Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
J Clin Oncol ; 39(7): 822-835, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33405951

ABSTRACT

PURPOSE: SJMB03 (ClinicalTrials.gov identifier: NCT00085202) was a phase III risk-adapted trial that aimed to determine the frequency and clinical significance of biological variants and genetic alterations in medulloblastoma. PATIENTS AND METHODS: Patients 3-21 years old were stratified into average-risk and high-risk treatment groups based on metastatic status and extent of resection. Medulloblastomas were molecularly classified into subgroups (Wingless [WNT], Sonic Hedgehog [SHH], group 3, and group 4) and subtypes based on DNA methylation profiles and overlaid with gene mutations from next-generation sequencing. Coprimary study end points were (1) to assess the relationship between ERBB2 protein expression in tumors and progression-free survival (PFS), and (2) to estimate the frequency of mutations associated with WNT and SHH tumors. Clinical and molecular risk factors were evaluated, and the most robust were used to model new risk-classification categories. RESULTS: Three hundred thirty eligible patients with medulloblastoma were enrolled. Five-year PFS was 83.2% (95% CI, 78.4 to 88.2) for average-risk patients (n = 227) and 58.7% (95% CI, 49.8 to 69.1) for high-risk patients (n = 103). No association was found between ERBB2 status and PFS in the overall cohort (P = .74) or when patients were stratified by clinical risk (P = .71). Mutations in CTNNB1 (96%), DDX3X (37%), and SMARCA4 (24%) were most common in WNT tumors and PTCH1 (38%), TP53 (21%), and DDX3X (19%) in SHH tumors. Methylome profiling classified 53 WNT (17.4%), 48 SHH (15.7%), 65 group 3 (21.3%), and 139 group 4 (45.6%) tumors. A comprehensive clinicomolecular risk factor analysis identified three low-risk groups (WNT, low-risk SHH, and low-risk combined groups 3 and 4) with excellent (5-year PFS > 90%) and two very high-risk groups (high-risk SHH and high-risk combined groups 3 and 4) with poor survival (5-year PFS < 60%). CONCLUSION: These results establish a new risk stratification for future medulloblastoma trials.


Subject(s)
Biomarkers, Tumor/genetics , Cerebellar Neoplasms/therapy , DNA Methylation , Medulloblastoma/therapy , Mutation , Adolescent , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/mortality , Cerebellar Neoplasms/pathology , Child , Child, Preschool , DNA Mutational Analysis , Epigenome , Epigenomics , Female , High-Throughput Nucleotide Sequencing , Humans , Magnetic Resonance Imaging , Male , Medulloblastoma/genetics , Medulloblastoma/mortality , Medulloblastoma/secondary , Predictive Value of Tests , Progression-Free Survival , Risk Assessment , Risk Factors , Time Factors , Young Adult
2.
Cell Immunol ; 346: 103996, 2019 12.
Article in English | MEDLINE | ID: mdl-31703914

ABSTRACT

Sex hormones are best known for their influences on reproduction, but they also have profound influences on the immune response. Examples of sex-specific differences include: (i) the relatively poor control of influenza virus infections in males compared to females, (ii) allergic asthma, an IgE-associated hypersensitivity reaction that is exacerbated in adolescent females compared to males, and (iii) systemic lupus erythematosus, a life-threatening autoimmune disease with a 9:1 female:male bias. Here we consider how estrogen and estrogen receptor α (ERα) may influence the immune response by modifying class switch recombination (CSR) and immunoglobulin expression patterns. We focus on ERα binding to enhancers (Eµ and the 3' regulatory region) and switch sites (Sµ and Sε) in the immunoglobulin heavy chain locus. Our preliminary data from ChIP-seq analyses of purified, activated B cells show estrogen-mediated changes in the positioning of ERα binding within and near Sµ and Sε. In the presence of estrogen, ERα is bound not only to estrogen response elements (ERE), but also to adenosine-cytidine (AC)-repeats and poly adenosine (poly A) sequences, in some cases within constant region gene introns. We propose that by binding these sites, estrogen and ERα directly participate in the DNA loop formation required for CSR. We further suggest that estrogen regulates immunoglobulin expression patterns and can thereby influence life-and-death outcomes of infection, hypersensitivity, and autoimmune disease.


