Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Nature ; 617(7959): 147-153, 2023 05.
Article in English | MEDLINE | ID: mdl-36949200

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is characterized by aggressive local invasion and metastatic spread, leading to high lethality. Although driver gene mutations during PDA progression are conserved, no specific mutation is correlated with the dissemination of metastases1-3. Here we analysed RNA splicing data of a large cohort of primary and metastatic PDA tumours to identify differentially spliced events that correlate with PDA progression. De novo motif analysis of these events detected enrichment of motifs with high similarity to the RBFOX2 motif. Overexpression of RBFOX2 in a patient-derived xenograft (PDX) metastatic PDA cell line drastically reduced the metastatic potential of these cells in vitro and in vivo, whereas depletion of RBFOX2 in primary pancreatic tumour cell lines increased the metastatic potential of these cells. These findings support the role of RBFOX2 as a potent metastatic suppressor in PDA. RNA-sequencing and splicing analysis of RBFOX2 target genes revealed enrichment of genes in the RHO GTPase pathways, suggesting a role of RBFOX2 splicing activity in cytoskeletal organization and focal adhesion formation. Modulation of RBFOX2-regulated splicing events, such as via myosin phosphatase RHO-interacting protein (MPRIP), is associated with PDA metastases, altered cytoskeletal organization and the induction of focal adhesion formation. Our results implicate the splicing-regulatory function of RBFOX2 as a tumour suppressor in PDA and suggest a therapeutic approach for metastatic PDA.


Subject(s)
Alternative Splicing , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Alternative Splicing/genetics , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Repressor Proteins/genetics , Repressor Proteins/metabolism , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Animals , Neoplasm Metastasis , Focal Adhesions
2.
Hum Mol Genet ; 32(15): 2455-2463, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37145099

ABSTRACT

Duchene muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) are genetic neuromuscular disorders that affect skeletal and cardiac muscle resulting from mutations in the dystrophin gene (DMD), coding for dystrophin protein. Read-through therapies hold great promise for the treatment of genetic diseases harboring nonsense mutations, such as DMD/BMD, as they enable a complete translation of the affected mRNA. However, to date, most read-through drugs have not achieved a cure for patients. One possible explanation for the limitation of these therapies for DMD/BMD is that they rely on the presence of mutant dystrophin mRNAs. However, the mutant mRNAs containing premature termination codons are identified by the cellular surveillance mechanism, the nonsense-mediated mRNA decay (NMD) process, and are degraded. Here, we show that the combination of read-through drugs together with known NMD inhibitors have a synergistic effect on the levels of nonsense-containing mRNAs, among them the mutant dystrophin mRNA. This synergistic effect may enhance read-through therapies' efficacy and improve the current treatment for patients.


Subject(s)
Muscular Dystrophy, Duchenne , Humans , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Dystrophin/genetics , Dystrophin/metabolism , Codon, Terminator/genetics , Nonsense Mediated mRNA Decay , Mutation
3.
RNA ; 29(4): 506-515, 2023 04.
Article in English | MEDLINE | ID: mdl-36697261

ABSTRACT

Alternative splicing (AS) of mRNAs is an essential regulatory mechanism in eukaryotic gene expression. AS misregulation, caused by either dysregulation or mutation of splicing factors, has been shown to be involved in cancer development and progression, making splicing factors suitable targets for cancer therapy. In recent years, various types of pharmacological modulators, such as small molecules and oligonucleotides, targeting distinct components of the splicing machinery, have been under development to treat multiple disorders. Although these approaches have promise, targeting the core spliceosome components disrupts the early stages of spliceosome assembly and can lead to nonspecific and toxic effects. New research directions have been focused on targeting specific splicing factors for a more precise effect. In this Perspective, we will highlight several approaches for targeting splicing factors and their functions and suggest ways to improve their specificity.


