Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
2.
J Hered ; 111(1): 21-32, 2020 02 05.
Article in English | MEDLINE | ID: mdl-31723957

ABSTRACT

The Hawai'ian honeycreepers (drepanids) are a classic example of adaptive radiation: they adapted to a variety of novel dietary niches, evolving a wide range of bill morphologies. Here we investigated genomic diversity, demographic history, and genes involved in bill morphology phenotypes in 2 honeycreepers: the 'akiapola'au (Hemignathus wilsoni) and the Hawai'i 'amakihi (Chlorodrepanis virens). The 'akiapola'au is an endangered island endemic, filling the "woodpecker" niche by using a unique bill morphology, while the Hawai'i 'amakihi is a dietary generalist common on the islands of Hawai'i and Maui. We de novo sequenced the 'akiapola'au genome and compared it to the previously sequenced 'amakihi genome. The 'akiapola'au is far less heterozygous and has a smaller effective population size than the 'amakihi, which matches expectations due to its smaller census population and restricted ecological niche. Our investigation revealed genomic islands of divergence, which may be involved in the honeycreeper radiation. Within these islands of divergence, we identified candidate genes (including DLK1, FOXB1, KIF6, MAML3, PHF20, RBP1, and TIMM17A) that may play a role in honeycreeper adaptations. The gene DLK1, previously shown to influence Darwin's finch bill size, may be related to honeycreeper bill morphology evolution, while the functions of the other candidates remain unknown.


Subject(s)
Adaptation, Biological , Genetic Speciation , Passeriformes/genetics , Animals , Ecosystem , Evolution, Molecular , Female , Genetic Variation , Genome , Male , Molecular Sequence Annotation , Passeriformes/anatomy & histology
3.
Blood ; 133(22): 2401-2412, 2019 05 30.
Article in English | MEDLINE | ID: mdl-30975638

ABSTRACT

Refractory or relapsed diffuse large B-cell lymphoma (DLBCL) often associates with the activated B-cell-like (ABC) subtype and genetic alterations that drive constitutive NF-κB activation and impair B-cell terminal differentiation. Here, we show that DNA damage response by p53 is a central mechanism suppressing the pathogenic cooperation of IKK2ca-enforced canonical NF-κB and impaired differentiation resulting from Blimp1 loss in ABC-DLBCL lymphomagenesis. We provide evidences that the interplay between these genetic alterations and the tumor microenvironment select for additional molecular addictions that promote lymphoma progression, including aberrant coexpression of FOXP1 and the B-cell mutagenic enzyme activation-induced deaminase, and immune evasion through major histocompatibility complex class II downregulation, PD-L1 upregulation, and T-cell exhaustion. Consistently, PD-1 blockade cooperated with anti-CD20-mediated B-cell cytotoxicity, promoting extended T-cell reactivation and antitumor specificity that improved long-term overall survival in mice. Our data support a pathogenic cooperation among NF-κB-driven prosurvival, genetic instability, and immune evasion mechanisms in DLBCL and provide preclinical proof of concept for including PD-1/PD-L1 blockade in combinatorial immunotherapy for ABC-DLBCL.


Subject(s)
B-Lymphocytes/immunology , B7-H1 Antigen/immunology , Gene Expression Regulation, Neoplastic , Lymphocyte Activation , Lymphoma, Large B-Cell, Diffuse/immunology , Programmed Cell Death 1 Receptor/immunology , Tumor Escape , Tumor Suppressor Protein p53/immunology , Animals , B-Lymphocytes/pathology , B7-H1 Antigen/genetics , Female , Humans , Immunotherapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Large B-Cell, Diffuse/therapy , Male , Mice , Mice, Transgenic , Programmed Cell Death 1 Receptor/genetics , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Suppressor Protein p53/genetics
4.
Am J Respir Crit Care Med ; 197(12): 1552-1564, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29509491

