssA and LTB was constructed. The chimeric gene was synthesized with codon bias of E. coli for enhanced expression of the protein. Recombinant proteins were expressed and purified. Mice were immunized with the recombinant protein. The antibody titer and specificity of the immune sera were analyzed by ELISA and Western blotting. Efficiency of the immune sera against ETEC was evaluated. RESULTS: Antibody induction was followed by immunization of mice with the chimeric protein. Pretreatment of the ETEC cells with immunized animal antisera remarkably decreased their adhesion to Caco-2 cells. DISCUSSION: The results indicate efficacy of the recombinant chimeric protein as an effective immunogen, which induces strong humoral response as well as protection against ETEC adherence and toxicity.


Subject(s)
Antigens, Bacterial/genetics , Chimerin Proteins/immunology , Enterotoxigenic Escherichia coli/genetics , Escherichia coli Infections/microbiology , Escherichia coli Proteins/genetics , Animals , Blotting, Western , Chimerin Proteins/chemistry , Enterotoxigenic Escherichia coli/immunology , Escherichia coli Infections/immunology , Female , Mice , Mice, Inbred BALB C
SELECTION OF CITATIONS
SEARCH DETAIL
...