Subject(s)
Neoplasms , RNA Splicing , Humans , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , RNA Splicing/genetics , Alternative Splicing , Spliceosomes/genetics , Spliceosomes/metabolism , Neoplasms/drug therapy , Neoplasms/genetics
4.
Nucleic Acids Res ; 46(21): 11396-11404, 2018 11 30.
Article in English | MEDLINE | ID: mdl-30329087

ABSTRACT

The gene encoding the kinase Mnk2 (MKNK2) is alternatively spliced to produce two isoforms-Mnk2a and Mnk2b. We previously showed that Mnk2a is downregulated in several types of cancer and acts as a tumor suppressor by activation of the p38-MAPK stress pathway, inducing apoptosis. Moreover, Mnk2a overexpression suppressed Ras-induced transformation in culture and in vivo. In contrast, the Mnk2b isoform acts as a pro-oncogenic factor. In this study, we designed modified-RNA antisense oligonucleotides and screened for those that specifically induce a strong switch in alternative splicing of the MKNK2 gene (splice switching oligonucleotides or SSOs), elevating the tumor suppressive isoform Mnk2a at the expense of the pro-oncogenic isoform Mnk2b. Induction of Mnk2a by SSOs in glioblastoma cells activated the p38-MAPK pathway, inhibited the oncogenic properties of the cells, re-sensitized the cells to chemotherapy and inhibited glioblastoma development in vivo. Moreover, inhibition of p38-MAPK partially rescued glioblastoma cells suggesting that most of the anti-oncogenic activity of the SSO is mediated by activation of this pathway. These results suggest that manipulation of MKNK2 alternative splicing by SSOs is a novel approach to inhibit glioblastoma tumorigenesis.


Subject(s)
Brain Neoplasms/therapy , Glioblastoma/therapy , Intracellular Signaling Peptides and Proteins/genetics , Oligonucleotides/genetics , Protein Serine-Threonine Kinases/genetics , Alternative Splicing , Animals , Apoptosis , Brain Neoplasms/genetics , Cell Line, Tumor , Genes, Tumor Suppressor , Glioblastoma/genetics , Humans , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Signaling System , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Oligonucleotides, Antisense , Phosphorylation , Protein Isoforms , Protein Serine-Threonine Kinases/metabolism , RNA Splicing , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Int J Cancer ; 143(1): 179-183, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29396858

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies. BRCA-associated PDAC comprises a clinically relevant subtype. A portion of these patients are highly susceptible to DNA damaging therapeutics, however, responses are heterogeneous and clinical resistance evolves. We have developed unique patient-derived xenograft (PDX) models from metastatic lesions of germline BRCA-mutated patients obtained at distinct time points; before treatment and at progression. Thus, closely mimicking clinical scenarios, to further investigate treatment naïve and resistant patients. DNA was isolated from six BRCA-mutated PDXs and classified by whole-genome sequencing to stable-genome or homologous recombination deficient (HRD)-genome. The sensitivity to DNA-damaging agents was evaluated in vivo in three BRCA-associated PDAC PDXs models: (1) HRD-genome naïve to treatments; (2) stable-genome naïve to treatment; (3) HRD-genome resistant to treatment. Correlation between disease course at tissue acquisition and response to PARP inhibitor (PARPi)/platinum was demonstrated in PDXs in vivo. Only the HRD-genome PDX, naïve to treatment, was sensitive to PARP inhibitor/cisplatin treatments. Our results demonstrate heterogeneous responses to DNA damaging agents/PARPi in BRCA-associated PDX thus reflecting the wide clinical spectrum. An HRD-genome PDX generated from a naïve to treatment biopsy was sensitive to platinum/PARPi whereas no benefit was observed in treating a HRD-genome PDXs generated from a patient that had acquired resistance nor stable-genome PDXs.


Subject(s)
BRCA1 Protein/genetics , BRCA2 Protein/genetics , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Platinum Compounds/administration & dosage , Poly(ADP-ribose) Polymerase Inhibitors/administration & dosage , Animals , Carcinoma, Pancreatic Ductal/genetics , Disease Progression , Drug Resistance, Neoplasm , Genomic Instability , Homologous Recombination , Humans , Mice , Mutation , Neoplasm Metastasis , Neoplasm Transplantation , Pancreatic Neoplasms/genetics , Platinum Compounds/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Prognosis , Whole Genome Sequencing
6.
Hum Genet ; 136(9): 1113-1127, 2017 09.
Article in English | MEDLINE | ID: mdl-28429085

