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










Publication year range
1.
Leuk Lymphoma ; 65(2): 209-218, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37921062

ABSTRACT

A large-scale genomic analysis of patients with ASXL1-mutated myeloid disease has not been performed to date. We reviewed comprehensive genomic profiling results from 6043 adults to characterize clinicopathologic features and co-mutation patterns by ASXL1 mutation status. ASXL1 mutations occurred in 1414 patients (23%). Mutation co-occurrence testing revealed strong co-occurrence (p < 0.01) between mutations in ASXL1 and nine genes (SRSF2, U2AF1, RUNX1, SETBP1, EZH2, STAG2, CUX1, CSF3R, CBL). Further analysis of patients with these co-mutations yielded several novel findings. Co-mutation patterns supported that ASXL1/SF3B1 co-mutation may be biologically distinct from ASXL1/non-SF3B1 spliceosome co-mutation. In AML, ASXL1/SRSF2 co-mutated patients frequently harbored STAG2 mutations (42%), which were dependent on the presence of both ASXL1 and SRSF2 mutation (p < 0.05). STAG2 and SETBP1 mutations were also exclusive in ASXL1/SRSF2 co-mutated patients and associated with divergent chronic myeloid phenotypes. Our findings support that certain multi-mutant genotypes may be biologically relevant in ASXL1-mutated myeloid disease.


Subject(s)
Leukemia, Myeloid, Acute , Myeloproliferative Disorders , Neoplasms , Adult , Humans , Myeloproliferative Disorders/genetics , Spliceosomes/genetics , Spliceosomes/pathology , Transcription Factors/genetics , Genomics , Mutation , Leukemia, Myeloid, Acute/genetics , Prognosis , Repressor Proteins/genetics
2.
J Comp Neurol ; 531(18): 2080-2108, 2023 Dec.
Article in English | MEDLINE | ID: mdl-36989381

ABSTRACT

Neurofibrillary tangles (NFTs) contain abnormally phosphorylated tau proteins, which spread within components of the medial temporal lobe (MTL) memory circuit in Alzheimer's disease (AD). Here, we used quantitative immunohistochemistry to determine the density of posttranslational oligomeric (TOC1 and TNT1), phosphorylated (AT8), and late truncated (TauC3) tau epitopes within the MTL subfields including entorhinal cortex (EC) layer II, subiculum, Cornu Ammonis (CA) subfields, and dentate gyrus (DG) in subjects who died with a clinical diagnosis of no cognitive impairment (NCI), mild cognitive impairment (MCI), and AD. We also examined whether alterations of the nuclear alternative splicing protein, SRSF2, are associated with tau pathology. Although a significant increase in TOC1, TNT1, and AT8 neuron density occurred in the EC in MCI and AD, subicular, DG granule cell, and CA1 and CA3 densities were only significantly higher in AD. TauC3 counts were not different between connectome regions and clinical groups. SRSF2 intensity in AT8-positive cells decreased significantly in all regions independent of the clinical groups examined. CA1 and subicular AT8, TauC3, and oligomeric densities correlated across clinical groups. EC AT8 counts correlated with CA subfields and subicular and DG values across clinical groups. Oligomeric and AT8 CA1, EC, and subicular density correlated with Braak stage. Decreased nuclear SRSF2 in the presence of cytoplasmic phosphorylated tau suggests a dual-hit process in NFT formation within the entorhinal hippocampal connectome during the onset of AD. Although oligomeric and phosphorylated tau follow a stereotypical pattern, clinical disease stage determined density of tau deposition and not anatomic location within the entorhinal-hippocampal connectome.


