Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 132(11)2022 06 01.
Article in English | MEDLINE | ID: mdl-35642640

ABSTRACT

The discovery of microRNAs and their role in diseases was a breakthrough that inspired research into microRNAs as drug targets. Cardiovascular diseases are an area in which limitations of conventional pharmacotherapy are highly apparent and where microRNA-based drugs have appreciably progressed into preclinical and clinical testing. In this Review, we summarize the current state of microRNAs as therapeutic targets in the cardiovascular system. We report recent advances in the identification and characterization of microRNAs, their manipulation and clinical translation, and discuss challenges and perspectives toward clinical application.


Subject(s)
Cardiovascular Diseases , MicroRNAs , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/therapy , Humans , MicroRNAs/genetics
2.
Nat Commun ; 8(1): 1614, 2017 11 20.
Article in English | MEDLINE | ID: mdl-29158499

ABSTRACT

Chronic cardiac stress induces pathologic hypertrophy and fibrosis of the myocardium. The microRNA-29 (miR-29) family has been found to prevent excess collagen expression in various organs, particularly through its function in fibroblasts. Here, we show that miR-29 promotes pathologic hypertrophy of cardiac myocytes and overall cardiac dysfunction. In a mouse model of cardiac pressure overload, global genetic deletion of miR-29 or antimiR-29 infusion prevents cardiac hypertrophy and fibrosis and improves cardiac function. Targeted deletion of miR-29 in cardiac myocytes in vivo also prevents cardiac hypertrophy and fibrosis, indicating that the function of miR-29 in cardiac myocytes dominates over that in non-myocyte cell types. Mechanistically, we found cardiac myocyte miR-29 to de-repress Wnt signaling by directly targeting four pathway factors. Our data suggests that, cell- or tissue-specific antimiR-29 delivery may have therapeutic value for pathological cardiac remodeling and fibrosis.


Subject(s)
Cardiomegaly/metabolism , MicroRNAs/metabolism , Myocytes, Cardiac/metabolism , Wnt Proteins/metabolism , Adult , Aged , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Middle Aged , Myocardium/metabolism , Myocardium/pathology , Signal Transduction , Wnt Proteins/genetics
3.
Mol Ther ; 24(11): 1939-1948, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27545313

ABSTRACT

Systemic inhibition of miR-21 has proven effective against myocardial fibrosis and dysfunction, while studies in cardiac myocytes suggested a protective role in this cell type. Considering potential implications for therapy, we aimed to determine the cell fraction where miR-21 exerts its pathological activity. We developed a viral vector-based strategy for gene targeting of nonmyocyte cardiac cells in vivo and compared global to cardiac myocyte-specific and nonmyocyte-specific deletion of miR-21 in chronic left ventricular pressure overload. Murine moloney virus and serotype 9 of adeno-associated virus were engineered to encode improved Cre recombinase for genetic deletion in miR-21fl/fl mice. Pericardial injection of murine moloney virus-improved Cre recombinase to neonates achieved highly selective genetic ablation of miR-21 in nonmyocyte cardiac cells, identified as cardiac fibroblasts and endothelial cells. Upon left ventricular pressure overload, cardiac function was only preserved in mice with miR-21 deficiency in nonmyocyte cardiac cells, but not in mice with global or cardiac myocyte-specific ablation. Our data demonstrate that miR-21 exerts its pathologic activity directly in cardiac nonmyocytes and encourage further development of antimiR-21 therapy toward cellular tropism.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/administration & dosage , Heart Diseases/therapy , Heart Ventricles/physiopathology , MicroRNAs/genetics , Ventricular Remodeling , Animals , Dependovirus/genetics , Disease Models, Animal , Gene Deletion , Gene Knockout Techniques , HEK293 Cells , Heart Diseases/genetics , Heart Failure , Humans , Mice , Moloney murine leukemia virus/genetics , Myocytes, Cardiac/metabolism
4.
J Mol Cell Cardiol ; 99: 57-64, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27539859