ABSTRACT

Tumor cells alter their metabolism by a wide array of mechanisms to promote growth and proliferation. Dysregulated expression and/or somatic mutations of key components of the glycolytic pathway/TCA cycle as well as other metabolic pathways allow tumor cells to improve their ability to survive harsh conditions such as hypoxia and the presence of reactive oxygen species, as well as the ability to obtain nutrients to increase lipids, protein, and nucleic acids biogenesis. Approximately 95% of the human protein encoding genes undergo alternative splicing (AS), a regulated process of gene expression that greatly diversifies the proteome by creating multiple proteins from a single gene. In recent years, a growing body of evidence suggests that unbalanced AS, the formation of certain pro-tumorigenic isoforms and the reduction of anti-tumorigenic isoforms, is implicated in a variety of cancers. It is becoming increasingly clear that cancer-associated AS contributes to increased growth and proliferation, partially due to effects on metabolic reprogramming. Here, we summarize the known roles of AS in regulating cancer metabolism. We present evidence supporting the idea that AS, in many types of cancer, acts as a molecular switch that alters metabolism to drive tumorigenesis. We propose that the elucidation of misregulated AS and its downstream effects on cancer metabolism emphasizes the need for new therapeutic approaches aiming to modulate the splicing machinery to selectively target cancer cells.


Subject(s)
Alternative Splicing , Citric Acid Cycle/genetics , Glycolysis/genetics , Neoplasms , RNA, Neoplasm , Animals , Humans , Neoplasms/genetics , Neoplasms/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
7.
Bioconjug Chem ; 28(12): 3036-3042, 2017 Dec 20.
Article in English | MEDLINE | ID: mdl-29211451

ABSTRACT

Efficient delivery of oligonucleotides still remains a challenge in the field of oligonucleotide based therapy. Peptide nucleic acid (PNA), a DNA analogue that is typically synthesized by solid phase peptide chemistry, has been conjugated to a variety of cell penetrating peptides (CPP) as a means of improving its cellular uptake. These CPPs typically deliver their cargoes into cells by an endosomal-dependent mechanism resulting in lower bioavailability of the cargo. Herein, we designed and synthesized PNA-peptide conjugates as splice switching oligonucleotides (SSO) targeting the Mnk2 gene, a therapeutic target in cancer. In humans, the MKNK2 gene, is alternatively spliced, generating isoforms with opposite biological activities: Mnk2a and Mnk2b. It was found that the Mnk2a isoform is down-regulated in breast, lung, brain, and colon tumors and is a tumor suppressor, whereas MnK2b is oncogenic. We have designed and synthesized PNAs that were conjugated to either of the following peptides: a nuclear localization sequence (NLS) or a cytosol localizing internalization peptide (CLIP6). CLIP6-PNA demonstrates effective cellular uptake and exclusively employs a nonendosomal mechanism to cross the cellular membranes of glioblastoma cells (U87). Simple incubation of PNA-peptide conjugates in human glioblastoma cells up-regulates the Mnk2a isoform leading to cancer cell death.


Subject(s)
Cell-Penetrating Peptides/metabolism , Drug Carriers/metabolism , Oligonucleotides/metabolism , Peptide Nucleic Acids/metabolism , Base Sequence , Cell Line, Tumor , HeLa Cells , Humans , Hydrogen-Ion Concentration , Oligonucleotides/genetics
8.
Nucleic Acids Res ; 43(10): 5130-44, 2015 May 26.
Article in English | MEDLINE | ID: mdl-25908786

ABSTRACT

Cancer is a complex disease that involves aberrant gene expression regulation. Discriminating the modified expression patterns driving tumor biology from the many that have no or little contribution is important for understanding cancer molecular basis. Recurrent deregulation patterns observed in multiple cancer types are enriched for such driver events. Here, we studied splicing alterations in hundreds of matched tumor and normal RNA-seq samples of eight solid cancer types. We found hundreds of cassette exons for which splicing was altered in multiple cancer types and identified a set of highly frequent altered splicing events. Specific splicing regulators, including RBFOX2, MBNL1/2 and QKI, appear to account for many splicing alteration events in multiple cancer types. Together, our results provide a first global analysis of regulated splicing alterations in cancer and identify common events with a potential causative role in solid tumor development.