Subject(s)
Alzheimer Disease , Connectome , Humans , Alzheimer Disease/pathology , Spliceosomes/metabolism , Spliceosomes/pathology , Hippocampus/metabolism , tau Proteins/metabolism , Neurofibrillary Tangles/pathology
3.
Am J Med Genet A ; 188(12): 3432-3447, 2022 12.
Article in English | MEDLINE | ID: mdl-36367278

ABSTRACT

Verheij syndrome (VRJS) is a rare craniofacial spliceosomopathy presenting with craniofacial dysmorphism, multiple congenital anomalies and variable neurodevelopmental delay. It is caused by single nucleotide variants (SNVs) in PUF60 or interstitial deletions of the 8q24.3 region. PUF60 encodes a splicing factor which forms part of the spliceosome. To date, 36 patients with a sole diagnosis of VRJS due to disease-causing PUF60 SNVs have been reported in peer-reviewed publications. Although the depth of their phenotyping has varied greatly, they exhibit marked phenotypic heterogeneity. We report 10 additional unrelated patients, including the first described patients of Khmer, Indian, and Vietnamese ethnicities, and the eldest patient to date, with 10 heterozygous PUF60 variants identified through exome sequencing, 8 previously unreported. All patients underwent deep phenotyping identifying variable dysmorphism, growth delay, neurodevelopmental delay, and multiple congenital anomalies, including several unique features. The eldest patient is the only reported individual with a germline variant and neither neurodevelopmental delay nor intellectual disability. In combining these detailed phenotypic data with that of previously reported patients (n = 46), we further refine the known frequencies of features associated with VRJS. These include neurodevelopmental delay/intellectual disability (98%), axial skeletal anomalies (74%), appendicular skeletal anomalies (73%), oral anomalies (68%), short stature (66%), cardiac anomalies (63%), brain malformations (48%), hearing loss (46%), microcephaly (41%), colobomata (38%), and other ocular anomalies (65%). This case series, incorporating three patients from previously unreported ethnic backgrounds, further delineates the broad pleiotropy and mutational spectrum of PUF60 pathogenic variants.


Subject(s)
Abnormalities, Multiple , Intellectual Disability , Microcephaly , RNA Splicing Factors , Repressor Proteins , Humans , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Microcephaly/diagnosis , Microcephaly/genetics , Phenotype , Repressor Proteins/genetics , RNA Splicing Factors/genetics , Spliceosomes/genetics , Spliceosomes/pathology
4.
Forensic Sci Int ; 340: 111471, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36162298

ABSTRACT

Sudden unexplained death in schizophrenia (SUD-SCZ) is not uncommon and its incidence is approximately three times higher than that in the general population. However, diagnosis of SUD-SCZ remains a great challenge in forensic pathology. This study designed a two-phase study to investigate whether three proteins, namely two potassium ion channel proteins (KCNJ3 and KCNAB1) and one spliceosome protein (SF3B3) that were identified in our previous work, could be applied in the postmortem diagnosis of SUD-SCZ. Immunohistochemical staining of the three biomarkers, followed by a rigorous quantitative analysis, was performed on heart specimens from both SUD-SCZ and control groups. A diagnostic software based on the logistic regression formula derived from the test phase data was then constructed. In the test phase, we found that the staining intensities of KCNJ3, KCNAB1, and SF3B3 were all significantly lower in the SUD-SCZ group (n = 20) as compared with the control group that died from non-natural causes (n = 25), with fold-changes being 14.85 (p < 0.001), 4.13 (p = 0.028) and 2.12 (p = 0.048), respectively. Receiver operating characteristic analysis further illustrated that combination of the three biomarkers achieved the optimal diagnostic specificity (92%) and area under the curve (0.886). In the validation phase, the diagnostic software was confirmed to be a promising tool for predicting the risk of SUD-SCZ in authentic cases. Our study provided a valid strategy towards the practical diagnosis of SUD-SCZ by using KCNJ3, KCNAB1, and SF3B3 proteins as diagnostic biomarkers.


Subject(s)
Death, Sudden, Cardiac , Schizophrenia , Death, Sudden, Cardiac/pathology , Humans , Incidence , Potassium Channels , Schizophrenia/complications , Schizophrenia/diagnosis , Spliceosomes/pathology
5.
Endocr Relat Cancer ; 29(9): R123-R142, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35728261

ABSTRACT

The dysregulation of the splicing process has emerged as a novel hallmark of metabolic and tumor pathologies. In breast cancer (BCa), which represents the most diagnosed cancer type among women worldwide, the generation and/or dysregulation of several oncogenic splicing variants have been described. This is the case of the splicing variants of HER2, ER, BRCA1, or the recently identified by our group, In1-ghrelin and SST5TMD4, which exhibit oncogenic roles, increasing the malignancy, poor prognosis, and resistance to treatment of BCa. This altered expression of oncogenic splicing variants has been closely linked with the dysregulation of the elements belonging to the macromolecular machinery that controls the splicing process (spliceosome components and the associated splicing factors). In this review, we compile the current knowledge demonstrating the altered expression of splicing variants and spliceosomal components in BCa, showing the existence of a growing body of evidence supporting the close implication of the alteration in the splicing process in mammary tumorigenesis.


