Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Development ; 151(2)2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38063851

ABSTRACT

Cornelia de Lange syndrome (CdLS) is a congenital disorder featuring facial dysmorphism, postnatal growth deficits, cognitive disability and upper limb abnormalities. CdLS is genetically heterogeneous, with cases arising from mutation of BRD4, a bromodomain protein that binds and reads acetylated histones. In this study, we have modeled CdLS facial pathology through mouse neural crest cell (NCC)-specific mutation of BRD4 to characterize cellular and molecular function in craniofacial development. Mice with BRD4 NCC loss of function died at birth with severe facial hypoplasia, cleft palate, mid-facial clefting and exencephaly. Following migration, BRD4 mutant NCCs initiated RUNX2 expression for differentiation to osteoblast lineages but failed to induce downstream RUNX2 targets required for lineage commitment. BRD4 bound to active enhancers to regulate expression of osteogenic transcription factors and extracellular matrix components integral for bone formation. RUNX2 physically interacts with a C-terminal domain in the long isoform of BRD4 and can co-occupy osteogenic enhancers. This BRD4 association is required for RUNX2 recruitment and appropriate osteoblast differentiation. We conclude that BRD4 controls facial bone development through osteoblast enhancer regulation of the RUNX2 transcriptional program.


Subject(s)
De Lange Syndrome , Transcription Factors , Animals , Mice , Cell Cycle Proteins/genetics , Cell Differentiation , Core Binding Factor Alpha 1 Subunit , De Lange Syndrome/genetics , Neural Crest/metabolism , Nuclear Proteins/metabolism , Osteoblasts/metabolism , Osteogenesis , Transcription Factors/metabolism
2.
Birth Defects Res ; 115(20): 1885-1898, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37800171

ABSTRACT

BACKGROUND: Kabuki syndrome is a congenital developmental disorder that is characterized by distinctive facial gestalt and skeletal abnormalities. Although rare, the disorder shares clinical features with several related craniofacial syndromes that manifest from mutations in chromatin-modifying enzymes. Collectively, these clinical studies underscore the crucial, concerted functions of chromatin factors in shaping developmental genome structure and driving cellular transcriptional states. Kabuki syndrome predominantly results from mutations in KMT2D, a histone H3 lysine 4 methylase, or KDM6A, a histone H3 lysine 27 demethylase. AIMS: In this review, we summarize the research efforts to model Kabuki syndrome in vivo to understand the cellular and molecular mechanisms that lead to the craniofacial and skeletal pathogenesis that defines the disorder. DISCUSSION: As several studies have indicated the importance of KMT2D and KDM6A function through catalytic-independent mechanisms, we highlight noncanonical roles for these enzymes as recruitment centers for alternative chromatin and transcriptional machinery.


Subject(s)
Developmental Disabilities , Histones , Lysine , Child , Humans , Chromatin/genetics , Developmental Disabilities/genetics , Genomics , Histone Demethylases/genetics , Histone Demethylases/metabolism , Histones/genetics , Lysine/genetics , Craniofacial Abnormalities/genetics
3.
Cell Rep ; 35(2): 108966, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33852868

ABSTRACT

Persistent virus infections can cause pathogenesis that is debilitating or lethal. During these infections, virus-specific T cells fail to protect due to weakened antiviral activity or failure to persist. These outcomes are governed by histone modifications, although it is unknown which enzymes contribute to T cell loss or impaired function over time. In this study, we show that T cell receptor-stimulated CD8+ T cells increase their expression of UTX (ubiquitously transcribed tetratricopeptide repeat, X chromosome) to enhance gene expression. During chronic lymphocytic choriomeningitis virus (LCMV) infection in mice, UTX binds to enhancers and transcription start sites of effector genes, allowing for improved cytotoxic T lymphocyte (CTL)-mediated protection, independent of its trimethylation of histone 3 lysine 27 (H3K27me3) demethylase activity. UTX also limits the frequency and durability of virus-specific CD8+ T cells, which correspond to increased expression of inhibitory receptors. Thus, UTX guides gene expression patterns in CD8+ T cells, advancing early antiviral defenses while reducing the longevity of CD8+ T cell responses.