Subject(s)
Alternative Splicing , Neoplasms/genetics , Exons , Humans , Neoplasms/metabolism , RNA Splicing Factors , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Regulatory Sequences, Ribonucleic Acid , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/metabolism
9.
Mol Microbiol ; 96(6): 1283-97, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25807998

ABSTRACT

Plasmodium species have evolved complex biology to adapt to different hosts and changing environments throughout their life cycle. Remarkably, these adaptations are achieved by a relatively small genome. One way by which the parasite expands its proteome is through alternative splicing (AS). We recently identified PfSR1 as a bona fide Ser/Arg-rich (SR) protein that shuttles between the nucleus and cytoplasm and regulates AS in Plasmodium falciparum. Here we show that PfSR1 is localized adjacent to the Nuclear Pore Complex (NPC) clusters in the nucleus of early stage parasites. To identify the endogenous RNA targets of PfSR1, we adapted an inducible overexpression system for tagged PfSR1 and performed RNA immunoprecipitation followed by microarray analysis (RIP-chip) to recover and identify the endogenous RNA targets that bind PfSR1. Bioinformatic analysis of these RNAs revealed common sequence motifs potentially recognized by PfSR1. RNA-EMSAs show that PfSR1 preferentially binds RNA molecules containing these motifs. Interestingly, we find that PfSR1 not only regulates AS but also the steady-state levels of mRNAs containing these motifs in vivo.


Subject(s)
Nucleotide Motifs , Plasmodium falciparum/genetics , RNA, Protozoan/genetics , Serine-Arginine Splicing Factors/genetics , Alternative Splicing , Base Sequence , Cytoplasm/metabolism , Molecular Sequence Data , Nuclear Pore/metabolism , Nuclear Proteins/genetics , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Protozoan/metabolism , RNA-Binding Proteins/metabolism , Serine-Arginine Splicing Factors/metabolism
10.
RNA ; 20(4): 505-15, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24572810

ABSTRACT

In recent years, it has become clear that splicing factors play a direct role in cancer development. We showed previously that splicing factors SRSF1, SRSF6, and hnRNP A2/B1 are up-regulated in several cancers and can act as oncogenes when up-regulated. Here we examined the role of splicing factors hnRNP A1/A1b and hnRNP A2/B1 in hepatocellular carcinoma (HCC). We show that the splicing factors hnRNP A1 and hnRNP A2 are up-regulated in HCC tumors derived from inflammation-induced liver cancer mouse model. Overexpression of hnRNP A1 or hnRNP A2, but not the splicing isoform hnRNP B1, induced tumor formation of immortalized liver progenitor cells, while knockdown of these proteins inhibited anchorage-independent growth and tumor growth of human liver cancer cell lines. In addition, we found that cells overexpressing hnRNP A2 showed constitutive activation of the Ras-MAPK-ERK pathway. In contrast, knockdown of hnRNP A2 inhibited the Ras-MAPK-ERK pathway and prevented ERK1/2 activation by EGF. Moreover, we found that hnRNP A2 regulates the splicing of A-Raf, reducing the production of a short dominant-negative isoform of A-Raf and elevating the full-length A-Raf transcript. Taken together, our data suggest that hnRNP A2 up-regulation in HCC induces an alternative splicing switch that down-regulates a dominant-negative isoform of A-Raf, leading to activation of the Raf-MEK-ERK pathway and cellular transformation.


Subject(s)
Alternative Splicing , Carcinoma, Hepatocellular/pathology , Cell Transformation, Neoplastic/pathology , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins A-raf/genetics , ras Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B/physiology , Animals , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/metabolism , Cells, Cultured , Hepatocytes/metabolism , Hepatocytes/pathology , Heterogeneous Nuclear Ribonucleoprotein A1 , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/antagonists & inhibitors , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Humans , Inflammation/complications , Inflammation/genetics , Inflammation/pathology , Liver Neoplasms/etiology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Knockout , Mice, Nude , Mice, SCID , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinases/genetics , RNA, Small Interfering/genetics , Tumor Suppressor Protein p53/physiology , Xenograft Model Antitumor Assays , ras Proteins/genetics , ATP-Binding Cassette Sub-Family B Member 4
11.
Blood ; 120(24): 4712-9, 2012 Dec 06.
Article in English | MEDLINE | ID: mdl-23086752

