Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
PLoS One ; 17(1): e0251286, 2022.
Article in English | MEDLINE | ID: mdl-35081118

ABSTRACT

Endometrioid endometrial carcinomas (EECs) are the most common histological subtype of uterine cancer. Late-stage disease is an adverse prognosticator for EEC. The purpose of this study was to analyze EEC exome mutation data to identify late-stage-specific statistically significantly mutated genes (SMGs), which represent candidate driver genes potentially associated with disease progression. We exome sequenced 15 late-stage (stage III or IV) non-ultramutated EECs and paired non-tumor DNAs; somatic variants were called using Strelka, Shimmer, SomaticSniper and MuTect. Additionally, somatic mutation calls were extracted from The Cancer Genome Atlas (TCGA) data for 66 late-stage and 270 early-stage (stage I or II) non-ultramutated EECs. MutSigCV (v1.4) was used to annotate SMGs in the two late-stage cohorts and to derive p-values for all mutated genes in the early-stage cohort. To test whether late-stage SMGs are statistically significantly mutated in early-stage tumors, q-values for late-stage SMGs were re-calculated from the MutSigCV (v1.4) early-stage p-values, adjusting for the number of late-stage SMGs tested. We identified 14 SMGs in the combined late-stage EEC cohorts. When the 14 late-stage SMGs were examined in the TCGA early-stage data, only Krüppel-like factor 3 (KLF3) and Paired box 6 (PAX6) failed to reach significance as early-stage SMGs, despite the inclusion of enough early-stage cases to ensure adequate statistical power. Within TCGA, nonsynonymous mutations in KLF3 and PAX6 were, respectively, exclusive or nearly exclusive to the microsatellite instability (MSI)-hypermutated molecular subgroup and were dominated by insertions-deletions at homopolymer tracts. In conclusion, our findings are hypothesis-generating and suggest that KLF3 and PAX6, which encode transcription factors, are MSI target genes and late-stage-specific SMGs in EEC.


Subject(s)
Carcinoma, Endometrioid , Kruppel-Like Transcription Factors , PAX6 Transcription Factor , Carcinoma, Endometrioid/genetics , Carcinoma, Endometrioid/pathology , Cohort Studies , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Exome , Female , Humans , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Microsatellite Instability , Middle Aged , Mutation , PAX6 Transcription Factor/genetics , PAX6 Transcription Factor/metabolism , Uterine Neoplasms/genetics , Uterine Neoplasms/pathology
2.
Cancer Genet ; 240: 5-14, 2020 01.
Article in English | MEDLINE | ID: mdl-31678638

ABSTRACT

Uterine cancer is the 6th leading cause of cancer death amongst American women. Most uterine cancers are endometrial carcinomas (ECs), which are classified into histological subtypes including endometrioid, serous, and clear cell ECs. Somatic copy number alterations (SCNAs) are frequent in serous EC, infrequent in endometrioid ECs, and poorly defined in clear cell ECs. The purpose of this study was to evaluate the occurrence of SCNAs in clinically diagnosed clear cell ECs. Paired tumor-normal DNAs for 51 ECs were hybridized to Illumina Infinium HumanHap650Y or Human660W-Quad Beadchips. Copy number calls were made using the Hidden Markov Model based SNP-FASST2 segmentation algorithm within Nexus Copy Number software (v.6.1). High-level SCNAs were defined as gain of ≥5 copies or homozygous deletion, both <10Mb. GISTIC 1.0, in Nexus, was used to identify statistically significant SCNAs, corrected for multiple testing. One or more high-level SCNAs were detected in 50% of 6 clear cell ECs, 78.6% of 28 serous ECs, and 17.6% of 17 endometrioid ECs. A positive association was found between high-level SCNAs and TP53 mutation across ECs (two-tailed p value<0.0001). Classifying tumors according to POLE, MSI, and TP53 status yielded four molecular subgroups; copy number altered tumors were more frequent in the TP53-mutated subgroup (95.8%) than in the unspecified subgroup (22.2%), and absent from the POLE and MSI subgroups. In conclusion, our study provides evidence of inter-tumor heterogeneity in the extent to which SCNAs occur in clinically diagnosed clear cell EC, and across molecular subgroups of EC. The co-occurrence of high-level SCNAs and TP53 mutations in some clear cell ECs is consistent with the view that a subset of clinically diagnosed clear cell ECs have molecular similarities to serous ECs.