Subject(s)
Cytotoxicity, Immunologic/genetics , Histone Demethylases/genetics , Immunologic Memory/genetics , Lymphocytic Choriomeningitis/genetics , Lymphocytic choriomeningitis virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Gene Expression Profiling , Gene Expression Regulation , Hepatitis A Virus Cellular Receptor 2/genetics , Hepatitis A Virus Cellular Receptor 2/immunology , Histone Demethylases/deficiency , Histone Demethylases/immunology , Histones/genetics , Histones/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/growth & development , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Signal Transduction , T-Lymphocytes, Cytotoxic/virology , Viral Load/genetics , Viral Load/immunology , Lymphocyte Activation Gene 3 Protein
4.
Development ; 147(21)2020 07 17.
Article in English | MEDLINE | ID: mdl-32541010

ABSTRACT

Kabuki syndrome (KS) is a congenital craniofacial disorder resulting from mutations in the KMT2D histone methylase (KS1) or the UTX histone demethylase (KS2). With small cohorts of KS2 patients, it is not clear whether differences exist in clinical manifestations relative to KS1. We mutated KMT2D in neural crest cells (NCCs) to study cellular and molecular functions in craniofacial development with respect to UTX. Similar to UTX, KMT2D NCC knockout mice demonstrate hypoplasia with reductions in frontonasal bone lengths. We have traced the onset of KMT2D and UTX mutant NCC frontal dysfunction to a stage of altered osteochondral progenitor differentiation. KMT2D NCC loss-of-function does exhibit unique phenotypes distinct from UTX mutation, including fully penetrant cleft palate, mandible hypoplasia and deficits in cranial base ossification. KMT2D mutant NCCs lead to defective secondary palatal shelf elevation with reduced expression of extracellular matrix components. KMT2D mutant chondrocytes in the cranial base fail to properly differentiate, leading to defective endochondral ossification. We conclude that KMT2D is required for appropriate cranial NCC differentiation and KMT2D-specific phenotypes may underlie differences between Kabuki syndrome subtypes.


Subject(s)
Abnormalities, Multiple/enzymology , Abnormalities, Multiple/pathology , Cell Differentiation , Face/abnormalities , Hematologic Diseases/enzymology , Hematologic Diseases/pathology , Histone-Lysine N-Methyltransferase/metabolism , Myeloid-Lymphoid Leukemia Protein/metabolism , Neural Crest/enzymology , Neural Crest/pathology , Vestibular Diseases/enzymology , Vestibular Diseases/pathology , Alleles , Animals , Cell Lineage , Cell Movement , Chondrocytes/pathology , Face/pathology , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Mutation/genetics , Osteogenesis , Palate/embryology , Palate/metabolism , Palate/pathology , Phenotype , Skull/pathology
5.
EMBO J ; 39(9): e102808, 2020 05 04.
Article in English | MEDLINE | ID: mdl-32154941

ABSTRACT

Defects in transcriptional regulators of pancreatic exocrine differentiation have been implicated in pancreatic tumorigenesis, but the molecular mechanisms are poorly understood. The locus encoding the transcription factor HNF1A harbors susceptibility variants for pancreatic ductal adenocarcinoma (PDAC), while KDM6A, encoding Lysine-specific demethylase 6A, carries somatic mutations in PDAC. Here, we show that pancreas-specific Hnf1a null mutant transcriptomes phenocopy those of Kdm6a mutations, and both defects synergize with KrasG12D to cause PDAC with sarcomatoid features. We combine genetic, epigenomic, and biochemical studies to show that HNF1A recruits KDM6A to genomic binding sites in pancreatic acinar cells. This remodels the acinar enhancer landscape, activates differentiated acinar cell programs, and indirectly suppresses oncogenic and epithelial-mesenchymal transition genes. We also identify a subset of non-classical PDAC samples that exhibit the HNF1A/KDM6A-deficient molecular phenotype. These findings provide direct genetic evidence that HNF1A deficiency promotes PDAC. They also connect the tumor-suppressive role of KDM6A deficiency with a cell-specific molecular mechanism that underlies PDAC subtype definition.


Subject(s)
Acinar Cells/metabolism , Carcinoma, Pancreatic Ductal/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Histone Demethylases/genetics , Pancreatic Neoplasms/genetics , Animals , Carcinoma, Pancreatic Ductal/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Hepatocyte Nuclear Factor 1-alpha/metabolism , Histone Demethylases/metabolism , Humans , Mice , Mutation , Organ Specificity , Pancreas/metabolism , Pancreatic Neoplasms/metabolism
6.
Mol Cell Neurosci ; 87: 4-17, 2018 03.
Article in English | MEDLINE | ID: mdl-29254825

ABSTRACT

The histone H3 lysine 27 (H3K27) demethylase Kdm6b (Jmjd3) can promote cellular differentiation, however its physiological functions in neurons remain to be fully determined. We studied the expression and function of Kdm6b in differentiating granule neurons of the developing postnatal mouse cerebellum. At postnatal day 7, Kdm6b is expressed throughout the layers of the developing cerebellar cortex, but its expression is upregulated in newborn cerebellar granule neurons (CGNs). Atoh1-Cre mediated conditional knockout of Kdm6b in CGN precursors either alone or in combination with Kdm6a did not disturb the gross morphological development of the cerebellum. Furthermore, RNAi-mediated knockdown of Kdm6b in cultured CGN precursors did not alter the induced expression of early neuronal marker genes upon cell cycle exit. By contrast, knockdown of Kdm6b significantly impaired the induction of a mature neuronal gene expression program, which includes gene products required for functional synapse maturation. Loss of Kdm6b also impaired the ability of Brain-Derived Neurotrophic Factor (BDNF) to induce expression of Grin2c and Tiam1 in maturing CGNs. Taken together, these data reveal a previously unknown role for Kdm6b in the postmitotic stages of CGN maturation and suggest that Kdm6b may work, at least in part, by a transcriptional mechanism that promotes gene sensitivity to regulation by BDNF.


