Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Cell ; 168(1-2): 59-72.e13, 2017 Jan 12.
Article in English | MEDLINE | ID: mdl-28065413

ABSTRACT

Chromosomal translocations of the mixed-lineage leukemia (MLL) gene with various partner genes result in aggressive leukemia with dismal outcomes. Despite similar expression at the mRNA level from the wild-type and chimeric MLL alleles, the chimeric protein is more stable. We report that UBE2O functions in regulating the stability of wild-type MLL in response to interleukin-1 signaling. Targeting wild-type MLL degradation impedes MLL leukemia cell proliferation, and it downregulates a specific group of target genes of the MLL chimeras and their oncogenic cofactor, the super elongation complex. Pharmacologically inhibiting this pathway substantially delays progression, and it improves survival of murine leukemia through stabilizing wild-type MLL protein, which displaces the MLL chimera from some of its target genes and, therefore, relieves the cellular oncogenic addiction to MLL chimeras. Stabilization of MLL provides us with a paradigm in the development of therapies for aggressive MLL leukemia and perhaps for other cancers caused by translocations.


Subject(s)
Leukemia, Biphenotypic, Acute/drug therapy , Leukemia, Biphenotypic, Acute/metabolism , Proteolysis/drug effects , Animals , Disease Models, Animal , Histone-Lysine N-Methyltransferase/metabolism , Humans , Interleukin-1/metabolism , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Interleukin-1 Receptor-Associated Kinases/metabolism , Mice , Mice, Inbred C57BL , Myeloid-Lymphoid Leukemia Protein/metabolism , Ubiquitin-Conjugating Enzymes
2.
Proc Natl Acad Sci U S A ; 121(29): e2313370121, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-38985769

ABSTRACT

Heat Shock Factor 1 (HSF1) is best known as the master transcriptional regulator of the heat-shock response (HSR), a conserved adaptive mechanism critical for protein homeostasis (proteostasis). Combining a genome-wide RNAi library with an HSR reporter, we identified Jumonji domain-containing protein 6 (JMJD6) as an essential mediator of HSF1 activity. In follow-up studies, we found that JMJD6 is itself a noncanonical transcriptional target of HSF1 which acts as a critical regulator of proteostasis. In a positive feedback circuit, HSF1 binds and promotes JMJD6 expression, which in turn reduces heat shock protein 70 (HSP70) R469 monomethylation to disrupt HSP70-HSF1 repressive complexes resulting in enhanced HSF1 activation. Thus, JMJD6 is intricately wired into the proteostasis network where it plays a critical role in cellular adaptation to proteotoxic stress.


Subject(s)
HSP70 Heat-Shock Proteins , Heat Shock Transcription Factors , Heat-Shock Response , Jumonji Domain-Containing Histone Demethylases , Proteostasis , Humans , Heat Shock Transcription Factors/metabolism , Heat Shock Transcription Factors/genetics , Heat-Shock Response/physiology , Jumonji Domain-Containing Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , HSP70 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/genetics , Proteostasis/physiology , Feedback, Physiological , Adaptation, Physiological , HEK293 Cells , Proteotoxic Stress
3.
Genes Dev ; 33(1-2): 61-74, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30573454

ABSTRACT

Chromosomal translocations of the Mixed-lineage leukemia 1 (MLL1) gene generate MLL chimeras that drive the pathogenesis of acute myeloid and lymphoid leukemia. The untranslocated MLL1 is a substrate for proteolytic cleavage by the endopeptidase threonine aspartase 1 (taspase1); however, the biological significance of MLL1 cleavage by this endopeptidase remains unclear. Here, we demonstrate that taspase1-dependent cleavage of MLL1 results in the destabilization of MLL. Upon loss of taspase1, MLL1 association with chromatin is markedly increased due to the stabilization of its unprocessed version, and this stabilization of the uncleaved MLL1 can result in the displacement of MLL chimeras from chromatin in leukemic cells. Casein kinase II (CKII) phosphorylates MLL1 proximal to the taspase1 cleavage site, facilitating its cleavage, and pharmacological inhibition of CKII blocks taspase1-dependent MLL1 processing, increases MLL1 stability, and results in the displacement of the MLL chimeras from chromatin. Accordingly, inhibition of CKII in a MLL-AF9 mouse model of leukemia delayed leukemic progression in vivo. This study provides insights into the direct regulation of the stability of MLL1 through its cleavage by taspase1, which can be harnessed for targeted therapeutic approaches for the treatment of aggressive leukemia as the result of MLL translocations.