ABSTRACT

Understanding the process of myeloid differentiation offers important insights into both normal and abnormal developmental processes but is limited by the dearth of experimental models. Here we show that myeloid progenitors can be derived from embryonic stem cells, immortalized, and applied to the study of the mechanisms underlying myeloid differentiation. The embryonic stem cell-derived myeloid progenitors, when immortalized with estrogen-regulated Hoxb8 protein, demonstrate normal karyotyping, are genetically tractable, and can be differentiated into functional neutrophils. Using this model, we identified mammalian target of rapamycin complex 1 as a critical regulator of myeloid differentiation. Together, our studies led to a convenient, karyotypically normal, and genetically manipulatable cellular system, which can be used to shed new light on the mechanisms for myeloid differentiation.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Myeloid Progenitor Cells/cytology , Signal Transduction/physiology , Animals , Blotting, Western , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Cells, Cultured , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Embryonic Stem Cells/metabolism , Estradiol/pharmacology , Flow Cytometry , Granulocyte Colony-Stimulating Factor/pharmacology , HEK293 Cells , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Karyotype , Mice , Mice, Inbred C57BL , Myeloid Progenitor Cells/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Phosphorylation/drug effects , RNA Interference , Signal Transduction/drug effects , Signal Transduction/genetics , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
12.
J Pathol ; 229(4): 630-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23132731

ABSTRACT

An increasing body of evidence connects alterations in the process of alternative splicing with cancer development and progression. However, a direct role of splicing factors as drivers of cancer development is mostly unknown. We analysed the gene copy number of several splicing factors in colon and lung tumours, and found that the gene encoding for the splicing factor SRSF6 is amplified and over-expressed in these cancers. Moreover, over-expression of SRSF6 in immortal lung epithelial cells enhanced proliferation, protected them from chemotherapy-induced cell death and converted them to be tumourigenic in mice. In contrast, knock-down of SRSF6 in lung and colon cancer cell lines inhibited their tumourigenic abilities. SRSF6 up- or down-regulation altered the splicing of several tumour suppressors and oncogenes to generate the oncogenic isoforms and reduce the tumour-suppressive isoforms. Our data suggest that the splicing factor SRSF6 is an oncoprotein that regulates the proliferation and survival of lung and colon cancer cells.


Subject(s)
Adenocarcinoma/genetics , Breast Neoplasms/genetics , Colonic Neoplasms/genetics , Lung Neoplasms/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Phosphoproteins/genetics , RNA-Binding Proteins/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Alternative Splicing , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Ductal/genetics , Carcinoma, Ductal/metabolism , Carcinoma, Ductal/pathology , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cell Transformation, Neoplastic , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Progression , Down-Regulation , Female , Gene Dosage , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Phosphoproteins/metabolism , Protein Isoforms , RNA Splicing , RNA-Binding Proteins/metabolism , Serine-Arginine Splicing Factors , Up-Regulation
13.
Nucleic Acids Res ; 40(19): 9903-16, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22885299

ABSTRACT

Malaria parasites have a complex life cycle, during which they undergo significant biological changes to adapt to different hosts and changing environments. Plasmodium falciparum, the species responsible for the deadliest form of human malaria, maintains this complex life cycle with a relatively small number of genes. Alternative splicing (AS) is an important post-transcriptional mechanisms that enables eukaryotic organisms to expand their protein repertoire out of relatively small number of genes. SR proteins are major regulators of AS in higher eukaryotes. Nevertheless, the regulation of splicing as well as the AS machinery in Plasmodium spp. are still elusive. Here, we show that PfSR1, a putative P. falciparum SR protein, can mediate RNA splicing in vitro. In addition, we show that PfSR1 functions as an AS factor in mini-gene in vivo systems similar to the mammalian SR protein SRSF1. Expression of PfSR1-myc in P. falciparum shows distinct patterns of cellular localization during intra erythrocytic development. Furthermore, we determine that the predicted RS domain of PfSR1 is essential for its localization to the nucleus. Finally, we demonstrate that proper regulation of pfsr1 is required for parasite proliferation in human RBCs and over-expression of pfsr1 influences AS activity of P. falciparum genes in vivo.


Subject(s)
Alternative Splicing , Erythrocytes/parasitology , Nuclear Proteins/metabolism , Plasmodium falciparum/genetics , Protozoan Proteins/metabolism , RNA-Binding Proteins/metabolism , Cell Line , Humans , Nuclear Localization Signals , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Plasmodium falciparum/growth & development , Plasmodium falciparum/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , RNA Precursors/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/genetics , Serine-Arginine Splicing Factors
14.
Carcinogenesis ; 34(11): 2498-504, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23843040

