Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Am J Physiol Lung Cell Mol Physiol ; 305(2): L141-53, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23686855

ABSTRACT

Alveolar epithelial cells (AECs) maintain the pulmonary blood-gas barrier integrity with gasketlike intercellular tight junctions (TJ) that are anchored internally to the actin cytoskeleton. We have previously shown that AEC monolayers stretched cyclically and equibiaxially undergo rapid magnitude- and frequency-dependent actin cytoskeletal remodeling to form perijunctional actin rings (PJARs). In this work, we show that even 10 min of stretch induced increases in the phosphorylation of Akt and LIM kinase (LIMK) and decreases in cofilin phosphorylation, suggesting that the Rac1/Akt pathway is involved in these stretch-mediated changes. We confirmed that Rac1 inhibitors wortmannin or EHT-1864 decrease stretch-stimulated Akt and LIMK phosphorylation and that Rac1 agonists PIP3 or PDGF increase phosphorylation of these proteins in unstretched cells. We also confirmed that Rac1 pathway inhibition during stretch modulated stretch-induced changes in occludin content and monolayer permeability, actin remodeling and PJAR formation, and cell death. As further validation, overexpression of Rac GTPase-activating protein ß2-chimerin also preserved monolayer barrier properties in stretched monolayers. In summary, our data suggest that constitutive activity of Rac1, which is necessary for stretch-induced activation of the Rac1 downstream proteins, mediates stretch-induced increases in permeability and PJAR formation.


Subject(s)
Epithelial Cells/enzymology , Pulmonary Alveoli/enzymology , Respiratory Mucosa/enzymology , Signal Transduction/physiology , rac1 GTP-Binding Protein/metabolism , Androstadienes/pharmacology , Animals , Chimerin Proteins/metabolism , Cytoskeleton/metabolism , Epithelial Cells/cytology , Lim Kinases/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Platelet-Derived Growth Factor/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Alveoli/cytology , Pyrones/pharmacology , Quinolines/pharmacology , Rats , Rats, Sprague-Dawley , Respiratory Mucosa/cytology , Signal Transduction/drug effects , Wortmannin
11.
PLoS One ; 7(12): e52258, 2012.
Article in English | MEDLINE | ID: mdl-23284959

ABSTRACT

Epithelial organs are made of a well-polarized monolayer of epithelial cells, and their morphology is maintained strictly for their proper functions. Previously, we showed that Rac1 activation is suppressed at the apical membrane in the mature organoid, and that such spatially biased Rac1 activity is required for the polarity maintenance. Here we identify Chimaerin, a GTPase activating protein for Rac1, as a suppressor of Rac1 activity at the apical membrane. Depletion of Chimaerin causes over-activation of Rac1 at the apical membrane in the presence of hepatocyte growth factor (HGF), followed by luminal cell accumulation. Importantly, Chimaerin depletion did not inhibit extension formation at the basal membrane. These observations suggest that Chimaerin functions as the apical-specific Rac1 GAP to maintain epithelial morphology.


Subject(s)
Chimerin Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Cell Line , Chimerin 1/metabolism , Dogs , Hepatocyte Growth Factor/pharmacology , Microscopy, Confocal , Polymerase Chain Reaction
12.
Immunobiology ; 214(9-10): 877-86, 2009.
Article in English | MEDLINE | ID: mdl-19625100

