Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Cancer Immunol Immunother ; 72(2): 339-350, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35881197

ABSTRACT

BACKGROUND: Adjuvant immune checkpoint blockade (ICB) following chemoradiotherapy and adding ICB to chemotherapy have been key advances for stages III-IV non-small cell lung cancer (NSCLC) treatment. However, known biomarkers like PD-L1 are not consistently indicative of ICB response. Other markers within the tumor immune microenvironment (TIME) may better reflect ICB response and/or resistance mechanisms, but an understanding of how TIMEs differ between stage III and IV NSCLC has not been explored. METHODS: Real-world data from unresectable, stage III-IV, non-squamous, pretreatment NSCLCs (stage III n = 106, stage IV n = 285) were retrospectively analyzed. PD-L1 immunohistochemistry (IHC) was compared to CD274 gene expression. Then, differential gene expression levels, pathway enrichment, and immune infiltrate between stages were calculated from whole-transcriptome RNA-seq. Analyses were stratified by EGFR status. RESULTS: PD-L1 IHC and CD274 expression in tumor cells were highly correlated (n = 295, P < 2.2e-16, ⍴ = 0.74). CTLA4 expression was significantly increased in stage III tumors (P = 1.32e-04), while no differences were observed for other ICB-related genes. Metabolic pathway activity was significantly enriched in stage IV tumors (P = 0.004), whereas several immune-related KEGG pathways were enriched in stage III. Stage IV tumors had significantly increased macrophage infiltration (P = 0.0214), and stage III tumors had a significantly higher proportion of CD4 + T cells (P = 0.017). CD4 + T cells were also relatively more abundant in EGFR-mutant tumors vs. wild-type (P = 0.0081). CONCLUSION: Directly comparing the TIMEs of stage III and IV NSCLC, these results carry implications for further studies of ICB response in non-resectable stage III NSCLC and guide further research of prognostic biomarkers and therapeutic targets.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , B7-H1 Antigen/metabolism , Retrospective Studies , Biomarkers , Tumor Microenvironment , Immune Checkpoint Inhibitors/therapeutic use , ErbB Receptors , Biomarkers, Tumor
2.
J Allergy Clin Immunol ; 144(5): 1391-1401.e10, 2019 11.
Article in English | MEDLINE | ID: mdl-31401285

ABSTRACT

OBJECTIVES: Amish children raised on traditional farms have lower atopy and asthma risk than Hutterite children raised on modern farms. In our previous study we established that the Amish environment affects the innate immune response to decrease asthma and atopy risk. Here we investigated T-cell phenotypes in the same Amish and Hutterite children as in our earlier study to elucidate how this altered innate immunity affects adaptive T cells. METHODS: Blood was collected from 30 Amish and 30 Hutterite age- and sex-matched children; cells were cryopreserved until analysis. Flow cytometry was used to analyze cell subsets. Atopy was determined based on allergen-specific and total IgE levels. RESULTS: Children exposed to Amish farms had increased activated regulatory CD4+ T-cell phenotypes, whereas conventional CD4 T cells expressed lower levels of costimulation molecules and other activation markers. The increase in numbers of circulating activated regulatory CD4+ T cells was associated with an increase in inhibitory receptors on monocytes in Amish, but not Hutterite, children. Strikingly, the Amish children had a higher proportion of CD28null CD8 T cells than the Hutterite children (P < .0001, nonparametric t test), a difference that remained even after accounting for the effects of age and sex (conditional log regression exponential ß = 1.08, P = .0053). The proportion of these cells correlated with high T-cell IFN-γ production (rs = 0.573, P = .005) and low serum IgE levels (rs = -0.417, P = .025). Furthermore, CD28null CD8 T-cell numbers were increased in Amish children, with high expression of the innate genes TNF and TNF-α-induced protein 3 (TNFAIP3) in peripheral blood leukocytes. CONCLUSION: Amish children's blood leukocytes are not only altered in their innate immune status but also have distinct T-cell phenotypes that are often associated with increased antigen exposure.