Subject(s)
Breast Neoplasms , Spliceosomes , Breast Neoplasms/pathology , Carcinogenesis/metabolism , Cell Transformation, Neoplastic/metabolism , Female , Humans , RNA Splicing , Spliceosomes/genetics , Spliceosomes/metabolism , Spliceosomes/pathology
6.
BMC Med Genomics ; 15(1): 89, 2022 04 18.
Article in English | MEDLINE | ID: mdl-35436926

ABSTRACT

BACKGROUND: Intellectual disability (ID) is a clinically important disease and a most prevalent neurodevelopmental disorder. The etiology and pathogenesis of ID are poorly recognized. Exome sequencing revealed a homozygous missense mutation in the POLR3B gene in a consanguineous family with three Intellectual disability with craniofacial anomalies patients. POLR3B gene encoding the second largest subunit of RNA polymerase III. METHODS: We performed RNA sequencing on blood samples to obtain insights into the biological pathways influenced by POLR3B mutation. We applied the results of our RNA-Seq analysis to several gene ontology programs such as ToppGene, Enrichr, KEGG. RESULTS: A significant decrease in expression of several spliceosomal RNAs, ribosomal proteins, and transcription factors was detected in the affected, compared to unaffected, family members. CONCLUSIONS: We hypothesize that POLR3B mutation dysregulates the expression of some important transcription factors, ribosomal and spliceosomal genes, and impairments in protein synthesis and splicing mediated in part by transcription factors such as FOXC2 and GATA1 contribute to impaired neuronal function and concurrence of intellectual disability and craniofacial anomalies in our patients. Our study highlights the emerging role of the spliceosome and ribosomal proteins in intellectual disability.


Subject(s)
Intellectual Disability , Humans , Intellectual Disability/pathology , Mutation , Mutation, Missense , Pedigree , RNA Polymerase III/genetics , Ribosomal Proteins/genetics , Spliceosomes/genetics , Spliceosomes/pathology , Transcription Factors/genetics
7.
Am J Med Genet A ; 185(5): 1515-1518, 2021 05.
Article in English | MEDLINE | ID: mdl-33559401

ABSTRACT

Nager syndrome epitomizes the acrofacial dysostoses, which are characterized by craniofacial and limb defects. The craniofacial defects include midfacial retrusion, downslanting palpebral fissures, prominent nasal bridge, and micrognathia. Limb malformations typically include hypoplasia or aplasia of radial elements including the thumb. Nager syndrome is caused by haploinsufficiency of SF3B4, encoding a spliceosomal protein called SAP49. Here, we report a patient with a loss of function variant in SF3B4 without acrofacial dysostosis or limb defects, whose reason for referral was developmental and growth delay. This patient is evidence of a broader phenotypic spectrum associated with SF3B4 variants than previously appreciated.


Subject(s)
Genetic Predisposition to Disease , Mandibulofacial Dysostosis/genetics , RNA Splicing Factors/genetics , Spliceosomes/genetics , Craniofacial Dysostosis/genetics , Craniofacial Dysostosis/pathology , Haploinsufficiency/genetics , Humans , Infant , Limb Deformities, Congenital/genetics , Limb Deformities, Congenital/pathology , Male , Mandibulofacial Dysostosis/pathology , Mutation/genetics , Phenotype , Spliceosomes/pathology
8.
Am J Med Genet A ; 185(2): 413-423, 2021 02.
Article in English | MEDLINE | ID: mdl-33247512