Subject(s)
Endopeptidases/metabolism , Leukemia/therapy , Myeloid-Lymphoid Leukemia Protein/genetics , Animals , Chromatin/metabolism , Disease Models, Animal , Disease Progression , Endopeptidases/genetics , Enzyme Inhibitors/pharmacology , Gene Knockout Techniques , HCT116 Cells , HEK293 Cells , Humans , Leukemia/enzymology , Leukemia/genetics , MCF-7 Cells , Mice , Myeloid-Lymphoid Leukemia Protein/metabolism , Protein Stability , Survival Analysis
4.
J Immunol ; 211(9): 1376-1384, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37702745

ABSTRACT

IFN-γ-producing invariant NKT (iNKT)1 cells are lipid-reactive innate-like lymphocytes that are resident in the thymus and peripheral tissues where they protect against pathogenic infection. The thymic functions of iNKT1 cells are not fully elucidated, but subsets of thymic iNKT cells modulate CD8 T cell, dendritic cell, B cell, and thymic epithelial cell numbers or function. In this study, we show that a subset of murine thymic iNKT1 cells required TGF-ß-induced signals for their postselection development, to maintain hallmark TGF-ß-induced genes, and for expression of the adhesion receptors CD49a and CD103. However, the residency-associated receptor CD69 was not TGF-ß signaling-dependent. Recently described CD244+ c2 thymic iNKT1 cells, which produce IFN-γ without exogenous stimulation and have NK-like characteristics, reside in this TGF-ß-responsive population. Liver and spleen iNKT1 cells do not share this TGF-ß gene signature, but nonetheless TGF-ß impacts liver iNKT1 cell phenotype and function. Our findings provide insight into the heterogeneity of mechanisms guiding iNKT1 cell development in different tissues and suggest a close association between a subset of iNKT1 cells and TGF-ß-producing cells in the thymus that support their development.


Subject(s)
Natural Killer T-Cells , Transforming Growth Factor beta , Animals , Mice , CD8-Positive T-Lymphocytes , Cell Differentiation/genetics , Mice, Inbred C57BL , Thymus Gland , Transforming Growth Factor beta/metabolism
5.
Mol Cell ; 65(3): 460-475.e6, 2017 Feb 02.
Article in English | MEDLINE | ID: mdl-28157506

ABSTRACT

The spatiotemporal regulation of gene expression is central for cell-lineage specification during embryonic development and is achieved through the combinatorial action of transcription factors/co-factors and epigenetic states at cis-regulatory elements. Here, we show that in addition to implementing H3K4me3 at promoters of bivalent genes, Mll2 (KMT2B)/COMPASS can also implement H3K4me3 at a subset of non-TSS regulatory elements, a subset of which shares epigenetic signatures of active enhancers. Our mechanistic studies reveal that association of Mll2's CXXC domain with CpG-rich regions plays an instrumental role for chromatin targeting and subsequent implementation of H3K4me3. Although Mll2/COMPASS is required for H3K4me3 implementation on thousands of loci, generation of catalytically mutant MLL2/COMPASS demonstrated that H3K4me3 implemented by this enzyme was essential for expression of a subset of genes, including those functioning in the control of transcriptional programs during embryonic development. Our findings suggest that not all H3K4 trimethylations implemented by MLL2/COMPASS are functionally equivalent.