ABSTRACT

RATIONALE: Albuterol, a bronchodilator medication, is the first-line therapy for asthma worldwide. There are significant racial/ethnic differences in albuterol drug response. OBJECTIVES: To identify genetic variants important for bronchodilator drug response (BDR) in racially diverse children. METHODS: We performed the first whole-genome sequencing pharmacogenetics study from 1,441 children with asthma from the tails of the BDR distribution to identify genetic association with BDR. MEASUREMENTS AND MAIN RESULTS: We identified population-specific and shared genetic variants associated with BDR, including genome-wide significant (P < 3.53 × 10-7) and suggestive (P < 7.06 × 10-6) loci near genes previously associated with lung capacity (DNAH5), immunity (NFKB1 and PLCB1), and ß-adrenergic signaling (ADAMTS3 and COX18). Functional analyses of the BDR-associated SNP in NFKB1 revealed potential regulatory function in bronchial smooth muscle cells. The SNP is also an expression quantitative trait locus for a neighboring gene, SLC39A8. The lack of other asthma study populations with BDR and whole-genome sequencing data on minority children makes it impossible to perform replication of our rare variant associations. Minority underrepresentation also poses significant challenges to identify age-matched and population-matched cohorts of sufficient sample size for replication of our common variant findings. CONCLUSIONS: The lack of minority data, despite a collaboration of eight universities and 13 individual laboratories, highlights the urgent need for a dedicated national effort to prioritize diversity in research. Our study expands the understanding of pharmacogenetic analyses in racially/ethnically diverse populations and advances the foundation for precision medicine in at-risk and understudied minority populations.


Subject(s)
Albuterol/therapeutic use , Asthma/drug therapy , Bronchodilator Agents/therapeutic use , Genome-Wide Association Study , Mexican Americans/genetics , Pharmacogenomic Variants/genetics , Race Factors , Adolescent , Black or African American/genetics , Child , Female , Hispanic or Latino/genetics , Humans , Male , Polymorphism, Single Nucleotide , United States
5.
Immunity ; 45(3): 497-512, 2016 09 20.
Article in English | MEDLINE | ID: mdl-27637145

ABSTRACT

During the humoral immune response, B cells undergo a dramatic change in phenotype to enable antibody affinity maturation in germinal centers (GCs). Using genome-wide chromosomal conformation capture (Hi-C), we found that GC B cells undergo massive reorganization of the genomic architecture that encodes the GC B cell transcriptome. Coordinate expression of genes that specify the GC B cell phenotype-most prominently BCL6-was achieved through a multilayered chromatin reorganization process involving (1) increased promoter connectivity, (2) formation of enhancer networks, (3) 5' to 3' gene looping, and (4) merging of gene neighborhoods that share active epigenetic marks. BCL6 was an anchor point for the formation of GC-specific gene and enhancer loops on chromosome 3. Deletion of a GC-specific, highly interactive locus control region upstream of Bcl6 abrogated GC formation in mice. Thus, large-scale and multi-tiered genomic three-dimensional reorganization is required for coordinate expression of phenotype-driving gene sets that determine the unique characteristics of GC B cells.


Subject(s)
Antibody Affinity/immunology , B-Lymphocytes/immunology , Genome/immunology , Germinal Center/immunology , Locus Control Region/immunology , Animals , Antibody Formation/immunology , Chromosomes, Human, Pair 3/immunology , Epigenesis, Genetic/immunology , Humans , Immunity, Humoral/immunology , Mice , Promoter Regions, Genetic/immunology , Proto-Oncogene Proteins c-bcl-6/immunology
6.
Curr Opin Immunol ; 25(3): 339-46, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23725655

ABSTRACT

The BCL6 oncogenic repressor is a master regulator of humoral immunity and B-cell lymphoma survival. Whereas much research has focused on its regulation and function in germinal center B-cells, its role in other mature lymphoid cell compartments is less clear. A novel role for BCL6 in follicular T helper cell development was recently uncovered. The latest discoveries reveal that BCL6 is also an important regulator of other specialized helper T-cell subsets within germinal centers, pre-germinal center events, and peripheral T-cell effector functions. Here, we review newly discovered roles for BCL6 in lymphocyte subsets residing within and outside of germinal centers, and discuss their implications with respect to the molecular mechanisms of BCL6 regulation and potential links to B and T-cell lymphomas.