ABSTRACT

Recent evidences suggest a significant role of Plasmacytoid dendritic cells (PDC) role in the pathogenesis of lupus erythematosus (LE) via production of type I IFN. Taking advantage on the availability of multiple reagents (CD123, BDCA2, and CD2ap) specifically recognizing PDC on fixed tissues, we investigated the occurrence of PDC in a cohort of 74 LE patients. The large majority of LE biopsies (67/74; 90.5%) showed cutaneous infiltration of PDC. PDC were more frequently observed (96.4 vs 72.2) and numerous in cutaneous LE compared to systemic LE (SLE) and correlated with the density of the inflammatory infiltrate (r=0.40; p<0.001). PDC reduction in SLE might be related to a broader tissue distribution of this cellular population, as indicated by their occurrence in kidneys in 11 out of 24 (45.8%) cases studied. The distribution of cutaneous PDC showed two distinct patterns. More commonly, PDC were observed within perivascular inflammatory nodules in the dermis, associated with CD208+ mature DC and T-bet+ cells [D-PDC]. A second component was observed along the dermal-epithelial junction [J-PDC], in association with cytotoxic T-cells in areas of severe epithelial damage. Notably, chemerin reactivity was observed in 64% of LE biopsies on endothelial cells and in the granular layer keratinocytes. Cutaneous PDC in LE strongly produced type I IFN, as indicated by the diffuse MxA expression, and the cytotoxic molecule granzyme B. This study confirms cutaneous PDC infiltration as hallmark of LE. The topographical segregation in D-PDC and J-PDC suggests a novel view of the role of these cells in skin autoimmunity.


Subject(s)
Apoptosis , Dendritic Cells/immunology , Lupus Erythematosus, Cutaneous/immunology , Skin/immunology , Cell Movement , Chimerin Proteins/metabolism , Dermis/immunology , Dermis/pathology , Humans , Immunohistochemistry , Interferon Type I/biosynthesis , Kidney/immunology , Kidney/pathology , Lupus Erythematosus, Cutaneous/pathology , Lysosomal Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Skin/pathology , T-Box Domain Proteins/metabolism
13.
Cell Signal ; 20(4): 758-70, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18249095

ABSTRACT

Chimaerins are GTPase-activating proteins that inactivate the GTP-hydrolase Rac1 in a diacylglycerol-dependent manner. To date, the study of chimaerins has been done mostly in neuronal cells. Here, we show that alpha2- and beta2-chimaerin are expressed at different levels in T-cells and that they participate in T-cell receptor signaling. In agreement with this, we have observed that alpha2- and beta2-chimaerins translocate to the T-cell/B-cell immune synapse and, using both gain- and loss-of-function approaches, demonstrated that their catalytic activity is important for the inhibition of the T-cell receptor- and Vav1-dependent stimulation of the transcriptional factor NF-AT. Mutagenesis-based approaches have revealed the molecular determinants that contribute to the biological program of chimaerins during T-cell responses. Unexpectedly, we have found that the translocation of chimaerins to the T-cell/B-cell immune synapse does not rely on the canonical binding of diacylglycerol to the C1 region of these GTPase-activating proteins. Taken together, these results identify chimaerins as candidates for the downmodulation of Rac1 in T-lymphocytes and, in addition, uncover a novel regulatory mechanism that mediates their activation in T-cells.


Subject(s)
Chimerin Proteins/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , T-Lymphocytes/metabolism , rac1 GTP-Binding Protein/metabolism , B-Lymphocytes/enzymology , B-Lymphocytes/metabolism , CD3 Complex/metabolism , Cell Membrane/enzymology , Cell Membrane/metabolism , Chimerin 1/metabolism , Chimerin Proteins/genetics , Diglycerides/metabolism , Down-Regulation , Enzyme Activation , Humans , Jurkat Cells , Mutation , NFATC Transcription Factors/metabolism , Neoplasm Proteins/metabolism , Protein Structure, Tertiary , Protein Transport , Proto-Oncogene Proteins c-vav/metabolism , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/metabolism , Signal Transduction/immunology , T-Lymphocytes/enzymology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Transfection
14.
Microb Pathog ; 44(6): 484-93, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18242046

