Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 52(8): 4167-4184, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38324473

ABSTRACT

Sam68 and SLM2 are paralog RNA binding proteins (RBPs) expressed in the cerebral cortex and display similar splicing activities. However, their relative functions during cortical development are unknown. We found that these RBPs exhibit an opposite expression pattern during development. Sam68 expression declines postnatally while SLM2 increases after birth, and this developmental pattern is reinforced by hierarchical control of Sam68 expression by SLM2. Analysis of Sam68:Slm2 double knockout (Sam68:Slm2dko) mice revealed hundreds of exons that respond to joint depletion of these proteins. Moreover, parallel analysis of single and double knockout cortices indicated that exons regulated mainly by SLM2 are characterized by a dynamic splicing pattern during development, whereas Sam68-dependent exons are spliced at relatively constant rates. Dynamic splicing of SLM2-sensitive exons is completely suppressed in the Sam68:Slm2dko developing cortex. Sam68:Slm2dko mice die perinatally with defects in neurogenesis and in neuronal differentiation, and develop a hydrocephalus, consistent with splicing alterations in genes related to these biological processes. Thus, our study reveals that developmental control of separate Sam68 and Slm2 paralog genes encoding homologous RBPs enables the orchestration of a dynamic splicing program needed for brain development and viability, while ensuring a robust redundant mechanism that supports proper cortical development.


Subject(s)
Cerebral Cortex , RNA Splicing , RNA-Binding Proteins , Animals , Mice , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Exons/genetics , Gene Expression Regulation, Developmental , Mice, Knockout , Neurogenesis/genetics , Neurons/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
3.
J Exp Clin Cancer Res ; 42(1): 214, 2023 Aug 21.
Article in English | MEDLINE | ID: mdl-37599362

ABSTRACT

BACKGROUND: Medulloblastoma (MB) is the most common cerebellar malignancy during childhood. Among MB, MYC-amplified Group 3 tumors display the worst prognosis. MYC is an oncogenic transcription factor currently thought to be undruggable. Nevertheless, targeting MYC-dependent processes (i.e. transcription and RNA processing regulation) represents a promising approach. METHODS: We have tested the sensitivity of MYC-driven Group 3 MB cells to a pool of transcription and splicing inhibitors that display a wide spectrum of targets. Among them, we focus on THZ531, an inhibitor of the transcriptional cyclin-dependent kinases (CDK) 12 and 13. High-throughput RNA-sequencing analyses followed by bioinformatics and functional analyses were carried out to elucidate the molecular mechanism(s) underlying the susceptibility of Group 3 MB to CDK12/13 chemical inhibition. Data from International Cancer Genome Consortium (ICGC) and other public databases were mined to evaluate the functional relevance of the cellular pathway/s affected by the treatment with THZ531 in Group 3 MB patients. RESULTS: We found that pharmacological inhibition of CDK12/13 is highly selective for MYC-high Group 3 MB cells with respect to MYC-low MB cells. We identified a subset of genes enriched in functional terms related to the DNA damage response (DDR) that are up-regulated in Group 3 MB and repressed by CDK12/13 inhibition. Accordingly, MYC- and CDK12/13-dependent higher expression of DDR genes in Group 3 MB cells limits the toxic effects of endogenous DNA lesions in these cells. More importantly, chemical inhibition of CDK12/13 impaired the DDR and induced irreparable DNA damage exclusively in MYC-high Group 3 MB cells. The augmented sensitivity of MYC-high MB cells to CDK12/13 inhibition relies on the higher elongation rate of the RNA polymerase II in DDR genes. Lastly, combined treatments with THZ531 and DNA damage-inducing agents synergically suppressed viability of MYC-high Group 3 MB cells. CONCLUSIONS: Our study demonstrates that CDK12/13 activity represents an exploitable vulnerability in MYC-high Group 3 MB and may pave the ground for new therapeutic approaches for this high-risk brain tumor.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Humans , Medulloblastoma/drug therapy , Medulloblastoma/genetics , Up-Regulation , Anilides , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/genetics , CDC2 Protein Kinase , Cyclin-Dependent Kinases/genetics
4.
Int J Mol Sci ; 23(5)2022 Mar 04.
Article in English | MEDLINE | ID: mdl-35269953