Subject(s)
DNA-Binding Proteins/metabolism , Fibroblasts/cytology , Germ Cells/cytology , Histones/metabolism , Mouse Embryonic Stem Cells/cytology , Myeloid-Lymphoid Leukemia Protein/metabolism , Neoplasm Proteins/metabolism , Animals , Cell Differentiation , Cells, Cultured , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Genetic Speciation , Germ Cells/metabolism , HEK293 Cells , Histone-Lysine N-Methyltransferase , Humans , Mice , Mouse Embryonic Stem Cells/metabolism , Myeloid-Lymphoid Leukemia Protein/chemistry , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Promoter Regions, Genetic , Protein Domains
6.
Genes Dev ; 31(8): 787-801, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28487406

ABSTRACT

The homeotic (Hox) genes are highly conserved in metazoans, where they are required for various processes in development, and misregulation of their expression is associated with human cancer. In the developing embryo, Hox genes are activated sequentially in time and space according to their genomic position within Hox gene clusters. Accumulating evidence implicates both enhancer elements and noncoding RNAs in controlling this spatiotemporal expression of Hox genes, but disentangling their relative contributions is challenging. Here, we identify two cis-regulatory elements (E1 and E2) functioning as shadow enhancers to regulate the early expression of the HoxA genes. Simultaneous deletion of these shadow enhancers in embryonic stem cells leads to impaired activation of HoxA genes upon differentiation, while knockdown of a long noncoding RNA overlapping E1 has no detectable effect on their expression. Although MLL/COMPASS (complex of proteins associated with Set1) family of histone methyltransferases is known to activate transcription of Hox genes in other contexts, we found that individual inactivation of the MLL1-4/COMPASS family members has little effect on early Hox gene activation. Instead, we demonstrate that SET1A/COMPASS is required for full transcriptional activation of multiple Hox genes but functions independently of the E1 and E2 cis-regulatory elements. Our results reveal multiple regulatory layers for Hox genes to fine-tune transcriptional programs essential for development.


Subject(s)
Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Genes, Homeobox/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Animals , Chromatin/genetics , Embryonic Stem Cells/cytology , Gene Deletion , Histone Methyltransferases , Mice , Protein Binding , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Transcriptional Activation/genetics
7.
J Biol Chem ; 299(7): 104867, 2023 07.
Article in English | MEDLINE | ID: mdl-37247756

ABSTRACT

Age-associated bone marrow changes include myeloid skewing and mutations that lead to clonal hematopoiesis. Molecular mechanisms for these events are ill defined, but decreased expression of Irf8/Icsbp (interferon regulatory factor 8/interferon consensus sequence binding protein) in aging hematopoietic stem cells may contribute. Irf8 functions as a leukemia suppressor for chronic myeloid leukemia, and young Irf8-/- mice have neutrophilia with progression to acute myeloid leukemia (AML) with aging. Irf8 is also required to terminate emergency granulopoiesis during the innate immune response, suggesting this may be the physiologic counterpart to leukemia suppression by this transcription factor. Identifying Irf8 effectors may define mediators of both events and thus contributors to age-related bone marrow disorders. In this study, we identified RASSF5 (encoding Nore1) as an Irf8 target gene and investigated the role of Nore1 in hematopoiesis. We found Irf8 activates RASSF5 transcription and increases Nore1a expression during emergency granulopoiesis. Similar to Irf8-/- mice, we found that young Rassf5-/- mice had increased neutrophils and progressed to AML with aging. We identified enhanced DNA damage, excess clonal hematopoiesis, and a distinct mutation profile in hematopoietic stem cells from aging Rassf5-/- mice compared with wildtype. We found sustained emergency granulopoiesis in Rassf5-/- mice, with repeated episodes accelerating AML, also similar to Irf8-/- mice. Identifying Nore1a downstream from Irf8 defines a pathway involved in leukemia suppression and the innate immune response and suggests a novel molecular mechanism contributing to age-related clonal myeloid disorders.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Leukemia, Myeloid, Acute , Animals , Mice , Cell Lineage , Clonal Hematopoiesis , Hematopoiesis , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
8.
J Virol ; 97(7): e0065223, 2023 07 27.
Article in English | MEDLINE | ID: mdl-37310263