Subject(s)
Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Immunoglobulin Class Switching/immunology , Autoimmune Diseases/immunology , Female , Humans , Hypersensitivity/genetics , Hypersensitivity/immunology , Male , Poly A/genetics , Response Elements/genetics
3.
Vaccines (Basel) ; 7(1)2019 Feb 27.
Article in English | MEDLINE | ID: mdl-30818795

ABSTRACT

Despite extraordinary advances in fields of immunology and infectious diseases, vaccine development remains a challenge. The development of a respiratory syncytial virus vaccine, for example, has spanned more than 50 years of research with studies of more than 100 vaccine candidates. Dozens of attractive vaccine products have entered clinical trials, but none have completed the path to licensing. Human immunodeficiency virus vaccine development has proven equally difficult, as there is no licensed product after more than 30 years of pre-clinical and clinical research. Here, we examine vaccine development with attention to the host. We discuss how nuclear hormones, including vitamins and sex hormones, can influence responses to vaccines. We show how nuclear hormones interact with regulatory elements of immunoglobulin gene loci and how the deletion of estrogen response elements from gene enhancers will alter patterns of antibody isotype expression. Based on these findings, and findings that nuclear hormone levels are often insufficient or deficient among individuals in both developed and developing countries, we suggest that failed vaccine studies may in some cases reflect weaknesses of the host rather than the product. We encourage analyses of nuclear hormone levels and immunocompetence among study participants in clinical trials to ensure the success of future vaccine programs.

4.
Int Immunol ; 31(3): 141-156, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30407507

ABSTRACT

Nuclear hormone receptors including the estrogen receptor (ERα) and the retinoic acid receptor regulate a plethora of biological functions including reproduction, circulation and immunity. To understand how estrogen and other nuclear hormones influence antibody production, we characterized total serum antibody isotypes in female and male mice of C57BL/6J, BALB/cJ and C3H/HeJ mouse strains. Antibody levels were higher in females compared to males in all strains and there was a female preference for IgG2b production. Sex-biased patterns were influenced by vitamin levels, and by antigen specificity toward influenza virus or pneumococcus antigens. To help explain sex biases, we examined the direct effects of estrogen on immunoglobulin heavy chain sterile transcript production among purified, lipopolysaccharide-stimulated B cells. Supplemental estrogen in B-cell cultures significantly increased immunoglobulin heavy chain sterile transcripts. Chromatin immunoprecipitation analyses of activated B cells identified significant ERα binding to estrogen response elements (EREs) centered within enhancer elements of the immunoglobulin heavy chain locus, including the Eµ enhancer and hypersensitive site 1,2 (HS1,2) in the 3' regulatory region. The ERE in HS1,2 was conserved across animal species, and in humans marked a site of polymorphism associated with the estrogen-augmented autoimmune disease, lupus. Taken together, the results highlight: (i) the important targets of ERα in regulatory regions of the immunoglobulin heavy chain locus that influence antibody production, and (ii) the complexity of mechanisms by which estrogen instructs sex-biased antibody production profiles.


Subject(s)
Antibody Formation/genetics , Enhancer Elements, Genetic , Immunoglobulin Heavy Chains/genetics , Receptors, Estrogen/metabolism , Response Elements/genetics , Sex Characteristics , Animals , Antibody Formation/immunology , Binding Sites , Immunoglobulin Heavy Chains/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Response Elements/immunology
5.
Cell ; 170(1): 142-157.e19, 2017 Jun 29.
Article in English | MEDLINE | ID: mdl-28648661

ABSTRACT

Immune-checkpoint-blockade (ICB)-mediated rejuvenation of exhausted T cells has emerged as a promising approach for treating various cancers and chronic infections. However, T cells that become fully exhausted during prolonged antigen exposure remain refractory to ICB-mediated rejuvenation. We report that blocking de novo DNA methylation in activated CD8 T cells allows them to retain their effector functions despite chronic stimulation during a persistent viral infection. Whole-genome bisulfite sequencing of antigen-specific murine CD8 T cells at the effector and exhaustion stages of an immune response identified progressively acquired heritable de novo methylation programs that restrict T cell expansion and clonal diversity during PD-1 blockade treatment. Moreover, these exhaustion-associated DNA-methylation programs were acquired in tumor-infiltrating PD-1hi CD8 T cells, and approaches to reverse these programs improved T cell responses and tumor control during ICB. These data establish de novo DNA-methylation programming as a regulator of T cell exhaustion and barrier of ICB-mediated T cell rejuvenation.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Epigenesis, Genetic , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Adenocarcinoma/drug therapy , Animals , CD8-Positive T-Lymphocytes/immunology , DNA Methylation , Female , Male , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Prostatic Neoplasms/drug therapy , Virus Diseases/drug therapy
6.
PLoS Pathog ; 12(8): e1005804, 2016 08.
Article in English | MEDLINE | ID: mdl-27505057