ABSTRACT

The advance of experimental and computational techniques has allowed us to highlight the existence of numerous different mechanisms of RNA maturation, which have been so far unknown. Besides canonical splicing, consisting of the removal of introns from pre-mRNA molecules, non-canonical splicing events may occur to further increase the regulatory and coding potential of the human genome. Among these, splicing of microexons, recursive splicing and biogenesis of circular and chimeric RNAs through back-splicing and trans-splicing processes, respectively, all contribute to expanding the repertoire of RNA transcripts with newly acquired regulatory functions. Interestingly, these non-canonical splicing events seem to occur more frequently in the central nervous system, affecting neuronal development and differentiation programs with important implications on brain physiology. Coherently, dysregulation of non-canonical RNA processing events is associated with brain disorders, including brain tumours. Herein, we summarize the current knowledge on molecular and regulatory mechanisms underlying canonical and non-canonical splicing events with particular emphasis on cis-acting elements and trans-acting factors that all together orchestrate splicing catalysis reactions and decisions. Lastly, we review the impact of non-canonical splicing on brain physiology and pathology and how unconventional splicing mechanisms may be targeted or exploited for novel therapeutic strategies in cancer.


Subject(s)
Neoplasms , RNA Splicing , Alternative Splicing/genetics , Brain/metabolism , Humans , Introns , Neoplasms/genetics , RNA/genetics , RNA Precursors/genetics , RNA Splicing/genetics
5.
Biomolecules ; 11(10)2021 10 07.
Article in English | MEDLINE | ID: mdl-34680108

ABSTRACT

Signal transduction pathways transmit the information received from external and internal cues and generate a response that allows the cell to adapt to changes in the surrounding environment. Signaling pathways trigger rapid responses by changing the activity or localization of existing molecules, as well as long-term responses that require the activation of gene expression programs. All steps involved in the regulation of gene expression, from transcription to processing and utilization of new transcripts, are modulated by multiple signal transduction pathways. This review provides a broad overview of the post-translational regulation of factors involved in RNA processing events by signal transduction pathways, with particular focus on the regulation of pre-mRNA splicing, cleavage and polyadenylation. The effects of several post-translational modifications (i.e., sumoylation, ubiquitination, methylation, acetylation and phosphorylation) on the expression, subcellular localization, stability and affinity for RNA and protein partners of many RNA-binding proteins are highlighted. Moreover, examples of how some of the most common signal transduction pathways can modulate biological processes through changes in RNA processing regulation are illustrated. Lastly, we discuss challenges and opportunities of therapeutic approaches that correct RNA processing defects and target signaling molecules.


Subject(s)
Polyadenylation/genetics , RNA Processing, Post-Transcriptional/genetics , RNA/genetics , Signal Transduction/genetics , Alternative Splicing/genetics , Humans , Methylation , Phosphorylation/genetics , RNA-Binding Proteins/genetics , Sumoylation/genetics , Ubiquitination/genetics
6.
J Neurochem ; 157(4): 1153-1166, 2021 05.
Article in English | MEDLINE | ID: mdl-32959393