Subject(s)
Amish , Ethnicity , Hypersensitivity, Immediate/immunology , Immunoglobulin E/blood , T-Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Adaptive Immunity , Adolescent , Allergens/immunology , Cells, Cultured , Child , Environmental Exposure/adverse effects , Female , Humans , Immunophenotyping , Male , Phenotype , Tumor Necrosis Factor alpha-Induced Protein 3/genetics
3.
Genes Immun ; 20(6): 462-472, 2019 07.
Article in English | MEDLINE | ID: mdl-29977032

ABSTRACT

The low affinity Fcγ receptor, FcγRIIA, harbors a common missense mutation, rs1801274 (G>A, Arg131His) that modifies binding affinity to human IgG2 and mouse IgG1 antibodies and is associated with increased risk of autoimmune disease. Despite the important role of the Arg131His variant, little is understood about heterozygous genotype effects on global gene expression and cytokine production during an FcγR-dependent response. To address this gap in knowledge, we treated human whole-blood samples from 130 individuals with mouse IgG1 anti-CD3 and anti-CD28 antibodies and characterized the genome-wide gene expression profiles and cytokine production among individuals stratified by rs1801274 genotype. Our analysis revealed that the levels of four cytokines (IFNγ, IL-12, IL-2, TNFα) and global gene expression patterns differed between all three genotype classes. Surprisingly, the heterozygotes showed suboptimal T cell activation compared to cells from individuals homozygous for the higher-affinity FcγRIIA allele (GG; Arg/Arg). The results of this study demonstrate that IgG response varies among all rs1801274 genotype classes and results in profound differences in both cytokine responses and gene expression patterns in blood leukocytes. Because even heterozygotes showed dampened global responses, our data may provide insight into the heterogeneity of outcomes in cytokine release assays and immunotherapy efficacy.


Subject(s)
Antibodies/pharmacology , CD28 Antigens/immunology , CD3 Complex/immunology , Leukocytes/immunology , Polymorphism, Genetic , Receptors, IgG/genetics , Adolescent , Adult , Aged , Alleles , CD28 Antigens/antagonists & inhibitors , CD3 Complex/antagonists & inhibitors , Child , Genotype , Heterozygote , Homozygote , Humans , Interferon-gamma/blood , Interferon-gamma/metabolism , Interleukin-12/blood , Interleukin-12/metabolism , Interleukin-2/blood , Interleukin-2/metabolism , Leukocytes/metabolism , Middle Aged , T-Lymphocytes/metabolism , Transcriptome/immunology , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism , Young Adult
4.
N Engl J Med ; 375(5): 411-421, 2016 Aug 04.
Article in English | MEDLINE | ID: mdl-27518660

ABSTRACT

BACKGROUND: The Amish and Hutterites are U.S. agricultural populations whose lifestyles are remarkably similar in many respects but whose farming practices, in particular, are distinct; the former follow traditional farming practices whereas the latter use industrialized farming practices. The populations also show striking disparities in the prevalence of asthma, and little is known about the immune responses underlying these disparities. METHODS: We studied environmental exposures, genetic ancestry, and immune profiles among 60 Amish and Hutterite children, measuring levels of allergens and endotoxins and assessing the microbiome composition of indoor dust samples. Whole blood was collected to measure serum IgE levels, cytokine responses, and gene expression, and peripheral-blood leukocytes were phenotyped with flow cytometry. The effects of dust extracts obtained from Amish and Hutterite homes on immune and airway responses were assessed in a murine model of experimental allergic asthma. RESULTS: Despite the similar genetic ancestries and lifestyles of Amish and Hutterite children, the prevalence of asthma and allergic sensitization was 4 and 6 times as low in the Amish, whereas median endotoxin levels in Amish house dust was 6.8 times as high. Differences in microbial composition were also observed in dust samples from Amish and Hutterite homes. Profound differences in the proportions, phenotypes, and functions of innate immune cells were also found between the two groups of children. In a mouse model of experimental allergic asthma, the intranasal instillation of dust extracts from Amish but not Hutterite homes significantly inhibited airway hyperreactivity and eosinophilia. These protective effects were abrogated in mice that were deficient in MyD88 and Trif, molecules that are critical in innate immune signaling. CONCLUSIONS: The results of our studies in humans and mice indicate that the Amish environment provides protection against asthma by engaging and shaping the innate immune response. (Funded by the National Institutes of Health and others.).