ABSTRACT

A key response of the myocardium to stress is the secretion of factors with paracrine or endocrine function. Intriguing in this respect is peptidase inhibitor 16 (PI16), a member of the CAP family of proteins which we found to be highly upregulated in cardiac disease. Up to this point, the mechanism of action and physiological function of PI16 remained elusive. Here, we show that PI16 is predominantly expressed by cardiac fibroblasts, which expose PI16 to the interstitium via a glycophosphatidylinositol (-GPI) membrane anchor. Based on a reported genetic association of PI16 and plasma levels of the chemokine chemerin, we investigated whether PI16 regulates post-translational processing of its precursor pro-chemerin. PI16-deficient mice were engineered and found to generate higher levels of processed chemerin than wildtype mice. Purified recombinant PI16 efficiently inhibited cathepsin K, a chemerin-activating protease, in vitro. Moreover, we show that conditioned medium from PI16-overexpressing cells impaired the activation of pro-chemerin. Together, our data indicate that PI16 suppresses chemerin activation in the myocardium and suggest that this circuit may be part of the cardiac stress response.


Subject(s)
Chemokines/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Myocardium/metabolism , Proteinase Inhibitory Proteins, Secretory/metabolism , Animals , Cathepsin K/metabolism , Cell Communication , Cell Membrane/metabolism , Chemokines/genetics , Fibroblasts/metabolism , Gene Knockout Techniques , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Models, Biological , Receptors, Chemokine , Receptors, G-Protein-Coupled/metabolism , Signal Transduction
5.
PLoS One ; 9(11): e111754, 2014.
Article in English | MEDLINE | ID: mdl-25383878

ABSTRACT

Pre-mRNA splicing by the spliceosome is an essential step in the maturation of nearly all human mRNAs. Mutations in six spliceosomal proteins, PRPF3, PRPF4, PRPF6, PRPF8, PRPF31 and SNRNP200, cause retinitis pigmentosa (RP), a disease characterized by progressive photoreceptor degeneration. All splicing factors linked to RP are constituents of the U4/U6.U5 tri-snRNP subunit of the spliceosome, suggesting that the compromised function of this particle may lead to RP. Here, we report the identification of the p.R192H variant of the tri-snRNP factor PRPF4 in a patient with RP. The mutation affects a highly conserved arginine residue that is crucial for PRPF4 function. Introduction of a corresponding mutation into the zebrafish homolog of PRPF4 resulted in a complete loss of function in vivo. A series of biochemical experiments suggested that p.R192H disrupts the binding interface between PRPF4 and its interactor PRPF3. This interferes with the ability of PRPF4 to integrate into the tri-snRNP, as shown in a human cell line and in zebrafish embryos. These data suggest that the p.R192H variant of PRPF4 represents a functional null allele. The resulting haploinsufficiency of PRPF4 compromises the function of the tri-snRNP, reinforcing the notion that this spliceosomal particle is of crucial importance in the physiology of the retina.


Subject(s)
Mutation, Missense/genetics , Retinitis Pigmentosa/genetics , Ribonucleoprotein, U4-U6 Small Nuclear/genetics , Spliceosomes/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Western , Embryo, Nonmammalian/metabolism , Gangliosides/metabolism , Gene Components , HEK293 Cells , Humans , Molecular Sequence Data , Nuclear Proteins/metabolism , Pedigree , Ribonucleoprotein, U4-U6 Small Nuclear/metabolism , Sequence Analysis, DNA , Spliceosomes/metabolism , Zebrafish
6.
J Clin Invest ; 124(12): 5385-97, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25401477