ABSTRACT

Neural Progenitor Cells (NPCs) are multipotent cells that are able to self-renew and differentiate into neurons. The size of the initial pool of NPCs during the brain development strongly affects the number of neurons that compose cortical multi-layer during development. Gonadal hormones can influence the balance between self-renewal and differentiation processes. Herein, we investigated the role of dihydrotestosterone (DHT), the active metabolite of testosterone, in the regulation of NPC stemness and differentiation. First, we evaluated the expression of the androgen receptor (AR), the transcription factor activated by DHT that mediates the physiological effects of androgens, in NPCs. Western blot analysis showed that DHT-mediated activation of AR induces mitogenic signaling pathways (PI3K/AKT and MAPK/ERK) in NPCs, whereas luciferase activity assays demonstrated the induction of AR transcriptional activity. AR activation mediated by DHT treatment strongly increased the proliferation of NPCs and reduced their propensity to differentiate into neurons. Furthermore, the effects of AR activation were mediated, at least in part, by increased expression of Aldehyde Dehydrogenase 1 Family Member A3 enzyme (ALDH1A3). Pharmacological inhibition of ALDH activity with N,N-diethylaminobenzaldehyde (DEAB) reduced the effect of DHT on NPC proliferation in vitro. Furthermore, inhibition of AR activity by Enzalutamide reduced the NPC pool in the developing cortex of male C57/BL6 mouse embryos. These findings indicate that androgens engage an AR-dependent signaling pathway that impact on neurogenesis by increasing the NPC pool in the developing mouse cortex.


Subject(s)
Cerebral Cortex/embryology , Neural Stem Cells/metabolism , Neurogenesis/physiology , Receptors, Androgen/metabolism , Signal Transduction/physiology , Androgens/pharmacology , Animals , Dihydrotestosterone/pharmacology , Male , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology
7.
Cell Rep ; 31(9): 107703, 2020 06 02.
Article in English | MEDLINE | ID: mdl-32492419

ABSTRACT

Tight coordination of gene expression in the developing cerebellum is crucial for establishment of neuronal circuits governing motor and cognitive function. However, transcriptional changes alone do not explain all of the switches underlying neuronal differentiation. Here we unveiled a widespread and highly dynamic splicing program that affects synaptic genes in cerebellar neurons. The motifs enriched in modulated exons implicated the splicing factor Sam68 as a regulator of this program. Sam68 controls splicing of exons with weak branchpoints by directly binding near the 3' splice site and competing with U2AF recruitment. Ablation of Sam68 disrupts splicing regulation of synaptic genes associated with neurodevelopmental diseases and impairs synaptic connections and firing of Purkinje cells, resulting in motor coordination defects, ataxia, and abnormal social behavior. These findings uncover an unexpectedly dynamic splicing regulatory network that shapes the synapse in early life and establishes motor and cognitive circuitry in the developing cerebellum.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cerebellum/metabolism , RNA Splicing , RNA-Binding Proteins/metabolism , Synapses/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Behavior, Animal , Cerebellum/cytology , Cerebellum/growth & development , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Purkinje Cells/metabolism , RNA Splice Sites , RNA-Binding Proteins/genetics , Splicing Factor U2AF/metabolism
8.
Cell Death Dis ; 11(4): 240, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32303676

ABSTRACT

Homologous recombination and chromosome segregation in meiosis rely on the timely expression of two splice variants of the endonuclease SPO11, named α and ß, which respectively skip or include exon 2. However, in spite of its physiological importance, the mechanism underlying Spo11 alternative splicing in meiosis is still unknown. By screening the activity of factors that are predicted to bind the alternatively spliced region of Spo11, we identified hnRNPH as a key regulator of SPO11α splicing in mouse spermatocytes. Although hnRNPH was not upregulated in meiosis concomitantly with the switch in splicing, its recruitment to Spo11 pre-mRNA was favored by selective modulation of RNA polymerase II (RNAPII) phosphorylation and processivity in proximity of exon 2. The hnRNPH binding sites were localized near those of splicing factors that promote SPO11ß splicing, suggesting that hnRNPH favors exon 2 skipping by competing out positive regulators. Indeed, hnRNPH binds proximal to a consensus motif for Sam68, a positive regulator of SPO11ß splicing in vitro and in vivo, and it interferes with Sam68 binding to the Spo11 pre-mRNA. Thus, our work reveals that modulation of RNAPII dynamics in concert with hnRNPH recruitment exerts a combinatorial control of the timely regulated Spo11 splicing during meiosis.


