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










Publication year range
1.
Arterioscler Thromb Vasc Biol ; 40(5): 1311-1324, 2020 05.
Article in English | MEDLINE | ID: mdl-32188273

ABSTRACT

OBJECTIVE: TMEM55B (transmembrane protein 55B) is a phosphatidylinositol-(4,5)-bisphosphate (PI[4,5]P2) phosphatase that regulates cellular cholesterol, modulates LDLR (low-density lipoprotein receptor) decay, and lysosome function. We tested the effects of Tmem55b knockdown on plasma lipids in mice and assessed the roles of LDLR lysosomal degradation and change in (PI[4,5]P2) in mediating these effects. Approach and Results: Western diet-fed C57BL/6J mice were treated with antisense oligonucleotides against Tmem55b or a nontargeting control for 3 to 4 weeks. Hepatic Tmem55b transcript and protein levels were reduced by ≈70%, and plasma non-HDL (high-density lipoprotein) cholesterol was increased ≈1.8-fold (P<0.0001). Immunoblot analysis of fast protein liquid chromatography (FPLC) fractions revealed enrichment of ApoE-containing particles in the LDL size range. In contrast, Tmem55b knockdown had no effect on plasma cholesterol in Ldlr-/- mice. In primary hepatocytes and liver tissues from Tmem55b knockdown mice, there was decreased LDLR protein. In the hepatocytes, there was increased lysosome staining and increased LDLR-lysosome colocalization. Impairment of lysosome function (incubation with NH4Cl or knockdown of the lysosomal proteins LAMP1 or RAB7) abolished the effect of TMEM55B knockdown on LDLR in HepG2 (human hepatoma) cells. Colocalization of the recycling endosome marker RAB11 (Ras-related protein 11) with LDLR in HepG2 cells was reduced by 50% upon TMEM55B knockdown. Finally, knockdown increased hepatic PI(4,5)P2 levels in vivo and in HepG2 cells, while TMEM55B overexpression in vitro decreased PI(4,5)P2. TMEM55B knockdown decreased, whereas overexpression increased, LDL uptake in HepG2 cells. Notably, the TMEM55B overexpression effect was reversed by incubation with PI(4,5)P2. Conclusions: These findings indicate a role for TMEM55B in regulating plasma cholesterol levels by affecting PI(4,5)P2-mediated LDLR lysosomal degradation.


Subject(s)
Cholesterol/blood , Hepatocytes/metabolism , Liver/metabolism , Lysosomes/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphoinositide Phosphatases/metabolism , Receptors, LDL/metabolism , Animals , Diet, High-Fat , Down-Regulation , Female , Hep G2 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Phosphoinositide Phosphatases/genetics , Protein Transport , Proteolysis , Receptors, LDL/genetics , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism
2.
Proc Natl Acad Sci U S A ; 116(38): 18943-18950, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31484776

ABSTRACT

Rapid advances in genomic technologies have led to a wealth of diverse data, from which novel discoveries can be gleaned through the application of robust statistical and computational methods. Here, we describe GeneFishing, a semisupervised computational approach to reconstruct context-specific portraits of biological processes by leveraging gene-gene coexpression information. GeneFishing incorporates multiple high-dimensional statistical ideas, including dimensionality reduction, clustering, subsampling, and results aggregation, to produce robust results. To illustrate the power of our method, we applied it using 21 genes involved in cholesterol metabolism as "bait" to "fish out" (or identify) genes not previously identified as being connected to cholesterol metabolism. Using simulation and real datasets, we found that the results obtained through GeneFishing were more interesting for our study than those provided by related gene prioritization methods. In particular, application of GeneFishing to the GTEx liver RNA sequencing (RNAseq) data not only reidentified many known cholesterol-related genes, but also pointed to glyoxalase I (GLO1) as a gene implicated in cholesterol metabolism. In a follow-up experiment, we found that GLO1 knockdown in human hepatoma cell lines increased levels of cellular cholesterol ester, validating a role for GLO1 in cholesterol metabolism. In addition, we performed pantissue analysis by applying GeneFishing on various tissues and identified many potential tissue-specific cholesterol metabolism-related genes. GeneFishing appears to be a powerful tool for identifying related components of complex biological systems and may be used across a wide range of applications.