ABSTRACT

Acute stimulation of cardiac ß-adrenoceptors is crucial to increasing cardiac function under stress; however, sustained ß-adrenergic stimulation has been implicated in pathological myocardial remodeling and heart failure. Here, we have demonstrated that export of cAMP from cardiac myocytes is an intrinsic cardioprotective mechanism in response to cardiac stress. We report that infusion of cAMP into mice averted myocardial hypertrophy and fibrosis in a disease model of cardiac pressure overload. The protective effect of exogenous cAMP required adenosine receptor signaling. This observation led to the identification of a potent paracrine mechanism that is dependent on secreted cAMP. Specifically, FRET-based imaging of cAMP formation in primary cells and in myocardial tissue from murine hearts revealed that cardiomyocytes depend on the transporter ABCC4 to export cAMP as an extracellular signal. Extracellular cAMP, through its metabolite adenosine, reduced cardiomyocyte cAMP formation and hypertrophy by activating A1 adenosine receptors while delivering an antifibrotic signal to cardiac fibroblasts by A2 adenosine receptor activation. Together, our data reveal a paracrine role for secreted cAMP in intercellular signaling in the myocardium, and we postulate that secreted cAMP may also constitute an important signal in other tissues.


Subject(s)
Cardiomegaly/metabolism , Cyclic AMP/metabolism , Myocytes, Cardiac/metabolism , Paracrine Communication , Receptor, Adenosine A1/metabolism , Receptors, Adenosine A2/metabolism , Signal Transduction , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cyclic AMP/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Mice , Mice, Knockout , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Myocytes, Cardiac/pathology , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A1/genetics , Receptors, Adenosine A2/genetics
7.
RNA Biol ; 10(7): 1125-35, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23696004

ABSTRACT

MiRNAs are short, non-coding RNAs that regulate gene expression post-transcriptionally through specific binding to mRNA. Deregulation of miRNAs is associated with various diseases and interference with miRNA function has proven therapeutic potential. Most mRNAs are thought to be regulated by multiple miRNAs and there is some evidence that such joint activity is enhanced if a short distance between sites allows for cooperative binding. Until now, however, the concept of cooperativity among miRNAs has not been addressed in a transcriptome-wide approach. Here, we computationally screened human mRNAs for distances between miRNA binding sites that are expected to promote cooperativity. We find that sites with a maximal spacing of 26 nucleotides are enriched for naturally occurring miRNAs compared with control sequences. Furthermore, miRNAs with similar characteristics as indicated by either co-expression within a specific tissue or co-regulation in a disease context are predicted to target a higher number of mRNAs cooperatively than unrelated miRNAs. These bioinformatic data were compared with genome-wide sets of biochemically validated miRNA targets derived by Argonaute crosslinking and immunoprecipitation (HITS-CLIP and PAR-CLIP). To ease further research into combined and cooperative miRNA function, we developed miRco, a database connecting miRNAs and respective targets involved in distance-defined cooperative regulation (mips.helmholtz-muenchen.de/mirco). In conclusion, our findings suggest that cooperativity of miRNA-target interaction is a widespread phenomenon that may play an important role in miRNA-mediated gene regulation.


Subject(s)
Binding Sites , MicroRNAs/genetics , RNA, Messenger/genetics , Transcriptome , 3' Untranslated Regions , Computational Biology/methods , Gene Expression Regulation , Humans , MicroRNAs/metabolism , RNA, Messenger/metabolism , Software
8.
Circulation ; 127(21): 2097-106, 2013 May 28.
Article in English | MEDLINE | ID: mdl-23625957

ABSTRACT

BACKGROUND: Several microRNAs (miRs) have been shown to regulate gene expression in the heart, and dysregulation of their expression has been linked to cardiac disease. miR-378 is strongly expressed in the mammalian heart but so far has been studied predominantly in cancer, in which it regulates cell survival and tumor growth. METHODS AND RESULTS: Here, we report tight control of cardiomyocyte hypertrophy through miR-378. In isolated primary cardiomyocytes, miR-378 was found to be both necessary and sufficient to repress cardiomyocyte hypertrophy. Bioinformatic prediction suggested that factors of the mitogen-activated protein kinase (MAPK) pathway are enriched among miR-378 targets. Using mRNA and protein expression analysis along with luciferase assays, we validated 4 key components of the MAPK pathway as targets of miR-378: MAPK1 itself, insulin-like growth factor receptor 1, growth factor receptor-bound protein 2, and kinase suppressor of ras 1. RNA interference with these targets prevented the prohypertrophic effect of antimiR-378, suggesting their functional relation with miR-378. Because miR-378 significantly decreases in cardiac disease, we sought to compensate for its loss through adeno-associated virus-mediated, cardiomyocyte-targeted expression of miR-378 in an in vivo model of cardiac hypertrophy (pressure overload by thoracic aortic constriction). Restoration of miR-378 levels significantly attenuated thoracic aortic constriction-induced cardiac hypertrophy and improved cardiac function. CONCLUSIONS: Our data identify miR-378 as a regulator of cardiomyocyte hypertrophy, which exerts its activity by suppressing the MAPK signaling pathway on several distinct levels. Restoration of disease-associated loss of miR-378 through cardiomyocyte-targeted adeno-associated virus-miR-378 may prove to be an effective therapeutic strategy in myocardial disease.