Subject(s)
Alternative Splicing/genetics , Endodeoxyribonucleases/metabolism , Meiosis/genetics , RNA Polymerase II/genetics , Spermatocytes/metabolism , Spermatogenesis/genetics , Animals , Humans , Male , Mice , RNA Polymerase II/metabolism , RNA Splicing Factors
9.
J Neurochem ; 153(2): 264-275, 2020 04.
Article in English | MEDLINE | ID: mdl-31811660

ABSTRACT

Spinal muscular atrophy (SMA) is a motor neuron disease caused by loss of function mutations in the Survival Motor Neuron 1 (SMN1) gene and reduced expression of the SMN protein, leading to spinal motor neuron death, muscle weakness and atrophy. Although humans harbour the highly homologous SMN2 gene, its defective splicing regulation yields a truncated and unstable SMN protein. The first therapy for SMA was recently approved by the Food and Drug Administration and consists of an antisense oligonucleotide (Nusinersen) rendering SMN2 functional and thus improving patients' motor activity and quality of life. Nevertheless, not all patients equally respond to this therapy and the long-term tolerability and safety of Nusinersen are still unknown. Herein, in vivo splicing assays indicated that the HDAC inhibitor LBH589 is particularly efficient in rescuing the SMN2 splicing defect in SMA fibroblasts and SMA type-I mice-derived neural stem cells. Western blot analyses showed that LBH589 also causes a significant increase in SMN protein expression in SMA cells. Moreover chromatin immunoprecipitation analyses revealed that LBH589 treatment induces widespread H4 acetylation of the entire SMN2 locus and selectively favors the inclusion of the disease-linked exon 7 in SMN2 mature mRNA. The combined treatment of SMA cells with sub-optimal doses of LBH589 and of an antisense oligonucleotide that mimic Nusinersen (ASO_ISSN1) elicits additive effects on SMN2 splicing and SMN protein expression. These findings suggest that HDAC inhibitors can potentiate the activity of Nusinersen and support the notion that 'SMN-plus' combinatorial therapeutic approaches might represent an enhanced opportunity in the scenario of SMA therapy.


Subject(s)
Muscular Atrophy, Spinal , Oligonucleotides/pharmacology , Panobinostat/pharmacology , RNA Splicing/drug effects , Survival of Motor Neuron 2 Protein/biosynthesis , Animals , Drug Therapy, Combination , Female , Fibroblasts/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Male , Mice , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/metabolism , Neural Stem Cells/drug effects , Oligonucleotides, Antisense/pharmacology , RNA Splicing/genetics , Survival of Motor Neuron 2 Protein/genetics
10.
Nucleic Acids Res ; 48(2): 633-645, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31777926

ABSTRACT

The Spinal Muscular Atrophy (SMA) gene SMN was recently duplicated (SMN1 and SMN2) in higher primates. Furthermore, invasion of the locus by repetitive elements almost doubled its size with respect to mouse Smn, in spite of an almost identical protein-coding sequence. Herein, we found that SMN ranks among the human genes with highest density of Alus, which are evolutionary conserved in primates and often occur in inverted orientation. Inverted repeat Alus (IRAlus) negatively regulate splicing of long introns within SMN, while promoting widespread alternative circular RNA (circRNA) biogenesis. Bioinformatics analyses revealed the presence of ultra-conserved Sam68 binding sites in SMN IRAlus. Cross-link-immunoprecipitation (CLIP), mutagenesis and silencing experiments showed that Sam68 binds in proximity of intronic Alus in the SMN pre-mRNA, thus favouring circRNA biogenesis in vitro and in vivo. These findings highlight a novel layer of regulation in SMN expression, uncover the crucial impact exerted by IRAlus and reveal a role for Sam68 in SMN circRNA biogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Alu Elements/genetics , DNA-Binding Proteins/genetics , Muscular Atrophy, Spinal/genetics , RNA, Circular/genetics , RNA-Binding Proteins/genetics , Alternative Splicing/genetics , Animals , Binding Sites/genetics , Exons/genetics , Humans , Introns/genetics , Mice , Muscular Atrophy, Spinal/pathology , RNA Precursors/genetics , SMN Complex Proteins/genetics , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 2 Protein/genetics
11.
Cells ; 9(1)2019 12 18.
Article in English | MEDLINE | ID: mdl-31861467