ABSTRACT

Mandibulofacial dysostosis with microcephaly (MFDM) is due to haploinsufficiency of spliceosomal GTPase EFTUD2. Features include microcephaly, craniofacial dysmorphology, developmental disability, and other anomalies. We surveyed parents of individuals with MFDM to expand knowledge about health, development, and parental concerns. Participants included attendees of the inaugural MFDM family conference in June 2019 and members of the MFDM online group. To explore MFDM variable expressivity, we offered targeted Sanger sequencing for untested parents. Forty-seven parents participated in the survey. 59% of individuals with MFDM were male, with mean age 6.4 years (range 8 months to 49 years). Similar to the literature (n = 123), common features include microcephaly, cleft palate, choanal stenosis, tracheoesophageal fistula, heart problems, and seizures. New information includes airway intervention details, age-based developmental outcomes, rate of vision refractive errors, and lower incidences of prematurity and IUGR. Family concerns focused on development, communication, and increased support. Targeted Sanger sequencing for families of seven individuals demonstrated de novo variants, for a total of 91.9% de novo EFTUD2 variants (n = 34/37). This study reports the largest single cohort of individuals with MFDM, expands phenotypic spectrum and inheritance patterns, improves understanding of developmental outcomes and care needs, and identifies development as the biggest concern for parents.


Subject(s)
Mandibulofacial Dysostosis/genetics , Microcephaly/genetics , Peptide Elongation Factors/genetics , Ribonucleoprotein, U5 Small Nuclear/genetics , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Adolescent , Adult , Child , Child, Preschool , Cohort Studies , Female , GTP Phosphohydrolases/genetics , Genetic Predisposition to Disease , Haploinsufficiency/genetics , Humans , Infant , Male , Mandibulofacial Dysostosis/complications , Mandibulofacial Dysostosis/pathology , Microcephaly/complications , Microcephaly/pathology , Middle Aged , Mutation/genetics , Phenotype , Spliceosomes/genetics , Spliceosomes/pathology , Young Adult
9.
Nat Commun ; 11(1): 5608, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33154379

ABSTRACT

The minor spliceosome is evolutionarily conserved in higher eukaryotes, but its biological significance remains poorly understood. Here, by precise CRISPR/Cas9-mediated disruption of the U12 and U6atac snRNAs, we report that a defective minor spliceosome is responsible for spinal muscular atrophy (SMA) associated phenotypes in Drosophila. Using a newly developed bioinformatic approach, we identified a large set of minor spliceosome-sensitive splicing events and demonstrate that three sensitive intron-containing neural genes, Pcyt2, Zmynd10, and Fas3, directly contribute to disease development as evidenced by the ability of their cDNAs to rescue the SMA-associated phenotypes in muscle development, neuromuscular junctions, and locomotion. Interestingly, many splice sites in sensitive introns are recognizable by both minor and major spliceosomes, suggesting a new mechanism of splicing regulation through competition between minor and major spliceosomes. These findings reveal a vital contribution of the minor spliceosome to SMA and to regulated splicing in animals.


Subject(s)
Drosophila Proteins/genetics , Introns , Muscular Atrophy, Spinal/genetics , Nerve Tissue Proteins/genetics , Spliceosomes/pathology , Animals , Disease Models, Animal , Drosophila , Muscular Atrophy, Spinal/pathology , Mutation , Phenotype , RNA Splice Sites , RNA Splicing/genetics , RNA, Small Nuclear/genetics , RNA, Small Nuclear/metabolism , RNA-Binding Proteins/genetics , Spliceosomes/genetics
10.
Dev Dyn ; 249(9): 1038-1046, 2020 09.
Article in English | MEDLINE | ID: mdl-32506634

ABSTRACT

The spliceosome is a complex of RNA and proteins that function together to identify intron-exon junctions in precursor messenger-RNAs, splice out the introns, and join the flanking exons. Mutations in any one of the genes encoding the proteins that make up the spliceosome may result in diseases known as spliceosomopathies. While the spliceosome is active in all cell types, with the majority of the proteins presumably expressed ubiquitously, spliceosomopathies tend to be tissue-specific as a result of germ line or somatic mutations, with phenotypes affecting primarily the retina in retinitis pigmentosa, hematopoietic lineages in myelodysplastic syndromes, or the craniofacial skeleton in mandibulofacial dysostosis. Here we describe the major spliceosomopathies, review the proposed mechanisms underlying retinitis pigmentosa and myelodysplastic syndromes, and discuss how this knowledge may inform our understanding of craniofacial spliceosomopathies.