ABSTRACT

HIV-1 (HIV) infects CD4+ T cells, the gradual depletion of which can lead to AIDS in the absence of antiretroviral therapy (ART). Some cells, however, survive HIV infection and persist as part of the latently infected reservoir that causes recurrent viremia after ART cessation. Improved understanding of the mechanisms of HIV-mediated cell death could lead to a way to clear the latent reservoir. Death induced by survival gene elimination (DISE), an RNA interference (RNAi)-based mechanism, kills cells through short RNAs (sRNAs) with toxic 6-mer seeds (positions 2 to 7 of sRNA). These toxic seeds target the 3' untranslated region (UTR) of mRNAs, decreasing the expression of hundreds of genes critical for cell survival. In most cells under normal conditions, highly expressed cell-encoded nontoxic microRNAs (miRNAs) block access of toxic sRNAs to the RNA-induced silencing complex (RISC) that mediates RNAi, promoting cell survival. HIV has been shown to inhibit the biogenesis of host miRNAs in multiple ways. We now report that HIV infection of cells deficient in miRNA expression or function results in enhanced RISC loading of an HIV-encoded miRNA HIV-miR-TAR-3p, which can kill cells by DISE through a noncanonical (positions 3 to 8) 6-mer seed. In addition, cellular RISC-bound sRNAs shift to lower seed viability. This also occurs after latent HIV provirus reactivation in J-Lat cells, suggesting independence of permissiveness of cells to viral infection. More precise targeting of the balance between protective and cytotoxic sRNAs could provide new avenues to explore novel cell death mechanisms that could be used to kill latent HIV. IMPORTANCE Several mechanisms by which initial HIV infection is cytotoxic to infected cells have been reported and involve various forms of cell death. Characterizing the mechanisms underlying the long-term survival of certain T cells that become persistent provirus reservoirs is critical to developing a cure. We recently discovered death induced by survival gene elimination (DISE), an RNAi-based mechanism of cell death whereby toxic short RNAs (sRNAs) containing 6-mer seed sequences (exerting 6-mer seed toxicity) targeting essential survival genes are loaded into RNA-induced silencing complex (RISC) complexes, resulting in inescapable cell death. We now report that HIV infection in cells with low miRNA expression causes a shift of mostly cellular RISC-bound sRNAs to more toxic seeds. This could prime cells to DISE and is further enhanced by the viral microRNA (miRNA) HIV-miR-TAR-3p, which carries a toxic noncanonical 6-mer seed. Our data provide multiple new avenues to explore novel cell death mechanisms that could be used to kill latent HIV.


Subject(s)
HIV Infections , HIV-1 , MicroRNAs , Humans , HIV-1/physiology , Virus Latency/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , RNA-Induced Silencing Complex/metabolism
9.
Genes Dev ; 30(1): 92-101, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26728555

ABSTRACT

Genomic imprinting is a critical developmental process characteristic of parent of origin-specific gene expression. It is well accepted that differentially DNA-methylated regions (DMRs) and enhancers are two major classes of cis-elements determining parent of origin-specific gene expression, with each recruiting different sets of transcription factors. Previously, we identified the AF4/FMR2 (AFF) family protein AFF3 within the transcription elongation complex SEC-L3. Here, we report that AFF3 can specifically bind both gametic DMRs (gDMRs) and enhancers within imprinted loci in an allele-specific manner. We identify the molecular regulators involved in the recruitment of AFF3 to gDMRs and provide mechanistic insight into the requirement of AFF3 at an enhancer for the expression of an ∼200-kb polycistronic transcript within the imprinted Dlk1-Dio3 locus. Our data suggest that the heterochromatic environment at the gDMR reinforces silencing of its related enhancer by controlling the binding and activity of AFF3 in an allele-specific manner. In summary, this study provides molecular details about the regulation of dosage-critical imprinted gene expression through the regulated binding of the transcription elongation factor AFF3 between a DMR and an enhancer.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation , Genomic Imprinting/genetics , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Alleles , Animals , Calcium-Binding Proteins , Cell Line , Chromatin Immunoprecipitation , DNA Methylation , Embryonic Stem Cells , Gene Silencing , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Sequence Analysis, DNA
10.
Nat Immunol ; 12(12): 1212-20, 2011 Oct 30.
Article in English | MEDLINE | ID: mdl-22037603