ABSTRACT

Brain tumors are a heterogeneous group of neoplasms ranging from almost benign to highly aggressive phenotypes. The malignancy of these tumors mostly relies on gene expression reprogramming, which is frequently accompanied by the aberrant regulation of RNA processing mechanisms. In brain tumors, defects in alternative splicing result either from the dysregulation of expression and activity of splicing factors, or from mutations in the genes encoding splicing machinery components. Aberrant splicing regulation can generate dysfunctional proteins that lead to modification of fundamental physiological cellular processes, thus contributing to the development or progression of brain tumors. Herein, we summarize the current knowledge on splicing abnormalities in brain tumors and how these alterations contribute to the disease by sustaining proliferative signaling, escaping growth suppressors, or establishing a tumor microenvironment that fosters angiogenesis and intercellular communications. Lastly, we review recent efforts aimed at developing novel splicing-targeted cancer therapies, which employ oligonucleotide-based approaches or chemical modulators of alternative splicing that elicit an impact on brain tumor biology.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , RNA Splicing/genetics , Alternative Splicing/genetics , Alternative Splicing/physiology , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Disease Progression , Gene Expression Regulation, Neoplastic/genetics , Humans , Mutation/genetics , Oncogenes/genetics , Oncogenes/physiology , RNA Splicing/physiology , RNA Splicing Factors/genetics , Tumor Microenvironment/genetics
12.
Nat Cell Biol ; 20(8): 917-927, 2018 08.
Article in English | MEDLINE | ID: mdl-30050118

ABSTRACT

Fibro-adipogenic progenitors (FAPs) are typically activated in response to muscle injury, and establish functional interactions with inflammatory and muscle stem cells (MuSCs) to promote muscle repair. We found that denervation causes progressive accumulation of FAPs, without concomitant infiltration of macrophages and MuSC-mediated regeneration. Denervation-activated FAPs exhibited persistent STAT3 activation and secreted elevated levels of IL-6, which promoted muscle atrophy and fibrosis. FAPs with aberrant activation of STAT3-IL-6 signalling were also found in mouse models of spinal cord injury, spinal muscular atrophy, amyotrophic lateral sclerosis (ALS) and in muscles of ALS patients. Inactivation of STAT3-IL-6 signalling in FAPs effectively countered muscle atrophy and fibrosis in mouse models of acute denervation and ALS (SODG93A mice). Activation of pathogenic FAPs following loss of integrity of neuromuscular junctions further illustrates the functional versatility of FAPs in response to homeostatic perturbations and suggests their potential contribution to the pathogenesis of neuromuscular diseases.


Subject(s)
Adipogenesis , Amyotrophic Lateral Sclerosis/metabolism , Denervation/methods , Interleukin-6/metabolism , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy/metabolism , Myoblasts, Skeletal/metabolism , Quadriceps Muscle/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Spinal Cord Injuries/metabolism , Adipogenesis/drug effects , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Amyotrophic Lateral Sclerosis/prevention & control , Animals , Cardiotoxins , Cell Line , Coculture Techniques , Disease Models, Animal , Fibrosis , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Male , Mice, Inbred C57BL , Mice, Transgenic , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Muscular Atrophy/prevention & control , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/prevention & control , Mutation , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/pathology , Neuromuscular Agents/pharmacology , Quadriceps Muscle/drug effects , Quadriceps Muscle/innervation , Quadriceps Muscle/pathology , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Sciatic Nerve/surgery , Spinal Cord Injuries/genetics , Spinal Cord Injuries/pathology , Spinal Cord Injuries/prevention & control , Superoxide Dismutase-1/genetics
13.
EMBO Rep ; 19(7)2018 07.
Article in English | MEDLINE | ID: mdl-29752334