Subject(s)
Agriculture , Asthma/immunology , Environmental Exposure , Immunity, Innate , Adaptor Proteins, Vesicular Transport/deficiency , Adolescent , Animals , Asthma/epidemiology , Child , Christianity , Cross-Sectional Studies , Cytokines/blood , Disease Models, Animal , Dust/immunology , Female , Gene Expression , Humans , Immunity, Innate/genetics , Immunity, Innate/immunology , Immunoglobulin E/blood , Leukocyte Count , Leukocytes/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Animal , Myeloid Differentiation Factor 88/deficiency , Prevalence
5.
J Allergy Clin Immunol ; 142(3): 749-764.e3, 2018 09.
Article in English | MEDLINE | ID: mdl-29307657

ABSTRACT

Chromosome 17q12-21 remains the most highly replicated and significant asthma locus. Genotypes in the core region defined by the first genome-wide association study correlate with expression of 2 genes, ORM1-like 3 (ORMDL3) and gasdermin B (GSDMB), making these prime candidate asthma genes, although recent studies have implicated gasdermin A (GSDMA) distal to and post-GPI attachment to proteins 3 (PGAP3) proximal to the core region as independent loci. We review 10 years of studies on the 17q12-21 locus and suggest that genotype-specific risks for asthma at the proximal and distal loci are not specific to early-onset asthma and mediated by PGAP3, ORMDL3, and/or GSDMA expression. We propose that the weak and inconsistent associations of 17q single nucleotide polymorphisms with asthma in African Americans is due to the high frequency of some 17q alleles, the breakdown of linkage disequilibrium on African-derived chromosomes, and possibly different early-life asthma endotypes in these children. Finally, the inconsistent association between asthma and gene expression levels in blood or lung cells from older children and adults suggests that genotype effects may mediate asthma risk or protection during critical developmental windows and/or in response to relevant exposures in early life. Thus studies of young children and ethnically diverse populations are required to fully understand the relationship between genotype and asthma phenotype and the gene regulatory architecture at this locus.


Subject(s)
Asthma/genetics , Chromosomes, Human, Pair 17 , Asthma/ethnology , Chromatin , DNA Methylation , Humans , Phenotype , Quantitative Trait Loci
6.
N Engl J Med ; 368(15): 1398-407, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23534543

ABSTRACT

BACKGROUND: Both genetic variation at the 17q21 locus and virus-induced respiratory wheezing illnesses are associated with the development of asthma. Our aim was to determine the effects of these two factors on the risk of asthma in the Childhood Origins of Asthma (COAST) and the Copenhagen Prospective Study on Asthma in Childhood (COPSAC) birth cohorts. METHODS: We tested genotypes at the 17q21 locus for associations with asthma and with human rhinovirus (HRV) and respiratory syncytial virus (RSV) wheezing illnesses and tested for interactions between 17q21 genotypes and HRV and RSV wheezing illnesses with respect to the risk of asthma. Finally, we examined genotype-specific expression of 17q21 genes in unstimulated and HRV-stimulated peripheral-blood mononuclear cells (PBMCs). RESULTS: The 17q21 variants were associated with HRV wheezing illnesses in early life, but not with RSV wheezing illnesses. The associations of 17q21 variants with asthma were restricted to children who had had HRV wheezing illnesses, resulting in a significant interaction effect with respect to the risk of asthma. Moreover, the expression levels of ORMDL3 and of GSDMB were significantly increased in HRV-stimulated PBMCs, as compared with unstimulated PBMCs. The expression of these genes was associated with 17q21 variants in both conditions, although the increase with exposure to HRV was not genotype-specific. CONCLUSIONS: Variants at the 17q21 locus were associated with asthma in children who had had HRV wheezing illnesses and with expression of two genes at this locus. The expression levels of both genes increased in response to HRV stimulation, although the relative increase was not associated with the 17q21 genotypes. (Funded by the National Institutes of Health.).