ABSTRACT

During B lymphopoiesis, recombination of the locus encoding the immunoglobulin κ-chain complex (Igk) requires expression of the precursor to the B cell antigen receptor (pre-BCR) and escape from signaling via the interleukin 7 receptor (IL-7R). By activating the transcription factor STAT5, IL-7R signaling maintains proliferation and represses Igk germline transcription by unknown mechanisms. We demonstrate that a STAT5 tetramer bound the Igk intronic enhancer (E(κi)), which led to recruitment of the histone methyltransferase Ezh2. Ezh2 marked trimethylation of histone H3 at Lys27 (H3K27me3) throughout the κ-chain joining region (J(κ)) to the κ-chain constant region (C(κ)). In the absence of Ezh2, IL-7 failed to repress Igk germline transcription. H3K27me3 modifications were lost after termination of IL-7R-STAT5 signaling, and the transcription factor E2A bound E(κi), which resulted in acquisition of H3K4me1 and acetylated histone H4 (H4Ac). Genome-wide analyses showed a STAT5 tetrameric binding motif associated with transcriptional repression. Our data demonstrate how IL-7R signaling represses Igk germline transcription and provide a general model for STAT5-mediated epigenetic transcriptional repression.


Subject(s)
Epigenesis, Genetic , Histone-Lysine N-Methyltransferase/metabolism , Immunoglobulins/genetics , STAT5 Transcription Factor/metabolism , Animals , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites/genetics , Binding, Competitive , COS Cells , Chlorocebus aethiops , Cluster Analysis , Enhancer of Zeste Homolog 2 Protein , Gene Expression Profiling , Gene Expression Regulation , Histones/metabolism , Methylation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Polycomb Repressive Complex 2 , Protein Binding , Receptors, Interleukin-7/metabolism , Signal Transduction
11.
PLoS Comput Biol ; 18(3): e1010022, 2022 03.
Article in English | MEDLINE | ID: mdl-35358200

ABSTRACT

microRNAs (miRNAs) are (18-22nt long) noncoding short (s)RNAs that suppress gene expression by targeting the 3' untranslated region of target mRNAs. This occurs through the seed sequence located in position 2-7/8 of the miRNA guide strand, once it is loaded into the RNA induced silencing complex (RISC). G-rich 6mer seed sequences can kill cells by targeting C-rich 6mer seed matches located in genes that are critical for cell survival. This results in induction of Death Induced by Survival gene Elimination (DISE), through a mechanism we have called 6mer seed toxicity. miRNAs are often quantified in cells by aligning the reads from small (sm)RNA sequencing to the genome. However, the analysis of any smRNA Seq data set for predicted 6mer seed toxicity requires an alternative workflow, solely based on the exact position 2-7 of any short (s)RNA that can enter the RISC. Therefore, we developed SPOROS, a semi-automated pipeline that produces multiple useful outputs to predict and compare 6mer seed toxicity of cellular sRNAs, regardless of their nature, between different samples. We provide two examples to illustrate the capabilities of SPOROS: Example one involves the analysis of RISC-bound sRNAs in a cancer cell line (either wild-type or two mutant lines unable to produce most miRNAs). Example two is based on a publicly available smRNA Seq data set from postmortem brains (either from normal or Alzheimer's patients). Our methods (found at https://github.com/ebartom/SPOROS and at Code Ocean: https://doi.org/10.24433/CO.1732496.v1) are designed to be used to analyze a variety of smRNA Seq data in various normal and disease settings.


Subject(s)
MicroRNAs , 3' Untranslated Regions , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Messenger/genetics , Seeds/genetics , Sequence Analysis, RNA/methods
12.
J Immunol ; 204(7): 1760-1769, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32094206

ABSTRACT

Lymphocyte lineage specification and commitment requires the activation of lineage-specific genes and repression of alternative lineage genes, respectively. The mechanisms governing alternative lineage gene repression and commitment in lymphocytes are largely unknown. In this study, we demonstrate that Ezh2, which represses gene expression through methylation of histone 3 lysine 27, was essential for repression of numerous genes, including genes encoding innate lymphocyte transcription factors, specifically in murine B lymphocyte progenitors, but these cells maintained their B lymphocyte identity. However, adult Ezh2-deficient B lymphocytes expressed Lin28b, which encodes an RNA-binding protein associated with fetal hematopoietic gene expression programs, and these cells acquired a fetal B-1 lymphocyte phenotype in vitro and in vivo. Therefore, Ezh2 coordinates the repression of multiple gene programs in B lymphocytes and maintains the adult B-2 cell fate.