ABSTRACT

Heat-shock factor 1 (HSF1) is the master transcription factor that regulates the response to proteotoxic stress by controlling the transcription of many stress-responsive genes including the heat-shock proteins. Here, we show a novel molecular mechanism controlling the activation of HSF1. We demonstrate that transglutaminase type 2 (TG2), dependent on its protein disulphide isomerase activity, triggers the trimerization and activation of HSF1 regulating adaptation to stress and proteostasis impairment. In particular, we find that TG2 loss of function correlates with a defect in the nuclear translocation of HSF1 and in its DNA-binding ability to the HSP70 promoter. We show that the inhibition of TG2 restores the unbalance in HSF1-HSP70 pathway in cystic fibrosis (CF), a human disorder characterized by deregulation of proteostasis. The absence of TG2 leads to an increase of about 40% in CFTR function in a new experimental CF mouse model lacking TG2. Altogether, these results indicate that TG2 plays a key role in the regulation of cellular proteostasis under stressful cellular conditions through the modulation of the heat-shock response.


Subject(s)
Cystic Fibrosis/genetics , DNA-Binding Proteins/genetics , GTP-Binding Proteins/genetics , Heat Shock Transcription Factors/genetics , Transglutaminases/genetics , Animals , Cystic Fibrosis/pathology , Gene Expression Regulation , Heat-Shock Response/genetics , Humans , Mice , Promoter Regions, Genetic , Protein Binding , Protein Disulfide-Isomerases/genetics , Protein Glutamine gamma Glutamyltransferase 2 , Protein Processing, Post-Translational/genetics , Proteostasis/genetics , Signal Transduction
14.
Hum Genet ; 136(9): 1215-1235, 2017 09.
Article in English | MEDLINE | ID: mdl-28434044

ABSTRACT

Alternative splicing is a powerful mechanism that largely expands the coding potential of eukaryotic genomes. Indeed, its complex and flexible regulation is exploited by cells to adapt to various environmental conditions, through production of protein variants displaying different functions. Such flexibility, however, is accompanied by high risk of errors, and dysregulation of splicing is now recognized as an important factor in human diseases. Notably, the RNA-based nature of splicing, which involves high specificity through base pair recognition, offers a remarkable therapeutic opportunity by allowing design of tools with elevated target selectivity. Herein, we illustrate examples of how defective splicing, obtained by mutations affecting multiple layers of regulation, can result in pathology. In particular, we focus on splicing-related defects occurring in brain and muscle diseases and describe therapeutic approaches currently available for these pathologies.


Subject(s)
Brain Diseases , Muscular Diseases , RNA Splicing , Animals , Brain Diseases/genetics , Brain Diseases/metabolism , Humans , Muscular Diseases/genetics , Muscular Diseases/metabolism
15.
Int J Cardiol ; 232: 233-242, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28089144