Subject(s)
Mandibulofacial Dysostosis , Mutation , Myelodysplastic Syndromes , Retinitis Pigmentosa , Spliceosomes , Animals , Humans , Mandibulofacial Dysostosis/genetics , Mandibulofacial Dysostosis/metabolism , Mandibulofacial Dysostosis/pathology , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Retinitis Pigmentosa/pathology , Spliceosomes/genetics , Spliceosomes/metabolism , Spliceosomes/pathology
11.
Am J Med Genet A ; 182(8): 1952-1956, 2020 08.
Article in English | MEDLINE | ID: mdl-32462814

ABSTRACT

Pathogenic variants in components of the minor spliceosome have been associated with several human diseases. Recently, it was reported that biallelic RNPC3 variants lead to severe isolated growth hormone deficiency and pituitary hypoplasia. The RNPC3 gene codes for the U11/U12-65K protein, a component of the minor spliceosome. The minor spliceosome plays a role in the splicing of minor (U12-type) introns, which are present in ~700-800 genes in humans and represent about 0.35% of all introns. Here, we report a second family with biallelic RNPC3 variants in three siblings with a growth hormone deficiency, central congenital hypothyroidism, congenital cataract, developmental delay/intellectual deficiency and delayed puberty. These cases further confirm the association between biallelic RNPC3 variants and severe postnatal growth retardation due to growth hormone deficiency. Furthermore, these cases show that the phenotype of this minor spliceosome-related disease might be broader than previously described.


Subject(s)
Congenital Hypothyroidism/genetics , Developmental Disabilities/genetics , Dwarfism, Pituitary/genetics , Nuclear Proteins/genetics , RNA-Binding Proteins/genetics , Adolescent , Adult , Cataract , Child , Child, Preschool , Congenital Hypothyroidism/complications , Congenital Hypothyroidism/pathology , Developmental Disabilities/complications , Developmental Disabilities/pathology , Dwarfism, Pituitary/complications , Dwarfism, Pituitary/diagnosis , Dwarfism, Pituitary/pathology , Female , Growth Hormone/deficiency , Growth Hormone/genetics , Humans , Introns/genetics , Male , Phenotype , Puberty, Delayed/complications , Puberty, Delayed/genetics , Puberty, Delayed/pathology , RNA Splicing/genetics , Spliceosomes/genetics , Spliceosomes/pathology , Young Adult
12.
Mol Cell ; 76(1): 82-95.e7, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31474574

ABSTRACT

SF3B1, which encodes an essential spliceosomal protein, is frequently mutated in myelodysplastic syndromes (MDS) and many cancers. However, the defect of mutant SF3B1 is unknown. Here, we analyzed RNA sequencing data from MDS patients and confirmed that SF3B1 mutants use aberrant 3' splice sites. To elucidate the underlying mechanism, we purified complexes containing either wild-type or the hotspot K700E mutant SF3B1 and found that levels of a poorly studied spliceosomal protein, SUGP1, were reduced in mutant spliceosomes. Strikingly, SUGP1 knockdown completely recapitulated the splicing errors, whereas SUGP1 overexpression drove the protein, which our data suggest plays an important role in branchsite recognition, into the mutant spliceosome and partially rescued splicing. Other hotspot SF3B1 mutants showed similar altered splicing and diminished interaction with SUGP1. Our study demonstrates that SUGP1 loss is a common defect of spliceosomes with disease-causing SF3B1 mutations and, because this defect can be rescued, suggests possibilities for therapeutic intervention.