Subject(s)
Lymphocytes/immunology , Lymphoma, B-Cell/immunology , Lymphoma, T-Cell/immunology , Proto-Oncogene Proteins c-bcl-6/immunology , Animals , Germinal Center/immunology , Humans , Transcription, Genetic
7.
Cancer Cell ; 23(5): 677-92, 2013 May 13.
Article in English | MEDLINE | ID: mdl-23680150

ABSTRACT

The EZH2 histone methyltransferase is highly expressed in germinal center (GC) B cells and targeted by somatic mutations in B cell lymphomas. Here, we find that EZH2 deletion or pharmacologic inhibition suppresses GC formation and functions. EZH2 represses proliferation checkpoint genes and helps establish bivalent chromatin domains at key regulatory loci to transiently suppress GC B cell differentiation. Somatic mutations reinforce these physiological effects through enhanced silencing of EZH2 targets. Conditional expression of mutant EZH2 in mice induces GC hyperplasia and accelerated lymphomagenesis in cooperation with BCL2. GC B cell (GCB)-type diffuse large B cell lymphomas (DLBCLs) are mostly addicted to EZH2 but not the more differentiated activated B cell (ABC)-type DLBCLs, thus clarifying the therapeutic scope of EZH2 targeting.


Subject(s)
B-Lymphocytes/metabolism , Cell Transformation, Neoplastic/genetics , Germinal Center/metabolism , Mutation , Polycomb Repressive Complex 2/physiology , Animals , Cell Differentiation , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein , Gene Deletion , Gene Expression Regulation, Neoplastic , Germinal Center/drug effects , Histones/metabolism , Methylation , Mice , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/physiology
8.
Blood ; 121(21): 4311-20, 2013 May 23.
Article in English | MEDLINE | ID: mdl-23580662

ABSTRACT

B-cell maturation and germinal center (GC) formation are dependent on the interplay between BCL6 and other transcriptional regulators. FOXP1 is a transcription factor that regulates early B-cell development, but whether it plays a role in mature B cells is unknown. Analysis of human tonsillar B-cell subpopulations revealed that FOXP1 shows the opposite expression pattern to BCL6, suggesting that FOXP1 regulates the transition from resting follicular B cell to activated GC B cell. Chromatin immunoprecipitation-on-chip and gene expression assays on B cells indicated that FOXP1 acts as a transcriptional activator and repressor of genes involved in the GC reaction, half of which are also BCL6 targets. To study FOXP1 function in vivo, we developed transgenic mice expressing human FOXP1 in lymphoid cells. These mice exhibited irregular formation of splenic GCs, showing a modest increase in naïve and marginal-zone B cells and a significant decrease in GC B cells. Furthermore, aberrant expression of FOXP1 impaired transcription of noncoding γ1 germline transcripts and inhibited efficient class switching to the immunoglobulin G1 isotype. These studies show that FOXP1 is physiologically downregulated in GC B cells and that aberrant expression of FOXP1 impairs mechanisms triggered by B-cell activation, potentially contributing to B-cell lymphomagenesis.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Forkhead Transcription Factors/metabolism , Germinal Center/cytology , Lymphoma/immunology , Repressor Proteins/metabolism , Animals , Cell Differentiation/immunology , Cell Line , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Down-Regulation/immunology , Forkhead Transcription Factors/immunology , Germinal Center/immunology , Humans , Lymphoma/metabolism , Mice , Mice, Transgenic , Palatine Tonsil/cytology , Proto-Oncogene Proteins c-bcl-6 , Repressor Proteins/immunology , Transcriptional Activation/immunology
9.
Transcription ; 3(3): 130-45, 2012.
Article in English | MEDLINE | ID: mdl-22771948

