Pre-mRNA splicing-associated diseases and therapies
暂无分享,去创建一个
[1] K. Nikolić,et al. Reversible Control of RNA Splicing by Photoswitchable Small Molecules. , 2023, Journal of the American Chemical Society.
[2] C. Dienemann,et al. Structural basis of catalytic activation in human splicing , 2023, Nature.
[3] Gregory A. Newby,et al. Base editing rescue of spinal muscular atrophy in cells and in mice , 2023, Science.
[4] Robert J. Weatheritt,et al. Global detection of human variants and isoforms by deep proteome sequencing , 2023, Nature Biotechnology.
[5] Sonya M. Hanson,et al. splice sites by noncanonical base-pairing to U1 snRNA involving , 2022 .
[6] Yue Huang,et al. DHX15 is involved in SUGP1-mediated RNA missplicing by mutant SF3B1 in cancer , 2022, Proceedings of the National Academy of Sciences of the United States of America.
[7] Yongmei Xie,et al. An overview of PROTACs: a promising drug discovery paradigm , 2022, Molecular biomedicine.
[8] A. Burlingame,et al. Targeted high throughput mutagenesis of the human spliceosome reveals its in vivo operating principles , 2022, bioRxiv.
[9] C. Burge,et al. Splicing quality control mediated by DHX15 and its G-patch activator, SUGP1 , 2022, bioRxiv.
[10] J. Winkler,et al. An alternative splicing modulator decreases mutant HTT and improves the molecular fingerprint in Huntington’s disease patient neurons , 2022, Nature Communications.
[11] M. Kolář,et al. Retinitis pigmentosa associated mutations in mouse Prpf8 cause misexpression of circRNAs and degeneration of cerebellar granule neurons , 2022, bioRxiv.
[12] M. Donahue,et al. Splicing modulators impair DNA damage response and induce killing of cohesin-mutant MDS/AML , 2022, bioRxiv.
[13] W. P. Galej,et al. Structural studies of the spliceosome: Bridging the gaps , 2022, Current opinion in structural biology.
[14] Jessica L. Childs-Disney,et al. Targeting RNA structures with small molecules , 2022, Nature Reviews Drug Discovery.
[15] S. Butcher,et al. Identification of transient intermediates during spliceosome activation by single molecule fluorescence microscopy , 2022, bioRxiv.
[16] O. Abdel-Wahab,et al. Synthetic introns enable splicing factor mutation-dependent targeting of cancer cells , 2022, Nature Biotechnology.
[17] S. Peltz,et al. Small molecule splicing modifiers with systemic HTT-lowering activity , 2021, Nature Communications.
[18] F. Weis,et al. Structural basis of branch site recognition by the human spliceosome , 2021, Science.
[19] Margaret A. Goralski,et al. Targeting the spliceosome through RBM39 degradation results in exceptional responses in high-risk neuroblastoma models , 2021, Science advances.
[20] Hong Zhang,et al. RNA-binding protein 39: a promising therapeutic target for cancer , 2021, Cell death discovery.
[21] B. Davidson,et al. Regulated control of gene therapies by drug-induced splicing , 2021, Nature.
[22] J. Al-Aama,et al. Pre-mRNA Processing Factors and Retinitis Pigmentosa: RNA Splicing and Beyond , 2021, Frontiers in Cell and Developmental Biology.
[23] Arun K. Ghosh,et al. Structural basis of intron selection by U2 snRNP in the presence of covalent inhibitors , 2021, Nature Communications.
[24] H. Dvinge,et al. Human spliceosomal snRNA sequence variants generate variant spliceosomes , 2021, RNA.
[25] I. Flinn,et al. Phase I First-in-Human Dose Escalation Study of the oral SF3B1 modulator H3B-8800 in myeloid neoplasms , 2021, Leukemia.
[26] Daniel G. Anderson,et al. mRNA therapeutics: beyond vaccine applications. , 2021, Trends in molecular medicine.
[27] J. Wolchok,et al. Pharmacologic modulation of RNA splicing enhances anti-tumor immunity , 2021, Cell.
[28] Kathryn E. Hentges,et al. The role of splicing factors in retinitis pigmentosa: links to cilia. , 2021, Biochemical Society transactions.
[29] J. Diedrich,et al. Coupling of spliceosome complexity to intron diversity , 2021, Current Biology.
[30] H. Ratni,et al. Risdiplam, the First Approved Small Molecule Splicing Modifier Drug as a Blueprint for Future Transformative Medicines. , 2021, ACS medicinal chemistry letters.
[31] W. Newman,et al. The Role of the U5 snRNP in Genetic Disorders and Cancer , 2021, Frontiers in Genetics.
[32] Jitendra Kumar Meena,et al. Spliceosome-targeted therapies trigger an antiviral immune response in triple-negative breast cancer , 2021, Cell.
[33] J. Boultwood,et al. SF3B1 mutant myelodysplastic syndrome: Recent advances. , 2020, Advances in biological regulation.
[34] R. Zhao,et al. DEAH-Box RNA Helicases in Pre-mRNA Splicing. , 2020, Trends in biochemical sciences.
[35] N. Gehring,et al. Anything but Ordinary - Emerging Splicing Mechanisms in Eukaryotic Gene Regulation. , 2020, Trends in genetics : TIG.
[36] G. Carvill,et al. Poison exons in neurodevelopment and disease. , 2020, Current opinion in genetics & development.
[37] J. Saint-Jeannet,et al. Spliceosomopathies: Diseases and mechanisms , 2020, Developmental dynamics : an official publication of the American Association of Anatomists.
[38] H. Stark,et al. Molecular architecture of the human 17S U2 snRNP , 2020, Nature.
[39] K. Horie-Inoue,et al. Roles of Splicing Factors in Hormone-Related Cancer Progression , 2020, International journal of molecular sciences.
[40] V. Hornak,et al. Structural basis of indisulam-mediated RBM39 recruitment to DCAF15 E3 ligase complex , 2019, Nature Chemical Biology.
[41] Paul M Wehn,et al. Structural Basis and Kinetic Pathway of RBM39 Recruitment to DCAF15 by a Sulfonamide Molecular Glue E7820. , 2019, Structure.
[42] A. Grossfield,et al. Structures of SF3b1 reveal a dynamic Achilles heel of spliceosome assembly: Implications for cancer-associated abnormalities and drug discovery. , 2019, Biochimica et biophysica acta. Gene regulatory mechanisms.
[43] H. Stark,et al. Structural Insights into Nuclear pre-mRNA Splicing in Higher Eukaryotes. , 2019, Cold Spring Harbor perspectives in biology.
[44] John A. Calarco,et al. Recurrent noncoding U1 snRNA mutations drive cryptic splicing in SHH medulloblastoma , 2019, Nature.
[45] Anna Knörlein,et al. Structural basis of a small molecule targeting RNA for a specific splicing correction , 2019, Nature Chemical Biology.
[46] R. Rabadán,et al. Disease-Causing Mutations in SF3B1 Alter Splicing by Disrupting Interaction with SUGP1. , 2019, Molecular cell.
[47] Julio Delgado,et al. The U1 spliceosomal RNA is recurrently mutated in multiple cancers , 2019, Nature.
[48] M. Carmo-Fonseca,et al. Targeting mRNA processing as an anticancer strategy , 2019, Nature Reviews Drug Discovery.
[49] Alexander V Penson,et al. Spliceosomal disruption of the non-canonical BAF complex in cancer , 2019, Nature.
[50] A. Hoskins,et al. Structural and functional modularity of the U2 snRNP in pre-mRNA splicing , 2019, Critical reviews in biochemistry and molecular biology.
[51] Rhiju Das,et al. A unified mechanism for intron and exon definition and back-splicing , 2019, Nature.
[52] N. Gray,et al. Structural complementarity facilitates E7820-mediated degradation of RBM39 by DCAF15 , 2019, bioRxiv.
[53] A. Krainer,et al. Antisense Oligonucleotide Therapies for Neurodegenerative Diseases. , 2019, Annual review of neuroscience.
[54] L. Maquat,et al. Quality and quantity control of gene expression by nonsense-mediated mRNA decay , 2019, Nature Reviews Molecular Cell Biology.
[55] A. Hoskins,et al. Chemical Inhibition of Pre-mRNA Splicing in Living Saccharomyces cerevisiae. , 2019, Cell chemical biology.
[56] A. Krainer,et al. Targeting an RNA-Binding Protein Network in Acute Myeloid Leukemia. , 2019, Cancer cell.
[57] K. Nagai,et al. Structural Basis of Nuclear pre-mRNA Splicing: Lessons from Yeast. , 2019, Cold Spring Harbor perspectives in biology.
[58] Dahui Qin. Next-generation sequencing and its clinical application , 2019, Cancer biology & medicine.
[59] Angela N. Brooks,et al. Splicing modulation sensitizes chronic lymphocytic leukemia cells to venetoclax by remodeling mitochondrial apoptotic dependencies. , 2018, JCI insight.
[60] B. Ebert,et al. Synthetic Lethal and Convergent Biological Effects of Cancer-Associated Spliceosomal Gene Mutations. , 2018, Cancer cell.
[61] Michael Reutlinger,et al. Discovery of Risdiplam, a Selective Survival of Motor Neuron-2 ( SMN2) Gene Splicing Modifier for the Treatment of Spinal Muscular Atrophy (SMA). , 2018, Journal of medicinal chemistry.
[62] Joerg E Braun,et al. Synergistic assembly of human pre-spliceosomes across introns and exons , 2018, eLife.
[63] H. Dvinge,et al. RNA components of the spliceosome regulate tissue- and cancer-specific alternative splicing , 2018, bioRxiv.
[64] A. Hoskins,et al. Functional analysis of Hsh155/SF3b1 interactions with the U2 snRNA/branch site duplex , 2018, RNA.
[65] Abramowicz Anna,et al. Splicing mutations in human genetic disorders: examples, detection, and confirmation , 2018, Journal of Applied Genetics.
[66] C. Will,et al. Structural Basis of Splicing Modulation by Antitumor Macrolide Compounds. , 2018, Molecular cell.
[67] Ping Zhu,et al. Somatic Mutational Landscape of Splicing Factor Genes and Their Functional Consequences across 33 Cancer Types. , 2018, Cell reports.
[68] M. Warmuth,et al. H3B-8800, an orally available small-molecule splicing modulator, induces lethality in spliceosome-mutant cancers , 2018, Nature Medicine.
[69] Peter G. Smith,et al. Targeting splicing abnormalities in cancer. , 2018, Current opinion in genetics & development.
[70] Qiang Zhou,et al. The cryo-EM structure of the SF3b spliceosome complex bound to a splicing modulator reveals a pre-mRNA substrate competitive mechanism of action , 2018, Genes & development.
[71] S. Butcher,et al. The life of U6 small nuclear RNA, from cradle to grave , 2018, RNA.
[72] J. Valcárcel,et al. Molecular basis of differential 3′ splice site sensitivity to anti-tumor drugs targeting U2 snRNP , 2017, Nature Communications.
[73] Adrian R. Krainer,et al. Binding to SMN2 pre-mRNA-protein complex elicits specificity for small molecule splicing modifiers , 2017, Nature Communications.
[74] R. J. Ramamurthi,et al. Nusinersen versus Sham Control in Infantile‐Onset Spinal Muscular Atrophy , 2017, The New England journal of medicine.
[75] M. Warmuth,et al. Splicing modulators act at the branch point adenosine binding pocket defined by the PHF5A–SF3b complex , 2017, Nature Communications.
[76] Margaret A. Goralski,et al. Anticancer sulfonamides target splicing by inducing RBM39 degradation via recruitment to DCAF15 , 2017, Science.
[77] T. Owa,et al. Selective degradation of splicing factor CAPERα by anticancer sulfonamides. , 2017, Nature chemical biology.
[78] Kamil J. Cygan,et al. Pathogenic variants that alter protein code often disrupt splicing , 2017, Nature Genetics.
[79] M. Jurica,et al. Modulating splicing with small molecular inhibitors of the spliceosome , 2017, Wiley interdisciplinary reviews. RNA.
[80] F. Slack,et al. MicroRNA therapeutics: towards a new era for the management of cancer and other diseases , 2017, Nature Reviews Drug Discovery.
[81] J. Karanicolas,et al. Musashi RNA-Binding Proteins as Cancer Drivers and Novel Therapeutic Targets , 2017, Clinical Cancer Research.
[82] A. Hoskins,et al. SF3b1 mutations associated with myelodysplastic syndromes alter the fidelity of branchsite selection in yeast , 2017, Nucleic acids research.
[83] Susana Rodríguez-Santiago,et al. SF3B1/Hsh155 HEAT motif mutations affect interaction with the spliceosomal ATPase Prp5, resulting in altered branch site selectivity in pre-mRNA splicing , 2016, Genes & development.
[84] Eunhee Kim,et al. RNA splicing factors as oncoproteins and tumour suppressors , 2016, Nature Reviews Cancer.
[85] M. Moore,et al. Single molecule analysis reveals reversible and irreversible steps during spliceosome activation , 2016, eLife.
[86] C. Guthrie,et al. Prp8 retinitis pigmentosa mutants cause defects in the transition between the catalytic steps of splicing , 2016, RNA.
[87] A. Godzik,et al. UHM-ULM interactions in the RBM39-U2AF65 splicing-factor complex. , 2016, Acta crystallographica. Section D, Structural biology.
[88] Arun K. Ghosh,et al. Interchangeable SF3B1 inhibitors interfere with pre-mRNA splicing at multiple stages , 2016, RNA.
[89] M. Swanson,et al. RNA mis-splicing in disease , 2015, Nature Reviews Genetics.
[90] M. Warmuth,et al. Cancer-Associated SF3B1 Hotspot Mutations Induce Cryptic 3' Splice Site Selection through Use of a Different Branch Point. , 2015, Cell reports.
[91] A. Krainer,et al. The spliceosome, a potential Achilles heel of MYC-driven tumors , 2015, Genome Medicine.
[92] C. Soneson,et al. Human Mpn1 promotes post‐transcriptional processing and stability of U6atac , 2015, FEBS letters.
[93] Thomas M. Smith,et al. SMN2 splice modulators enhance U1-pre-mRNA association and rescue SMA mice. , 2015, Nature chemical biology.
[94] P. Patil,et al. Incomplete splicing of neutrophil-specific genes affects neutrophil development in a zebrafish model of poikiloderma with neutropenia , 2015, RNA biology.
[95] T. Misteli,et al. Inhibition of vemurafenib-resistant melanoma by interference with pre-mRNA splicing , 2015, Nature Communications.
[96] Dennis Carson,et al. Transcriptome Sequencing Reveals Potential Mechanism of Cryptic 3’ Splice Site Selection in SF3B1-mutated Cancers , 2015, PLoS Comput. Biol..
[97] R. Ferris,et al. Regulation of HPV16 E6 and MCL1 by SF3B1 inhibitor in head and neck cancer cells , 2014, Scientific Reports.
[98] Anirvan Ghosh,et al. SMN2 splicing modifiers improve motor function and longevity in mice with spinal muscular atrophy , 2014, Science.
[99] C. J. O’Donnell,et al. Spliceostatin hemiketal biosynthesis in Burkholderia spp. is catalyzed by an iron/α-ketoglutarate–dependent dioxygenase , 2014, Proceedings of the National Academy of Sciences.
[100] R. Padgett,et al. Biochemical defects in minor spliceosome function in the developmental disorder MOPD I , 2014, RNA.
[101] Paul K. Korir,et al. A mutation in a splicing factor that causes retinitis pigmentosa has a transcriptome-wide effect on mRNA splicing , 2014, BMC Research Notes.
[102] Michael R. Green,et al. Cancer-relevant Splicing Factor CAPERα Engages the Essential Splicing Factor SF3b155 in a Specific Ternary Complex* , 2014, The Journal of Biological Chemistry.
[103] S. Daiger,et al. Genes and mutations causing retinitis pigmentosa , 2013, Clinical genetics.
[104] C. Will,et al. Inhibition of RNA Helicase Brr2 by the C-Terminal Tail of the Spliceosomal Protein Prp8 , 2013, Science.
[105] C. Azzalin,et al. The Mpn1 RNA exonuclease: Cellular functions and implication in disease , 2013, FEBS letters.
[106] Yang Gao,et al. Chemical perturbation of Mcl-1 pre-mRNA splicing to induce apoptosis in cancer cells. , 2013, ACS chemical biology.
[107] P. Campbell,et al. Aberrant 3' oligoadenylation of spliceosomal U6 small nuclear RNA in poikiloderma with neutropenia. , 2013, Blood.
[108] C. Soneson,et al. Mpn1, mutated in poikiloderma with neutropenia protein 1, is a conserved 3'-to-5' RNA exonuclease processing U6 small nuclear RNA. , 2012, Cell reports.
[109] M. Frilander,et al. The significant other: splicing by the minor spliceosome , 2012, Wiley interdisciplinary reviews. RNA.
[110] K. Ginalski,et al. C16orf57, a gene mutated in poikiloderma with neutropenia, encodes a putative phosphodiesterase responsible for the U6 snRNA 3' end modification. , 2012, Genes & development.
[111] T. Cooper,et al. Pre-mRNA splicing in disease and therapeutics. , 2012, Trends in molecular medicine.
[112] A. Krainer,et al. Widespread recognition of 5' splice sites by noncanonical base-pairing to U1 snRNA involving bulged nucleotides. , 2012, Genes & development.
[113] H. Scott,et al. Spliceosome mutations in hematopoietic malignancies , 2011, Nature Genetics.
[114] E. Giné,et al. Exome sequencing identifies recurrent mutations of the splicing factor SF3B1 gene in chronic lymphocytic leukemia , 2011, Nature Genetics.
[115] Scott L. Pomeroy,et al. Molecular subgroups of medulloblastoma: the current consensus , 2011, Acta Neuropathologica.
[116] S. Sugano,et al. Frequent pathway mutations of splicing machinery in myelodysplasia , 2011, Nature.
[117] D. Staněk,et al. Where splicing joins chromatin , 2011, Nucleus.
[118] Sandya Liyanarachchi,et al. Mutations in U4atac snRNA, a Component of the Minor Spliceosome, in the Developmental Disorder MOPD I , 2011, Science.
[119] M. Moore,et al. Ordered and Dynamic Assembly of Single Spliceosomes , 2011, Science.
[120] Miaosheng Li,et al. Structural requirements for the antiproliferative activity of pre-mRNA splicing inhibitor FR901464. , 2011, Chemistry.
[121] S. Horinouchi,et al. Identification of SAP155 as the target of GEX1A (Herboxidiene), an antitumor natural product. , 2011, ACS chemical biology.
[122] R. O’Keefe,et al. The function of the NineTeen Complex (NTC) in regulating spliceosome conformations and fidelity during pre-mRNA splicing. , 2010, Biochemical Society transactions.
[123] M. Estelle,et al. Auxin perception--structural insights. , 2010, Cold Spring Harbor perspectives in biology.
[124] Steven L Salzberg,et al. Between a chicken and a grape: estimating the number of human genes , 2010, Genome Biology.
[125] C. Guthrie,et al. Conformational dynamics of single pre-mRNA molecules during in vitro splicing , 2010, Nature Structural &Molecular Biology.
[126] Fabio Macciardi,et al. Targeted next-generation sequencing appoints c16orf57 as clericuzio-type poikiloderma with neutropenia gene. , 2010, American journal of human genetics.
[127] T. Nilsen,et al. Expansion of the eukaryotic proteome by alternative splicing , 2010, Nature.
[128] Henning Urlaub,et al. The evolutionarily conserved core design of the catalytic activation step of the yeast spliceosome. , 2009, Molecular cell.
[129] Jason M. Edmonds,et al. Tetracyclines That Promote SMN2 Exon 7 Splicing as Therapeutics for Spinal Muscular Atrophy , 2009, Science Translational Medicine.
[130] C. Will,et al. The Spliceosome: Design Principles of a Dynamic RNP Machine , 2009, Cell.
[131] C. Guthrie,et al. ATP-dependent unwinding of U4/U6 snRNAs by the Brr2 helicase requires the C-terminus of Prp8 , 2008, Nature Structural &Molecular Biology.
[132] Eric T. Wang,et al. Alternative Isoform Regulation in Human Tissue Transcriptomes , 2008, Nature.
[133] Soo-Chen Cheng,et al. Both Catalytic Steps of Nuclear Pre-mRNA Splicing Are Reversible , 2008, Science.
[134] J. Beggs,et al. prp8 mutations that cause human retinitis pigmentosa lead to a U5 snRNP maturation defect in yeast , 2007, Nature Structural &Molecular Biology.
[135] M. Hagiwara,et al. Spliceostatin A targets SF3b and inhibits both splicing and nuclear retention of pre-mRNA , 2007, Nature Chemical Biology.
[136] T. Owa,et al. Splicing factor SF3b as a target of the antitumor natural product pladienolide , 2007, Nature Chemical Biology.
[137] A. Kornblihtt,et al. The connection between splicing and cancer , 2006, Journal of Cell Science.
[138] P. Francis. Genetics of Inherited Retinal Disease , 2006 .
[139] F. Clark,et al. Understanding alternative splicing: towards a cellular code , 2005, Nature Reviews Molecular Cell Biology.
[140] J. Beggs,et al. Prp8 protein: at the heart of the spliceosome. , 2005, RNA.
[141] B. O’Malley,et al. Steroid Hormone Receptor Coactivation and Alternative RNA Splicing by U2AF65-Related Proteins CAPERα and CAPERβ , 2005 .
[142] T. Yamori,et al. Pladienolides, new substances from culture of Streptomyces platensis Mer-11107. III. In vitro and in vivo antitumor activities. , 2004, The Journal of antibiotics.
[143] S. Horinouchi,et al. GEX1 compounds, novel antitumor antibiotics related to herboxidiene, produced by Streptomyces sp. II. The effects on cell cycle progression and gene expression. , 2002, The Journal of antibiotics.
[144] D. Krause,et al. Alternative first exons of PTCH1 are differentially regulated in vivo and may confer different functions to the PTCH1 protein , 2002, Oncogene.
[145] A. Krainer,et al. Listening to silence and understanding nonsense: exonic mutations that affect splicing , 2002, Nature Reviews Genetics.
[146] T. Kitahara,et al. A synthesis of FR901464 , 2001 .
[147] D. Hunt,et al. A human homolog of yeast pre-mRNA splicing gene, PRP31, underlies autosomal dominant retinitis pigmentosa on chromosome 19q13.4 (RP11). , 2001, Molecular cell.
[148] D. Mackey,et al. Mutations in the pre-mRNA splicing factor gene PRPC8 in autosomal dominant retinitis pigmentosa (RP13). , 2001, Human molecular genetics.
[149] R. Deshaies,et al. Protacs: Chimeric molecules that target proteins to the Skp1–Cullin–F box complex for ubiquitination and degradation , 2001, Proceedings of the National Academy of Sciences of the United States of America.
[150] M. Nakafuku,et al. Regulation of Gli2 and Gli3 activities by an amino-terminal repression domain: implication of Gli2 and Gli3 as primary mediators of Shh signaling. , 1999, Development.
[151] T. Owa,et al. Discovery of novel antitumor sulfonamides targeting G1 phase of the cell cycle. , 1999, Journal of medicinal chemistry.
[152] C. Guthrie,et al. Mechanical Devices of the Spliceosome: Motors, Clocks, Springs, and Things , 1998, Cell.
[153] M. Okuhara,et al. New antitumor substances, FR901463, FR901464 and FR901465. II. Activities against experimental tumors in mice and mechanism of action. , 1996, The Journal of antibiotics.
[154] P. Sharp,et al. Evidence for two active sites in the spliceosome provided by stereochemistry of pre-mRNA splicing , 1993, Nature.
[155] D. Mccormick. Sequence the Human Genome , 1986, Bio/Technology.
[156] James P. Orengo,et al. Alternative splicing in disease. , 2007, Advances in experimental medicine and biology.
[157] D. Brow,et al. Allosteric cascade of spliceosome activation. , 2002, Annual review of genetics.
[158] S. Berget,et al. Exon definition may facilitate splice site selection in RNAs with multiple exons. , 1990, Molecular and cellular biology.