Subject(s)
Biological Phenomena/genetics , Computational Biology/methods , Gene Expression Profiling , Genomics/methods , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cholesterol/metabolism , Databases, Genetic , Humans , Lactoylglutathione Lyase/genetics , Lipid Metabolism/genetics , Organ Specificity/genetics , Reproducibility of Results , Workflow
3.
Sci Rep ; 8(1): 12443, 2018 08 20.
Article in English | MEDLINE | ID: mdl-30127457

ABSTRACT

Statins are the most commonly prescribed cardiovascular disease drug, but their inter-individual efficacy varies considerably. Genetic factors uncovered to date have only explained a small proportion of variation in low-density lipoprotein cholesterol (LDLC) lowering. To identify novel markers and determinants of statin response, we used whole transcriptome sequence data collected from simvastatin and control incubated lymphoblastoid cell lines (LCLs) established from participants of the Cholesterol and Pharmacogenetics (CAP) simvastatin clinical trial. We looked for genes whose statin-induced expression changes were most different between LCLs derived from individuals with high versus low plasma LDLC statin response during the CAP trial. We created a classification model of 82 "signature" gene expression changes that distinguished high versus low LDLC statin response. One of the most differentially changing genes was zinc finger protein 542 pseudogene (ZNF542P), the signature gene with changes most correlated with statin-induced change in cellular cholesterol ester, an in vitro marker of statin response. ZNF542P knock-down in a human hepatoma cell line increased intracellular cholesterol ester levels upon simvastatin treatment. Together, these findings imply a role for ZNF542P in LDLC response to simvastatin and, importantly, highlight the potential significance of noncoding RNAs as a contributing factor to variation in drug response.


Subject(s)
Cholesterol, LDL/genetics , Pseudogenes/genetics , Simvastatin/pharmacology , Cell Line , Cell Line, Tumor , Cholesterol/genetics , Humans , Pharmacogenetics/methods , Transcriptome/genetics
4.
Circ Cardiovasc Genet ; 9(3): 223-30, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27071970

ABSTRACT

BACKGROUND: Numerous genetic contributors to cardiovascular disease risk have been identified through genome-wide association studies; however, identifying the molecular mechanism underlying these associations is not straightforward. The Justification for the Use of Statins in Primary Prevention: An Intervention Trial Evaluating Rosuvastatin (JUPITER) trial of rosuvastatin users identified a sub-genome-wide association of rs6924995, a single-nucleotide polymorphism ≈10 kb downstream of myosin regulatory light chain interacting protein (MYLIP, aka IDOL and inducible degrader of low-density lipoprotein receptor [LDLR]), with LDL cholesterol statin response. Interestingly, although this signal was initially attributed to MYLIP, rs6924995 lies within RP1-13D10.2, an uncharacterized long noncoding RNA. METHODS AND RESULTS: Using simvastatin and sham incubated lymphoblastoid cell lines from participants of the Cholesterol and Pharmacogenetics (CAP) simvastatin clinical trial, we found that statin-induced change in RP1-13D10.2 levels differed between cell lines from the tails of the white and black low-density lipoprotein cholesterol response distributions, whereas no difference in MYLIP was observed. RP1-13D10.2 overexpression in Huh7 and HepG2 increased LDLR transcript levels, increased LDL uptake, and decreased media levels of apolipoprotein B. In addition, we found a trend of slight differences in the effects of RP1-13D10.2 overexpression on LDLR transcript levels between hepatoma cells transfected with the rs6924995 A versus G allele and a suggestion of an association between rs6924995 and RP1-10D13.2 expression levels in the CAP lymphoblastoid cell lines. Finally, RP1-13D10.2 expression levels seem to be sterol regulated, consistent with its potential role as a novel lipid regulator. CONCLUSIONS: RP1-13D10.2 is a long noncoding RNA that regulates LDLR and may contribute to low-density lipoprotein cholesterol response to statin treatment. These findings highlight the potential role of noncoding RNAs as determinants of interindividual variation in drug response.


Subject(s)
Cholesterol, LDL/metabolism , Dyslipidemias/drug therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lipid Metabolism/drug effects , RNA, Long Noncoding/genetics , Simvastatin/pharmacology , Adult , Aged , Apolipoprotein B-100/metabolism , Biomarkers/blood , Clinical Trials as Topic , Dyslipidemias/blood , Dyslipidemias/diagnosis , Dyslipidemias/genetics , Female , Hep G2 Cells , Humans , Lipid Metabolism/genetics , Male , Middle Aged , Polymorphism, Single Nucleotide , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Time Factors , Transcription, Genetic , Transfection , Up-Regulation
5.
Bone ; 81: 97-103, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26151122