Subject(s)
Cardiomegaly/pathology , Cardiomegaly/physiopathology , MicroRNAs/physiology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/physiology , Signal Transduction/physiology , Adenoviridae/genetics , Animals , Cells, Cultured , Disease Models, Animal , Down-Regulation/physiology , GRB2 Adaptor Protein/antagonists & inhibitors , GRB2 Adaptor Protein/physiology , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/physiology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Protein Kinases/physiology , RNA Interference , Rats , Rats, Sprague-Dawley , Receptors, Somatomedin/antagonists & inhibitors , Receptors, Somatomedin/physiology
9.
J Mol Cell Cardiol ; 52(1): 13-20, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21801730

ABSTRACT

MicroRNAs (miRNAs) are small non-coding RNAs that control expression of complementary target mRNAs. A growing number of miRNAs has been implicated in the pathogenesis of cardiac diseases, mostly based not on functional data, but on the observation that they are dysregulated in diseased myocardium. Consequently, our knowledge regarding a potential cardiac role of the majority of miRNAs is limited. Here, we report the development of an assay format that allows the simultaneous analysis of several hundred molecules with regard to their phenotypic effect on primary rat cardiomyocytes. Using automated microscopy and an edge detection algorithm, this assay achieved high reproducibility and a robust assessment of cardiomyocyte size as a key parameter. Screening a library of synthetic miRNAs revealed several miRNAs previously not recognized as pro- or anti-hypertrophic. Out of these, we selected nine miRNAs and confirmed the pro-hypertrophic potential of miR-22, miR-30c, miR-30d, miR-212, miR-365 and the anti-hypertrophic potential of miR-27a, miR-27b and miR-133a. Quantitative analysis of the expression level of pro-hypertrophic miRNAs in primary cardiomyocytes indicated a rather low level of correlation of the phenotypic effects of individual miRNAs and their expression level. This assay allows the automated determination of cell size in primary cardiomyocytes and permitted the identification of a set of miRNAs capable of regulating cardiomyocyte hypertrophy. Elucidating their mechanism of action should provide insight into mechanisms underlying the cardiomyocyte hypertrophic response. This article is part of a Special Issue entitled 'Possible Editorial'.


Subject(s)
High-Throughput Screening Assays , MicroRNAs/genetics , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Phenotype , Animals , Cell Enlargement , Cell Separation , Cells, Cultured , Gene Expression Profiling , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Transfection
10.
Circulation ; 124(7): 796-805, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21810664