Subject(s)
B-Lymphocytes/immunology , Enhancer of Zeste Homolog 2 Protein/immunology , Immunity, Innate/immunology , Precursor Cells, B-Lymphoid/immunology , Transcription, Genetic/immunology , Animals , Cell Differentiation/immunology , Cell Lineage/immunology , Gene Expression/immunology , Histones/immunology , Methylation , Mice , Mice, Inbred C57BL , RNA-Binding Proteins/immunology , Transcription Factors/immunology
13.
J Cell Sci ; 132(12)2019 06 19.
Article in English | MEDLINE | ID: mdl-31118235

ABSTRACT

Receptor of activated protein C kinase 1 (RACK1) is a highly conserved eukaryotic protein that regulates several aspects of mRNA translation; yet, how it does so, remains poorly understood. Here we show that, although RACK1 consists largely of conserved ß-propeller domains that mediate binding to several other proteins, a short interconnecting loop between two of these blades varies across species to control distinct RACK1 functions during translation. Mutants and chimeras revealed that the amino acid composition of the loop is optimized to regulate interactions with eIF6, a eukaryotic initiation factor that controls 60S biogenesis and 80S ribosome assembly. Separately, phylogenetics revealed that, despite broad sequence divergence of the loop, there is striking conservation of negatively charged residues amongst protists and dicot plants, which is reintroduced to mammalian RACK1 by poxviruses through phosphorylation. Although both charged and uncharged loop mutants affect eIF6 interactions, only a negatively charged plant - but not uncharged yeast or human loop - enhances translation of mRNAs with adenosine-rich 5' untranslated regions (UTRs). Our findings reveal how sequence plasticity within the RACK1 loop confers multifunctionality in translational control across species.


Subject(s)
Neoplasm Proteins/metabolism , Protein Binding , Receptors for Activated C Kinase/metabolism , Ribosomes/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Eukaryotic Initiation Factors/metabolism , GTP-Binding Proteins/metabolism , Humans , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/metabolism
14.
EMBO Rep ; 19(3)2018 03.
Article in English | MEDLINE | ID: mdl-29440125

ABSTRACT

Trinucleotide repeat (TNR) expansions in the genome cause a number of degenerative diseases. A prominent TNR expansion involves the triplet CAG in the huntingtin (HTT) gene responsible for Huntington's disease (HD). Pathology is caused by protein and RNA generated from the TNR regions including small siRNA-sized repeat fragments. An inverse correlation between the length of the repeats in HTT and cancer incidence has been reported for HD patients. We now show that siRNAs based on the CAG TNR are toxic to cancer cells by targeting genes that contain long reverse complementary TNRs in their open reading frames. Of the 60 siRNAs based on the different TNRs, the six members in the CAG/CUG family of related TNRs are the most toxic to both human and mouse cancer cells. siCAG/CUG TNR-based siRNAs induce cell death in vitro in all tested cancer cell lines and slow down tumor growth in a preclinical mouse model of ovarian cancer with no signs of toxicity to the mice. We propose to explore TNR-based siRNAs as a novel form of anticancer reagents.


Subject(s)
Huntingtin Protein/genetics , Neoplasms/genetics , RNA, Small Interfering/pharmacology , Trinucleotide Repeats/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Disease Models, Animal , Humans , Huntingtin Protein/antagonists & inhibitors , Huntington Disease/genetics , Huntington Disease/pathology , Mice , Neoplasms/pathology , Neoplasms/therapy , Open Reading Frames , RNA, Small Interfering/genetics , Trinucleotide Repeat Expansion/genetics , Trinucleotide Repeats/drug effects
15.
J Immunol ; 198(12): 4682-4691, 2017 06 15.
Article in English | MEDLINE | ID: mdl-28490575