Subject(s)
Leukemia, Erythroblastic, Acute/metabolism , Mutation , Myelodysplastic Syndromes/metabolism , Phosphoproteins/metabolism , RNA Splicing Factors/metabolism , RNA Splicing , Spliceosomes/metabolism , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , HEK293 Cells , Humans , K562 Cells , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/pathology , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Phenotype , Phosphoproteins/genetics , Protein Binding , RNA Splicing Factors/genetics , Spliceosomes/genetics , Spliceosomes/pathology
13.
Exp Hematol ; 70: 10-23, 2019 02.
Article in English | MEDLINE | ID: mdl-30408513

ABSTRACT

Myelodysplastic syndromes (MDS) and related myelodysplastic/myeloproliferative neoplasms (MDS/MPNs) are clonal stem cell disorders, primarily affecting patients over 65 years of age. Mapping of the MDS and MDS/MPN genome identified recurrent heterozygous mutations in the RNA splicing machinery, with the SF3B1, SRSF2, and U2AF1 genes being frequently mutated. To better understand how spliceosomal mutations contribute to MDS pathogenesis in vivo, numerous groups have sought to establish conditional murine models of SF3B1, SRSF2, and U2AF1 mutations. The high degree of conservation of hematopoiesis between mice and human and the well-established phenotyping and genetic modification approaches make murine models an effective tool with which to study how a gene mutation contributes to disease pathogenesis. The murine models of spliceosomal mutations described to date recapitulate human MDS or MDS/MPN to varying extents. Reasons for the differences in phenotypes reported between alleles of the same mutation are varied, but the nature of the genetic modification itself and subsequent analysis methods are important to consider. In this review, we summarize recently reported murine models of SF3B1, SRSF2, and U2AF1 mutations, with a particular focus on the genetically engineered modifications underlying the models and the experimental approaches applied.


Subject(s)
Hematologic Neoplasms , Hematopoiesis/genetics , Mutation , Myelodysplastic Syndromes , Neoplasms, Experimental , RNA Splicing Factors , Spliceosomes , Animals , Hematologic Neoplasms/genetics , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/pathology , Humans , Mice , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Spliceosomes/genetics , Spliceosomes/metabolism , Spliceosomes/pathology
14.
Cancer Cell ; 34(1): 119-135.e10, 2018 07 09.
Article in English | MEDLINE | ID: mdl-29937354

ABSTRACT

Aggressive cancers such as glioblastoma (GBM) contain intermingled apoptotic cells adjacent to proliferating tumor cells. Nonetheless, intercellular signaling between apoptotic and surviving cancer cells remain elusive. In this study, we demonstrate that apoptotic GBM cells paradoxically promote proliferation and therapy resistance of surviving tumor cells by secreting apoptotic extracellular vesicles (apoEVs) enriched with various components of spliceosomes. apoEVs alter RNA splicing in recipient cells, thereby promoting their therapy resistance and aggressive migratory phenotype. Mechanistically, we identified RBM11 as a representative splicing factor that is upregulated in tumors after therapy and shed in extracellular vesicles upon induction of apoptosis. Once internalized in recipient cells, exogenous RBM11 switches splicing of MDM4 and Cyclin D1 toward the expression of more oncogenic isoforms.


Subject(s)
Apoptosis , Brain Neoplasms/metabolism , Extracellular Vesicles/metabolism , Glioblastoma/metabolism , RNA-Binding Proteins/metabolism , Spliceosomes/metabolism , Animals , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Communication , Cell Cycle Proteins , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cyclin D1/genetics , Cyclin D1/metabolism , Drug Resistance, Neoplasm , Extracellular Vesicles/drug effects , Extracellular Vesicles/genetics , Extracellular Vesicles/pathology , Female , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mice, Inbred NOD , Mice, SCID , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phenotype , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Splicing , RNA-Binding Proteins/genetics , Signal Transduction , Spliceosomes/drug effects , Spliceosomes/genetics , Spliceosomes/pathology , Tumor Burden
15.
Clin Exp Metastasis ; 35(5-6): 393-402, 2018 08.
Article in English | MEDLINE | ID: mdl-29845349

ABSTRACT

Metastatic cells exhibit an extraordinary phenotypic plasticity, not only in adapting to unfamiliar microenvironments but also in surviving aggressive treatments and immune responses. A major source of phenotypic variability is alternative splicing (AS) of the pre-messenger RNA. This process is catalyzed by one of the most complex pieces of cellular molecular regulatory events, the spliceosome, which is composed of ribonucleoproteins and polypeptides termed spliceosome factors. With strong evidence indicating that AS affects nearly all genes encoded by the human genome, aberrant AS programs have a significant impact on cancer cell development and progression. In this review, we present insights about the genomic and epigenomic factors affecting AS, summarize the most recent findings linking aberrant AS to metastatic progression, and highlight potential prognostic and therapeutic applications.