ABSTRACT

BACKGROUND: Cardiomyocytes use Ca2+ not only in excitation-contraction coupling but also as a signaling molecule promoting, for example, cardiac hypertrophy. It is largely unclear how Ca2+ triggers signaling in cardiomyocytes in the presence of the rapid and large Ca2+ fluctuations that occur during excitation-contraction coupling. A potential route is store-operated Ca2+ entry, a drug-inducible mechanism for Ca2+ signaling that requires stromal interaction molecule 1 (STIM1). Store-operated Ca2+ entry can also be induced in cardiomyocytes, which prompted us to study STIM1-dependent Ca2+ entry with respect to cardiac hypertrophy in vitro and in vivo. METHODS AND RESULTS: Consistent with earlier reports, we found drug-inducible store-operated Ca2+ entry in neonatal rat cardiomyocytes, which was dependent on STIM1. Although this STIM1-dependent, drug-inducible store-operated Ca2+ entry was only marginal in adult cardiomyocytes isolated from control hearts, it increased significantly in cardiomyocytes isolated from adult rats that had developed compensated cardiac hypertrophy after abdominal aortic banding. Moreover, we detected an inwardly rectifying current in hypertrophic cardiomyocytes that occurs under native conditions (i.e., in the absence of drug-induced store depletion) and is dependent on STIM1. By manipulating its expression, we found STIM1 to be both sufficient and necessary for cardiomyocyte hypertrophy in vitro and in the adult heart in vivo. Stim1 silencing by adeno-associated viruses of serotype 9-mediated gene transfer protected rats from pressure overload-induced cardiac hypertrophy. CONCLUSION: By controlling a previously unrecognized sarcolemmal current, STIM1 promotes cardiac hypertrophy.


Subject(s)
Calcium Signaling/physiology , Cardiomegaly/physiopathology , Membrane Glycoproteins/physiology , Myocytes, Cardiac/physiology , Adenoviridae/genetics , Age Factors , Animals , Animals, Newborn , Caffeine/pharmacology , Calcium/metabolism , Calcium Channels , Calcium Signaling/drug effects , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Gene Silencing , Gene Transfer Techniques , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Patch-Clamp Techniques , Phosphodiesterase Inhibitors/pharmacology , Rats , Sarcolemma/metabolism , Stromal Interaction Molecule 1 , Thapsigargin/pharmacology
11.
Hum Mol Genet ; 20(2): 368-77, 2011 Jan 15.
Article in English | MEDLINE | ID: mdl-21051334

ABSTRACT

Retinitis pigmentosa (RP) is a common hereditary eye disease that causes blindness due to a progressive loss of photoreceptors in the retina. RP can be elicited by mutations that affect the tri-snRNP subunit of the pre-mRNA splicing machinery, but how defects in this essential macromolecular complex transform into a photoreceptor-specific phenotype is unknown. We have modeled the disease in zebrafish by silencing the RP-associated splicing factor Prpf31 and observed detrimental effects on visual function and photoreceptor morphology. Despite reducing the level of a constitutive splicing factor, no general defects in gene expression were found. Instead, retinal genes were selectively affected, providing the first in vivo link between mutations in splicing factors and the RP phenotype. Silencing of Prpf4, a splicing factor hitherto unrelated to RP, evoked the same defects in vision, photoreceptor morphology and retinal gene expression. Hence, various routes affecting the tri-snRNP can elicit tissue-specific gene expression defects and lead to the RP phenotype.


Subject(s)
Disease Models, Animal , Eye Proteins/genetics , Eye Proteins/metabolism , Retinitis Pigmentosa/pathology , Zebrafish , Animals , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Haploinsufficiency/genetics , Mutation , Organ Specificity , Phenotype , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , RNA Splicing/genetics , Retina/metabolism , Retina/physiopathology , Retinitis Pigmentosa/genetics
12.
Hum Mol Genet ; 18(7): 1288-300, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19158098

ABSTRACT

Distal spinal muscular atrophy type 1 (DSMA1) is an autosomal recessive disease that is clinically characterized by distal limb weakness and respiratory distress. In this disease, the degeneration of alpha-motoneurons is caused by mutations in the immunoglobulin mu-binding protein 2 (IGHMBP2). This protein has been implicated in DNA replication, pre-mRNA splicing and transcription, but its precise function in all these processes has remained elusive. We have purified catalytically active recombinant IGHMBP2, which has enabled us to assess its enzymatic properties and to identify its cellular targets. Our data reveal that IGHMBP2 is an ATP-dependent 5' --> 3' helicase, which unwinds RNA and DNA duplices in vitro. Importantly, this helicase localizes predominantly to the cytoplasm of neuronal and non-neuronal cells and associates with ribosomes. DSMA1-causing amino acid substitutions in IGHMBP2 do not affect ribosome binding yet severely impair ATPase and helicase activity. We propose that IGHMBP2 is functionally linked to translation, and that mutations in its helicase domain interfere with this function in DSMA1 patients.