ABSTRACT

Osteoblasts perceive and respond to changes in their pericellular environment, including biophysical signals and oxygen availability, to elicit an anabolic or catabolic response. Parathyroid hormone (PTH) affects each arm of skeletal remodeling, with net anabolic or catabolic effects dependent upon duration of exposure. Similarly, the capacity of osteoblastic cells to perceive pericellular oxygen has a profound effect on skeletal mass and architecture, as mice expressing stable hypoxia-inducible factor (HIF)-1α and -2α demonstrate age-dependent increases in bone volume per tissue volume and osteoblast number. Further, HIF levels and signaling can be influenced in an oxygen-independent manner. Because the cellular mechanisms involved in PTH regulation of the skeleton remain vague, we sought whether PTH could influence HIF-1α expression and HIF-α-driven luciferase activity independently of altered oxygen availability. Using UMR106.01 mature osteoblasts, we observed that 100nM hPTH(1-34) decreased HIF-1α and HIF-responsive luciferase activity in a process involving heat shock protein 90 (Hsp90) and cyclic AMP but not intracellular calcium. Altering activity of the small GTPase RhoA and its effector kinase ROCK altered HIF-α-driven luciferase activity in the absence and presence of PTH. Taken together, these data introduce PTH as a regulator of oxygen-independent HIF-1α levels through a mechanism involving cyclic AMP, Hsp90, and the cytoskeleton.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Osteoblasts/metabolism , Osteogenesis/physiology , Parathyroid Hormone/metabolism , Signal Transduction/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Blotting, Western , Cell Line , Humans , Mutagenesis, Site-Directed , Rats , Transfection
6.
Nature ; 512(7515): 449-52, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25164756

ABSTRACT

Genome function is dynamically regulated in part by chromatin, which consists of the histones, non-histone proteins and RNA molecules that package DNA. Studies in Caenorhabditis elegans and Drosophila melanogaster have contributed substantially to our understanding of molecular mechanisms of genome function in humans, and have revealed conservation of chromatin components and mechanisms. Nevertheless, the three organisms have markedly different genome sizes, chromosome architecture and gene organization. On human and fly chromosomes, for example, pericentric heterochromatin flanks single centromeres, whereas worm chromosomes have dispersed heterochromatin-like regions enriched in the distal chromosomal 'arms', and centromeres distributed along their lengths. To systematically investigate chromatin organization and associated gene regulation across species, we generated and analysed a large collection of genome-wide chromatin data sets from cell lines and developmental stages in worm, fly and human. Here we present over 800 new data sets from our ENCODE and modENCODE consortia, bringing the total to over 1,400. Comparison of combinatorial patterns of histone modifications, nuclear lamina-associated domains, organization of large-scale topological domains, chromatin environment at promoters and enhancers, nucleosome positioning, and DNA replication patterns reveals many conserved features of chromatin organization among the three organisms. We also find notable differences in the composition and locations of repressive chromatin. These data sets and analyses provide a rich resource for comparative and species-specific investigations of chromatin composition, organization and function.


Subject(s)
Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Chromatin/genetics , Chromatin/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Animals , Cell Line , Centromere/genetics , Centromere/metabolism , Chromatin/chemistry , Chromatin Assembly and Disassembly/genetics , DNA Replication/genetics , Enhancer Elements, Genetic/genetics , Epigenesis, Genetic , Heterochromatin/chemistry , Heterochromatin/genetics , Heterochromatin/metabolism , Histones/chemistry , Histones/metabolism , Humans , Molecular Sequence Annotation , Nuclear Lamina/metabolism , Nucleosomes/chemistry , Nucleosomes/genetics , Nucleosomes/metabolism , Promoter Regions, Genetic/genetics , Species Specificity
7.
PLoS Genet ; 8(9): e1002933, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23028348

ABSTRACT

The Caenorhabditis elegans dosage compensation complex (DCC) equalizes X-chromosome gene dosage between XO males and XX hermaphrodites by two-fold repression of X-linked gene expression in hermaphrodites. The DCC localizes to the X chromosomes in hermaphrodites but not in males, and some subunits form a complex homologous to condensin. The mechanism by which the DCC downregulates gene expression remains unclear. Here we show that the DCC controls the methylation state of lysine 20 of histone H4, leading to higher H4K20me1 and lower H4K20me3 levels on the X chromosomes of XX hermaphrodites relative to autosomes. We identify the PR-SET7 ortholog SET-1 and the Suv4-20 ortholog SET-4 as the major histone methyltransferases for monomethylation and di/trimethylation of H4K20, respectively, and provide evidence that X-chromosome enrichment of H4K20me1 involves inhibition of SET-4 activity on the X. RNAi knockdown of set-1 results in synthetic lethality with dosage compensation mutants and upregulation of X-linked gene expression, supporting a model whereby H4K20me1 functions with the condensin-like C. elegans DCC to repress transcription of X-linked genes. H4K20me1 is important for mitotic chromosome condensation in mammals, suggesting that increased H4K20me1 on the X may restrict access of the transcription machinery to X-linked genes via chromatin compaction.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Dosage Compensation, Genetic , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Methyltransferases/genetics , Animals , Chromatin/genetics , Disorders of Sex Development/genetics , Gene Expression Regulation, Developmental , Genes, X-Linked , Histone-Lysine N-Methyltransferase/metabolism , Male , Methylation , RNA Interference , X Chromosome/genetics
8.
Curr Biol ; 22(4): 320-5, 2012 Feb 21.
Article in English | MEDLINE | ID: mdl-22264607