ABSTRACT

Botulinum neurotoxin (BoNT) binds to presynaptic neuronal cells and blocks neurotransmitter release. The carboxyl-terminal half of the heavy chain (H(C)) of the neurotoxin recognizes its specific receptor on the plasma membrane. We have previously demonstrated that BoNT/C binds to gangliosides GD1b and GT1b under physiological conditions, while BoNT/D interacts with phosphatidylethanolamine (PE). Here we report that the recognition sites for gangliosides and PE are present in the carboxyl-terminal domain of H(C). Chimeric mutants and site-directed mutants of BoNT/C-H(C) and BoNT/D-H(C) were generated and their binding activities evaluated. The chimeric H(C) that consisted of the amino-terminal half of BoNT/D-H(C) and the carboxyl-terminal half of BoNT/C-H(C) possessed activity similar to the authentic BoNT/C-H(C), suggesting that the carboxyl-terminal region of H(C) is involved in the receptor recognition of BoNT/C. Moreover, analysis using site-directed mutants indicated that the peptide motif W(1257)Ycdots, three dots, centeredG(1270)cdots, three dots, centeredH(1282) plays an important role in the interaction between BoNT/C and gangliosides. In contrast, we revealed that two lysine residues of BoNT/D-H(C) are involved in the formation of the critical binding site for receptor binding.


Subject(s)
Botulinum Toxins/chemistry , Botulinum Toxins/metabolism , Clostridium botulinum/metabolism , SNARE Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Botulinum Toxins/genetics , Chimerin Proteins/chemistry , Chimerin Proteins/genetics , Chimerin Proteins/metabolism , Clostridium botulinum/chemistry , Clostridium botulinum/genetics , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Rats , Sequence Alignment , Synaptosomes/metabolism
15.
Immunity ; 26(5): 593-604, 2007 May.
Article in English | MEDLINE | ID: mdl-17509909

ABSTRACT

The ability of natural-killer cells to regulate adaptive immunity is not well understood. Here we define an interaction between the class Ib major histocompatibility complex (MHC) molecule Qa-1-Qdm on activated T cells responsible for adaptive immunity and CD94-NKG2A inhibitory receptors expressed by natural-killer cells by using Qa-1-deficient and Qa-1 knockin mice containing a point mutation that selectively abolishes Qa-1-Qdm binding to CD94-NKG2A receptors. The Qa-1-NKG2A interaction protected activated CD4+ T cells from lysis by a subset of NKG2A+ NK cells and was essential for T cell expansion and development of immunologic memory. Antibody-dependent blockade of this Qa-1-NKG2A interaction resulted in potent NK-dependent elimination of activated autoreactive T cells and amelioration of experimental autoimmune encephalomyelitis. These findings extend the functional reach of the NK system to include regulation of adaptive T cell responses and suggest a new clinical strategy for elimination of antigen-activated T cells in the context of autoimmune disease and transplantation.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Gene Expression Regulation/immunology , Histocompatibility Antigens Class I/metabolism , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Receptors, Immunologic/metabolism , Signal Transduction/immunology , Animals , Bone Marrow/immunology , Bone Marrow/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cells, Cultured , Chimerin Proteins/genetics , Chimerin Proteins/immunology , Chimerin Proteins/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Genetic Predisposition to Disease , Histocompatibility Antigens Class I/genetics , Immunologic Memory/immunology , Interferon-gamma/biosynthesis , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Lentivirus/genetics , Mice , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily C , NK Cell Lectin-Like Receptor Subfamily D/genetics , NK Cell Lectin-Like Receptor Subfamily D/metabolism , Phenotype , Receptors, Immunologic/genetics , Receptors, Natural Killer Cell
16.
Proc Natl Acad Sci U S A ; 104(17): 7098-103, 2007 Apr 24.
Article in English | MEDLINE | ID: mdl-17438281

ABSTRACT

The chimaerin family of Rac GTPase-activating proteins (GAPs) has been implicated in neural development and tumor progression, although the cellular mechanisms of their effects are poorly understood. To study their physiologic function, we used the Drosophila retina as a model system. Reduced expression of the fly chimaerin ortholog RhoGAP5a in the pupal eye led to an excess of interommatidial pigment cells, aberrant cell contacts, and an increase in activated ERK that localized specifically to the plasma membrane. Reducing RhoGAP5A levels suppressed the effects of disrupted EGF receptor signaling. Perturbation of Rac activity led to similar phenotypes, whereas coexpression of Rac and RhoGAP5A-dsRNAi resulted in the elimination of adherens junctions between interommatidial cells. Our results reveal a role for chimaerin in the regulation of ERK signaling and cell-cell adhesion and have implications for its participation in epithelial development and tumor progression.