ABSTRACT

OBJECTIVE: Elevated aldosterone is associated with increased risk of atherosclerosis complications, whereas treatment with mineralocorticoid receptor (MR) antagonists decreases the rate of cardiovascular events. Here we test the hypothesis that aldosterone promotes early atherosclerosis by modulating intercellular adhesion molecule-1 (ICAM-1) expression and investigate the molecular mechanisms by which aldosterone regulates ICAM-1 expression. METHODS AND RESULTS: Apolipoprotein-E (ApoE)-/- mice fed an atherogenic diet and treated with aldosterone for 4weeks showed increased vascular expression of ICAM-1, paralleled by enhanced atherosclerotic plaque size in the aortic root. Moreover, aldosterone treatment resulted in increased plaque lipid and inflammatory cell content, consistent with an unstable plaque phenotype. ApoE/ICAM-1 double knockout (ApoE-/-/ICAM-1-/-) littermates were protected from the aldosterone-induced increase in plaque size, lipid content and macrophage infiltration. Since aldosterone is known to regulate ICAM-1 transcription via MR in human endothelial cells, we explored MR regulation of the ICAM-1 promoter. Luciferase reporter assays performed in HUVECs using deletion constructs of the human ICAM-1 gene promoter showed that a region containing a predicted MR-responsive element (MRE) is required for MR-dependent transcriptional regulation of ICAM-1. CONCLUSIONS: Pro-atherogenic effects of aldosterone are mediated by increased ICAM-1 expression, through transcriptional regulation by endothelial MR. These data enhance our understanding of the molecular mechanism by which MR activation promotes atherosclerosis complications.


Subject(s)
Atherosclerosis/genetics , Gene Expression Regulation , Intercellular Adhesion Molecule-1/genetics , RNA/genetics , Aldosterone/toxicity , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Blotting, Western , Cells, Cultured , Disease Models, Animal , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Flow Cytometry , Genotype , Immunohistochemistry , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Mineralocorticoid/metabolism
16.
Nucleic Acids Res ; 45(7): 4120-4130, 2017 04 20.
Article in English | MEDLINE | ID: mdl-27994030

ABSTRACT

SLM2 and Sam68 are splicing regulator paralogs that usually overlap in function, yet only SLM2 and not Sam68 controls the Neurexin2 AS4 exon important for brain function. Herein we find that SLM2 and Sam68 similarly bind to Neurexin2 pre-mRNA, both within the mouse cortex and in vitro. Protein domain-swap experiments identify a region including the STAR domain that differentiates SLM2 and Sam68 activity in splicing target selection, and confirm that this is not established via the variant amino acids involved in RNA contact. However, far fewer SLM2 and Sam68 RNA binding sites flank the Neurexin2 AS4 exon, compared with those flanking the Neurexin1 and Neurexin3 AS4 exons under joint control by both Sam68 and SLM2. Doubling binding site numbers switched paralog sensitivity, by placing the Neurexin2 AS4 exon under joint splicing control by both Sam68 and SLM2. Our data support a model where the density of shared RNA binding sites around a target exon, rather than different paralog-specific protein-RNA binding sites, controls functional target specificity between SLM2 and Sam68 on the Neurexin2 AS4 exon. Similar models might explain differential control by other splicing regulators within families of paralogs with indistinguishable RNA binding sites.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Nerve Tissue Proteins/genetics , RNA-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Alternative Splicing , Animals , Binding Sites , Exons , Introns , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Protein Domains , RNA Precursors/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Substrate Specificity
17.
Cell Rep ; 17(12): 3269-3280, 2016 12 20.
Article in English | MEDLINE | ID: mdl-28009295

ABSTRACT

The brain is made up of trillions of synaptic connections that together form neural networks needed for normal brain function and behavior. SLM2 is a member of a conserved family of RNA binding proteins, including Sam68 and SLM1, that control splicing of Neurexin1-3 pre-mRNAs. Whether SLM2 affects neural network activity is unknown. Here, we find that SLM2 levels are maintained by a homeostatic feedback control pathway that predates the divergence of SLM2 and Sam68. SLM2 also controls the splicing of Tomosyn2, LysoPLD/ATX, Dgkb, Kif21a, and Cask, each of which are important for synapse function. Cortical neural network activity dependent on synaptic connections between SLM2-expressing-pyramidal neurons and interneurons is decreased in Slm2-null mice. Additionally, these mice are anxious and have a decreased ability to recognize novel objects. Our data reveal a pathway of SLM2 homeostatic auto-regulation controlling brain network activity and behavior.