ABSTRACT

Alternative splicing regulators have emerged as new players in cancer development, modulating the activities of many tumor suppressors and oncogenes and regulating the signaling pathways. However, little is known about the mechanisms by which these oncogenic splicing factors lead to cellular transformation. We have shown previously that the splicing factor serine and arginine splicing factor 1 (SRSF1; SF2/ASF) is a proto-oncogene, which is amplified in breast cancer and transforms immortal cells when overexpressed. In this study, we performed a structure-function analysis of SRSF1 and found that the RNA recognition motif 1 (RRM1) domain is required for its oncogenic activity. Deletion of RRM1 eliminated the splicing activity of SRSF1 on some of its endogenous targets. Moreover, we found that SRSF1 elevates the expression of B-Raf and activates the mitogen-activated protein kinase kinase (MEK) extracellular signal-regulated kinase (ERK) pathway and that RRM1 is required for this activation as well. B-Raf-MEK-ERK activation by SRSF1 contributes to transformation as pharmacological inhibition of MEK1 inhibits SRSF1-mediated transformation. In conclusion, RRM1 of SRSF1 is both required (and when tethered to the RS domain) also sufficient to activate the Raf-MEK-ERK pathway and to promote cellular transformation.


Subject(s)
Cell Transformation, Neoplastic/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Nuclear Proteins/physiology , RNA Splicing/genetics , RNA-Binding Proteins/physiology , Ribonucleotide Reductases/physiology , Amino Acid Motifs , Animals , Blotting, Western , Cell Adhesion , Cell Proliferation , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/genetics , HEK293 Cells , Humans , Liver/metabolism , Liver/pathology , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinases/genetics , Mutation , Protein Binding , Protein Structure, Tertiary/physiology , Proto-Oncogene Mas , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Ribonucleoside Diphosphate Reductase , Serine-Arginine Splicing Factors , Signal Transduction , Stem Cells/metabolism , Stem Cells/pathology , Tumor Suppressor Protein p53/physiology
15.
Mol Cell Proteomics ; 10(1): M110.002998, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20938052

ABSTRACT

Alternative splicing is a crucial step in the generation of protein diversity and its misregulation is observed in many human cancer types. By analyzing 143 colorectal samples using exon arrays, SLC39A14, a divalent cation transporter, was identified as being aberrantly spliced in tumor samples. SLC39A14 contains two mutually exclusive exons 4A and 4B and the exon 4A/4B ratio was significantly altered in adenomas (p = 3.6 × 10(-10)) and cancers (p = 9.4 × 10(-11)), independent of microsatellite stability status. The findings were validated in independent exon array data sets and by quantitative real-time reverse-transcription PCR (qRT-PCR). Aberrant Wnt signaling is a hallmark of colorectal tumorigenesis and is characterized by nuclear ß-catenin. Experimental inactivation of Wnt signaling in DLD1 and Ls174T cells by knockdown of ß-catenin or overexpression of dominant negative TCFs (TCF1 and TCF4) altered the 4A/4B ratio, indicating that SLC39A14 splicing is regulated by the Wnt pathway. An altered 4A/4B ratio was also observed in gastric and lung cancer where Wnt signaling is also known to be aberrantly activated. The splicing factor SRSF1 and its regulator, the kinase SRPK1, were found to be deregulated upon Wnt inactivation in colorectal carcinoma cells. SRPK1 was also found up-regulated in both adenoma samples (p = 1.5 × 10(-5)) and cancer samples (p = 5 × 10(-4)). In silico splicing factor binding analysis predicted SRSF1 to bind predominantly to the cancer associated exon 4B, hence, it was hypothesized that SRPK1 activates SRSF1 through phosphorylation, followed by SRSF1 binding to exon 4B and regulation of SLC39A14 splicing. Indeed, siRNA-mediated knockdown of SRPK1 and SRSF1 in DLD1 and SW480 colorectal cancer cells led to a change in the 4A/4B isoform ratio, supporting a role of these factors in the regulation of SLC39A14 splicing. In conclusion, alternative splicing of SLC39A14 was identified in colorectal tumors and found to be regulated by the Wnt pathway, most likely through regulation of SRPK1 and SRSF1.