ABSTRACT

The healthy lung maintains a steady state of immune readiness to rapidly respond to injury from invaders. Integrins are important for setting the parameters of this resting state, particularly the epithelial-restricted αVß6 integrin, which is upregulated during injury. Once expressed, αVß6 moderates acute lung injury (ALI) through as yet undefined molecular mechanisms. We show that the upregulation of ß6 during influenza infection is involved in disease pathogenesis. ß6-deficient mice (ß6 KO) have increased survival during influenza infection likely due to the limited viral spread into the alveolar spaces leading to reduced ALI. Although the ß6 KO have morphologically normal lungs, they harbor constitutively activated lung CD11b+ alveolar macrophages (AM) and elevated type I IFN signaling activity, which we traced to the loss of ß6-activated transforming growth factor-ß (TGF-ß). Administration of exogenous TGF-ß to ß6 KO mice leads to reduced numbers of CD11b+ AMs, decreased type I IFN signaling activity and loss of the protective phenotype during influenza infection. Protection extended to other respiratory pathogens such as Sendai virus and bacterial pneumonia. Our studies demonstrate that the loss of one epithelial protein, αVß6 integrin, can alter the lung microenvironment during both homeostasis and respiratory infection leading to reduced lung injury and improved survival.


Subject(s)
Antigens, Neoplasm/immunology , Integrins/immunology , Interferon Type I/biosynthesis , Interferon Type I/immunology , Lung/immunology , Respiratory Tract Infections/immunology , Adoptive Transfer , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Fluorescent Antibody Technique , Immunoblotting , Lung/microbiology , Macrophages, Alveolar/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction
7.
Mol Immunol ; 77: 97-102, 2016 09.
Article in English | MEDLINE | ID: mdl-27494228

ABSTRACT

Females and males differ in antibody isotype expression patterns and in immune responses to foreign- and self-antigens. For example, systemic lupus erythematosus is a condition that associates with the production of isotype-skewed anti-self antibodies, and exhibits a 9:1 female:male disease ratio. To explain differences between B cell responses in males and females, we sought to identify direct interactions of the estrogen receptor (ER) with the immunoglobulin heavy chain locus. This effort was encouraged by our previous identification of estrogen response elements (ERE) in heavy chain switch (S) regions. We conducted a full-genome chromatin immunoprecipitation analysis (ChIP-seq) using DNA from LPS-activated B cells and an ERα-specific antibody. Results revealed ER binding to a wide region of DNA, spanning sequences from the JH cluster to Cδ, with peaks in Eµ and Sµ sites. Additional peaks of ERα binding were coincident with hs1,2 and hs4 sites in the 3' regulatory region (3'RR) of the heavy chain locus. This first demonstration of direct binding of ER to key regulatory elements in the immunoglobulin locus supports our hypothesis that estrogen and other nuclear hormone receptors and ligands may directly influence antibody expression and class switch recombination (CSR). Our hypothesis encourages the conduct of new experiments to evaluate the consequences of ER binding. A better understanding of ER:DNA interactions in the immunoglobulin heavy chain locus, and respective mechanisms, may ultimately translate to better control of antibody expression, better protection against pathogens, and prevention of pathologies caused by auto-immune disease.


Subject(s)
B-Lymphocytes/immunology , Gene Expression Regulation/immunology , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Switch Region/immunology , Receptors, Estrogen/immunology , Response Elements/immunology , Animals , Antibody Formation/genetics , Antibody Formation/immunology , Chromatin Immunoprecipitation , Female , Humans , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Receptors, Estrogen/metabolism
8.
Nat Commun ; 6: 7553, 2015 Jun 24.
Article in English | MEDLINE | ID: mdl-26104880

ABSTRACT

There is increasing evidence from genome-wide association studies for a strong inherited genetic basis of susceptibility to acute lymphoblastic leukaemia (ALL) in children, yet the effects of protein-coding variants on ALL risk have not been systematically evaluated. Here we show a missense variant in CDKN2A associated with the development of ALL at genome-wide significance (rs3731249, P=9.4 × 10(-23), odds ratio=2.23). Functional studies indicate that this hypomorphic variant results in reduced tumour suppressor function of p16(INK4A), increases the susceptibility to leukaemic transformation of haematopoietic progenitor cells, and is preferentially retained in ALL tumour cells. Resequencing the CDKN2A-CDKN2B locus in 2,407 childhood ALL cases reveals 19 additional putative functional germline variants. These results provide direct functional evidence for the influence of inherited genetic variation on ALL risk, highlighting the important and complex roles of CDKN2A-CDKN2B tumour suppressors in leukaemogenesis.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Case-Control Studies , Child , Cyclin-Dependent Kinase Inhibitor p15/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA-Binding Proteins/genetics , Genetic Predisposition to Disease , Genetic Variation , Genome-Wide Association Study , Genotype , Germ-Line Mutation , HEK293 Cells , Hematopoietic Stem Cells/metabolism , Humans , Ikaros Transcription Factor/genetics , Mice , Mutagenesis, Site-Directed , Mutation, Missense , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transcription Factors/genetics
9.
Blood ; 120(20): 4197-204, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23007406