Subject(s)
Carcinoma, Endometrioid/genetics , DNA Copy Number Variations , Endometrial Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Carcinoma, Endometrioid/mortality , Carcinoma, Endometrioid/pathology , Endometrial Neoplasms/mortality , Endometrial Neoplasms/pathology , Endometrium/pathology , Female , Humans , Kaplan-Meier Estimate , Polymorphism, Single Nucleotide , Prognosis , Progression-Free Survival
3.
Cancer ; 124(1): 65-73, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28940304

ABSTRACT

BACKGROUND: Uterine carcinosarcomas (UCSs) are a rare but clinically aggressive form of cancer. They are biphasic tumors consisting of both epithelial and sarcomatous components. The majority of uterine carcinosarcomas are clonal, with the carcinomatous cells undergoing metaplasia to give rise to the sarcomatous component. The objective of the current study was to identify novel somatically mutated genes in UCSs. METHODS: We whole exome sequenced paired tumor and nontumor DNAs from 14 UCSs and orthogonally validated 464 somatic variants using Sanger sequencing. Fifteen genes that were somatically mutated in at least 2 tumor exomes were Sanger sequenced in another 39 primary UCSs. RESULTS: Overall, among 53 UCSs in the current study, the most frequently mutated of these 15 genes were tumor protein p53 (TP53) (75.5%), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) (34.0%), protein phosphatase 2, regulatory subunit A, alpha (PPP2R1A) (18.9%), F-box and WD repeat domain containing 7 (FBXW7) (18.9%), chromodomain helicase DNA binding protein 4 (CHD4) (17.0%), and forkhead box A2 (FOXA2) (15.1%). FOXA2 has not previously been implicated in UCSs and was predominated by frameshift and nonsense mutations. One UCS with a FOXA2 frameshift mutation expressed truncated FOXA2 protein by immunoblotting. Sequencing of FOXA2 in 160 primary endometrial carcinomas revealed somatic mutations in 5.7% of serous, 22.7% of clear cell, 9% of endometrioid, and 11.1% of mixed endometrial carcinomas, the majority of which were frameshift mutations. CONCLUSIONS: Collectively, the findings of the current study provide compelling genetic evidence that FOXA2 is a pathogenic driver gene in the etiology of primary uterine cancers, including UCSs. Cancer 2018;124:65-73. © 2017 American Cancer Society.


Subject(s)
Carcinoma/genetics , Carcinosarcoma/genetics , Uterine Neoplasms/genetics , Adenocarcinoma, Clear Cell/genetics , Autoantigens/genetics , Carcinoma, Endometrioid/genetics , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases/genetics , Endometrial Neoplasms/genetics , F-Box-WD Repeat-Containing Protein 7/genetics , Female , Hepatocyte Nuclear Factor 3-beta/genetics , Humans , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Microsatellite Instability , Mutation , Neoplasms, Cystic, Mucinous, and Serous/genetics , Polymerase Chain Reaction , Protein Phosphatase 2/genetics , Sequence Analysis, DNA , Tumor Suppressor Protein p53/genetics
4.
Cancer ; 123(17): 3261-3268, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28485815

ABSTRACT

BACKGROUND: The molecular pathogenesis of clear cell endometrial cancer (CCEC), a tumor type with a relatively unfavorable prognosis, is not well defined. We searched exome-wide for novel somatically mutated genes in CCEC and assessed the mutational spectrum of known and candidate driver genes in a large cohort of cases. METHODS: We conducted whole exome sequencing of paired tumor-normal DNAs from 16 cases of CCEC (12 CCECs and the CCEC components of 4 mixed histology tumors). Twenty-two genes-of-interest were Sanger-sequenced from another 47 cases of CCEC. Microsatellite instability (MSI) and microsatellite stability (MSS) were determined by genotyping 5 mononucleotide repeats. RESULTS: Two tumor exomes had relatively high mutational loads and MSI. The other 14 tumor exomes were MSS and had 236 validated nonsynonymous or splice junction somatic mutations among 222 protein-encoding genes. Among the 63 cases of CCEC in this study, we identified frequent somatic mutations in TP53 (39.7%), PIK3CA (23.8%), PIK3R1 (15.9%), ARID1A (15.9%), PPP2R1A (15.9%), SPOP (14.3%), and TAF1 (9.5%), as well as MSI (11.3%). Five of 8 mutations in TAF1, a gene with no known role in CCEC, localized to the putative histone acetyltransferase domain and included 2 recurrently mutated residues. Based on patterns of MSI and mutations in 7 genes, CCEC subsets molecularly resembled serous endometrial cancer (SEC) or endometrioid endometrial cancer (EEC). CONCLUSION: Our findings demonstrate molecular similarities between CCEC and SEC and EEC and implicate TAF1 as a novel candidate CCEC driver gene. Cancer 2017;123:3261-8. © 2017 American Cancer Society.