ABSTRACT

The histone methyltransferase EZH2 is required for B and T cell development; however, the molecular mechanisms underlying this requirement remain elusive. In a murine model of lymphoid-specific EZH2 deficiency we found that EZH2 was required for proper development of adaptive, but not innate, lymphoid cells. In adaptive lymphoid cells EZH2 prevented the premature expression of Cdkn2a and the consequent stabilization of p53, an effector of the pre-Ag receptor checkpoints. Deletion of Cdkn2a in EZH2-deficient lymphocytes prevented p53 stabilization, extended lymphocyte survival, and restored differentiation resulting in the generation of mature B and T lymphocytes. Our results uncover a crucial role for EZH2 in adaptive lymphocytes to control the developmental timing of effectors of the pre-Ag receptor checkpoints.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/metabolism , Receptors, Antigen/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Adaptive Immunity , Animals , B-Lymphocytes/immunology , Cyclin-Dependent Kinase Inhibitor p16/deficiency , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Enhancer of Zeste Homolog 2 Protein/deficiency , Enhancer of Zeste Homolog 2 Protein/genetics , Gene Expression Regulation , Genes, p53 , Killer Cells, Natural/immunology , Lymphopoiesis , Mice , Receptors, Antigen/genetics , Receptors, Antigen/immunology
16.
Am J Respir Cell Mol Biol ; 59(2): 145-157, 2018 08.
Article in English | MEDLINE | ID: mdl-29624415

ABSTRACT

Since the first publications coining the term RNA-seq (RNA sequencing) appeared in 2008, the number of publications containing RNA-seq data has grown exponentially, hitting an all-time high of 2,808 publications in 2016 (PubMed). With this wealth of RNA-seq data being generated, it is a challenge to extract maximal meaning from these datasets, and without the appropriate skills and background, there is risk of misinterpretation of these data. However, a general understanding of the principles underlying each step of RNA-seq data analysis allows investigators without a background in programming and bioinformatics to critically analyze their own datasets as well as published data. Our goals in the present review are to break down the steps of a typical RNA-seq analysis and to highlight the pitfalls and checkpoints along the way that are vital for bench scientists and biomedical researchers performing experiments that use RNA-seq.


Subject(s)
Data Analysis , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Sequence Analysis, RNA , Animals , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing/methods , Male , Mice, Inbred C57BL , Quality Control , Sequence Analysis, RNA/methods , Software , Transcriptome/genetics
17.
Blood ; 121(6): 975-83, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23212519

ABSTRACT

Loss of chromosome 7 and del(7q) [-7/del(7q)] are recurring cytogenetic abnormalities in hematologic malignancies, including acute myeloid leukemia and therapy-related myeloid neoplasms, and associated with an adverse prognosis. Despite intensive effort by many laboratories, the putative myeloid tumor suppressor(s) on chromosome 7 has not yet been identified.We performed transcriptome sequencing and SNP array analysis on de novo and therapy-related myeloid neoplasms, half with -7/del(7q). We identified a 2.17-Mb commonly deleted segment on chromosome band 7q22.1 containing CUX1, a gene encoding a homeodomain-containing transcription factor. In 1 case, CUX1 was disrupted by a translocation, resulting in a loss-of-function RNA fusion transcript. CUX1 was the most significantly differentially expressed gene within the commonly deleted segment and was expressed at haploinsufficient levels in -7/del(7q) leukemias. Haploinsufficiency of the highly conserved ortholog, cut, led to hemocyte overgrowth and tumor formation in Drosophila melanogaster. Similarly, haploinsufficiency of CUX1 gave human hematopoietic cells a significant engraftment advantage on transplantation into immunodeficient mice. Within the RNA-sequencing data, we identified a CUX1-associated cell cycle transcriptional gene signature, suggesting that CUX1 exerts tumor suppressor activity by regulating proliferative genes. These data identify CUX1 as a conserved, haploinsufficient tumor suppressor frequently deleted in myeloid neoplasms.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 7/genetics , Homeodomain Proteins/genetics , Leukemia, Myeloid/genetics , Nuclear Proteins/genetics , Repressor Proteins/genetics , Acute Disease , Animals , Blotting, Western , Cell Line, Tumor , Drosophila melanogaster/genetics , Gene Expression Profiling , Haploinsufficiency , HeLa Cells , Homeodomain Proteins/metabolism , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , K562 Cells , Leukemia, Myeloid/metabolism , Leukemia, Myeloid/pathology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Nuclear Proteins/metabolism , RNA Interference , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors , Translocation, Genetic , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , U937 Cells , Xenograft Model Antitumor Assays
18.
Leukemia ; 38(3): 491-501, 2024 03.
Article in English | MEDLINE | ID: mdl-38155245