Subject(s)
Asthma/genetics , Common Cold/complications , Respiratory Sounds , Rhinovirus , Asthma/virology , Child , Chromosomes, Human, Pair 17 , DNA/isolation & purification , Gene Expression , Genetic Predisposition to Disease , Genotype , Humans , Leukocytes, Mononuclear , Polymorphism, Single Nucleotide , RNA/isolation & purification , Respiratory Sounds/genetics , Respiratory Syncytial Virus Infections/complications , Respiratory Syncytial Viruses , Risk
7.
JCO Precis Oncol ; 7: e2300378, 2023 Sep.
Article in English | MEDLINE | ID: mdl-38061006

ABSTRACT

PURPOSE: Homologous recombination deficiency (HRD) is a well-described phenotype of some prostate cancers; however, current biomarkers for HRD are imperfect and rely on detection of single gene alterations in the homologous recombination repair (HRR) pathway, which may not capture the complexity of HRD biology. RNA signature-based methods of HRD identification present a potentially dynamic assessment of the HRD phenotype; however, its relationship with HRR gene alterations is not well characterized in prostate cancer. METHODS: A HRD assay on the basis of an RNA signature associated with biallelic BRCA1/2 loss was applied to a retrospective cohort study of 985 men with prostate cancer analyzed on the Tempus xT platform. HRD status was defined by a binary threshold on a continuous scale. RESULTS: In this cohort, of the 126 (13%) patients found to be HRD+ by RNA signature (HRD-RNA+), 100 (79%) had no coexisting HRR gene alteration. Among samples with biallelic BRCA1/2 loss, 78% (7/9) were classified as HRD-RNA+, while 8% (2/25) of samples with BRCA1/2 monoallelic loss were HRD-RNA+. Biallelic and monoallelic ATM loss exhibited HRD-RNA+ at a lower prevalence: 6.7% (1/15) and 7.1% (1/14), respectively, compared with HRD-RNA+ prevalence among samples without any HRR gene loss (13%; 100/782). HRD-RNA+ was associated with a significantly higher prevalence of TP53 and AR gene alterations relative to HRD-RNA- after correction for multiple comparisons, 59% versus 39% (q = 0.003) and 23% versus 12% (q = 0.024), respectively. CONCLUSION: Use of an RNA-based HRD signature significantly expands the fraction of patients with prostate cancer who may derive benefit from poly (ADP-ribose) polymerase inhibitors (PARPis) compared with using HRR gene mutations alone. Further studies are needed to evaluate functional HRD significance and inform future usage as a predictive biomarker for PARPi selection.


Subject(s)
BRCA1 Protein , Prostatic Neoplasms , Male , Humans , BRCA1 Protein/genetics , Recombinational DNA Repair/genetics , Homologous Recombination/genetics , Retrospective Studies , BRCA2 Protein/genetics , Prostatic Neoplasms/genetics , Poly(ADP-ribose) Polymerase Inhibitors
8.
Nat Commun ; 13(1): 4053, 2022 07 13.
Article in English | MEDLINE | ID: mdl-35831288

ABSTRACT

The efficacy of immune checkpoint blockade (ICB) varies greatly among metastatic non-small cell lung cancer (NSCLC) patients. Loss of heterozygosity at the HLA-I locus (HLA-LOH) has been identified as an important immune escape mechanism. However, despite HLA-I disruptions in their tumor, many patients have durable ICB responses. Here we seek to identify HLA-I-independent features associated with ICB response in NSCLC. We use single-cell profiling to identify tumor-infiltrating, clonally expanded CD4+ T cells that express a canonical cytotoxic gene program and NSCLC cells with elevated HLA-II expression. We postulate cytotoxic CD4+ T cells mediate anti-tumor activity via HLA-II on tumor cells and augment HLA-I-dependent cytotoxic CD8+ T cell interactions to drive ICB response in NSCLC. We show that integrating tumor extrinsic cytotoxic gene expression with tumor mutational burden is associated with longer time to progression in a real-world cohort of 123 NSCLC patients treated with ICB regimens, including those with HLA-LOH.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Biomarkers, Tumor/genetics , CD8-Positive T-Lymphocytes , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Humans , Immunotherapy , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics
9.
Sci Rep ; 11(1): 1107, 2021 01 13.
Article in English | MEDLINE | ID: mdl-33441806

ABSTRACT

Sex-specific differences in prevalence are well documented for many common, complex diseases, especially for immune-mediated diseases, yet the precise mechanisms through which factors associated with biological sex exert their effects throughout life are not well understood. We interrogated sex-specific transcriptional responses of peripheral blood leukocytes (PBLs) to innate immune stimulation by lipopolysaccharide (LPS) in 46 male and 66 female members of the Hutterite community, who practice a communal lifestyle. We identified 1217 autosomal and 54 X-linked genes with sex-specific responses to LPS, as well as 71 autosomal and one X-linked sex-specific expression quantitative trait loci (eQTLs). Despite a similar proportion of the 15 HLA genes responding to LPS compared to all expressed autosomal genes, there was a significant over-representation of genes with sex by treatment interactions among HLA genes. We also observed an enrichment of sex-specific differentially expressed genes in response to LPS for X-linked genes compared to autosomal genes, suggesting that HLA and X-linked genes may disproportionately contribute to sex disparities in risk for immune-mediated diseases.


Subject(s)
Genes, MHC Class II , Genes, MHC Class I , Genes, X-Linked , Leukocytes/immunology , Leukocytes/metabolism , Sex Characteristics , Transcription, Genetic , Adolescent , Adult , Aged , Aged, 80 and over , Child , Ethnicity , Female , Gene Expression Profiling , Humans , Immunity, Innate , Lipopolysaccharides/immunology , Male , Middle Aged , Quantitative Trait Loci , Young Adult
10.
Nat Commun ; 12(1): 6115, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34675193

ABSTRACT

Genome-wide association studies (GWAS) have implicated the IL33 locus in asthma, but the underlying mechanisms remain unclear. Here, we identify a 5 kb region within the GWAS-defined segment that acts as an enhancer-blocking element in vivo and in vitro. Chromatin conformation capture showed that this 5 kb region loops to the IL33 promoter, potentially regulating its expression. We show that the asthma-associated single nucleotide polymorphism (SNP) rs1888909, located within the 5 kb region, is associated with IL33 gene expression in human airway epithelial cells and IL-33 protein expression in human plasma, potentially through differential binding of OCT-1 (POU2F1) to the asthma-risk allele. Our data demonstrate that asthma-associated variants at the IL33 locus mediate allele-specific regulatory activity and IL33 expression, providing a mechanism through which a regulatory SNP contributes to genetic risk of asthma.


Subject(s)
Asthma/genetics , Enhancer Elements, Genetic , Interleukin-33/genetics , Alleles , Animals , Asthma/metabolism , Chromatin/genetics , Chromatin/metabolism , Female , Genetic Predisposition to Disease , Humans , Interleukin-33/metabolism , Male , Mice, Transgenic , Octamer Transcription Factor-1/genetics , Octamer Transcription Factor-1/metabolism , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Zebrafish
11.
Cell Rep ; 36(4): 109429, 2021 07 27.
Article in English | MEDLINE | ID: mdl-34320344

ABSTRACT

Patient-derived tumor organoids (TOs) are emerging as high-fidelity models to study cancer biology and develop novel precision medicine therapeutics. However, utilizing TOs for systems-biology-based approaches has been limited by a lack of scalable and reproducible methods to develop and profile these models. We describe a robust pan-cancer TO platform with chemically defined media optimized on cultures acquired from over 1,000 patients. Crucially, we demonstrate tumor genetic and transcriptomic concordance utilizing this approach and further optimize defined minimal media for organoid initiation and propagation. Additionally, we demonstrate a neural-network-based high-throughput approach for label-free, light-microscopy-based drug assays capable of predicting patient-specific heterogeneity in drug responses with applicability across solid cancers. The pan-cancer platform, molecular data, and neural-network-based drug assay serve as resources to accelerate the broad implementation of organoid models in precision medicine research and personalized therapeutic profiling programs.


Subject(s)
Neoplasms/pathology , Organoids/pathology , Precision Medicine , Cell Proliferation , Drug Screening Assays, Antitumor , Female , Fluorescence , Genomics , HLA Antigens/genetics , Humans , Loss of Heterozygosity , Male , Middle Aged , Models, Biological , Neoplasms/genetics , Neural Networks, Computer , Transcriptome/genetics
12.
PLoS One ; 13(9): e0203906, 2018.
Article in English | MEDLINE | ID: mdl-30204804

ABSTRACT

Genomic imprinting is the phenomena that leads to silencing of one copy of a gene inherited from a specific parent. Mutations in imprinted regions have been involved in diseases showing parent of origin effects. Identifying genes with evidence of parent of origin expression patterns in family studies allows the detection of more subtle imprinting. Here, we use allele specific expression in lymphoblastoid cell lines from 306 Hutterites related in a single pedigree to provide formal evidence for parent of origin effects. We take advantage of phased genotype data to assign parent of origin to RNA-seq reads in individuals with gene expression data. Our approach identified known imprinted genes, two putative novel imprinted genes, PXDC1 and PWAR6, and 14 genes with asymmetrical parent of origin gene expression. We used gene expression in peripheral blood leukocytes (PBL) to validate our findings, and then confirmed imprinting control regions (ICRs) using DNA methylation levels in the PBLs.


Subject(s)
Founder Effect , Genomic Imprinting , Cell Line , DNA Methylation , Ethnicity/genetics , Female , Gene Expression , Gene Expression Profiling , Haplotypes , Humans , Lymphocytes/metabolism , Male , Pedigree , Polymorphism, Single Nucleotide , Sequence Analysis, RNA
13.
PLoS One ; 6(8): e23121, 2011.
Article in English | MEDLINE | ID: mdl-21858005

ABSTRACT

The retina of the mosquito Aedes aegypti can be divided into four regions based on the non-overlapping expression of a UV sensitive Aaop8 rhodopsin and a long wavelength sensitive Aaop2 type rhodopsin in the R7 photoreceptors. We show here that another rhodopsin, Aaop9, is expressed in all R7 photoreceptors and a subset of R8 photoreceptors. In the dorsal region, Aaop9 is expressed in both the cell body and rhabdomere of R7 and R8 cells. In other retinal regions Aaop9 is expressed only in R7 cells, being localized to the R7 rhabdomere in the central and ventral regions and in both the cell body and rhabdomere within the ventral stripe. Within the dorsal-central transition area ommatidia do not show a strict pairing of R7-R8 cell types. Thus, Aaop9 is coexpressed in the two classes of R7 photoreceptors previously distinguished by the non-overlapping expression of Aaop8 and Aaop2 rhodopsins. Electroretinogram analysis of transgenic Drosophila shows that Aaop9 is a short wavelength rhodopsin with an optimal response to 400-450 nm light. The coexpressed Aaop2 rhodopsin has dual wavelength sensitivity of 500-550 nm and near 350 nm in the UV region. As predicted by the spectral properties of each rhodopsin, Drosophila photoreceptors expressing both Aaop9 and Aaop2 rhodopsins exhibit a uniform sensitivity across the broad 350-550 nm light range. We propose that rhodopsin coexpression is an adaptation within the R7 cells to improve visual function in the low-light environments in which Ae. aegypti is active.


Subject(s)
Aedes/metabolism , Photoreceptor Cells, Invertebrate/metabolism , Retina/metabolism , Rhodopsin/metabolism , Aedes/cytology , Aedes/genetics , Amino Acid Sequence , Animals , Animals, Genetically Modified , Blotting, Western , Drosophila/cytology , Drosophila/genetics , Drosophila/metabolism , Fluorescent Antibody Technique , Molecular Sequence Data , Photoreceptor Cells, Invertebrate/cytology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Retina/cytology , Rhodopsin/genetics , Species Specificity , Spectrophotometry
SELECTION OF CITATIONS
SEARCH DETAIL