Subject(s)
DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Muscular Atrophy, Spinal/enzymology , Ribosomes/enzymology , Transcription Factors/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Extracts , Cell Line, Tumor , DNA Helicases/chemistry , DNA-Binding Proteins/chemistry , Enzyme Activation , Humans , Mice , Mutant Proteins/metabolism , Protein Binding , Ribonucleoproteins/metabolism , Transcription Factors/chemistry
13.
Hum Mol Genet ; 17(20): 3236-46, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18664458

ABSTRACT

Tudor domains are widespread among proteins involved in RNA metabolism, but only in a few cases their cellular function has been analyzed in detail. Here, we report on the characterization of the ubiquitously expressed Tudor domain containing protein Tdrd3. Apart from its Tudor domain, we show that Tdrd3 possesses an oligosaccharide/nucleotide binding fold (OB-fold) and an ubiquitin associated domain capable of binding tetra-ubiquitin. A set of biochemical experiments revealed an interaction of Tdrd3 with FMRP, the product of the gene affected in Fragile X syndrome, and its autosomal homologs FXR1 and FXR2. FMRP has been implicated in the translational regulation of target mRNAs and shown to be a component of stress granules (SG). We demonstrate that overexpression of Tdrd3 in cells induces the formation of SGs and as a result leads to its co-localization with endogenous FMRP in these structures. Interestingly, the disease-associated FMRP missense mutation I304N identified in a Fragile X patient severely impairs the interaction with Tdrd3 in biochemical experiments. We propose a contribution of Tdrd3 to FMRP-mediated translational repression and suggest that the loss of the FMRP-Tdrd3 interaction caused by the I304N mutation might contribute to the pathogenesis of Fragile X syndrome.


Subject(s)
Fragile X Mental Retardation Protein/genetics , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/genetics , Fragile X Syndrome/metabolism , RNA-Binding Proteins/metabolism , Amino Acid Sequence , Amino Acid Substitution , Cytosol/metabolism , Fragile X Syndrome/etiology , HeLa Cells , Humans , In Vitro Techniques , Molecular Sequence Data , Mutation, Missense , Protein Binding , Protein Interaction Domains and Motifs , Protein Interaction Mapping , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
14.
Trends Mol Med ; 12(3): 113-21, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16473550

ABSTRACT

Degenerated motor neurons in the spinal cord are the pathological hallmark of spinal muscular atrophy (SMA). SMA is caused by mutations in the ubiquitously expressed survival motor neuron 1 (SMN1) gene, which lead to reduced levels of functional SMN protein. Many different functions have been assigned to SMN, including assembly of ribonucleoproteins (RNPs), splicing, transcription and axonal mRNA transport. Recently, tissue from SMA patients and animal models has been used to determine which function of SMN is affected in SMA patients. A surprising picture has emerged: the impaired assembly of RNP subunits of the spliceosome seems to be responsible for SMA pathogenesis. Here, we present a model of how this defect might cause motor-neuron degeneration and consider potential therapies.


Subject(s)
Cyclic AMP Response Element-Binding Protein , Muscular Atrophy, Spinal , Nerve Tissue Proteins , RNA-Binding Proteins , Ribonucleoproteins/metabolism , Alternative Splicing , Animals , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Humans , Motor Neurons/cytology , Motor Neurons/metabolism , Motor Neurons/pathology , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/physiopathology , Muscular Atrophy, Spinal/therapy , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribonucleoproteins/genetics , SMN Complex Proteins , Survival of Motor Neuron 1 Protein
15.
Genes Dev ; 19(19): 2320-30, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16204184