Subject(s)
Adherens Junctions/metabolism , Chimerin Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Extracellular Signal-Regulated MAP Kinases/metabolism , Eye/enzymology , MAP Kinase Signaling System , rac GTP-Binding Proteins/metabolism , Animals , Cell Count , Drosophila melanogaster/enzymology , Drosophila melanogaster/growth & development , Enzyme Activation , ErbB Receptors/metabolism , Eye/growth & development , Eye/pathology , Eye Proteins/metabolism , Models, Biological , Nerve Tissue Proteins/metabolism
17.
Laryngoscope ; 117(6): 1013-8, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17440426

ABSTRACT

OBJECTIVES: In advanced head and neck squamous cell carcinoma (HNSCC), there is a need for an adjuvant treatment. We aim to evaluate the biodistribution and therapeutic effect of radioimmunotherapy using the alpha emitting, astatine-211-labeled, chimeric monoclonal antibody U36 (U36) on the HNSCC cell line UT-SCC7 in vivo. STUDY DESIGN: Xenograft tumors were inoculated subcutaneously in nude mice. Astatine-211-labeled U36 was injected intravenously with or without blocking of target with nonlabeled U36. METHODS: In the biodistribution experiments, radioactivity was measured in tumors and various organs at set time points. In the therapeutic experiments, two groups (with or without blocking) received therapy, and the tumor growth was compared with that of controls. In addition, one group received nonlabeled U36 only. RESULTS: The biodistribution experiments demonstrated that astatine-211-labeled U36 could target UT-SCC7 xenografts in nude mice. With time, uptake increased in tumors and decreased in normal organs. Nonlabeled U36 did not influence tumor growth. In the two therapy groups, 18 of 20 tumors responded to therapy by decreasing or stabilizing their volumes. Significant difference was seen between the treated groups and the controls (P < .05). CONCLUSION: The study illustrates the specific binding of astatine-211-labeled U36 to HNSCC and suggests radioimmunotherapy with the alpha emitting radionuclide to be a useful treatment modality.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Astatine/therapeutic use , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/therapy , Chimerin Proteins/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/therapy , Radioimmunotherapy/methods , Animals , Carcinoma, Squamous Cell/pathology , Cell Growth Processes/immunology , Cell Growth Processes/radiation effects , Cell Line, Tumor/metabolism , Cell Line, Tumor/radiation effects , Cell Line, Tumor/transplantation , Cell Movement , Disease Models, Animal , Head and Neck Neoplasms/pathology , Mice , Mice, Nude , Random Allocation , Staining and Labeling
18.
J Immunol ; 177(5): 3283-93, 2006 Sep 01.
Article in English | MEDLINE | ID: mdl-16920969

ABSTRACT

The relationship between EBV infection and sensitivity to death receptor (DR)-induced apoptosis is poorly understood. Using EBV- and EBV+ BJAB cells, we provide the first evidence that EBV can protect latently infected B cell lymphomas from apoptosis triggered through Fas or TRAIL receptors. Caspase 8 activation was impaired and cellular FLIP recruitment was enriched in death-inducing signaling complexes formed in EBV-infected BJAB cells relative to parent BJAB cells. Furthermore, latent membrane protein 1 expression alone could reduce caspase activation and confer partial resistance to DR apoptosis in BJAB cells. This protective effect was dependent on C-terminal activating region 2-driven NF-kappaB activation, which in turn up-regulated cellular FLIP expression in latent membrane protein 1+ BJAB cells. Thus, the ability of latent EBV to block DR apoptosis may help to ensure the survival of host cells during B cell differentiation, and contribute to the development of B cell lymphomas, especially in immunocompromised individuals.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Herpesvirus 4, Human/physiology , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , Membrane Glycoproteins/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factors/metabolism , Caspases/metabolism , Cell Line , Chimerin Proteins/genetics , Chimerin Proteins/metabolism , Enzyme Activation , Fas Ligand Protein , Humans , NF-kappa B/metabolism , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand , Viral Matrix Proteins/genetics , Viral Matrix Proteins/metabolism
19.
Biochim Biophys Acta ; 1754(1-2): 296-304, 2005 Dec 30.
Article in English | MEDLINE | ID: mdl-16202672