Subject(s)
Histone Demethylases/genetics , Histones/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Neurons/metabolism , Animals , Cytoplasmic Granules/metabolism , Gene Expression/genetics , Histones/metabolism , Humans
7.
Proc Natl Acad Sci U S A ; 114(43): E9046-E9055, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29073101

ABSTRACT

Kabuki syndrome, a congenital craniofacial disorder, manifests from mutations in an X-linked histone H3 lysine 27 demethylase (UTX/KDM6A) or a H3 lysine 4 methylase (KMT2D). However, the cellular and molecular etiology of histone-modifying enzymes in craniofacial disorders is unknown. We now establish Kabuki syndrome as a neurocristopathy, whereby the majority of clinical features are modeled in mice carrying neural crest (NC) deletion of UTX, including craniofacial dysmorphism, cardiac defects, and postnatal growth retardation. Female UTX NC knockout (FKO) demonstrates enhanced phenotypic severity over males (MKOs), due to partial redundancy with UTY, a Y-chromosome demethylase-dead homolog. Thus, NC cells may require demethylase-independent UTX activity. Consistently, Kabuki causative point mutations upstream of the JmjC domain do not disrupt UTX demethylation. We have isolated primary NC cells at a phenocritical postmigratory timepoint in both FKO and MKO mice, and genome-wide expression and histone profiling have revealed UTX molecular function in establishing appropriate chromatin structure to regulate crucial NC stem-cell signaling pathways. However, the majority of UTX regulated genes do not experience aberrations in H3K27me3 or H3K4me3, implicating alternative roles for UTX in transcriptional control. These findings are substantiated through demethylase-dead knockin mutation of UTX, which supports appropriate facial development.


Subject(s)
Abnormalities, Multiple/etiology , Face/abnormalities , Hematologic Diseases/etiology , Histone Demethylases/metabolism , Neural Crest/physiopathology , Vestibular Diseases/etiology , Animals , Cell Survival/genetics , Disease Models, Animal , Female , Gene Expression Regulation, Developmental , HEK293 Cells , Histone Demethylases/genetics , Humans , Lysine/metabolism , Male , Mice, Knockout , Mice, Transgenic , Mutation , Neural Crest/metabolism , Nuclear Proteins/genetics , Skull/embryology
8.
Immunity ; 43(4): 703-14, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26431949

ABSTRACT

Epigenetic changes, including histone methylation, control T cell differentiation and memory formation, though the enzymes that mediate these processes are not clear. We show that UTX, a histone H3 lysine 27 (H3K27) demethylase, supports T follicular helper (Tfh) cell responses that are essential for B cell antibody generation and the resolution of chronic viral infections. Mice with a T cell-specific UTX deletion had fewer Tfh cells, reduced germinal center responses, lacked virus-specific immunoglobulin G (IgG), and were unable to resolve chronic lymphocytic choriomeningitis virus infections. UTX-deficient T cells showed decreased expression of interleukin-6 receptor-α and other Tfh cell-related genes that were associated with increased H3K27 methylation. Additionally, Turner Syndrome subjects, who are predisposed to chronic ear infections, had reduced UTX expression in immune cells and decreased circulating CD4(+) CXCR5(+) T cell frequency. Thus, we identify a critical link between UTX in T cells and immunity to infection.


Subject(s)
Histone Demethylases/deficiency , Histone Demethylases/physiology , Lymphocytic choriomeningitis virus/immunology , Nuclear Proteins/deficiency , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Helper-Inducer/immunology , Viremia/immunology , Animals , Antibodies, Viral/biosynthesis , Cell Differentiation , Female , Gene Dosage , Gene Expression Regulation/immunology , Genetic Predisposition to Disease , Histones/metabolism , Humans , Immunologic Memory , Interleukin-6 Receptor alpha Subunit/biosynthesis , Interleukin-6 Receptor alpha Subunit/genetics , Lymphocyte Cooperation , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/pathogenicity , Methylation , Mice , Models, Immunological , Otitis Media/etiology , Protein Processing, Post-Translational , Receptors, CXCR5/analysis , Species Specificity , T-Lymphocyte Subsets/enzymology , T-Lymphocyte Subsets/virology , T-Lymphocytes, Helper-Inducer/enzymology , T-Lymphocytes, Helper-Inducer/virology , Transcription, Genetic , Turner Syndrome/complications , Turner Syndrome/enzymology , Virulence , X Chromosome Inactivation
9.
PLoS Genet ; 10(8): e1004507, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25101834