Subject(s)
Alternative Splicing/genetics , Neoplasms/genetics , Prognosis , Disease Progression , Humans , Mutation , Neoplasm Metastasis , Neoplasms/pathology , Neoplasms/therapy , RNA, Messenger/genetics , Spliceosomes/genetics , Spliceosomes/pathology
16.
Adv Biol Regul ; 67: 13-29, 2018 01.
Article in English | MEDLINE | ID: mdl-28986033

ABSTRACT

Mutations in splicing factor genes (SF3B1, SRSF2, U2AF1 and ZRSR2) are frequently found in patients with myelodysplastic syndromes (MDS), suggesting that aberrant spliceosome function plays a key role in the pathogenesis of MDS. Splicing factor mutations have been shown to result in aberrant splicing of many downstream target genes. Recent functional studies have begun to characterize the splicing dysfunction in MDS, identifying some key aberrantly spliced genes that are implicated in disease pathophysiology. These findings have led to the development of therapeutic strategies using splicing-modulating agents and rapid progress is being made in this field. Splicing inhibitors are promising agents that exploit the preferential sensitivity of splicing factor-mutant cells to these compounds. Here, we review the known target genes associated with splicing factor mutations in MDS, and discuss the potential of splicing-modulating therapies for these disorders.


Subject(s)
Mutation , Myelodysplastic Syndromes , RNA Splicing Factors , RNA Splicing , Spliceosomes , Humans , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Myelodysplastic Syndromes/therapy , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Spliceosomes/genetics , Spliceosomes/metabolism , Spliceosomes/pathology
17.
Adv Biol Regul ; 63: 59-70, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27639445

ABSTRACT

Splicing factor gene mutations are the most frequent mutations found in patients with the myeloid malignancy myelodysplastic syndrome (MDS), suggesting that spliceosomal dysfunction plays a major role in disease pathogenesis. The aberrantly spliced target genes and deregulated cellular pathways associated with the commonly mutated splicing factor genes in MDS (SF3B1, SRSF2 and U2AF1) are being identified, illuminating the molecular mechanisms underlying MDS. Emerging data from mouse modeling studies indicate that the presence of splicing factor gene mutations can lead to bone marrow hematopoietic stem/myeloid progenitor cell expansion, impaired hematopoiesis and dysplastic differentiation that are hallmarks of MDS. Importantly, recent evidence suggests that spliceosome inhibitors and splicing modulators may have therapeutic value in the treatment of splicing factor mutant myeloid malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Mutation , Myelodysplastic Syndromes/genetics , Phosphoproteins/antagonists & inhibitors , Pyrans/pharmacology , RNA Splicing Factors/antagonists & inhibitors , Serine-Arginine Splicing Factors/antagonists & inhibitors , Spiro Compounds/pharmacology , Splicing Factor U2AF/antagonists & inhibitors , Animals , Cell Proliferation , Gene Expression , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Mice , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Phenotype , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA Splicing/drug effects , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism , Spliceosomes/drug effects , Spliceosomes/metabolism , Spliceosomes/pathology , Splicing Factor U2AF/genetics , Splicing Factor U2AF/metabolism
18.
Eur J Haematol ; 94(5): 379-90, 2015 May.
Article in English | MEDLINE | ID: mdl-25186093

ABSTRACT

Myelodysplastic syndromes are a heterogeneous group of clonal haematological stem cell disorders. Allogeneic stem cells transplantation remains the only curative treatment but only a minority of patients are eligible for this treatment. In spite of this, it has become clear that treatment with lenalidomide and azanucleotides can lead to increased overall survival in particular subsets of patients with MDS. The relative silence on the therapeutic side is counter-balanced by major advances in the understanding of this heterogeneous disease. The introduction of high-throughput molecular techniques has resulted in the discovery that most patients harbour molecular aberrations, including pathways such as the spliceosome machinery previously not known to be involved. These newly discovered pathways will undoubtedly result in new therapeutic strategies for this difficult to treat disease.