Subject(s)
Alternative Splicing/genetics , Cation Transport Proteins/genetics , Colorectal Neoplasms/genetics , Signal Transduction , Wnt Proteins/metabolism , Base Sequence , Binding Sites , Cation Transport Proteins/metabolism , Cell Line, Tumor , Colorectal Neoplasms/enzymology , Exons/genetics , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Intestinal Mucosa/enzymology , Intestinal Mucosa/pathology , Introns/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Serine-Arginine Splicing Factors
16.
Nat Struct Mol Biol ; 14(3): 185-93, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17310252

ABSTRACT

Alternative splicing modulates the expression of many oncogene and tumor-suppressor isoforms. We have tested whether some alternative splicing factors are involved in cancer. We found that the splicing factor SF2/ASF is upregulated in various human tumors, in part due to amplification of its gene, SFRS1. Moreover, slight overexpression of SF2/ASF is sufficient to transform immortal rodent fibroblasts, which form sarcomas in nude mice. We further show that SF2/ASF controls alternative splicing of the tumor suppressor BIN1 and the kinases MNK2 and S6K1. The resulting BIN1 isoforms lack tumor-suppressor activity; an isoform of MNK2 promotes MAP kinase-independent eIF4E phosphorylation; and an unusual oncogenic isoform of S6K1 recapitulates the transforming activity of SF2/ASF. Knockdown of either SF2/ASF or isoform-2 of S6K1 is sufficient to reverse transformation caused by the overexpression of SF2/ASF in vitro and in vivo. Thus, SF2/ASF can act as an oncoprotein and is a potential target for cancer therapy.


Subject(s)
Alternative Splicing , Nuclear Proteins/genetics , Proto-Oncogenes/genetics , Animals , Apoptosis , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Eukaryotic Initiation Factor-4E/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Mice , Mice, Nude , NIH 3T3 Cells , Neoplasms/genetics , Phosphorylation , Protein Isoforms/metabolism , Proto-Oncogene Mas , RNA-Binding Proteins , Rats , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Serine-Arginine Splicing Factors , Up-Regulation/genetics
17.
Elife ; 112022 10 03.
Article in English | MEDLINE | ID: mdl-36189922

ABSTRACT

The mTORC1 substrate, S6 Kinase 1 (S6K1), is involved in the regulation of cell growth, ribosome biogenesis, glucose homeostasis, and adipogenesis. Accumulating evidence has suggested a role for mTORC1 signaling in the DNA damage response. This is mostly based on the findings that mTORC1 inhibitors sensitized cells to DNA damage. However, a direct role of the mTORC1-S6K1 signaling pathway in DNA repair and the mechanism by which this signaling pathway regulates DNA repair is unknown. In this study, we discovered a novel role for S6K1 in regulating DNA repair through the coordinated regulation of the cell cycle, homologous recombination (HR) DNA repair (HRR) and mismatch DNA repair (MMR) mechanisms. Here, we show that S6K1 orchestrates DNA repair by phosphorylation of Cdk1 at serine 39, causing G2/M cell cycle arrest enabling homologous recombination and by phosphorylation of MSH6 at serine 309, enhancing MMR. Moreover, breast cancer cells harboring RPS6KB1 gene amplification show increased resistance to several DNA damaging agents and S6K1 expression is associated with poor survival of breast cancer patients treated with chemotherapy. Our findings reveal an unexpected function of S6K1 in the DNA repair pathway, serving as a tumorigenic barrier by safeguarding genomic stability.


Damage to the DNA in our cells can cause harmful changes that, if unchecked, can lead to the development of cancer. To help prevent this, cellular mechanisms are in place to repair defects in the DNA. A particular process, known as the mTORC1-S6K1 pathway is suspected to be important for repair because when this pathway is blocked, cells become more sensitive to DNA damage. It is still unknown how the various proteins involved in the mTORC1-S6K1 pathway contribute to repairing DNA. One of these proteins, S6K1, is an enzyme involved in coordinating cell growth and survival. The tumor cells in some forms of breast cancer produce more of this protein than normal, suggesting that S6K1 benefits these cells' survival. However, it is unclear exactly how the enzyme does this. Amar-Schwartz, Ben-Hur, Jbara et al. studied the role of S6K1 using genetically manipulated mouse cells and human cancer cells. These experiments showed that the protein interacts with two other proteins involved in DNA repair and activates them, regulating two different repair mechanisms and protecting cells against damage. These results might explain why some breast cancer tumors are resistant to radiotherapy and chemotherapy treatments, which aim to kill tumor cells by damaging their DNA. If this is the case, these findings could help clinicians choose more effective treatment options for people with cancers that produce additional S6K1. In the future, drugs that block the activity of the enzyme could make cancer cells more susceptible to chemotherapy.