ABSTRACT

Myosin IIIA (MYO3A) targets actin protrusion tips using a motility mechanism dependent on both motor and tail actin-binding activity [1]. We show that myosin IIIB (MYO3B) lacks tail actin-binding activity and is unable to target COS7 cell filopodia tips, yet is somehow able to target stereocilia tips. Strikingly, when MYO3B is coexpressed with espin-1 (ESPN1), a MYO3A cargo protein endogenously expressed in stereocilia [2], MYO3B targets and carries ESPN1 to COS7 filopodia tips. We show that this tip localization is lost when we remove the ESPN1 C terminus actin-binding site. We also demonstrate that, like MYO3A [2], MYO3B can elongate filopodia by transporting ESPN1 to the polymerizing end of actin filaments. The mutual dependence of MYO3B and ESPN1 for tip localization reveals a novel mechanism for the cell to regulate myosin tip localization via a reciprocal relationship with cargo that directly participates in actin binding for motility. Our results are consistent with a novel form of motility for class III myosins that requires both motor and tail domain actin-binding activity and show that the actin-binding tail can be replaced by actin-binding cargo. This study also provides a framework to better understand the late-onset hearing loss phenotype in patients with MYO3A mutations.


Subject(s)
Actin Cytoskeleton/metabolism , Microfilament Proteins/metabolism , Myosin Heavy Chains/metabolism , Myosin Type III/metabolism , Actins/metabolism , Amino Acid Motifs , Animals , COS Cells , Chlorocebus aethiops , Humans , Mice , Microfilament Proteins/chemistry , Microscopy, Confocal , Microscopy, Fluorescence , Polymerase Chain Reaction , Protein Binding , Protein Transport , Pseudopodia/metabolism , Pseudopodia/ultrastructure , Rats , Stereocilia/metabolism , Transfection
9.
J Neurochem ; 119(4): 772-84, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21895655

ABSTRACT

As class III unconventional myosins are motor proteins with an N-terminal kinase domain, it seems likely they play a role in both signaling and actin based transport. A growing body of evidence indicates that the motor functions of human class IIIA myosin, which has been implicated in progressive hearing loss, are modulated by intermolecular autophosphorylation. However, the phosphorylation sites have not been identified. We studied the kinase activity and phosphorylation sites of mouse class III myosins, mMyo3A and 3B, which are highly similar to their human orthologs. We demonstrate that the kinase domains of mMyo3A and 3B are active kinases, and that they have similar, if not identical, substrate specificities. We show that the kinase domains of these proteins autophosphorylate, and that they can phosphorylate sites within their myosin and tail domains. Using liquid chromatography-mass spectrometry, we identified phosphorylated sites in the kinase, myosin motor and tail domains of both mMyo3A and 3B. Most of the phosphorylated sites we identified and their consensus phosphorylation motifs are highly conserved among vertebrate class III myosins, including human class III myosins. Our findings are a major step toward understanding how the functions of class III myosins are regulated by phosphorylation.


Subject(s)
Myosin Type III/chemistry , Myosin Type III/metabolism , Protein Kinases/metabolism , Amino Acid Sequence , Amino Acids , Animals , Humans , Mass Spectrometry , Mice , Myosin Type III/classification , Myosin Type III/genetics , Peptides/metabolism , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Analysis, Protein , Substrate Specificity
10.
Science ; 330(6012): 1775-87, 2010 Dec 24.
Article in English | MEDLINE | ID: mdl-21177976

ABSTRACT

We systematically generated large-scale data sets to improve genome annotation for the nematode Caenorhabditis elegans, a key model organism. These data sets include transcriptome profiling across a developmental time course, genome-wide identification of transcription factor-binding sites, and maps of chromatin organization. From this, we created more complete and accurate gene models, including alternative splice forms and candidate noncoding RNAs. We constructed hierarchical networks of transcription factor-binding and microRNA interactions and discovered chromosomal locations bound by an unusually large number of transcription factors. Different patterns of chromatin composition and histone modification were revealed between chromosome arms and centers, with similarly prominent differences between autosomes and the X chromosome. Integrating data types, we built statistical models relating chromatin, transcription factor binding, and gene expression. Overall, our analyses ascribed putative functions to most of the conserved genome.