Subject(s)
Gene Expression Regulation, Neoplastic , Hematopoietic Stem Cell Transplantation , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/therapy , Neoplasm Proteins/genetics , Angiogenesis Inhibitors/therapeutic use , Antimetabolites, Antineoplastic/therapeutic use , Azacitidine/therapeutic use , Blood Transfusion , Erythropoietin/therapeutic use , Humans , Lenalidomide , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Prognosis , Spliceosomes/drug effects , Spliceosomes/metabolism , Spliceosomes/pathology , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use , Transplantation, Homologous
19.
J Mol Cell Biol ; 6(4): 312-23, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24872507

ABSTRACT

Different from canonical ubiquitin-like proteins, Hub1 does not form covalent conjugates with substrates but binds proteins non-covalently. In Saccharomyces cerevisiae, Hub1 associates with spliceosomes and mediates alternative splicing of SRC1, without affecting pre-mRNA splicing generally. Human Hub1 is highly similar to its yeast homolog, but its cellular function remains largely unexplored. Here, we show that human Hub1 binds to the spliceosomal protein Snu66 as in yeast; however, unlike its S. cerevisiae homolog, human Hub1 is essential for viability. Prolonged in vivo depletion of human Hub1 leads to various cellular defects, including splicing speckle abnormalities, partial nuclear retention of mRNAs, mitotic catastrophe, and consequently cell death by apoptosis. Early consequences of Hub1 depletion are severe splicing defects, however, only for specific splice sites leading to exon skipping and intron retention. Thus, the ubiquitin-like protein Hub1 is not a canonical spliceosomal factor needed generally for splicing, but rather a modulator of spliceosome performance and facilitator of alternative splicing.


Subject(s)
Alternative Splicing/genetics , RNA Precursors/genetics , RNA, Messenger/genetics , Ribonucleoproteins, Small Nuclear/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Spliceosomes/metabolism , Ubiquitins/metabolism , Cell Survival , Cells, Cultured , Exons/genetics , Humans , Introns/genetics , RNA, Small Interfering/genetics , Ribonucleoproteins, Small Nuclear/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Spliceosomes/genetics , Spliceosomes/pathology , Ubiquitins/antagonists & inhibitors , Ubiquitins/genetics
20.
Immunol Allergy Clin North Am ; 34(2): 239-62, 2014 May.
Article in English | MEDLINE | ID: mdl-24745672

ABSTRACT

In all variants of mastocytosis, activating KIT mutations are frequently found. In adults, neoplastic mast cells (MCs) cells show the KIT mutation D816V, whereas in children, MCs invading the skin are frequently positive for non-KIT D816V mutations. The clinical course and prognosis of the disease vary among patients with systemic mastocytosis (SM). Additional KIT-independent molecular defects might cause progression. Additional oncogenic lesions have recently been identified in advanced SM. In advanced SM the presence of additional genetic lesions or altered signaling worsening the prognosis might lead to the use of alternative therapies such as combined antisignaling targeted treatments or stem cell transplantation.


Subject(s)
Gene Expression Regulation, Neoplastic , Hematologic Neoplasms/genetics , Mast Cells/metabolism , Mastocytosis/genetics , Proto-Oncogene Proteins c-kit/genetics , Spliceosomes/genetics , Bone Marrow/drug effects , Bone Marrow/metabolism , Bone Marrow/pathology , Cell Proliferation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases , Exons , Hematologic Neoplasms/diagnosis , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/pathology , Humans , Mast Cells/drug effects , Mast Cells/pathology , Mastocytosis/diagnosis , Mastocytosis/drug therapy , Mastocytosis/pathology , Mutation , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Spliceosomes/metabolism , Spliceosomes/pathology , Stem Cell Factor/genetics , Stem Cell Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...