ABSTRACT

The early mammalian embryo utilizes histone H3 lysine 27 trimethylation (H3K27me3) to maintain essential developmental genes in a repressive chromatin state. As differentiation progresses, H3K27me3 is removed in a distinct fashion to activate lineage specific patterns of developmental gene expression. These rapid changes in early embryonic chromatin environment are thought to be dependent on H3K27 demethylases. We have taken a mouse genetics approach to remove activity of both H3K27 demethylases of the Kdm6 gene family, Utx (Kdm6a, X-linked gene) and Jmjd3 (Kdm6b, autosomal gene). Male embryos null for active H3K27 demethylation by the Kdm6 gene family survive to term. At mid-gestation, embryos demonstrate proper patterning and activation of Hox genes. These male embryos retain the Y-chromosome UTX homolog, UTY, which cannot demethylate H3K27me3 due to mutations in catalytic site of the Jumonji-C domain. Embryonic stem (ES) cells lacking all enzymatic KDM6 demethylation exhibit a typical decrease in global H3K27me3 levels with differentiation. Retinoic acid differentiations of these ES cells demonstrate loss of H3K27me3 and gain of H3K4me3 to Hox promoters and other transcription factors, and induce expression similar to control cells. A small subset of genes exhibit decreased expression associated with reduction of promoter H3K4me3 and some low-level accumulation of H3K27me3. Finally, Utx and Jmjd3 mutant mouse embryonic fibroblasts (MEFs) demonstrate dramatic loss of H3K27me3 from promoters of several Hox genes and transcription factors. Our results indicate that early embryonic H3K27me3 repression can be alleviated in the absence of active demethylation by the Kdm6 gene family.


Subject(s)
Cell Differentiation/genetics , Embryonic Development/genetics , Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Animals , Chromatin/genetics , Embryo, Mammalian , Embryonic Stem Cells , Female , Gene Expression Regulation, Developmental , Histone Demethylases/biosynthesis , Jumonji Domain-Containing Histone Demethylases/biosynthesis , Male , Mice , Mutation , Pregnancy
10.
PLoS Genet ; 8(9): e1002964, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23028370

ABSTRACT

UTX (KDM6A) and UTY are homologous X and Y chromosome members of the Histone H3 Lysine 27 (H3K27) demethylase gene family. UTX can demethylate H3K27; however, in vitro assays suggest that human UTY has lost enzymatic activity due to sequence divergence. We produced mouse mutations in both Utx and Uty. Homozygous Utx mutant female embryos are mid-gestational lethal with defects in neural tube, yolk sac, and cardiac development. We demonstrate that mouse UTY is devoid of in vivo demethylase activity, so hemizygous X(Utx-) Y(+) mutant male embryos should phenocopy homozygous X(Utx-) X(Utx-) females. However, X(Utx-) Y(+) mutant male embryos develop to term; although runted, approximately 25% survive postnatally reaching adulthood. Hemizygous X(+) Y(Uty-) mutant males are viable. In contrast, compound hemizygous X(Utx-) Y(Uty-) males phenocopy homozygous X(Utx-) X(Utx-) females. Therefore, despite divergence of UTX and UTY in catalyzing H3K27 demethylation, they maintain functional redundancy during embryonic development. Our data suggest that UTX and UTY are able to regulate gene activity through demethylase independent mechanisms. We conclude that UTX H3K27 demethylation is non-essential for embryonic viability.


Subject(s)
Embryonic Development/genetics , Gene Expression Regulation, Developmental , Histone Demethylases , Proteins , Animals , Female , Hemizygote , Histone Demethylases/genetics , Histone Demethylases/metabolism , Homozygote , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Male , Methylation , Mice , Minor Histocompatibility Antigens , Mutation , Proteins/genetics , Proteins/metabolism
11.
J Neurosci ; 32(34): 11706-15, 2012 Aug 22.
Article in English | MEDLINE | ID: mdl-22915113