Subject(s)
Caenorhabditis elegans/genetics , Chromosomes , Gene Expression Profiling , Gene Expression Regulation , Genome, Helminth , Molecular Sequence Annotation , Animals , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Chromatin/genetics , Chromatin/metabolism , Chromatin/ultrastructure , Chromosomes/genetics , Chromosomes/metabolism , Chromosomes/ultrastructure , Computational Biology/methods , Conserved Sequence , Evolution, Molecular , Gene Regulatory Networks , Genes, Helminth , Genomics/methods , Histones/metabolism , Models, Genetic , RNA, Helminth/genetics , RNA, Helminth/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Regulatory Sequences, Nucleic Acid , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Exp Eye Res ; 89(2): 224-37, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19332056

ABSTRACT

Class III myosins are important for the function and survival of photoreceptors and ciliary hair cells. Although vertebrates possess two class III myosin genes, myo3A and myo3B, recent studies have focused on Myo3A because mutations in the human gene are implicated in progressive hearing loss. Myo3B may compensate for defects in Myo3A, yet little is known about its distribution and function. This study focuses on Myo3B expression in the mouse retina. We cloned two variants of myo3B from mouse retina and determined that they are expressed early in retinal development. In this study we show for the first time in a mammal that both Myo3B and Myo3A proteins are present in inner segments of all photoreceptors. Myo3B is also present in outer segments of S opsin-immunoreactive cones but not M opsin dominant cones. Myo3B is also detected in rare cells of the inner nuclear layer and some ganglion cells. Myo3B may have diverse roles in retinal neurons. In photoreceptor inner segments Myo3B is positioned appropriately to prevent photoreceptor loss of function caused by Myo3A defects.


Subject(s)
Eye Proteins/metabolism , Myosin Heavy Chains/metabolism , Myosin Type III/metabolism , Retina/metabolism , Amino Acid Sequence , Animals , Cloning, Molecular , Eye Proteins/genetics , Eye Proteins/immunology , Immune Sera , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Myosin Heavy Chains/genetics , Myosin Heavy Chains/immunology , Myosin Type III/genetics , Myosin Type III/immunology , Retina/growth & development , Retinal Cone Photoreceptor Cells/metabolism , Retinal Ganglion Cells/metabolism , Retinal Photoreceptor Cell Outer Segment/metabolism , Tissue Distribution
12.
Nat Cell Biol ; 11(4): 443-50, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19287378

ABSTRACT

Two proteins implicated in inherited deafness, myosin IIIa, a plus-end-directed motor, and espin, an actin-bundling protein containing the actin-monomer-binding motif WH2, have been shown to influence the length of mechanosensory stereocilia. Here we report that espin 1, an ankyrin repeat-containing isoform of espin, colocalizes with myosin IIIa at stereocilia tips and interacts with a unique conserved domain of myosin IIIa. We show that combined overexpression of these proteins causes greater elongation of stereocilia, compared with overexpression of either myosin IIIa alone or espin 1 alone. When these two proteins were co-expressed in the fibroblast-like COS-7 cell line they induced a tenfold elongation of filopodia. This extraordinary filopodia elongation results from the transport of espin 1 to the plus ends of F-actin by myosin IIIa and depends on espin 1 WH2 activity. This study provides the basis for understanding the role of myosin IIIa and espin 1 in regulating stereocilia length, and presents a physiological example where myosins can boost elongation of actin protrusions by transporting actin regulatory factors to the plus ends of actin filaments.


Subject(s)
Actin Cytoskeleton/metabolism , Cilia/metabolism , Microfilament Proteins/metabolism , Myosin Type III/metabolism , Actin Cytoskeleton/ultrastructure , Animals , Ankyrin Repeat , COS Cells , Chlorocebus aethiops , Cilia/ultrastructure , Mice , Microfilament Proteins/chemistry , Protein Binding , Protein Transport , Pseudopodia/metabolism , Pseudopodia/ultrastructure , Rats , Transfection
13.
J Comp Neurol ; 513(2): 209-23, 2009 Mar 10.
Article in English | MEDLINE | ID: mdl-19137585

ABSTRACT

Myosins are cytoskeletal motors critical for generating the forces necessary for establishing cell structure and mediating actin-dependent cell motility. In each cell type a multitude of myosins are expressed, each myosin contributing to aspects of morphogenesis, transport, or motility occurring in that cell type. To examine the roles of myosins in individual retinal cell types, we first used polymerase chain reaction (PCR) screening to identify myosins expressed in retina and retinal pigmented epithelium (RPE), followed by immunohistochemistry to examine the cellular and subcellular localizations of seven of these expressed myosins. In the myosin PCR screen of cDNA from striped bass retina and striped bass RPE, we amplified 17 distinct myosins from eight myosin classes from retinal cDNA and 11 distinct myosins from seven myosin classes from RPE cDNA. By using antibodies specific for myosins IIA, IIB, IIIA, IIIB, VI, VIIA, and IXB, we examined the localization patterns of these myosins in retinas and RPE of fish, and in isolated inner/outer segment fragments of green sunfish photoreceptors. Each of the myosins exhibited unique expression patterns in fish retina. Individual cell types expressed multiple myosin family members, some of which colocalized within a particular cell type. Because much is known about the functions and properties of these myosins from studies in other systems, their cellular and subcellular localization patterns in the retina help us understand which roles they might play in the vertebrate retina and RPE.