ABSTRACT

Phorbol esters, natural compounds that mimic the action of the lipid second messenger diacylglycerol (DAG), are known to exert their biological actions through the activation of classical and novel protein kinase C (PKC) isozymes. Phorbol esters, via binding to the PKC C1 domains, cause major effects on mitogenesis by controlling the activity of cyclin-cdk complexes and the expression of cdk inhibitors. In the last years it became clear that phorbol esters activate other molecules having a C1 domain in addition to PKCs. One of the most interesting families of "non-kinase" phorbol ester receptors is represented by the chimaerins, lipid-regulated Rac-GAPs that modulate actin cytoskeleton reorganization, migration, and proliferation. The discovery of the chimaerins and other "non-kinase" phorbol ester receptors has major implications in the design of agents for cancer therapy.


Subject(s)
Protein Kinase C/metabolism , Receptors, Drug/metabolism , Animals , Anticarcinogenic Agents/therapeutic use , Apoptosis , Cell Cycle/drug effects , Cell Cycle/physiology , Chimerin Proteins/metabolism , Diacylglycerol Kinase/metabolism , Diglycerides/metabolism , Humans , Isoenzymes/metabolism , Models, Biological , Phorbol Esters/metabolism , Phorbol Esters/pharmacology , Phorbol Esters/therapeutic use , Protein Binding , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/pharmacology , Protein Structure, Tertiary , Second Messenger Systems , Signal Transduction/drug effects
20.
Bioorg Med Chem ; 12(17): 4575-83, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15358285

ABSTRACT

Recent investigations discovered nonkinase-type phorbol ester receptors, RasGRPs, chimaerins, and Unc13s. Phorbol ester binding occurs at the cysteine-rich sequences of about 50 residues in the C1 domains of these receptors. Fifty-one-residue RasGRP C1 peptides except for RasGRP2 showed significant phorbol 12,13-dibutyrate (PDBu) binding, but the K(d) values of the RasGRP1 and RasGRP3 C1 peptides were about 10-fold larger than those for the corresponding whole enzymes. Addition of the C-terminal basic amino acid cluster decreased their K(d) values about 10-fold, suggesting that the positive charges of these C1 peptides play an important role in the PDBu binding in the presence of negatively-charged phosphatidylserine. The 51-mer chimaerin C1 peptides showed potent PDBu binding, while the Unc13 and Munc13-1 C1 peptides without sufficient positive charges hardly bound PDBu. By the rapid screening system using this C1 peptide library, 5-prenyl-indolactam-V was identified as a promising lead for the novel protein kinase C isozyme specific ligands.


Subject(s)
Carcinogens/pharmacology , Chimerin Proteins/metabolism , DNA-Binding Proteins/metabolism , Glycogen Synthase/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Nerve Tissue Proteins/metabolism , Peptide Fragments/metabolism , Protein Kinase C/metabolism , Amino Acid Sequence , Binding Sites/drug effects , Chimerin Proteins/chemistry , DNA-Binding Proteins/chemistry , Glycogen Synthase/chemistry , Guanine Nucleotide Exchange Factors/chemistry , Humans , Indoles/pharmacology , Isoenzymes/drug effects , Isoenzymes/metabolism , Lactams/pharmacology , Ligands , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Peptide Fragments/chemistry , Peptide Fragments/drug effects , Phorbol 12,13-Dibutyrate/pharmacology , Protein Kinase C/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...