Subject(s)
Breast Neoplasms , DNA Repair , DNA-Binding Proteins/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Breast Neoplasms/genetics , CDC2 Protein Kinase/metabolism , DNA , Female , G2 Phase Cell Cycle Checkpoints , Glucose , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/genetics , Serine/genetics
18.
Proc Natl Acad Sci U S A ; 105(40): 15323-7, 2008 Oct 07.
Article in English | MEDLINE | ID: mdl-18832178

ABSTRACT

The splicing factor SF2/ASF is an oncoprotein that is up-regulated in many cancers and can transform immortal rodent fibroblasts when slightly overexpressed. The mTOR signaling pathway is activated in many cancers, and pharmacological blockers of this pathway are in clinical trials as anticancer drugs. We examined the activity of the mTOR pathway in cells transformed by SF2/ASF and found that this splicing factor activates the mTORC1 branch of the pathway, as measured by S6K and eIF4EBP1 phosphorylation. This activation is specific to mTORC1 because no activation of Akt, an mTORC2 substrate, was detected. mTORC1 activation by SF2/ASF bypasses upstream PI3K/Akt signaling and is essential for SF2/ASF-mediated transformation, as inhibition of mTOR by rapamycin blocked transformation by SF2/ASF in vitro and in vivo. Moreover, shRNA-mediated knockdown of mTOR, or of the specific mTORC1 and mTORC2 components Raptor and Rictor, abolished the tumorigenic potential of cells overexpressing SF2/ASF. These results suggest that clinical tumors with SF2/ASF up-regulation could be especially sensitive to mTOR inhibitors.


Subject(s)
Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Protein Kinases/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , HeLa Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Multiprotein Complexes , NIH 3T3 Cells , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Proteins , RNA Splicing , RNA-Binding Proteins , Rats , Serine-Arginine Splicing Factors , Signal Transduction , TOR Serine-Threonine Kinases , Transfection , Up-Regulation
19.
Curr Opin Pharmacol ; 59: 140-148, 2021 08.
Article in English | MEDLINE | ID: mdl-34217945

ABSTRACT

In light of recent advances in RNA splicing modulation as therapy for specific genetic diseases, there is great optimism that this approach can be applied to treatment of cancer as well. Dysregulation of alternative RNA splicing is a common aberration detected in many cancers and thus, provides an attractive target for therapeutics. Here, we present and compare two promising approaches that are currently being investigated to manipulate alternative splicing and their potential use in therapy. The first strategy makes use of splice-switching oligonucleotides, whereas the second strategy uses CRISPR (clustered regularly interspaced short palindromic repeat Cas (CRISPR-associated) technology. We will discuss both the challenges and limitations of these technologies and progress being made to implement splice-switching as a potential cancer therapy.


Subject(s)
Gene Editing , Neoplasms , CRISPR-Cas Systems/genetics , Genetic Therapy , Humans , Neoplasms/drug therapy , Neoplasms/genetics , RNA Splicing
20.
Cancer Immunol Res ; 9(6): 637-650, 2021 06.
Article in English | MEDLINE | ID: mdl-33762352

ABSTRACT

SLAMF6 is a homotypic receptor of the Ig-superfamily associated with progenitor-exhausted T cells. Here we show that in humans, SLAMF6 has three splice isoforms involving its V-domain. Although the canonical receptor inhibited T-cell activation through SAP recruitment, the short isoform SLAMF6Δ17-65 had a strong agonistic effect. The costimulatory action depended on protein phosphatase SHP1 and led to a cytotoxic molecular profile mediated by the expression of TBX21 and RUNX3. Patients treated with immune checkpoint blockade showed a shift toward SLAMF6Δ17-65 in peripheral blood T cells. We developed splice-switching antisense oligonucleotides (ASO) designed to target the relevant SLAMF6 splice junction. Our ASOs enhanced SLAMF6Δ17-65 expression in human tumor-infiltrating lymphocytes and improved their capacity to inhibit human melanoma in mice. The yin-yang relationship of SLAMF6 splice isoforms may represent a balancing mechanism that could be exploited to improve cancer immunotherapy.


Subject(s)
Alternative Splicing/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/genetics , Melanoma/immunology , Signaling Lymphocytic Activation Molecule Family/genetics , Animals , Female , HEK293 Cells , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy , Jurkat Cells , Lymphocyte Activation/immunology , Melanoma/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Nude
SELECTION OF CITATIONS
SEARCH DETAIL