Subject(s)
Myosins/genetics , Myosins/metabolism , Retina/metabolism , Retinal Pigment Epithelium/metabolism , Animals , Blotting, Northern , DNA, Complementary/metabolism , Immunohistochemistry , Molecular Sequence Data , Myosin Type II/genetics , Myosin Type II/metabolism , Myosin Type III/genetics , Myosin Type III/metabolism , Myosins/classification , Retina/cytology , Retinal Pigment Epithelium/cytology , Reverse Transcriptase Polymerase Chain Reaction , Zebrafish
14.
Biochemistry ; 47(8): 2485-96, 2008 Feb 26.
Article in English | MEDLINE | ID: mdl-18229949

ABSTRACT

Myosin IIIA is unique among myosin proteins in that it contains an N-terminal kinase domain capable of autophosphorylating sites on the motor domain. A construct of myosin IIIA lacking the kinase domain localizes more efficiently to the stereocilia tips and alters the morphology of the tips in inner ear hair cells. Therefore, we performed a kinetic analysis of myosin IIIA without the kinase domain (MIII DeltaK) and compared these results with our reported analysis of myosin IIIA containing the kinase domain (MIII). The steady-state kinetic properties of MIII DeltaK indicate that it has a 2-fold higher maximum actin-activated ATPase rate (kcat = 1.5 +/- 0.1 s-1) and a 5-fold tighter actin affinity (KATPase = 6.0 +/- 1.4 microM, and KActin = 1.4 +/- 0.4 microM) compared to MIII. The rate of ATP binding to the motor domain is enhanced in MIII DeltaK (K1k+2 approximately 0.10 +/- 0.01 microM-1.s-1) to a level similar to the rate of binding to MIII in the presence of actin. The rate of ATP hydrolysis in the absence of actin is slow and may be rate limiting. Actin-activated phosphate release is identical with and without the kinase domain. The transition between actomyosin.ADP states, which is rate limiting in MIII, is enhanced in MIII DeltaK. MIII DeltaK accumulates more efficiently at the tips of filopodia in HeLa cells. Our results suggest a model in which the activity and concentration of myosin IIIA localized to the tips of actin bundles mediates the morphology of the tips in sensory cells.


Subject(s)
Movement , Myosin Heavy Chains/chemistry , Myosin Heavy Chains/physiology , Myosin Type III/chemistry , Myosin Type III/physiology , Phosphotransferases , Actins/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Animals , Cells, Cultured , Enzyme Activation , HeLa Cells , Humans , Hydrolysis , Kinetics , Models, Biological , Movement/physiology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Myosin Type III/genetics , Myosin Type III/metabolism , Neurons, Afferent/metabolism , Phosphotransferases/metabolism , Phosphotransferases/physiology , Protein Binding , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , Rabbits , Spodoptera , Transfection
15.
J Biol Chem ; 282(1): 216-31, 2007 Jan 05.
Article in English | MEDLINE | ID: mdl-17074769

ABSTRACT

Myosin IIIA is specifically expressed in photoreceptors and cochlea and is important for the phototransduction and hearing processes. In addition, myosin IIIA contains a unique N-terminal kinase domain and C-terminal tail actin-binding motif. We examined the kinetic properties of baculovirus expressed human myosin IIIA containing the kinase, motor, and two IQ domains. The maximum actin-activated ATPase rate is relatively slow (k(cat) = 0.77 +/- 0.08 s(-1)), and high actin concentrations are required to fully activate the ATPase rate (K(ATPase) = 34 +/- 11 microm). However, actin co-sedimentation assays suggest that myosin III has a relatively high steady-state affinity for actin in the presence of ATP (K(actin) approximately 7 microm). The rate of ATP binding to the motor domain is quite slow both in the presence and absence of actin (K(1)k(+2) = 0.020 and 0.001 microm(-1).s(-1), respectively). The rate of actin-activated phosphate release is more than 100-fold faster (85 s(-1)) than the k(cat), whereas ADP release in the presence of actin follows a two-step mechanism (7.0 and 0.6 s(-1)). Thus, our data suggest a transition between two actomyosin-ADP states is the rate-limiting step in the actomyosin III ATPase cycle. Our data also suggest the myosin III motor spends a large fraction of its cycle in an actomyosin ADP state that has an intermediate affinity for actin (K(d) approximately 5 microm). The long lived actomyosin-ADP state may be important for the ability of myosin III to function as a cellular transporter and actin cross-linker in the actin bundles of sensory cells.


Subject(s)
Myosin Type III/chemistry , Actins/chemistry , Amino Acid Motifs , Dose-Response Relationship, Drug , Humans , Kinetics , Light , Models, Chemical , Myosins/chemistry , Phosphates/chemistry , Phosphorylation , Protein Conformation , Protein Structure, Tertiary , Scattering, Radiation , Spectrometry, Fluorescence
16.
J Neurosci ; 26(40): 10243-52, 2006 Oct 04.
Article in English | MEDLINE | ID: mdl-17021180

ABSTRACT

Class III myosins are motor proteins that contain an N-terminal kinase domain and a C-terminal actin-binding domain. We show that myosin IIIa, which has been implicated in nonsyndromic progressive hearing loss, is localized at stereocilia tips. Myosin IIIa progressively accumulates during stereocilia maturation in a thimble-like pattern around the stereocilia tip, distinct from the cap-like localization of myosin XVa and the shaft localization of myosin Ic. Overexpression of deletion mutants for functional domains of green fluorescent protein (GFP)-myosin IIIa shows that the motor domain, but not the actin-binding tail domain, is required for stereocilia tip localization. Deletion of the kinase domain produces stereocilia elongation and bulging of the stereocilia tips. The thimble-like localization and the influence myosin IIIa has on stereocilia shape reveal a previously unrecognized molecular compartment at the distal end of stereocilia, the site of actin polymerization as well as operation of the mechanoelectrical transduction apparatus.


Subject(s)
Ear, Inner/metabolism , Gene Expression Regulation, Developmental/physiology , Myosin Heavy Chains/biosynthesis , Myosin Type III/biosynthesis , Animals , Anura , Bass , COS Cells , Cells, Cultured , Chickens , Chlorocebus aethiops , Cilia/genetics , Cilia/metabolism , Guinea Pigs , Humans , Mice , Myosin Heavy Chains/genetics , Myosin Type III/genetics , Rats , Time Factors
17.
J Cell Sci ; 117(Pt 24): 5825-34, 2004 Nov 15.
Article in English | MEDLINE | ID: mdl-15522885

ABSTRACT

Myo3A, a class III myosin, localizes to the distal (plus) ends of inner segment actin filament bundles that form the core of microvillus-like calycal processes encircling the base of the photoreceptor outer segment. To investigate Myo3A localization and function, we expressed green fluorescent protein-tagged bass Myo3A and related constructs in transgenic Xenopus rods using a modified opsin promoter. Tagged intact Myo3A localized to rod calycal processes, as previously reported for native bass Myo3A. Transgenic rods developed abnormally large calycal processes and subsequently degenerated. Modified Myo3A expression constructs demonstrated that calycal process localization required an active motor domain and the tail domain. Expressed tail domain alone localized to actin bundles along the entire inner segment length, rather than to the distal end. This tail domain localization required the conserved C-terminal domain (3THDII) previously shown to possess an actin-binding motif. Our findings suggest that Myo3A plays a role in the morphogenesis and maintenance of calycal processes of vertebrate photoreceptors.


Subject(s)
Actins/metabolism , Myosin Heavy Chains/genetics , Myosins/genetics , Photoreceptor Cells, Vertebrate/metabolism , Transgenes , Actin Cytoskeleton/metabolism , Amino Acid Motifs , Animals , Animals, Genetically Modified , Bass , Cytoskeleton/metabolism , Green Fluorescent Proteins/metabolism , Myosin Type III , Promoter Regions, Genetic , Protein Structure, Tertiary , Rod Opsins/genetics , Xenopus laevis
18.
Mol Biol Cell ; 14(10): 4173-80, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14517327

ABSTRACT

Bass Myo3A, a class III myosin, was expressed in HeLa cells as a GFP fusion in order to study its cellular localization. GFP-Myo3A localized to the cytoplasm and to the tips of F-actin bundles in filopodia, a localization that is consistent with the observed concentration toward the distal ends of F-actin bundles in photoreceptor cells. A mutation in the motor active site resulted in a loss of filopodia localization, suggesting that Myo3A motor activity is required for filopodial tip localization. Deletion analyses showed that the NH2-terminal kinase domain is not required but the CO2H-terminal 22 amino acids of the Myo3A tail are required for filopodial localization. Expression of this tail fragment alone produced fluorescence associated with F-actin throughout the cytoplasm and filopodia and a recombinant tail fragment bound to F-actin in vitro. An actin-binding motif was identified within this tail fragment, and a mutation within this motif abolished both filopodia localization by Myo3A and F-actin binding by the tail fragment alone. Calmodulin localized to filopodial tips when coexpressed with Myo3A but not in the absence of Myo3A, an observation consistent with the previous proposal that class III myosins bind calmodulin and thereby localize it in certain cell types.


Subject(s)
Actins/metabolism , Calmodulin/metabolism , Myosin Type III/metabolism , Pseudopodia/metabolism , Amino Acid Sequence , DNA Mutational Analysis , DNA Primers/genetics , Fluorescent Antibody Technique , Green Fluorescent Proteins , HeLa Cells , Humans , Luminescent Proteins/metabolism , Molecular Sequence Data , Myosin Type III/genetics , Protein Binding , Protein Structure, Tertiary , Pseudopodia/genetics , Recombinant Fusion Proteins/metabolism
19.
Mol Biol Cell ; 14(3): 1058-73, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12631723

ABSTRACT

The striped bass has two retina-expressed class III myosin genes, each composed of a kinase, motor, and tail domain. We report the cloning, sequence analysis, and expression patterns of the long (Myo3A) and short (Myo3B) class III myosins, as well as cellular localization and biochemical characterization of the long isoform, Myo3A. Myo3A (209 kDa) is expressed in the retina, brain, testis, and sacculus, and Myo3B (155 kDa) is expressed in the retina, intestine, and testis. The tails of these two isoforms contain two highly conserved domains, 3THDI and 3THDII. Whereas Myo3B has three IQ motifs, Myo3A has nine IQ motifs, four in its neck and five in its tail domain. Myo3A localizes to actin filament bundles of photoreceptors and is concentrated in the calycal processes. An anti-Myo3A antibody decorates the actin cytoskeleton of rod inner/outer segments, and this labeling is reduced by the presence of ATP. The ATP-sensitive actin association is a feature characteristic of myosin motors. The numerous IQ motifs may play a structural or signaling role in the Myo3A, and its localization to calycal processes indicates that this myosin mediates a local function at this site in vertebrate photoreceptors.


Subject(s)
Myosin Heavy Chains/metabolism , Protein Isoforms/metabolism , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Saccule and Utricle/metabolism , Actins/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Animals , Bass/metabolism , Calmodulin/metabolism , Cytoskeleton/metabolism , Humans , Molecular Sequence Data , Myosin Heavy Chains/classification , Myosin Heavy Chains/genetics , Myosin Type III , Phylogeny , Protein Isoforms/genetics , Retina/chemistry , Retina/cytology , Retinal Cone Photoreceptor Cells/chemistry , Retinal Cone Photoreceptor Cells/ultrastructure , Retinal Rod Photoreceptor Cells/chemistry , Retinal Rod Photoreceptor Cells/ultrastructure , Saccule and Utricle/chemistry , Sequence Alignment , Tissue Distribution , Tubulin/metabolism
20.
Genomics ; 79(5): 621-4, 2002 May.
Article in English | MEDLINE | ID: mdl-11991710

ABSTRACT

Class III myosins are actin-based motors with amino-terminal kinase domains. Expression of these motors is highly enhanced in retinal photoreceptors. As mutations in the gene encoding NINAC, a Drosophila melanogaster class III myosin, cause retinal degeneration, human homologs of this gene are potential candidates for human retinal disease. We have recently reported the cloning of MYO3A, a human myosin III expressed predominantly in the retina and retinal pigmented epithelium [1]. The map locus of MYO3A is close to, but does not overlap, that of human Usher's 1F [2]. Here we introduce a shorter class III myosin isoform, MYO3B, which is expressed in the retina, kidney, and testis. We describe the cDNA sequence, genomic organization, and splice variants of MYO3B expressed in the human retina. A product of 36 exons, MYO3B has several splice variants containing either one or two calmodulin binding (IQ) motifs in the neck domain and one of three predominant tail variations: a short tail ending just past the second IQ motif, or two alternatively spliced longer tails. MYO3B maps to 2q31.1-q31.2, a region that overlaps the locus for a Bardet-Biedl syndrome (BBS5) linked to markers at 2q31 [3].


Subject(s)
Bardet-Biedl Syndrome/genetics , Genetic Predisposition to Disease/genetics , Myosin Heavy Chains/genetics , Retina/metabolism , Alternative Splicing , Amino Acid Sequence , Base Sequence , Blotting, Northern , Cell Line , Exons , Female , Gene Expression , Genes/genetics , Humans , Introns , Male , Molecular Sequence Data , Myosin Type III , Protein Isoforms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...