ABSTRACT

T lymphocyte acute lymphoblastic leukemia (T-ALL) is frequently associated with increased expression of the E protein transcription factor inhibitors TAL1 and LYL1. In mouse models, ectopic expression of TAL1 or LYL1 in T cell progenitors, or inactivation of E2A, is sufficient to predispose mice to develop T-ALL. How E2A suppresses thymocyte transformation is currently unknown. Here, we show that early deletion of E2a, prior to the DN3 stage, was required for robust leukemogenesis and was associated with alterations in thymus cellularity, T cell differentiation, and gene expression in immature CD4+CD8+ thymocytes. Introduction of wild-type thymocytes into mice with early deletion of E2a prevented leukemogenesis, or delayed disease onset, and impacted the expression of multiple genes associated with transformation and genome instability. Our data indicate that E2A suppresses leukemogenesis by promoting T cell development and enforcing inter-thymocyte competition, a mechanism that is emerging as a safeguard against thymocyte transformation. These studies have implications for understanding how multiple essential regulators of T cell development suppress T-ALL and support the hypothesis that thymocyte competition suppresses leukemogenesis.


Subject(s)
Precursor Cell Lymphoblastic Leukemia-Lymphoma , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Mice , Animals , Transcription Factors/genetics , Thymocytes/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Thymus Gland/metabolism , Cell Differentiation/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics
19.
Cell Rep ; 43(2): 113680, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38241148

ABSTRACT

Extracellular vesicles (EVs) facilitate intercellular communication by transferring cargo between cells in a variety of tissues. However, how EVs achieve cell-type-specific intercellular communication is still largely unknown. We found that Notch1 and Notch2 proteins are expressed on the surface of neuronal EVs that have been generated in response to neuronal excitatory synaptic activity. Notch ligands bind these EVs on the neuronal plasma membrane, trigger their internalization, activate the Notch signaling pathway, and drive the expression of Notch target genes. The generation of these neuronal EVs requires the endosomal sorting complex required for transport-associated protein Alix. Adult Alix conditional knockout mice have reduced hippocampal Notch signaling activation and glutamatergic synaptic protein expression. Thus, EVs facilitate neuron-to-neuron communication via the Notch receptor-ligand system in the brain.


Subject(s)
Extracellular Vesicles , Neurons , Animals , Mice , Ligands , Protein Transport , Signal Transduction , Mice, Knockout
20.
Nat Commun ; 15(1): 264, 2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38238311

ABSTRACT

Alzheimer's disease (AD) is characterized by progressive neurodegeneration, but the specific events that cause cell death remain poorly understood. Death Induced by Survival gene Elimination (DISE) is a cell death mechanism mediated by short (s) RNAs acting through the RNA-induced silencing complex (RISC). DISE is thus a form of RNA interference, in which G-rich 6mer seed sequences in the sRNAs (position 2-7) target hundreds of C-rich 6mer seed matches in genes essential for cell survival, resulting in the activation of cell death pathways. Here, using Argonaute precipitation and RNAseq (Ago-RP-Seq), we analyze RISC-bound sRNAs to quantify 6mer seed toxicity in several model systems. In mouse AD models and aging brain, in induced pluripotent stem cell-derived neurons from AD patients, and in cells exposed to Aß42 oligomers, RISC-bound sRNAs show a shift to more toxic 6mer seeds compared to controls. In contrast, in brains of "SuperAgers", humans over age 80 who have superior memory performance, RISC-bound sRNAs are shifted to more nontoxic 6mer seeds. Cells depleted of nontoxic sRNAs are sensitized to Aß42-induced cell death, and reintroducing nontoxic RNAs is protective. Altogether, the correlation between DISE and Aß42 toxicity suggests that increasing the levels of nontoxic miRNAs in the brain or blocking the activity of toxic RISC-bound sRNAs could ameliorate neurodegeneration.


Subject(s)
Alzheimer Disease , MicroRNAs , Mice , Animals , Humans , Aged, 80 and over , Alzheimer Disease/genetics , MicroRNAs/genetics , RNA-Induced Silencing Complex/genetics , RNA Interference , Aging/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL