Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
G3 (Bethesda) ; 14(4)2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38366796

ABSTRACT

Aging is a multifactorial process that disturbs homeostasis, increases disease susceptibility, and ultimately results in death. Although the definitive set of molecular mechanisms responsible for aging remain to be discovered, epigenetic change over time is proving to be a promising piece of the puzzle. Several post-translational histone modifications have been linked to the maintenance of longevity. Here, we focus on lysine-36 of the replication-independent histone protein, H3.3 (H3.3K36). To interrogate the role of this residue in Drosophila developmental gene regulation, we generated a lysine-to-arginine mutant that blocks the activity of its cognate-modifying enzymes. We found that an H3.3BK36R mutation causes a significant reduction in adult lifespan, accompanied by dysregulation of the genomic and transcriptomic architecture. Transgenic co-expression of wild-type H3.3B completely rescues the longevity defect. Because H3.3 is known to accumulate in nondividing tissues, we carried out transcriptome profiling of young vs aged adult fly heads. The data show that loss of H3.3K36 results in age-dependent misexpression of NF-κB and other innate immune target genes, as well as defects in silencing of heterochromatin. We propose H3.3K36 maintains the postmitotic epigenomic landscape, supporting longevity by regulating both pericentric and telomeric retrotransposons and by suppressing aberrant immune signaling.


Subject(s)
Drosophila , Histones , Longevity , Animals , Drosophila/genetics , Drosophila/metabolism , Heterochromatin , Histones/genetics , Histones/metabolism , Longevity/genetics , Lysine/metabolism
2.
Sci Immunol ; 8(88): eadg7597, 2023 10 20.
Article in English | MEDLINE | ID: mdl-37831759

ABSTRACT

Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS) caused by CNS-infiltrating leukocytes, including TH17 cells that are critical mediators of disease pathogenesis. Although targeting leukocyte trafficking is effective in treating autoimmunity, there are currently no therapeutic interventions that specifically block encephalitogenic TH17 cell migration. Here, we report integrin α3 as a TH17 cell-selective determinant of pathogenicity in experimental autoimmune encephalomyelitis. CNS-infiltrating TH17 cells express high integrin α3, and its deletion in CD4+ T cells or Il17a fate-mapped cells attenuated disease severity. Mechanistically, integrin α3 enhanced the immunological synapse formation to promote the polarization and proliferation of TH17 cells. Moreover, the transmigration of TH17 cells into the CNS was dependent on integrin α3, and integrin α3 deficiency enhanced the retention of CD4+ T cells in the perivascular space of the blood-brain barrier. Integrin α3-dependent interactions continuously maintain TH17 cell identity and effector function. The requirement of integrin α3 in TH17 cell pathogenicity suggests integrin α3 as a therapeutic target for MS treatment.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Humans , Integrin alpha3 , Neuroinflammatory Diseases , Central Nervous System
3.
Genetics ; 224(4)2023 08 09.
Article in English | MEDLINE | ID: mdl-37279945

ABSTRACT

The chromatin of animal cells contains two types of histones: canonical histones that are expressed during S phase of the cell cycle to package the newly replicated genome, and variant histones with specialized functions that are expressed throughout the cell cycle and in non-proliferating cells. Determining whether and how canonical and variant histones cooperate to regulate genome function is integral to understanding how chromatin-based processes affect normal and pathological development. Here, we demonstrate that variant histone H3.3 is essential for Drosophila development only when canonical histone gene copy number is reduced, suggesting that coordination between canonical H3.2 and variant H3.3 expression is necessary to provide sufficient H3 protein for normal genome function. To identify genes that depend upon, or are involved in, this coordinate regulation we screened for heterozygous chromosome 3 deficiencies that impair development of flies bearing reduced H3.2 and H3.3 gene copy number. We identified two regions of chromosome 3 that conferred this phenotype, one of which contains the Polycomb gene, which is necessary for establishing domains of facultative chromatin that repress master regulator genes during development. We further found that reduction in Polycomb dosage decreases viability of animals with no H3.3 gene copies. Moreover, heterozygous Polycomb mutations result in de-repression of the Polycomb target gene Ubx and cause ectopic sex combs when either canonical or variant H3 gene copy number is reduced. We conclude that Polycomb-mediated facultative heterochromatin function is compromised when canonical and variant H3 gene copy number falls below a critical threshold.


Subject(s)
Drosophila melanogaster , Gene Dosage , Histones , Polycomb-Group Proteins , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Epigenetic Repression , Gene Expression Regulation , Histones/genetics , Histones/metabolism , Larva/genetics , Larva/metabolism , Polycomb-Group Proteins/metabolism , RNA, Messenger/metabolism , Animals
4.
bioRxiv ; 2023 Mar 28.
Article in English | MEDLINE | ID: mdl-37034607

ABSTRACT

The chromatin of animal cells contains two types of histones: canonical histones that are expressed during S phase of the cell cycle to package the newly replicated genome, and variant histones with specialized functions that are expressed throughout the cell cycle and in non-proliferating cells. Determining whether and how canonical and variant histones cooperate to regulate genome function is integral to understanding how chromatin-based processes affect normal and pathological development. Here, we demonstrate that variant histone H3.3 is essential for Drosophila development only when canonical histone gene copy number is reduced, suggesting that coordination between canonical H3.2 and variant H3.3 expression is necessary to provide sufficient H3 protein for normal genome function. To identify genes that depend upon, or are involved in, this coordinate regulation we screened for heterozygous chromosome 3 deficiencies that impair development of flies bearing reduced H3.2 and H3.3 gene copy number. We identified two regions of chromosome 3 that conferred this phenotype, one of which contains the Polycomb gene, which is necessary for establishing domains of facultative chromatin that repress master regulator genes during development. We further found that reduction in Polycomb dosage decreases viability of animals with no H3.3 gene copies. Moreover, heterozygous Polycomb mutations result in de-repression of the Polycomb target gene Ubx and cause ectopic sex combs when either canonical or variant H3 gene copy number is also reduced. We conclude that Polycomb-mediated facultative heterochromatin function is compromised when canonical and variant H3 gene copy number falls below a critical threshold.

5.
Sci Adv ; 9(9): eadf2451, 2023 03.
Article in English | MEDLINE | ID: mdl-36857457

ABSTRACT

Polycomb complexes regulate cell type-specific gene expression programs through heritable silencing of target genes. Trimethylation of histone H3 lysine 27 (H3K27me3) is essential for this process. Perturbation of H3K36 is thought to interfere with H3K27me3. We show that mutants of Drosophila replication-dependent (H3.2K36R) or replication-independent (H3.3K36R) histone H3 genes generally maintain Polycomb silencing and reach later stages of development. In contrast, combined (H3.3K36RH3.2K36R) mutants display widespread Hox gene misexpression and fail to develop past the first larval stage. Chromatin profiling revealed that the H3.2K36R mutation disrupts H3K27me3 levels broadly throughout silenced domains, whereas these regions are mostly unaffected in H3.3K36R animals. Analysis of H3.3 distributions showed that this histone is enriched at presumptive Polycomb response elements located outside of silenced domains but relatively depleted from those inside. We conclude that H3.2 and H3.3 K36 residues collaborate to repress Hox genes using different mechanisms.


Subject(s)
Drosophila Proteins , Histones , Animals , Lysine , Chromatin , Drosophila , Polycomb-Group Proteins
6.
bioRxiv ; 2023 Dec 13.
Article in English | MEDLINE | ID: mdl-38196611

ABSTRACT

Aging is a multifactorial process that disturbs homeostasis, increases disease susceptibility, and ultimately results in death. Although the definitive set of molecular mechanisms responsible for aging remain to be discovered, epigenetic change over time is proving to be a promising piece of the puzzle. Several posttranslational histone modifications (PTMs) have been linked to the maintenance of longevity. Here, we focus on lysine-36 of the replication-independent histone protein, H3.3 (H3.3K36). To interrogate the role of this residue in Drosophila developmental gene regulation, we generated a lysine to arginine mutant that blocks the activity of its cognate modifying enzymes. We found that an H3.3BK36R mutation causes a significant reduction in adult lifespan, accompanied by dysregulation of the genomic and transcriptomic architecture. Transgenic co-expression of wild-type H3.3B completely rescues the longevity defect. Because H3.3 is known to accumulate in non-dividing tissues, we carried out transcriptome profiling of young vs aged adult fly heads. The data show that loss of H3.3K36 results in age-dependent misexpression of NF-κB and other innate immune target genes, as well as defects in silencing of heterochromatin. We propose H3.3K36 maintains the postmitotic epigenomic landscape, supporting longevity by regulating both pericentric and telomeric retrotransposons and by suppressing aberrant immune signaling.

7.
Biol Open ; 11(3)2022 03 15.
Article in English | MEDLINE | ID: mdl-35132432

ABSTRACT

Mature transfer (t)RNAs are generated by multiple RNA processing events, which can include the excision of intervening sequences. The tRNA splicing endonuclease (TSEN) complex is responsible for cleaving these intron-containing pre-tRNA transcripts. In humans, TSEN copurifies with CLP1, an RNA kinase. Despite extensive work on CLP1, its in vivo connection to tRNA splicing remains unclear. Interestingly, mutations in CLP1 or TSEN genes cause neurological diseases in humans that are collectively termed Pontocerebellar Hypoplasia (PCH). In mice, loss of Clp1 kinase activity results in premature death, microcephaly and progressive loss of motor function. To determine if similar phenotypes are observed in Drosophila, we characterized mutations in crowded-by-cid (cbc), the CLP1 ortholog, as well as in the fly ortholog of human TSEN54. Analyses of organismal viability, larval locomotion and brain size revealed that mutations in both cbc and Tsen54 phenocopy those in mammals in several details. In addition to an overall reduction in brain lobe size, we also found increased cell death in mutant larval brains. Ubiquitous or tissue-specific knockdown of cbc in neurons and muscles reduced viability and locomotor function. These findings indicate that we can successfully model PCH in a genetically-tractable invertebrate.


Subject(s)
Drosophila , RNA Processing, Post-Transcriptional , Animals , Cerebellar Diseases , Drosophila/genetics , Drosophila/metabolism , Mammals/genetics , Mammals/metabolism , Mice , Mutation , Phenotype , RNA, Transfer/genetics , RNA, Transfer/metabolism
8.
Nat Immunol ; 20(5): 663, 2019 May.
Article in English | MEDLINE | ID: mdl-30816306

ABSTRACT

In the version of this article initially published, the top right plot in Figure 4a was aligned incorrectly. The error has been corrected in the HTML and PDF versions of the article. The original and corrected figures are provided in the accompanying Publisher Correction.

9.
Nat Immunol ; 20(1): 73-85, 2019 01.
Article in English | MEDLINE | ID: mdl-30538336

ABSTRACT

γδ T cells that produce the cytokine IL-17 (Tγδ17 cells) are innate-like mediators of immunity that undergo effector programming in the thymus. While regulators of Tγδ17 specialization restricted to various Vγ subsets are known, a commitment factor essential to all Tγδ17 cells has remained undefined. In this study, we identified the transcription factor c-Maf as a universal regulator of Tγδ17 cell differentiation and maintenance. Maf deficiency caused an absolute lineage block at the immature CD24+CD45RBlo γδ thymocyte stage, which revealed a critical checkpoint in the acquisition of effector functions. Here, c-Maf enforced Tγδ17 cell identity by promoting chromatin accessibility and expression of key type 17 program genes, notably Rorc and Blk, while antagonizing the transcription factor TCF1, which promotes interferon-γ-producing γδ T cells (Tγδ1 cells). Furthermore, γδ T cell antigen receptor (γδTCR) signal strength tuned c-Maf expression, which indicates that c-Maf is a core node that connects γδTCR signals to Tγδ17 cell transcriptional programming.


Subject(s)
Interleukin-17/metabolism , Proto-Oncogene Proteins c-maf/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Th17 Cells/physiology , Thymocytes/physiology , Animals , CD24 Antigen/metabolism , Cell Differentiation/genetics , Cell Lineage/genetics , Cells, Cultured , Immunity, Innate , Leukocyte Common Antigens/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Proto-Oncogene Proteins c-maf/genetics , Signal Transduction , src-Family Kinases/genetics
10.
Dev Cell ; 24(6): 623-34, 2013 Mar 25.
Article in English | MEDLINE | ID: mdl-23537633

ABSTRACT

Compartmentalization of RNA biosynthetic factors into nuclear bodies (NBs) is a ubiquitous feature of eukaryotic cells. How NBs initially assemble and ultimately affect gene expression remains unresolved. The histone locus body (HLB) contains factors necessary for replication-coupled histone messenger RNA transcription and processing and associates with histone gene clusters. Using a transgenic assay for ectopic Drosophila HLB assembly, we show that a sequence located between, and transcription from, the divergently transcribed H3-H4 genes nucleates HLB formation and activates other histone genes in the histone gene cluster. In the absence of transcription from the H3-H4 promoter, "proto-HLBs" (containing only a subset of HLB components) form, and the adjacent histone H2a-H2b genes are not expressed. Proto-HLBs also transiently form in mutant embryos with the histone locus deleted. We conclude that HLB assembly occurs through a stepwise process involving stochastic interactions of individual components that localize to a specific sequence in the H3-H4 promoter.


Subject(s)
Drosophila/genetics , Histones/genetics , RNA, Messenger/biosynthesis , Animals , DNA Replication , Drosophila/metabolism , Gene Expression , Histones/metabolism , Promoter Regions, Genetic , RNA, Messenger/genetics , Transcription, Genetic , Transcriptional Activation
11.
RNA ; 17(6): 1132-47, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21525146

ABSTRACT

Metazoan replication-dependent histone mRNAs are the only nonpolyadenylated cellular mRNAs. Formation of the histone mRNA 3' end requires the U7 snRNP, which contains Lsm10 and Lsm11, and FLASH, a processing factor that binds Lsm11. Here, we identify sequences in Drosophila FLASH (dFLASH) that bind Drosophila Lsm11 (dLsm11), allow localization of dFLASH to the nucleus and histone locus body (HLB), and participate in histone pre-mRNA processing in vivo. Amino acids 105-154 of dFLASH bind to amino acids 1-78 of dLsm11. A two-amino acid mutation of dLsm11 that prevents dFLASH binding but does not affect localization of U7 snRNP to the HLB cannot rescue the lethality or histone pre-mRNA processing defects resulting from an Lsm11 null mutation. The last 45 amino acids of FLASH are required for efficient localization to the HLB in Drosophila cultured cells. Removing the first 64 amino acids of FLASH has no effect on processing in vivo. Removal of 13 additional amino acids of dFLASH results in a dominant negative protein that binds Lsm11 but inhibits processing of histone pre-mRNA in vivo. Inhibition requires the Lsm11 binding site, suggesting that the mutant dFLASH protein sequesters the U7 snRNP in an inactive complex and that residues between 64 and 77 of dFLASH interact with a factor required for processing. Together, these studies demonstrate that direct interaction between dFLASH and dLsm11 is essential for histone pre-mRNA processing in vivo and for proper development and viability in flies.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/metabolism , Drosophila/genetics , Histones/genetics , RNA Precursors/metabolism , RNA, Messenger/metabolism , Ribonucleoproteins, Small Nuclear/chemistry , Ribonucleoproteins, Small Nuclear/metabolism , Animals , Binding Sites , Carrier Proteins/genetics , Cells, Cultured , Drosophila/metabolism , Drosophila Proteins/genetics , Histones/metabolism , RNA Processing, Post-Transcriptional , RNA, Heterogeneous Nuclear/genetics , RNA, Heterogeneous Nuclear/metabolism , Ribonucleoprotein, U7 Small Nuclear/genetics , Ribonucleoprotein, U7 Small Nuclear/metabolism , Ribonucleoproteins, Small Nuclear/genetics
12.
PLoS One ; 4(12): e8168, 2009 Dec 04.
Article in English | MEDLINE | ID: mdl-19997601

ABSTRACT

BACKGROUND: Metazoan replication-dependent histone mRNAs terminate in a conserved stem-loop structure rather than a polyA tail. Formation of this unique mRNA 3' end requires Stem-loop Binding Protein (SLBP), which directly binds histone pre-mRNA and stimulates 3' end processing. The 3' end stem-loop is necessary for all aspects of histone mRNA metabolism, including replication coupling, but its importance to organism fitness and genome maintenance in vivo have not been characterized. METHODOLOGY/PRINCIPAL FINDINGS: In Drosophila, disruption of the Slbp gene prevents normal histone pre-mRNA processing and causes histone pre-mRNAs to utilize the canonical 3' end processing pathway, resulting in polyadenylated histone mRNAs that are no longer properly regulated. Here we show that Slbp mutants display genomic instability, including loss of heterozygosity (LOH), increased presence of chromosome breaks, tetraploidy, and changes in position effect variegation (PEV). During imaginal disc growth, Slbp mutant cells show defects in S phase and proliferate more slowly than control cells. CONCLUSIONS/SIGNIFICANCE: These data are consistent with a model in which changing the 3' end of histone mRNA disrupts normal replication-coupled histone mRNA biosynthesis and alters chromatin assembly, resulting in genomic instability, inhibition of cell proliferation, and impaired development.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Genomic Instability/genetics , Histones/metabolism , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional/genetics , RNA-Binding Proteins/metabolism , Animals , Cell Proliferation , Chromosomal Position Effects/genetics , DNA Breaks, Double-Stranded , Euchromatin/metabolism , Genetic Markers , Heterochromatin/metabolism , Loss of Heterozygosity/genetics , Mitosis , Mutation/genetics , Polyploidy , S Phase
13.
Mol Cell ; 28(4): 692-9, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-18042462

ABSTRACT

Metazoan replication-dependent histone mRNAs are not polyadenylated and instead end in a conserved stem loop that is the cis element responsible for coordinate posttranscriptional regulation of these mRNAs. Using biochemical approaches, only a limited number of factors required for cleavage of histone pre-mRNA have been identified. We therefore performed a genome-wide RNA interference screen in Drosophila cells using a GFP reporter that is expressed only when histone pre-mRNA processing is disrupted. Four of the 24 genes identified encode proteins also necessary for cleavage/polyadenylation, indicating mechanistic conservation in formation of different mRNA 3' ends. We also unexpectedly identified the histone variants H2Av and H3.3A/B. In H2Av mutant cells, U7 snRNP remains active but fails to accumulate at the histone locus, suggesting there is a regulatory pathway that coordinates the production of variant and canonical histones that acts via localization of essential histone pre-mRNA processing factors.


Subject(s)
DNA Replication , Genome, Insect/genetics , Histones/metabolism , Mutant Proteins/metabolism , RNA Interference , RNA Precursors/metabolism , RNA Processing, Post-Transcriptional , Animals , Base Sequence , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Genes, Reporter , Histones/genetics , Molecular Sequence Data , Polyadenylation , Protein Transport , RNA Precursors/genetics , Ribonucleoprotein, U7 Small Nuclear/metabolism
14.
Hypertension ; 43(2): 445-51, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14718357

ABSTRACT

We previously showed that the phenotype of mice with targeted disruption of the gene encoding the AT1A receptor (Agtr1a), the major murine AT1 receptor isoform, is strongly influenced by recessive genetic modifiers derived from the C57BL/6 or 129 inbred strains. To further evaluate the genetic modifiers on the C57BL/6 background, we performed backcrosses between F1(C57BL/6x129) and C57BL/6 Agtr1a-/- mice and analyzed the progeny, focusing on the development of structural lesions in the renal vasculature. In affected animals, these lesions are characterized by medial thickening of small arteries and arterioles in the kidney that are reminiscent of vascular lesions in patients with nephrosclerosis. Among 180 consecutive progeny, 170 (94%) survived to completion of the study. On masked pathological examination at age 8 months, 86 had intermediate to severe vascular lesions whereas 84 had no detectable lesions. Based on a hypothetical model of a single recessive modifier locus arising from the C57BL/6 background, the observed proportion of affected animals among the backcross progeny was not statistically different from that predicted by chi2 analysis (51% versus 50%; P=0.88). We next performed genomic microsatellite analysis in a subset of 121 backcross progeny using a panel of markers spanning approximately 15 cM intervals across the mouse genome. By 2-point analysis, we found a region spanning 5 cM on chromosome 3, with significant linkage to the development of renal vascular lesions (LOD score: 3.3 to 3.8).


Subject(s)
Chromosomes, Mammalian , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney/pathology , Receptor, Angiotensin, Type 1/genetics , Animals , Blood Pressure , Crosses, Genetic , Genetic Linkage , Kidney/blood supply , Kidney Diseases/physiopathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...