ABSTRACT

Intraperitoneal injection of the Gram-negative bacterial endotoxin lipopolysaccharide (LPS) elicits a rapid innate immune response. While this systemic inflammatory response can be destructive, tolerable low doses of LPS render the brain transiently resistant to subsequent injuries. However, the mechanism by which microglia respond to LPS stimulation and participate in subsequent neuroprotection has not been documented. In this study, we first established a novel LPS treatment paradigm where mice were injected intraperitoneally with 1.0 mg/kg LPS for four consecutive days to globally activate CNS microglia. By using a reciprocal bone marrow transplantation procedure between wild-type and Toll-like receptor 4 (TLR4) mutant mice, we demonstrated that the presence of LPS receptor (TLR4) is not required on hematogenous immune cells but is required on cells that are not replaced by bone marrow transplantation, such as vascular endothelia and microglia, to transduce microglial activation and neuroprotection. Furthermore, we showed that activated microglia physically ensheathe cortical projection neurons, which have reduced axosomatic inhibitory synapses from the neuronal perikarya. In line with previous reports that inhibitory synapse reduction protects neurons from degeneration and injury, we show here that neuronal cell death and lesion volumes are significantly reduced in LPS-treated animals following experimental brain injury. Together, our results suggest that activated microglia participate in neuroprotection and that this neuroprotection is likely achieved through reduction of inhibitory axosomatic synapses. The therapeutic significance of these findings rests not only in identifying neuroprotective functions of microglia, but also in establishing the CNS location of TLR4 activation.


Subject(s)
Brain Injuries/drug therapy , Lipopolysaccharides/administration & dosage , Microglia/drug effects , Neuroprotective Agents/administration & dosage , Toll-Like Receptor 4/metabolism , Animals , Antigens, CD/metabolism , Apoptosis/drug effects , Bone Marrow Transplantation , Brain Injuries/pathology , Brain Injuries/surgery , Bromodeoxyuridine/metabolism , Cell Proliferation/drug effects , Central Nervous System/cytology , Central Nervous System/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Flow Cytometry , Immunity, Innate/drug effects , In Situ Nick-End Labeling , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Microglia/metabolism , Microglia/ultrastructure , Microscopy, Immunoelectron , Motor Cortex/pathology , Motor Cortex/ultrastructure , Synapses/genetics , Synapses/metabolism , Synapses/ultrastructure , Toll-Like Receptor 4/deficiency , Transplantation Chimera
12.
PLoS Genet ; 7(9): e1002307, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21980308

ABSTRACT

Heterozygous Twirler (Tw) mice develop obesity and circling behavior associated with malformations of the inner ear, whereas homozygous Tw mice have cleft palate and die shortly after birth. Zeb1 is a zinc finger protein that contributes to mesenchymal cell fate by repression of genes whose expression defines epithelial cell identity. This developmental pathway is disrupted in inner ears of Tw/Tw mice. The purpose of our study was to comprehensively characterize the Twirler phenotype and to identify the causative mutation. The Tw/+ inner ear phenotype includes irregularities of the semicircular canals, abnormal utricular otoconia, a shortened cochlear duct, and hearing loss, whereas Tw/Tw ears are severely malformed with barely recognizable anatomy. Tw/+ mice have obesity associated with insulin-resistance and have lymphoid organ hypoplasia. We identified a noncoding nucleotide substitution, c.58+181G>A, in the first intron of the Tw allele of Zeb1 (Zeb1(Tw)). A knockin mouse model of c.58+181G>A recapitulated the Tw phenotype, whereas a wild-type knockin control did not, confirming the mutation as pathogenic. c.58+181G>A does not affect splicing but disrupts a predicted site for Myb protein binding, which we confirmed in vitro. In comparison, homozygosity for a targeted deletion of exon 1 of mouse Zeb1, Zeb1(ΔEx1), is associated with a subtle abnormality of the lateral semicircular canal that is different than those in Tw mice. Expression analyses of E13.5 Twirler and Zeb1(ΔEx1) ears confirm that Zeb1(ΔEx1) is a null allele, whereas Zeb1(Tw) RNA is expressed at increased levels in comparison to wild-type Zeb1. We conclude that a noncoding point mutation of Zeb1 acts via a gain-of-function to disrupt regulation of Zeb1(Tw) expression, epithelial-mesenchymal cell fate or interactions, and structural development of the inner ear in Twirler mice. This is a novel mechanism underlying disorders of hearing or balance.


Subject(s)
Abnormalities, Multiple/genetics , Ear, Inner/abnormalities , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Introns/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Obesity/genetics , Animals , Binding Sites/genetics , Carrier Proteins/genetics , Chromosome Mapping , DNA-Binding Proteins/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Phenotype , Point Mutation/genetics , RNA, Untranslated/genetics , RNA-Binding Proteins , Transcription Factors , Zinc Finger E-box-Binding Homeobox 1
13.
Mol Biol Cell ; 20(1): 90-101, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18923150