Subject(s)
Adenocarcinoma, Clear Cell/genetics , Endometrial Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Histone Acetyltransferases/genetics , Mutation , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics , Adenocarcinoma, Clear Cell/pathology , Aged , Cohort Studies , DNA Mutational Analysis , Endometrial Neoplasms/pathology , Exome , Female , Genome-Wide Association Study , Humans , Immunoblotting/methods , Microsatellite Instability , Middle Aged , Molecular Sequence Data , Prognosis
5.
BMC Cancer ; 14: 884, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25427824

ABSTRACT

BACKGROUND: Endometrial cancer (EC) is the 8th leading cause of cancer death amongst American women. Most ECs are endometrioid, serous, or clear cell carcinomas, or an admixture of histologies. Serous and clear ECs are clinically aggressive tumors for which alternative therapeutic approaches are needed. The purpose of this study was to search for somatic mutations in the tyrosine kinome of serous and clear cell ECs, because mutated kinases can point to potential therapeutic targets. METHODS: In a mutation discovery screen, we PCR amplified and Sanger sequenced the exons encoding the catalytic domains of 86 tyrosine kinases from 24 serous, 11 clear cell, and 5 mixed histology ECs. For somatically mutated genes, we next sequenced the remaining coding exons from the 40 discovery screen tumors and sequenced all coding exons from another 72 ECs (10 clear cell, 21 serous, 41 endometrioid). We assessed the copy number of mutated kinases in this cohort of 112 tumors using quantitative real time PCR, and we used immunoblotting to measure expression of these kinases in endometrial cancer cell lines. RESULTS: Overall, we identified somatic mutations in TNK2 (tyrosine kinase non-receptor, 2) and DDR1 (discoidin domain receptor tyrosine kinase 1) in 5.3% (6 of 112) and 2.7% (3 of 112) of ECs. Copy number gains of TNK2 and DDR1 were identified in another 4.5% and 0.9% of 112 cases respectively. Immunoblotting confirmed TNK2 and DDR1 expression in endometrial cancer cell lines. Three of five missense mutations in TNK2 and one of two missense mutations in DDR1 are predicted to impact protein function by two or more in silico algorithms. The TNK2(P761Rfs*72) frameshift mutation was recurrent in EC, and the DDR1(R570Q) missense mutation was recurrent across tumor types. CONCLUSIONS: This is the first study to systematically search for mutations in the tyrosine kinome in clear cell endometrial tumors. Our findings indicate that high-frequency somatic mutations in the catalytic domains of the tyrosine kinome are rare in clear cell ECs. We uncovered ten new mutations in TNK2 and DDR1 within serous and endometrioid ECs, thus providing novel insights into the mutation spectrum of each gene in EC.


Subject(s)
Adenocarcinoma, Clear Cell/genetics , Cystadenocarcinoma, Serous/genetics , Endometrial Neoplasms/genetics , Mutation , Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/genetics , Adenocarcinoma, Clear Cell/pathology , Cell Line, Tumor , Cystadenocarcinoma, Serous/pathology , DNA Copy Number Variations , DNA Mutational Analysis , DNA Polymerase II/genetics , Discoidin Domain Receptor 1 , Endometrial Neoplasms/pathology , Female , Humans , Microsatellite Instability , Neoplasm Grading , Poly-ADP-Ribose Binding Proteins , Protein Interaction Domains and Motifs/genetics
6.
PLoS One ; 8(6): e63313, 2014.
Article in English | MEDLINE | ID: mdl-23755103

ABSTRACT

Most endometrial cancers can be classified histologically as endometrioid, serous, or clear cell. Non-endometrioid endometrial cancers (NEECs; serous and clear cell) are the most clinically aggressive of the three major histotypes and are characterized by aneuploidy, a feature of chromosome instability. The genetic alterations that underlie chromosome instability in endometrial cancer are poorly understood. In the present study, we used Sanger sequencing to search for nucleotide variants in the coding exons and splice junctions of 21 candidate chromosome instability genes, including 19 genes implicated in sister chromatid cohesion, from 24 primary, microsatellite-stable NEECs. Somatic mutations were verified by sequencing matched normal DNAs. We subsequently resequenced mutated genes from 41 additional NEECs as well as 42 endometrioid ECs (EECs). We uncovered nonsynonymous somatic mutations in ESCO1, CHTF18, and MRE11A in, respectively, 3.7% (4 of 107), 1.9% (2 of 107), and 1.9% (2 of 107) of endometrial tumors. Overall, 7.7% (5 of 65) of NEECs and 2.4% (1 of 42) of EECs had somatically mutated one or more of the three genes. A subset of mutations are predicted to impact protein function. The co-occurrence of somatic mutations in ESCO1 and CHTF18 was statistically significant (P = 0.0011, two-tailed Fisher's exact test). This is the first report of somatic mutations within ESCO1 and CHTF18 in endometrial tumors and of MRE11A mutations in microsatellite-stable endometrial tumors. Our findings warrant future studies to determine whether these mutations are driver events that contribute to the pathogenesis of endometrial cancer.


Subject(s)
Acetyltransferases/genetics , Carrier Proteins/genetics , Chromosomal Instability/genetics , DNA-Binding Proteins/genetics , Endometrial Neoplasms/genetics , Genetic Association Studies , Mutation/genetics , Nuclear Proteins/genetics , ATPases Associated with Diverse Cellular Activities , Cell Line, Tumor , DNA Mutational Analysis , Female , Genetic Predisposition to Disease , Humans , MRE11 Homologue Protein
7.
Nat Genet ; 44(12): 1310-5, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23104009

ABSTRACT

Endometrial cancer is the sixth most commonly diagnosed cancer in women worldwide, causing ~74,000 deaths annually. Serous endometrial cancers are a clinically aggressive subtype with a poorly defined genetic etiology. We used whole-exome sequencing to comprehensively search for somatic mutations within ~22,000 protein-encoding genes in 13 primary serous endometrial tumors. We subsequently resequenced 18 genes, which were mutated in more than 1 tumor and/or were components of an enriched functional grouping, from 40 additional serous tumors. We identified high frequencies of somatic mutations in CHD4 (17%), EP300 (8%), ARID1A (6%), TSPYL2 (6%), FBXW7 (29%), SPOP (8%), MAP3K4 (6%) and ABCC9 (6%). Overall, 36.5% of serous tumors had a mutated chromatin-remodeling gene, and 35% had a mutated ubiquitin ligase complex gene, implicating frequent mutational disruption of these processes in the molecular pathogenesis of one of the deadliest forms of endometrial cancer.


Subject(s)
Adenocarcinoma, Clear Cell/genetics , Carcinoma, Endometrioid/genetics , Chromatin Assembly and Disassembly/genetics , Endometrial Neoplasms/genetics , Exome/genetics , Ubiquitin-Protein Ligase Complexes/genetics , ATP-Binding Cassette Transporters/genetics , Adult , Autoantigens/genetics , Base Sequence , Cell Cycle Proteins/genetics , DNA-Binding Proteins , E1A-Associated p300 Protein/genetics , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Female , Gene Frequency , Humans , MAP Kinase Kinase Kinase 4/genetics , Mi-2 Nucleosome Remodeling and Deacetylase Complex/genetics , Molecular Sequence Data , Mutation , Nuclear Proteins/genetics , Potassium Channels, Inwardly Rectifying/genetics , Receptors, Drug/genetics , Repressor Proteins/genetics , Sequence Analysis, DNA , Sulfonylurea Receptors , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics
8.
PLoS Genet ; 7(8): e1002245, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21901109

ABSTRACT

ATAD5, the human ortholog of yeast Elg1, plays a role in PCNA deubiquitination. Since PCNA modification is important to regulate DNA damage bypass, ATAD5 may be important for suppression of genomic instability in mammals in vivo. To test this hypothesis, we generated heterozygous (Atad5(+/m)) mice that were haploinsuffficient for Atad5. Atad5(+/m) mice displayed high levels of genomic instability in vivo, and Atad5(+/m) mouse embryonic fibroblasts (MEFs) exhibited molecular defects in PCNA deubiquitination in response to DNA damage, as well as DNA damage hypersensitivity and high levels of genomic instability, apoptosis, and aneuploidy. Importantly, 90% of haploinsufficient Atad5(+/m) mice developed tumors, including sarcomas, carcinomas, and adenocarcinomas, between 11 and 20 months of age. High levels of genomic alterations were evident in tumors that arose in the Atad5(+/m) mice. Consistent with a role for Atad5 in suppressing tumorigenesis, we also identified somatic mutations of ATAD5 in 4.6% of sporadic human endometrial tumors, including two nonsense mutations that resulted in loss of proper ATAD5 function. Taken together, our findings indicate that loss-of-function mutations in mammalian Atad5 are sufficient to cause genomic instability and tumorigenesis.


Subject(s)
Adenosine Triphosphatases/genetics , Cell Transformation, Neoplastic/genetics , DNA-Binding Proteins/genetics , Neoplasms/genetics , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/metabolism , Aneuploidy , Animals , Cell Line , DNA Damage/genetics , DNA-Binding Proteins/metabolism , Endometrial Neoplasms/genetics , Female , Genetic Predisposition to Disease , Genomic Instability , Humans , Male , Mice , Mutation/genetics , Proliferating Cell Nuclear Antigen/metabolism , Ubiquitination
9.
Cancer Res ; 71(12): 4061-7, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21478295

ABSTRACT

Phosphoinositide 3-kinase (PI3K) is an important therapeutic target. Mutations in PIK3CA, which encodes p110α, the catalytic subunit of PI3K, occur in endometrioid endometrial cancers (EEC) and nonendometrioid endometrial cancers (NEEC). The goal of this study was to determine whether PIK3R1, which encodes p85α, the inhibitory subunit of PI3K, is mutated in endometrial carcinoma. We carried out exonic sequencing of PIK3R1 from 42 EECs and 66 NEECs. The pattern of PIK3R1 mutations was compared with the patterns of PIK3CA, PTEN, and KRAS mutations. The biochemical effect of seven PIK3R1 mutations was examined by stable expression in U2OS cells, followed by coimmunoprecipitation analysis of p110α, and Western blotting of phospho-AKT(Ser473) (p-AKT(Ser473)). We found that PIK3R1 was somatically mutated in 43% of EECs and 12% of NEECs. The majority of mutations (93.3%) were localized to the p85α-nSH2 and -iSH2 domains. Several mutations were recurrent. PIK3R1 mutations were significantly (P = 0.0015) more frequent in PIK3CA-wild type EECs (70%) than in PIK3CA mutant EECs (18%). Introduction of wild-type p85α into U2OS cells reduced the level of p-AKT(Ser473) compared with the vector control. Five p85α mutants, p85αdelH450-E451, p85αdelK459, p85αdelY463-L466, p85αdelR574-T576, and the p85αN564D positive control, were shown to bind p110α and led to increased levels of p-AKT(Ser473). The p85αR348X and p85αK511VfsX2 mutants did not bind p110α and showed no appreciable change in p-AKT(Ser473) levels. In conclusion, our study has revealed a new mode of PI3K alteration in primary endometrial tumors and warrants future studies to determine whether PIK3R1 mutations correlate with clinical outcome to targeted therapies directed against the PI3K pathway in EEC and NEEC.


Subject(s)
Carcinoma, Endometrioid/genetics , Class Ia Phosphatidylinositol 3-Kinase/genetics , Endometrial Neoplasms/genetics , Mutation , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Female , Humans , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras) , ras Proteins/genetics
10.
Clin Cancer Res ; 17(6): 1331-40, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21266528

ABSTRACT

PURPOSE: The goal of this study was to comprehensively define the incidence of mutations in all exons of PIK3CA in both endometrioid endometrial cancer (EEC) and nonendometrioid endometrial cancer (NEEC). EXPERIMENTAL DESIGN: We resequenced all coding exons of PIK3CA and PTEN, and exons 1 and 2 of KRAS, from 108 primary endometrial tumors. Somatic mutations were confirmed by sequencing matched normal DNAs. The biochemical properties of a subset of novel PIK3CA mutations were determined by exogenously expressing wild type and mutant constructs in U2OS cells and measuring levels of AKT(Ser473) phosphorylation. RESULTS: Somatic PIK3CA mutations were detected in 52.4% of 42 EECs and 33.3% of 66 NEECs. Half (29 of 58) of all nonsynonymous PIK3CA mutations were in exons 1-7 and half were in exons 9 and 20. The exons 1-7 mutations localized to the ABD, ABD-RBD linker and C2 domains of p110α. Within these regions, Arg88, Arg93, Gly106, Lys111, Glu365, and Glu453, were recurrently mutated; Arg88, Arg93, and Lys111 formed mutation hotspots. The p110α-R93W, -G106R, -G106V, -K111E, -delP449-L455, and -E453K mutants led to increased levels of phospho-AKT(Ser473) compared to wild-type p110α. Overall, 62% of exons 1-7 PIK3CA mutants and 64% of exons 9-20 PIK3CA mutants were activating; 72% of exon 1-7 mutations have not previously been reported in endometrial cancer. CONCLUSIONS: Our study identified a new subgroup of endometrial cancer patients with activating mutations in the amino-terminal domains of p110α; these patients might be appropriate for consideration in clinical trials of targeted therapies directed against the PI3K pathway.


Subject(s)
Carcinoma/genetics , Endometrial Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Mutation , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/physiology , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , DNA Mutational Analysis , Exons , Female , Humans , PTEN Phosphohydrolase/genetics , Phosphorylation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras) , ras Proteins/genetics
11.
Mol Cell Biol ; 26(21): 7892-900, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16923964

ABSTRACT

Thymocyte development is shaped by signals from the T-cell antigen receptor. The strength of receptor signaling determines developmental progression as well as deletion of self-reactive T cells. Receptor stimulation of the extracellular signal-regulated kinase (ERK) pathway plays an important regulatory role during thymocyte development. However, it is unclear how differences in receptor signaling are translated into distinctive activation of the ERK pathway. We have investigated the potential role of the Lck tyrosine kinase in regulating intracellular signaling during thymocyte development. While Lck is known to be critical for initial T-cell receptor signaling events, it may have an independent role in regulating intracellular signaling through the function of its SH3 domain. To determine whether such a regulatory mechanism functions during thymocyte development, we generated mice in which the normal lck allele is replaced with an lck SH3 domain mutant. Analysis of these mice revealed that both early thymocyte development and maturation of CD4(+) and CD8(+) lineages is impaired. Investigation of thymocyte responses to antigen receptor stimulation showed a significant reduction in proliferation and ERK pathway activation, although initial signaling events were intact. These findings indicate that Lck SH3 domain function may provide a means to independently couple receptor signaling to regulation of the ERK pathway during thymocyte development.


Subject(s)
Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction/physiology , Thymus Gland , src Homology Domains , Animals , Antigens, CD/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/chemistry , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell/genetics , Thymus Gland/cytology , Thymus Gland/growth & development
12.
J Neuroimmunol ; 160(1-2): 87-91, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15710461

ABSTRACT

Previous studies indicate that norepinephrine and epinephrine modulate production of interleukin-1(beta) (IL-1(beta)) by activated macrophages, but it is not known if macrophage-derived catecholamines affect IL-1(beta). In this study, recruited peritoneal macrophages from CBA/J female mice were activated with lipopolysaccharide (LPS) and treated with vehicle or adrenergic receptor antagonists for 24 h. Extracellular and intracellular levels of IL-1(beta) were measured with ELISA. Treatment with the beta-adrenergic receptor antagonists propranolol or ICI 118,551 increased LPS-induced production of IL-1(beta), whereas treatment with the alpha-adrenergic antagonists phentolamine or yohimbine decreased IL-1(beta). These findings demonstrate that adrenergic receptor antagonists unmask autocrine actions of macrophage-derived catecholamines on IL-1(beta) that may influence the inflammatory response.


Subject(s)
Autocrine Communication/immunology , Catecholamines/physiology , Interleukin-1/metabolism , Macrophages, Peritoneal/metabolism , Adjuvants, Immunologic/pharmacology , Adrenergic alpha-Antagonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Animals , Autocrine Communication/drug effects , Cell Survival/drug effects , Cell Survival/immunology , Extracellular Space/drug effects , Extracellular Space/immunology , Extracellular Space/metabolism , Interleukin-1/antagonists & inhibitors , Interleukin-1/biosynthesis , Intracellular Fluid/drug effects , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Lipopolysaccharides/pharmacology , Macrophage Activation/drug effects , Macrophage Activation/immunology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/immunology , Mice , Mice, Inbred CBA , Propanolamines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...