Subject(s)
Alternative Splicing/genetics , Nerve Net , Pyramidal Cells/metabolism , RNA-Binding Proteins/genetics , Synapses/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Behavior, Animal/physiology , Calcium-Binding Proteins , Homeostasis/genetics , Mice , Mice, Knockout , Neural Cell Adhesion Molecules/genetics , RNA Precursors/genetics , RNA-Binding Proteins/metabolism , Synapses/physiology
18.
J Cell Biol ; 211(1): 77-90, 2015 Oct 12.
Article in English | MEDLINE | ID: mdl-26438828

ABSTRACT

Spinal muscular atrophy (SMA) is a neurodegenerative disease caused by loss of motor neurons in patients with null mutations in the SMN1 gene. The almost identical SMN2 gene is unable to compensate for this deficiency because of the skipping of exon 7 during pre-messenger RNA (mRNA) processing. Although several splicing factors can modulate SMN2 splicing in vitro, the physiological regulators of this disease-causing event are unknown. We found that knockout of the splicing factor SAM68 partially rescued body weight and viability of SMAΔ7 mice. Ablation of SAM68 function promoted SMN2 splicing and expression in SMAΔ7 mice, correlating with amelioration of SMA-related defects in motor neurons and skeletal muscles. Mechanistically, SAM68 binds to SMN2 pre-mRNA, favoring recruitment of the splicing repressor hnRNP A1 and interfering with that of U2AF65 at the 3' splice site of exon 7. These findings identify SAM68 as the first physiological regulator of SMN2 splicing in an SMA mouse model.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Muscular Atrophy, Spinal/metabolism , RNA-Binding Proteins/physiology , Survival of Motor Neuron 2 Protein/metabolism , Animals , Base Sequence , Female , HEK293 Cells , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Motor Neurons/metabolism , Muscular Atrophy, Spinal/genetics , RNA Splicing , Spinal Cord/pathology
19.
Biomed Res Int ; 2015: 543067, 2015.
Article in English | MEDLINE | ID: mdl-26273627

ABSTRACT

Alternative splicing (AS) represents a major resource for eukaryotic cells to expand the coding potential of their genomes and to finely regulate gene expression in response to both intra- and extracellular cues. Cancer cells exploit the flexible nature of the mechanisms controlling AS in order to increase the functional diversity of their proteome. By altering the balance of splice isoforms encoded by human genes or by promoting the expression of aberrant oncogenic splice variants, cancer cells enhance their ability to adapt to the adverse growth conditions of the tumoral microenvironment. Herein, we will review the most relevant cancer-related splicing events and the underlying regulatory mechanisms allowing tumour cells to rapidly adapt to the harsh conditions they may face during the occurrence and development of cancer.


Subject(s)
Adaptation, Physiological/genetics , Alternative Splicing/genetics , Cell Plasticity/genetics , Gene Expression Regulation/genetics , Models, Genetic , Neoplasms/genetics , Animals , Humans
20.
Front Genet ; 6: 142, 2015.
Article in English | MEDLINE | ID: mdl-25926848

ABSTRACT

Genome integrity is constantly threatened by endogenous and exogenous factors. However, its preservation is ensured by a network of pathways that prevent and/or repair the lesion, which constitute the DNA damage response (DDR). Expression of the key proteins involved in the DDR is controlled by numerous post-transcriptional mechanisms, among which pre-mRNA splicing stands out. Intriguingly, several splicing factors (SFs) have been recently shown to play direct functions in DNA damage prevention and repair, which go beyond their expected splicing activity. At the same time, evidence is emerging that DNA repair proteins (DRPs) can actively sustain the DDR by acting as post-transcriptional regulator of gene expression, in addition to their well-known role in the mechanisms of signaling and repair of the lesion. Herein, we will review these non-canonical functions of both SFs and DRPs, which suggest the existence of a tight interplay between splicing regulation and canonical DNA safeguard mechanisms ensuring genome stability.

SELECTION OF CITATIONS
SEARCH DETAIL
...