ABSTRACT

Spinal muscular atrophy (SMA) is a motoneuron disease caused by reduced levels of survival motoneuron (SMN) protein. Previous studies have assigned SMN to uridine-rich small nuclear ribonucleoprotein particle (U snRNP) assembly, splicing, transcription, and RNA localization. Here, we have used gene silencing to assess the effect of SMN protein deficiency on U snRNP metabolism in living cells and organisms. In HeLa cells, we show that reduction of SMN to levels found in SMA patients impairs U snRNP assembly. In line with this, induced silencing of SMN expression in Xenopus laevis or zebrafish arrested embryonic development. Under less severe knock-down conditions, zebrafish embryos proceeded through development yet exhibited dramatic SMA-like motor axon degeneration. The same was observed after silencing two other essential factors in the U snRNP assembly pathway, Gemin2 and pICln. Importantly, the injection of purified U snRNPs into either SMN- or Gemin2-deficient embryos of Xenopus and zebrafish prevented developmental arrest and motoneuron degeneration, respectively. These findings suggest that motoneuron degeneration in SMA patients is a direct consequence of impaired production of U snRNPs.


Subject(s)
Muscular Atrophy, Spinal/metabolism , Nerve Tissue Proteins/metabolism , RNA Interference , Ribonucleoproteins, Small Nuclear/metabolism , Zebrafish/embryology , Animals , Axons/metabolism , Axons/pathology , Disease Models, Animal , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/pathology , Fibroblasts/metabolism , HeLa Cells , Humans , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Nerve Tissue Proteins/genetics , RNA-Binding Proteins , Ribonucleoproteins, Small Nuclear/genetics , Xenopus laevis , Zebrafish/genetics
16.
RNA ; 11(5): 598-608, 2005 May.
Article in English | MEDLINE | ID: mdl-15840814

ABSTRACT

The U5 snRNP plays an essential role in both U2- and U12-dependent splicing. Here, we have characterized a 52-kDa protein associated with the human U5 snRNP, designated U5-52K. Protein sequencing revealed that U5-52K is identical to the CD2BP2, which interacts with the cytoplasmic portion of the human T-cell surface protein CD2. Consistent with it associating with an snRNP, immunofluorescence studies demonstrated that the 52K protein is predominantly located in the nucleoplasm of HeLa cells, where it overlaps, at least in part, with splicing-factor compartments (or "speckles"). We further demonstrate that the 52K protein is a constituent of the 20S U5 snRNP, but is not found in U4/U6.U5 tri-snRNPs. Thus, it is the only 20S U5-specific protein that is not integrated into the tri-snRNP and resembles, in this respect, the U4/U6 di-snRNP assembly factor Prp24p/p110. Yeast two-hybrid screening and pulldown assays revealed that the 52K protein interacts with the U5-specific 102K and 15K proteins, suggesting that these interactions are responsible for its integration into the U5 particle. The N-terminal two-thirds of 52K interact with the 102K protein, whereas its C-terminal GYF-domain binds the 15K protein. As the latter lacks a proline-rich tract, our data indicate that a GYF-domain can also engage in specific protein-protein interactions in a polyproline-independent manner. Interestingly, the U5-102K protein has been shown previously to play an essential role in tri-snRNP formation, binding the U4/U6-61K protein. The interaction of 52K with a tri-snRNP bridging protein, coupled with its absence from the tri-snRNP, suggests it might function in tri-snRNP assembly.


Subject(s)
Carrier Proteins/metabolism , Ribonucleoprotein, U4-U6 Small Nuclear/metabolism , Ribonucleoprotein, U5 Small Nuclear/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Carrier Proteins/chemistry , Cell Nucleus/metabolism , Conserved Sequence , HeLa Cells , Humans , Mitochondrial Precursor Protein Import Complex Proteins , Molecular Sequence Data , Molecular Weight , Multiprotein Complexes/chemistry , Multiprotein Complexes/metabolism , Protein Binding , Protein Transport , Ribonucleoprotein, U4-U6 Small Nuclear/chemistry , Ribonucleoprotein, U5 Small Nuclear/chemistry , Substrate Specificity , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...