ABSTRACT

The methylation of histones is a fundamental epigenetic process regulating gene expression programs in mammalian cells. Dysregulated patterns of histone methylation are directly implicated in malignant transformation. Here, we report the unexpected finding that the invasive extracellular matrix degrading endoglycosidase heparanase enters the nucleus of activated human T lymphocytes and regulates the transcription of a cohort of inducible immune response genes by controlling histone H3 methylation patterns. It was found that nuclear heparanase preferentially associates with euchromatin. Genome-wide ChIP-on-chip analyses showed that heparanase is recruited to both the promoter and transcribed regions of a distinct cohort of transcriptionally active genes. Knockdown and overexpression of the heparanase gene also showed that chromatin-bound heparanase is a prerequisite for the transcription of a subset of inducible immune response genes in activated T cells. Furthermore, the actions of heparanase seem to influence gene transcription by associating with the demethylase LSD1, preventing recruitment of the methylase MLL and thereby modifying histone H3 methylation patterns. These data indicate that heparanase belongs to an emerging class of proteins that play an important role in regulating transcription in addition to their well-recognized extra-nuclear functions.


Subject(s)
Chromatin/metabolism , Glucuronidase/metabolism , Histones/metabolism , T-Lymphocytes/metabolism , Transcriptional Activation , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , Epigenesis, Genetic , Fluorescent Antibody Technique , Glucuronidase/genetics , Histone Demethylases/genetics , Histone Demethylases/metabolism , Histones/genetics , Humans , Methylation , Mice , Mice, Inbred C57BL , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction
10.
Mol Cell ; 41(6): 704-19, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21419345

ABSTRACT

Studies in yeast demonstrate that signaling kinases have a surprisingly active role in the nucleus, where they tether to chromatin and modulate gene expression programs. Despite these seminal studies, the nuclear mechanism of how signaling kinases control transcription of mammalian genes is in its infancy. Here, we provide evidence for a hitherto unknown function of protein kinase C-theta (PKC-θ), which physically associates with the regulatory regions of inducible immune response genes in human T cells. Chromatin-anchored PKC-θ forms an active nuclear complex by interacting with RNA polymerase II, the histone kinase MSK-1, and the adaptor molecule 14-3-3ζ. ChIP-on-chip reveals that PKC-θ binds to promoters and transcribed regions of genes, as well as to microRNA promoters that are crucial for cytokine regulation. Our results provide a molecular explanation for the role of PKC-θ not only in normal T cell function, but also in circumstances of its ectopic expression in cancer.


Subject(s)
Chromatin/metabolism , Gene Expression Regulation , Isoenzymes/metabolism , MicroRNAs/metabolism , Protein Kinase C/metabolism , T-Lymphocytes/physiology , Cell Line, Tumor , Cell Nucleus/metabolism , Humans , Interleukin-2/genetics , Isoenzymes/genetics , Jurkat Cells , MicroRNAs/genetics , Promoter Regions, Genetic , Protein Kinase C/genetics , Protein Kinase C-theta , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , T-Lymphocytes/cytology , Transcription, Genetic
11.
Cancer Cell ; 17(4): 400-11, 2010 Apr 13.
Article in English | MEDLINE | ID: mdl-20385364

ABSTRACT

The BCL6 transcriptional repressor is the most frequently involved oncogene in diffuse large B cell lymphoma (DLBCL). We combined computer-aided drug design with functional assays to identify low-molecular-weight compounds that bind to the corepressor binding groove of the BCL6 BTB domain. One such compound disrupted BCL6/corepressor complexes in vitro and in vivo, and was observed by X-ray crystallography and NMR to bind the critical site within the BTB groove. This compound could induce expression of BCL6 target genes and kill BCL6-positive DLBCL cell lines. In xenotransplantation experiments, the compound was nontoxic and potently suppressed DLBCL tumors in vivo. The compound also killed primary DLBCLs from human patients.