ABSTRACT

The assembly of metazoan Sm-class small nuclear ribonucleoproteins (snRNPs) is an elaborate, step-wise process that takes place in multiple subcellular compartments. The initial steps, including formation of the core RNP, are mediated by the survival motor neuron (SMN) protein complex. Loss-of-function mutations in human SMN1 result in a neuromuscular disease called spinal muscular atrophy. The SMN complex is comprised of SMN and a number of tightly associated proteins, collectively called Gemins. In this report, we identify and characterize the fruitfly ortholog of the DEAD box protein, Gemin3. Drosophila Gemin3 (dGem3) colocalizes and interacts with dSMN in vitro and in vivo. RNA interference for dGem3 codepletes dSMN and inhibits efficient Sm core assembly in vitro. Transposon insertion mutations in Gemin3 are larval lethals and also codeplete dSMN. Transgenic overexpression of dGem3 rescues lethality, but overexpression of dSMN does not, indicating that loss of dSMN is not the primary cause of death. Gemin3 mutant larvae exhibit motor defects similar to previously characterized Smn alleles. Remarkably, appreciable numbers of Gemin3 mutants (along with one previously undescribed Smn allele) survive as larvae for several weeks without pupating. Our results demonstrate the conservation of Gemin3 protein function in metazoan snRNP assembly and reveal that loss of either Smn or Gemin3 can contribute to neuromuscular dysfunction.


Subject(s)
DEAD Box Protein 20/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster , Larva/physiology , Motor Activity/physiology , Amino Acid Sequence , Animals , DEAD Box Protein 20/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Drosophila melanogaster/growth & development , Ecdysone/metabolism , Humans , Larva/anatomy & histology , Molecular Sequence Data , RNA Interference , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , SMN Complex Proteins/genetics , SMN Complex Proteins/metabolism , Sequence Alignment , Signal Transduction/physiology , Transgenes , snRNP Core Proteins/genetics , snRNP Core Proteins/metabolism
14.
Cleve Clin J Med ; 75 Suppl 2: S77-82, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18540152

ABSTRACT

Preconditioning is a phenomenon in which the brain protects itself against future injury by adapting to low doses of noxious insults. Preconditioning stimuli include ischemia, low doses of endotoxin, hypoxia, hypothermia and hyperthermia, cortical spreading depression, anesthetics, and 3-nitropropionic acid, among others. Understanding of the mechanisms underlying preconditioning has been elusive, but NMDA receptor activation, nitric oxide, inflammatory cytokines, and suppression of the innate immune system appear to have a role. Elucidation of the endogenous cell survival pathways involved in preconditioning has significant clinical implications for preventing neuronal damage in susceptible patients.


Subject(s)
Brain Ischemia/metabolism , Ischemic Preconditioning/methods , Brain Ischemia/prevention & control , Cytokines/metabolism , Humans , Immune System/metabolism , Microglia/metabolism , Nitric Oxide/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
15.
J Cell Biol ; 176(6): 831-41, 2007 Mar 12.
Article in English | MEDLINE | ID: mdl-17353360

ABSTRACT

Mutations in human survival motor neurons 1 (SMN1) cause spinal muscular atrophy (SMA) and are associated with defects in assembly of small nuclear ribonucleoproteins (snRNPs) in vitro. However, the etiological link between snRNPs and SMA is unclear. We have developed a Drosophila melanogaster system to model SMA in vivo. Larval-lethal Smn-null mutations show no detectable snRNP reduction, making it unlikely that these animals die from global snRNP deprivation. Hypomorphic mutations in Smn reduce dSMN protein levels in the adult thorax, causing flightlessness and acute muscular atrophy. Mutant flight muscle motoneurons display pronounced axon routing and arborization defects. Moreover, Smn mutant myofibers fail to form thin filaments and phenocopy null mutations in Act88F, which is the flight muscle-specific actin isoform. In wild-type muscles, dSMN colocalizes with sarcomeric actin and forms a complex with alpha-actinin, the thin filament crosslinker. The sarcomeric localization of Smn is conserved in mouse myofibrils. These observations suggest a muscle-specific function for SMN and underline the importance of this tissue in modulating SMA severity.


Subject(s)
Cyclic AMP Response Element-Binding Protein/physiology , Disease Models, Animal , Drosophila Proteins/physiology , Drosophila melanogaster/metabolism , Muscle, Skeletal/metabolism , Muscular Atrophy, Spinal/metabolism , Nerve Tissue Proteins/physiology , RNA-Binding Proteins/physiology , Actinin/metabolism , Actins/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/analysis , Cyclic AMP Response Element-Binding Protein/genetics , Drosophila Proteins/analysis , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/physiology , Muscular Atrophy, Spinal/genetics , Mutation , Myofibrils/metabolism , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , Phenotype , Protein Isoforms/metabolism , RNA-Binding Proteins/analysis , RNA-Binding Proteins/genetics , Ribonucleoproteins, Small Nuclear/metabolism , SMN Complex Proteins , Sarcomeres/metabolism , Survival of Motor Neuron 1 Protein
16.
Curr Opin Cell Biol ; 18(3): 317-24, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16632338

ABSTRACT

Cajal bodies (CBs) are nuclear subdomains involved in the biogenesis of several classes of small ribonucleoproteins (RNPs). A number of recent advances highlight progress in the understanding of the organization and dynamics of CB components. For example, a class of small Cajal body-specific (sca) RNPs has been discovered. Localization of scaRNPs to CBs was shown to depend on a conserved RNA motif. Intriguingly, this motif is also present in mammalian telomerase RNA and the evidence suggests that assembly of the active form of telomerase RNP occurs in and around CBs during S phase. Important steps in the assembly and modification of spliceosomal RNPs have also been shown to take place in CBs. Additional experiments have revealed the existence of kinetically distinct subclasses of CB components. Finally, the recent identification of novel markers for CBs in both Drosophila and Arabidopsis not only lays to rest questions about the evolutionary conservation of these nuclear suborganelles, but also should enable forward genetic screens for the identification of new components and pathways involved in their assembly, maintenance and function.


Subject(s)
Coiled Bodies/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Animals , Conserved Sequence/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Humans , Models, Biological , Nerve Tissue Proteins/metabolism , RNA Precursors/metabolism , RNA Splicing , RNA, Small Nuclear/metabolism , RNA-Binding Proteins/metabolism , Ribonucleoprotein, U2 Small Nuclear/metabolism , S Phase , SMN Complex Proteins , Spliceosomes/metabolism , Telomerase/metabolism
17.
Proc Natl Acad Sci U S A ; 102(48): 17372-7, 2005 Nov 29.
Article in English | MEDLINE | ID: mdl-16301532

ABSTRACT

Spinal muscular atrophy (SMA) is a neurodegenerative disease characterized by loss of spinal motor neurons. The gene encoding the survival of motor neurons (SMN) protein is mutated in >95% of SMA cases. SMN is the central component of a large oligomeric complex, including Gemins2-7, that is necessary and sufficient for the in vivo assembly of Sm proteins onto the small nuclear (sn)RNAs that mediate pre-mRNA splicing. After cytoplasmic assembly of the Sm core, both SMN and splicing snRNPs are imported into the nucleus, accumulating in Cajal bodies for additional snRNA maturation steps before targeting to splicing factor compartments known as "speckles." In this study, we analyzed the function of individual SMN complex members by RNA interference (RNAi). RNAi-mediated knockdown of SMN, Gemin2, Gemin3, and Gemin4 each disrupted Sm core assembly, whereas knockdown of Gemin5 and Snurportin1 had no effect. Assembly activity was rescued by expression of a GFP-SMN construct that is refractive to RNAi but not by similar constructs that contain SMA patient-derived mutations. Our results also demonstrate that Cajal body homeostasis requires SMN and ongoing snRNP biogenesis. Perturbation of SMN function results in disassembly of Cajal bodies and relocalization of the marker protein, coilin, to nucleoli. Moreover, in SMN-deficient cells, newly synthesized SmB proteins fail to associate with U2 snRNA or accumulate in Cajal bodies. Collectively, our results identify a previously uncharacterized function for Gemin3 and Gemin4 in Sm core assembly and correlate the activity of this pathway with SMA.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Muscular Atrophy, Spinal/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , RNA Helicases/metabolism , RNA-Binding Proteins/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , Biological Transport/physiology , Blotting, Western , Coiled Bodies/metabolism , DEAD Box Protein 20 , DEAD-box RNA Helicases , DNA Primers , Fluorescent Antibody Technique , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Minor Histocompatibility Antigens , Muscular Atrophy, Spinal/metabolism , Mutagenesis , RNA Interference , SMN Complex Proteins
18.
Chromosoma ; 114(3): 155-66, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16003501

ABSTRACT

Coilin is the signature protein of the Cajal body (CB), a nuclear suborganelle involved in the biogenesis of small nuclear ribonucleoproteins (snRNPs). Newly imported Sm-class snRNPs are thought to traffic through CBs before proceeding to their final nuclear destinations. Loss of coilin function in mice leads to significant viability and fertility problems. Coilin interacts directly with the spinal muscular atrophy (SMA) protein via dimethylarginine residues in its C-terminal domain. Although coilin hypomethylation results in delocalization of survival of motor neurons (SMN) from CBs, high concentrations of snRNPs remain within these structures. Thus, CBs appear to be involved in snRNP maturation, but factors that tether snRNPs to CBs have not been described. In this report, we demonstrate that the coilin C-terminal domain binds directly to various Sm and Lsm proteins via their Sm motifs. We show that the region of coilin responsible for this binding activity is separable from that which binds to SMN. Interestingly, U2, U4, U5, and U6 snRNPs interact with the coilin C-terminal domain in a glutathione S-transferase (GST)-pulldown assay, whereas U1 and U7 snRNPs do not. Thus, the ability to interact with free Sm (and Lsm) proteins as well as with intact snRNPs, indicates that coilin and CBs may facilitate the modification of newly formed snRNPs, the regeneration of 'mature' snRNPs, or the reclamation of unassembled snRNP components.


Subject(s)
Nuclear Proteins/metabolism , Protein Structure, Tertiary , Ribonucleoproteins, Small Nuclear/metabolism , Binding Sites , Escherichia coli/genetics , Escherichia coli/metabolism , Green Fluorescent Proteins/analysis , HeLa Cells/cytology , HeLa Cells/metabolism , Humans , Immunoprecipitation , Methylation , Nuclear Proteins/genetics , Protein Binding , Protein Processing, Post-Translational , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ribonucleoproteins, Small Nuclear/genetics , Transfection
19.
Acta Otolaryngol ; 124(3): 242-8, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15141750

ABSTRACT

OBJECTIVE: Mutations in the fibrillar collagen genes COL11A1 and COL11A2 can cause sensorineural hearing loss associated with Stickler syndrome. There is a correlation of hearing loss severity, onset, progression and affected frequencies with the underlying mutated collagen gene. We sought to determine whether differences in spatial or temporal expression of these genes underlie this correlation, and to identify the cochlear cell populations expressing these genes and the structures likely to be affected by mutations. MATERIALS AND METHODS: We used in situ hybridization analysis of C57BL/6J mouse temporal bones. RESULTS: Similar, diffuse expression of Col11a1 and Col11a2 mRNA was first observed in the cochlear duct at embryonic Day 15.5, with increasingly focal hybridization being noted at postnatal Days 1 and 5 in the greater epithelial ridge and lateral wall of the cochlea. The greater epithelial ridge appeared to be the main, if not only, source of mRNA encoding Col11a1 and Col11a2 in the tectorial membrane. At postnatal Day 13, Col11a1 and Col11a2 expression became more focal and co-localized in the inner sulcus, Claudius' cells and cells of Boettcher. CONCLUSIONS: We did not observe spatial or temporal differences in mRNA expression that could account for the auditory phenotype genotype correlation. The expression patterns suggest essential roles for Col11a1 and Col11a2 in the basilar or tectorial membranes.


Subject(s)
Cochlea/physiology , Collagen Type XI/genetics , Gene Expression Regulation, Developmental , Hearing Loss, Sensorineural/genetics , RNA, Messenger/biosynthesis , Animals , Basilar Membrane/embryology , Basilar Membrane/physiology , Cochlea/embryology , Collagen Type XI/metabolism , Extracellular Matrix/physiology , Genotype , In Situ Hybridization , Mice , Mice, Inbred C57BL , Mutation , Phenotype , Regression Analysis , Tectorial Membrane/embryology , Tectorial Membrane/physiology
20.
J Cell Biol ; 164(6): 831-42, 2004 Mar 15.
Article in English | MEDLINE | ID: mdl-15024031

ABSTRACT

Cajal bodies (CBs) are subnuclear domains implicated in small nuclear ribonucleoprotein (snRNP) biogenesis. In most cell types, CBs coincide with nuclear gems, which contain the survival of motor neurons (SMN) complex, an essential snRNP assembly factor. Here, we analyze the exchange kinetics of multiple components of CBs and gems in living cells using photobleaching microscopy. We demonstrate differences in dissociation kinetics of CB constituents and relate them to their functions. Coilin and SMN complex members exhibit relatively long CB residence times, whereas components of snRNPs, small nucleolar RNPs, and factors shared with the nucleolus have significantly shorter residence times. Comparison of the dissociation kinetics of these shared proteins from either the nucleolus or the CB suggests the existence of compartment-specific retention mechanisms. The dynamic properties of several CB components do not depend on their interaction with coilin because their dissociation kinetics are unaltered in residual nuclear bodies of coilin knockout cells. Photobleaching and fluorescence resonance energy transfer experiments demonstrate that coilin and SMN can interact within CBs, but their interaction is not the major determinant of their residence times. These results suggest that CBs and gems are kinetically independent structures.


Subject(s)
Coiled Bodies/metabolism , Animals , Cell Nucleolus/chemistry , Cell Nucleolus/metabolism , Coiled Bodies/chemistry , Cyclic AMP Response Element-Binding Protein , Fluorescence Recovery After Photobleaching , Fluorescence Resonance Energy Transfer , HeLa Cells , Humans , Macromolecular Substances , Mice , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA Splicing , RNA-Binding Proteins , Recombinant Fusion Proteins/metabolism , Ribonucleoproteins, Small Nuclear/metabolism , SMN Complex Proteins , Spliceosomes/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...