ABSTRACT

With the use of risk-directed therapy for childhood acute lymphoblastic leukemia (ALL), outcome has improved dramatically in the past 40 years. However, a substantial portion of patients, many of whom have no known risk factors, experience relapse. Taking a genome-wide approach, in the present study, we evaluated the relationships between genotypes at 444 044 single nucleotide polymorphisms (SNPs) with the risk of relapse in 2535 children with newly diagnosed ALL after adjusting for genetic ancestry and treatment regimen. We identified 134 SNPs that were reproducibly associated with ALL relapse. Of 134 relapse SNPs, 133 remained prognostic after adjusting for all known relapse risk factors, including minimal residual disease, and 111 were significant even among patients who were negative for minimal residual disease after remission induction therapy. The C allele at rs7142143 in the PYGL gene was associated with 3.6-fold higher risk of relapse than the T allele (P = 6.7 × 10(-9)). Fourteen of the 134 relapse SNPs, including variants in PDE4B and ABCB1, were also associated with antileukemic drug pharmacokinetics and/or pharmacodynamics. In the present study, we systematically identified host genetic variations related to treatment outcome of childhood ALL, most of which were prognostic independent of known risk factors for relapse, and some of which also influenced outcome by affecting host dis-position of antileukemic drugs. All trials are registered at www.clinicaltrials.gov or www.cancer.gov (COG P9904: NCT00005585; COG P9905: NCT00005596; COG P9906: NCT00005603; St Jude Total XIIIB: NCI-T93-0101D; and St Jude Total XV: NCT00137111).


Subject(s)
DNA, Neoplasm/genetics , Genome-Wide Association Study , Polymorphism, Single Nucleotide , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Alleles , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biotransformation/genetics , Genotype , Germ-Line Mutation , Glycogen Phosphorylase/genetics , Humans , Neoplasm Proteins/genetics , Neoplasm, Residual , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Prognosis , Recurrence , Remission Induction , Risk
10.
J Biol Chem ; 287(18): 14456-66, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22375007

ABSTRACT

The physiological role of multidrug resistance protein 4 (Mrp4, Abcc4) in the testes is unknown. We found that Mrp4 is expressed primarily in mouse and human Leydig cells; however, there is no current evidence that Mrp4 regulates testosterone production. We investigated its role in Leydig cells, where testosterone production is regulated by cAMP, an intracellular messenger formed when the luteinizing hormone (LH) receptor is activated. Because Mrp4 regulates cAMP, we compared testosterone levels in Mrp4(-/-) and Mrp4(+/+) mice. Young Mrp4(-/-) mice had significantly impaired gametogenesis, reduced testicular testosterone, and disruption of Leydig cell cAMP homeostasis. Both young and adult mice had impaired testosterone production. In Mrp4(-/-) primary Leydig cells treated with LH, intracellular cAMP production was impaired and cAMP-response element-binding protein (CREB) phosphorylation was strongly attenuated. Notably, expression of CREB target genes that regulate testosterone biosynthesis was reduced in Mrp4(-/-) Leydig cells in vivo. Therefore, Mrp4 is required for normal Leydig cell testosterone production. However, adult Mrp4(-/-) mice are fertile, with a normal circulating testosterone concentration. The difference is that in 3-week-old Mrp4(-/-) mice, disruption of gonadal testosterone production up-regulates hepatic Cyp2b10, a known testosterone-metabolizing enzyme. Therefore, defective testicular testosterone production de-regulates hepatic Cyp-mediated testosterone metabolism to disrupt gametogenesis. These findings have important implications for understanding the side effects of therapeutics that disrupt Mrp4 function and are reported to alter androgen production.


Subject(s)
Aryl Hydrocarbon Hydroxylases/biosynthesis , Leydig Cells/metabolism , Liver/enzymology , Multidrug Resistance-Associated Proteins/metabolism , Spermatogenesis/physiology , Steroid Hydroxylases/biosynthesis , Testosterone/biosynthesis , Animals , Aryl Hydrocarbon Hydroxylases/genetics , Cyclic AMP/genetics , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Cytochrome P450 Family 2 , Gene Expression Regulation, Enzymologic/physiology , Humans , Male , Mice , Mice, Knockout , Multidrug Resistance-Associated Proteins/genetics , Receptors, LH/genetics , Receptors, LH/metabolism , Steroid Hydroxylases/genetics , Testosterone/genetics , Up-Regulation/physiology
11.
J Clin Oncol ; 30(7): 751-7, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22291082

ABSTRACT

PURPOSE: Recent genome-wide screens have identified genetic variations in ARID5B associated with susceptibility to childhood acute lymphoblastic leukemia (ALL). We sought to determine the contribution of ARID5B single nucleotide polymorphisms (SNPs) to racial disparities in ALL susceptibility and treatment outcome. PATIENTS AND METHODS: We compared the association between ARID5B SNP genotype and ALL susceptibility in whites (> 95% European genetic ancestry; 978 cases and 1,046 controls) versus in Hispanics (> 10% Native American ancestry; 330 cases and 541 controls). We determined the relationships between ARID5B SNP genotype and ALL relapse risk in 1,605 children treated on the Children's Oncology Group (COG) P9904/9905 clinical trials. RESULTS: Among 49 ARID5B SNPs interrogated, 10 were significantly associated with ALL susceptibility in both whites and Hispanics (P < .05), with risk alleles consistently more frequent in Hispanics than in whites. rs10821936 exhibited the most significant association in both races (P = 8.4 × 10(-20) in whites; P = 1 × 10(-6) in Hispanics), and genotype at this SNP was highly correlated with local Native American genetic ancestry (P = 1.8 × 10(-8)). Multivariate analyses in Hispanics identified an additional SNP associated with ALL susceptibility independent of rs10821936. Eight ARID5B SNPs were associated with both ALL susceptibility and relapse hazard; the alleles related to higher ALL incidence were always linked to poorer treatment outcome and were more frequent in Hispanics. CONCLUSION: ARID5B polymorphisms are important determinants of childhood ALL susceptibility and treatment outcome, and they contribute to racial disparities in this disease.


Subject(s)
DNA-Binding Proteins/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/ethnology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transcription Factors/genetics , Case-Control Studies , Child , Female , Genetic Predisposition to Disease/ethnology , Genotype , Health Status Disparities , Hispanic or Latino/genetics , Humans , Incidence , Indians, North American/genetics , Male , Polymorphism, Single Nucleotide , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Treatment Outcome , White People/genetics
12.
Nature ; 481(7381): 329-34, 2012 Jan 11.
Article in English | MEDLINE | ID: mdl-22237022

ABSTRACT

Retinoblastoma is an aggressive childhood cancer of the developing retina that is initiated by the biallelic loss of RB1. Tumours progress very quickly following RB1 inactivation but the underlying mechanism is not known. Here we show that the retinoblastoma genome is stable, but that multiple cancer pathways can be epigenetically deregulated. To identify the mutations that cooperate with RB1 loss, we performed whole-genome sequencing of retinoblastomas. The overall mutational rate was very low; RB1 was the only known cancer gene mutated. We then evaluated the role of RB1 in genome stability and considered non-genetic mechanisms of cancer pathway deregulation. For example, the proto-oncogene SYK is upregulated in retinoblastoma and is required for tumour cell survival. Targeting SYK with a small-molecule inhibitor induced retinoblastoma tumour cell death in vitro and in vivo. Thus, retinoblastomas may develop quickly as a result of the epigenetic deregulation of key cancer pathways as a direct or indirect result of RB1 loss.


Subject(s)
Epigenesis, Genetic/genetics , Genomics , Molecular Targeted Therapy , Protein Kinase Inhibitors/pharmacology , Retinoblastoma/drug therapy , Retinoblastoma/genetics , Aneuploidy , Animals , Cell Death/drug effects , Cell Line , Cell Survival/drug effects , Chromosomal Instability/genetics , Gene Expression Regulation, Neoplastic , Genes, Retinoblastoma/genetics , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mutation/genetics , Protein Kinase Inhibitors/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Mas , Retinoblastoma/pathology , Retinoblastoma Protein/deficiency , Retinoblastoma Protein/genetics , Sequence Analysis, DNA , Syk Kinase , Xenograft Model Antitumor Assays
13.
Nat Genet ; 43(3): 237-41, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21297632

ABSTRACT

Although five-year survival rates for childhood acute lymphoblastic leukemia (ALL) are now over 80% in most industrialized countries, not all children have benefited equally from this progress. Ethnic differences in survival after childhood ALL have been reported in many clinical studies, with poorer survival observed among African Americans or those with Hispanic ethnicity when compared with European Americans or Asians. The causes of ethnic differences remain uncertain, although both genetic and non-genetic factors are likely important. Interrogating genome-wide germline SNP genotypes in an unselected large cohort of children with ALL, we observed that the component of genomic variation that co-segregated with Native American ancestry was associated with risk of relapse (P = 0.0029) even after adjusting for known prognostic factors (P = 0.017). Ancestry-related differences in relapse risk were abrogated by the addition of a single extra phase of chemotherapy, indicating that modifications to therapy can mitigate the ancestry-related risk of relapse.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Black or African American , Asian People , Child , Child, Preschool , Ethnicity/genetics , Female , Hispanic or Latino , Humans , Indians, North American , Male , Pharmacogenetics , Polymorphism, Single Nucleotide , Precursor Cell Lymphoblastic Leukemia-Lymphoma/ethnology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Principal Component Analysis , Recurrence , Risk , Survival Rate
14.
Acta Neuropathol ; 121(3): 381-96, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21267586

ABSTRACT

Medulloblastoma is heterogeneous, being characterized by molecular subgroups that demonstrate distinct gene expression profiles. Activation of the WNT or SHH signaling pathway characterizes two of these molecular subgroups, the former associated with low-risk disease and the latter potentially targeted by novel SHH pathway inhibitors. This manuscript reports the validation of a novel diagnostic immunohistochemical method to distinguish SHH, WNT, and non-SHH/WNT tumors and details their associations with clinical, pathological and cytogenetic variables. A cohort (n = 235) of medulloblastomas from patients aged 0.4-52 years was studied for expression of four immunohistochemical markers: GAB1, ß-catenin, filamin A, and YAP1. Immunoreactivity (IR) for GAB1 characterizes only SHH tumors and nuclear IR for ß-catenin only WNT tumors. IRs for filamin A and YAP1 identify SHH and WNT tumors. SHH, WNT, and non-SHH/WNT tumors contributed 31, 14, and 55% to the series. All desmoplastic/nodular (D/N) medulloblastomas were SHH tumors, while most WNT tumors (94%) had a classic phenotype. Monosomy 6 was strongly associated with WNT tumors, while PTCH1 loss occurred almost exclusively among SHH tumors. MYC or MYCN amplification and chromosome 17 imbalance occurred predominantly among non-SHH/WNT tumors. Among patients aged 3-16 years and entered onto the SIOP PNET3 trial, outcome was significantly better for children with WNT tumors, when compared to SHH or non-SHH/WNT tumors, which showed similar survival curves. However, high-risk factors (M+ disease, LC/A pathology, MYC amplification) significantly influenced survival in both SHH and non-SHH/WNT groups. We describe a robust method for detecting SHH, WNT, and non-SHH/WNT molecular subgroups in formalin-fixed medulloblastoma samples. In corroborating other studies that indicate the value of combining clinical, pathological, and molecular variables in therapeutic stratification schemes for medulloblastoma, we also provide the first outcome data based on a clinical trial cohort and novel data on how molecular subgroups are distributed across the range of disease.


Subject(s)
Cerebellar Neoplasms/metabolism , Cerebellar Neoplasms/pathology , Hedgehog Proteins/metabolism , Medulloblastoma/metabolism , Medulloblastoma/pathology , Wnt Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adolescent , Adult , Biomarkers, Tumor/metabolism , Child , Child, Preschool , Contractile Proteins/metabolism , Female , Filamins , Gene Expression Regulation, Neoplastic/physiology , Humans , Infant , Kaplan-Meier Estimate , Male , Microfilament Proteins/metabolism , Middle Aged , Phosphoproteins/metabolism , Retrospective Studies , Signal Transduction/physiology , Transcription Factors , YAP-Signaling Proteins , Young Adult , beta Catenin/metabolism
15.
Acta Neuropathol ; 120(6): 731-43, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21046410

ABSTRACT

Recent studies of genetic abnormalities in pediatric low-grade gliomas (LGGs) have focused on activation of the ERK/MAPK pathway by KIAA1549-BRAF gene fusions in the majority of pilocytic astrocytomas (PAs) and by rare mutations in elements of the pathway across histopathologically diverse LGGs. This study reports that MYB, an oncogene not previously implicated in gliomagenesis, is activated in a diverse subset of pediatric LGGs. The study cohort comprised 57 pediatric LGGs and a comparative cohort of 59 pediatric high-grade gliomas (HGGs). The LGG cohort included 34 PAs and 23 diffuse gliomas; fibrillary astrocytomas (n = 14), oligodendroglial tumors (n = 7), and angiocentric gliomas (n = 2). MYB copy number abnormalities were disclosed using Affymetrix 6.0 SNP arrays and confirmed using interphase fluorescence in situ hybridization. Novel MYB amplifications that upregulate MYB RNA and protein expression were demonstrated in 2/14 diffuse astrocytomas. In addition, focal deletion of the terminal region of MYB was seen in 1 of 2 angiocentric gliomas (AGs). Increased expression of MYB was demonstrated by quantitative RT-PCR and immunohistochemistry. MYB upregulation at the protein level was demonstrated in a proportion of diffuse LGGs (60%), pilocytic astrocytomas (41%), and HGGs (19%), but abnormalities at the genomic level were only a feature of diffuse gliomas. Our data suggest that MYB may have a role in a subset of pediatric gliomas, through a variety of mechanisms in addition to MYB amplification and deletion.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Chromosome Aberrations , Glioma/genetics , Mutation/genetics , Oncogene Proteins v-myb/genetics , Up-Regulation/genetics , Adolescent , Age Distribution , Age Factors , Child , Child, Preschool , Cohort Studies , Female , Gene Amplification/genetics , Gene Deletion , Genetic Predisposition to Disease/genetics , Glioma/metabolism , Humans , Male , Oncogene Proteins v-myb/biosynthesis
16.
PLoS One ; 5(9)2010 Sep 02.
Article in English | MEDLINE | ID: mdl-20824063

ABSTRACT

BACKGROUND: Inadequate extracellular conditions can adversely affect the environment of the ER and impinge on the maturation of nascent proteins. The resultant accumulation of unfolded proteins activates a signal transduction pathway, known as the unfolded protein response, which serves primarily to protect the cell during stress and helps restore homeostasis to the ER. PRINCIPAL FINDINGS: Microarray analysis of the unfolded protein response in a human medulloblastoma cell line treated with thapsigargin revealed that, in addition to known targets, a large number of proangiogenic factors were up-regulated. Real-Time PCR analyses confirmed that four of these factors, VEGFA, FGF2, angiogenin and IL8, were transcriptionally up-regulated in multiple cell lines by various ER stress inducers. Our studies on VEGFA regulation revealed that XBP-1(S), a UPR-inducible transcription factor, bound to two regions on the VEGFA promoter, and analysis of XBP-1 null mouse embryonic fibroblasts revealed that it contributes to VEGFA expression in response to ER stress. ATF4, another UPR-inducible transcription factor, also binds to the VEGFA gene, although its contribution to VEGFA transcription appeared to be fairly modest. We also found that VEGFA mRNA stability is increased in response to UPR activation, via activation of AMP kinase, demonstrating that increased mRNA levels occur at two regulatory points. In keeping with the mRNA levels, we found that VEGFA protein is secreted at levels as high as or higher than that achieved in response to hypoxia. CONCLUSIONS AND SIGNIFICANCE: Our results indicate that the UPR plays a significant role in inducing positive regulators of angiogenesis. It also regulates VEGFA expression at transcriptional, post-transcriptional and post-translational levels and is likely to have widespread implications for promoting angiogenesis in response to normal physiological cues as well as in pathological conditions like cancer.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Fibroblast Growth Factor 2/genetics , Transcription, Genetic , Unfolded Protein Response , Animals , Cell Line , Endoplasmic Reticulum/metabolism , Fibroblast Growth Factor 2/metabolism , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Mice , Mice, Knockout , Neovascularization, Physiologic , Promoter Regions, Genetic , Protein Processing, Post-Translational , Rats , Ribonuclease, Pancreatic/genetics , Ribonuclease, Pancreatic/metabolism , Transcriptional Activation , Up-Regulation , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
17.
J Pathol ; 218(2): 172-81, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19373855

ABSTRACT

We report genetic aberrations that activate the ERK/MAP kinase pathway in 100% of posterior fossa pilocytic astrocytomas, with a high frequency of gene fusions between KIAA1549 and BRAF among these tumours. These fusions were identified from analysis of focal copy number gains at 7q34, detected using Affymetrix 250K and 6.0 SNP arrays. PCR and sequencing confirmed the presence of five KIAA1549-BRAF fusion variants, along with a single fusion between SRGAP3 and RAF1. The resulting fusion genes lack the auto-inhibitory domains of BRAF and RAF1, which are replaced in-frame by the beginning of KIAA1549 and SRGAP3, respectively, conferring constitutive kinase activity. An activating mutation of KRAS was identified in the single pilocytic astrocytoma without a BRAF or RAF1 fusion. Further fusions and activating mutations in BRAF were identified in 28% of grade II astrocytomas, highlighting the importance of the ERK/MAP kinase pathway in the development of paediatric low-grade gliomas.


Subject(s)
Astrocytoma/enzymology , Brain Neoplasms/enzymology , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Adolescent , Adult , Astrocytoma/genetics , Brain Neoplasms/genetics , Child , Child, Preschool , DNA Mutational Analysis , DNA, Complementary/analysis , Enzyme Activation , GTPase-Activating Proteins/genetics , Humans , Infant , Mitogen-Activated Protein Kinases/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-raf/genetics , Young Adult
18.
Cancer ; 113(6): 1453-61, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18704985

ABSTRACT

BACKGROUND: Osteosarcoma cell lines and tumors have been shown to express epidermal growth factor receptor (EGFR) and harbor amplifications at the EGFR locus. In this study, the authors further analyzed the genomic features of EGFR in osteosarcoma tumors and investigated whether they correlate with phosphatase and tensin homolog (PTEN) expression and copy number status. METHODS: EGFR and PTEN expression was assessed by immunohistochemistry (n = 28), and copy number alterations at the EGFR and PTEN loci were surveyed using Affymetrix (Santa Clara, Calif) 50K single nucleotide polymorphism (SNP) arrays (n = 31) and fluorescence in situ hybridization (FISH) (n = 27). The EGFR tyrosine kinase domain was sequenced to survey for activating mutations (n = 34). In addition, EGFRvIII expression was assessed using reverse transcriptase polymerase chain reaction (n = 24). Results were correlated with available clinical information on 59 patients, with a median age of 14.1 years (range, 5-23 years) and median follow-up of 4.4 years. RESULTS: EGFR expression was detected in the majority of osteosarcoma tumors surveyed (23 of 28; 82%). SNP arrays revealed evidence of frequent copy number gains at 7p11.2 and losses at 10q23.21. A sizeable subset (16 of 27 cases; 59%) showed gains at the EGFR locus using FISH (amplification in 4 of 27 [15%] and balanced chromosome 7 polysomy in 12 of 27 [44%]), and 12 cases showed deletions at the PTEN locus (biallelic deletions in 4 of 27 [15%] and monoallelic deletion in 9 of 27 [33%]). No activating mutations in the EGFR tyrosine kinase domain, EGFRvIII expression, or association with clinical findings were detected. CONCLUSIONS: EGFR expression and genomic gains at the EGFR locus are prevalent in osteosarcoma tumors, which also commonly harbor deletions at the PTEN locus.


Subject(s)
Bone Neoplasms/genetics , ErbB Receptors/genetics , Gene Dosage , Gene Expression Regulation, Neoplastic , Osteosarcoma/genetics , PTEN Phosphohydrolase/genetics , Adolescent , Adult , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Child , Child, Preschool , ErbB Receptors/metabolism , Female , Gene Amplification , Humans , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Male , Mutation/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology , PTEN Phosphohydrolase/metabolism , Polymorphism, Single Nucleotide , Prognosis , Survival Rate
19.
Proc Natl Acad Sci U S A ; 102(17): 6074-9, 2005 Apr 26.
Article in English | MEDLINE | ID: mdl-15831586

ABSTRACT

The restriction of influenza A virus replication to mouse respiratory epithelium means that this host response is anatomically compartmentalized, on the one hand, to sites of T cell stimulation and proliferation in the secondary lymphoid tissue and, on the other hand, to the site of effector T cell function and pathology in the pneumonic lung. Thus, it is hardly surprising that virus-specific CD8(+) T cells recovered by bronchoalveolar lavage (BAL) from the infected respiratory tract seem more "activated" in terms of both cytolytic activity and cytokine production than those cells isolated from the spleen. The present analysis uses Affymetrix microarray technology to compare profiles of gene expression in these two lineage-related, yet anatomically separate, lymphocyte populations. Ninety differentially expressed genes were identified for influenza-specific CD8(+)D(b)NP(366)(+) T cells obtained directly ex vivo by BAL or spleen disruption, with nine genes being further analyzed by quantitative, real-time PCR at the population level. Integrin alphaE, for example, was shown by Affymetrix and real-time mRNA analyses and then by single-cell PCR and protein staining to be present at a much higher prevalence on the BAL CD8(+)D(b)NP(366)(+) set. The unpredicted finding, however, was that mRNA expression for 75% of the 90 genes was lower in T cells from the BAL than from the spleen. Apparently, the localization of virus-specific CD8(+) T cells to the site of virus-induced pathology is associated with a narrowing, or "focusing," of gene expression that favors enhanced effector function in the damaged, "high-antigen load" environment of the pneumonic lung.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Gene Expression Regulation/immunology , Influenza A virus/genetics , Influenza A virus/immunology , Orthomyxoviridae Infections/immunology , Animals , Bronchoalveolar Lavage Fluid/cytology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/physiology , Cell Differentiation , Female , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Orthomyxoviridae Infections/genetics , Pneumonia, Viral/genetics , Pneumonia, Viral/immunology , T-Lymphocytes, Cytotoxic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...