Subject(s)
Lymphoma, Large B-Cell, Diffuse/pathology , Proto-Oncogene Proteins/antagonists & inhibitors , Repressor Proteins/antagonists & inhibitors , Animals , Cell Division , Cell Survival/drug effects , Crystallography, X-Ray , Humans , Lymphoma, Large B-Cell, Diffuse/drug therapy , Mice , Mice, Inbred C57BL , Models, Molecular , Protein Conformation , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/toxicity , Repressor Proteins/chemistry , Repressor Proteins/genetics , Repressor Proteins/toxicity , Transcription, Genetic , Zinc Fingers
12.
Nat Med ; 15(12): 1369-76, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19966776

ABSTRACT

We report that heat shock protein 90 (Hsp90) inhibitors selectively kill diffuse large B cell lymphomas (DLBCLs) that depend on the BCL-6 transcriptional repressor. We found that endogenous Hsp90 interacts with BCL-6 in DLBCL cells and can stabilize BCL-6 mRNA and protein. Hsp90 formed a complex with BCL-6 at its target promoters, and Hsp90 inhibitors derepressed BCL-6 target genes. A stable mutant of BCL-6 rescued DLBCL cells from Hsp90 inhibitor-induced apoptosis. BCL-6 and Hsp90 were almost invariantly coexpressed in the nuclei of primary DLBCL cells, suggesting that their interaction is relevant in this disease. We examined the pharmacokinetics, toxicity and efficacy of PU-H71, a recently developed purine-derived Hsp90 inhibitor. PU-H71 preferentially accumulated in lymphomas compared to normal tissues and selectively suppressed BCL-6-dependent DLBCLs in vivo, inducing reactivation of key BCL-6 target genes and apoptosis. PU-H71 also induced cell death in primary human DLBCL specimens.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Lymphoma, B-Cell/metabolism , DNA-Binding Proteins/metabolism , Humans , Lymphoma, B-Cell/pathology , Protein Binding , Proto-Oncogene Proteins c-bcl-6
13.
Genome Biol ; 10(10): R107, 2009.
Article in English | MEDLINE | ID: mdl-19807913

ABSTRACT

BACKGROUND: Specific chromatin characteristics, especially the modification status of the core histone proteins, are associated with active and inactive genes. There is growing evidence that genes that respond to environmental or developmental signals may possess distinct chromatin marks. Using a T cell model and both genome-wide and gene-focused approaches, we examined the chromatin characteristics of genes that respond to T cell activation. RESULTS: To facilitate comparison of genes with similar basal expression levels, we used expression-profiling data to bin genes according to their basal expression levels. We found that inducible genes in the lower basal expression bins, especially rapidly induced primary response genes, were more likely than their non-responsive counterparts to display the histone modifications of active genes, have RNA polymerase II (Pol II) at their promoters and show evidence of ongoing basal elongation. There was little or no evidence for the presence of active chromatin marks in the absence of promoter Pol II on these inducible genes. In addition, we identified a subgroup of genes with active promoter chromatin marks and promoter Pol II but no evidence of elongation. Following T cell activation, we find little evidence for a major shift in the active chromatin signature around inducible gene promoters but many genes recruit more Pol II and show increased evidence of elongation. CONCLUSIONS: These results suggest that the majority of inducible genes are primed for activation by having an active chromatin signature and promoter Pol II with or without ongoing elongation.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Chromatin/metabolism , Gene Expression Regulation , Acetylation , Base Composition/genetics , Chromatin Immunoprecipitation , CpG Islands/genetics , Histones/metabolism , Humans , Lymphocyte Activation/genetics , Methylation , Promoter Regions, Genetic , Protein Binding , RNA Polymerase II/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...