Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Mol Cell ; 83(7): 1165-1179.e11, 2023 04 06.
Article in English | MEDLINE | ID: mdl-36944332

ABSTRACT

SF3B1 is the most mutated splicing factor (SF) in myelodysplastic syndromes (MDSs), which are clonal hematopoietic disorders with variable risk of leukemic transformation. Although tumorigenic SF3B1 mutations have been extensively characterized, the role of "non-mutated" wild-type SF3B1 in cancer remains largely unresolved. Here, we identify a conserved epitranscriptomic program that steers SF3B1 levels to counteract leukemogenesis. Our analysis of human and murine pre-leukemic MDS cells reveals dynamic regulation of SF3B1 protein abundance, which affects MDS-to-leukemia progression in vivo. Mechanistically, ALKBH5-driven 5' UTR m6A demethylation fine-tunes SF3B1 translation directing splicing of central DNA repair and epigenetic regulators during transformation. This impacts genome stability and leukemia progression in vivo, supporting an integrative analysis in humans that SF3B1 molecular signatures may predict mutational variability and poor prognosis. These findings highlight a post-transcriptional gene expression nexus that unveils unanticipated SF3B1-dependent cancer vulnerabilities.


Subject(s)
Leukemia , Myelodysplastic Syndromes , Phosphoproteins , RNA Splicing Factors , Animals , Humans , Mice , Carcinogenesis/genetics , Leukemia/genetics , Mutation , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , RNA Splicing/genetics , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism
2.
Nat Cell Biol ; 24(3): 299-306, 2022 03.
Article in English | MEDLINE | ID: mdl-35292784

ABSTRACT

Transfer RNA-derived fragments (tRFs) are emerging small noncoding RNAs that, although commonly altered in cancer, have poorly defined roles in tumorigenesis1. Here we show that pseudouridylation (Ψ) of a stem cell-enriched tRF subtype2, mini tRFs containing a 5' terminal oligoguanine (mTOG), selectively inhibits aberrant protein synthesis programmes, thereby promoting engraftment and differentiation of haematopoietic stem and progenitor cells (HSPCs) in patients with myelodysplastic syndrome (MDS). Building on evidence that mTOG-Ψ targets polyadenylate-binding protein cytoplasmic 1 (PABPC1), we employed isotope exchange proteomics to reveal critical interactions between mTOG and functional RNA-recognition motif (RRM) domains of PABPC1. Mechanistically, this hinders the recruitment of translational co-activator PABPC1-interacting protein 1 (PAIP1)3 and strongly represses the translation of transcripts sharing pyrimidine-enriched sequences (PES) at the 5' untranslated region (UTR), including 5' terminal oligopyrimidine tracts (TOP) that encode protein machinery components and are frequently altered in cancer4. Significantly, mTOG dysregulation leads to aberrantly increased translation of 5' PES messenger RNA (mRNA) in malignant MDS-HSPCs and is clinically associated with leukaemic transformation and reduced patient survival. These findings define a critical role for tRFs and Ψ in difficult-to-treat subsets of MDS characterized by high risk of progression to acute myeloid leukaemia (AML).


Subject(s)
Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Hematopoietic Stem Cells/metabolism , Humans , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Peptide Initiation Factors/metabolism , Pseudouridine , RNA, Transfer/genetics , RNA, Transfer/metabolism , RNA-Binding Proteins/genetics
3.
Mol Cell ; 81(7): 1453-1468.e12, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33662273

ABSTRACT

Splicing is a central RNA-based process commonly altered in human cancers; however, how spliceosomal components are co-opted during tumorigenesis remains poorly defined. Here we unravel the core splice factor SF3A3 at the nexus of a translation-based program that rewires splicing during malignant transformation. Upon MYC hyperactivation, SF3A3 levels are modulated translationally through an RNA stem-loop in an eIF3D-dependent manner. This ensures accurate splicing of mRNAs enriched for mitochondrial regulators. Altered SF3A3 translation leads to metabolic reprogramming and stem-like properties that fuel MYC tumorigenic potential in vivo. Our analysis reveals that SF3A3 protein levels predict molecular and phenotypic features of aggressive human breast cancers. These findings unveil a post-transcriptional interplay between splicing and translation that governs critical facets of MYC-driven oncogenesis.


Subject(s)
Breast Neoplasms/metabolism , Carcinogenesis/metabolism , Neoplastic Stem Cells/metabolism , Protein Biosynthesis , RNA Splicing Factors/biosynthesis , Spliceosomes/metabolism , Adult , Aged , Aged, 80 and over , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinogenesis/genetics , Female , Humans , Mice , Mice, Nude , Middle Aged , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Splicing Factors/genetics , Spliceosomes/genetics
4.
Sci Rep ; 10(1): 19142, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33154511

ABSTRACT

The under-representation of several ethnic groups in existing genetic databases and studies have undermined our understanding of the genetic variations and associated traits or diseases in many populations. Cost and technology limitations remain the challenges in performing large-scale genome sequencing projects in many developing countries, including Vietnam. As one of the most rapidly adopted genetic tests, non-invasive prenatal testing (NIPT) data offers an alternative untapped resource for genetic studies. Here we performed a large-scale genomic analysis of 2683 pregnant Vietnamese women using their NIPT data and identified a comprehensive set of 8,054,515 single-nucleotide polymorphisms, among which 8.2% were new to the Vietnamese population. Our study also revealed 24,487 disease-associated genetic variants and their allele frequency distribution, especially 5 pathogenic variants for prevalent genetic disorders in Vietnam. We also observed major discrepancies in the allele frequency distribution of disease-associated genetic variants between the Vietnamese and other populations, thus highlighting a need for genome-wide association studies dedicated to the Vietnamese population. The resulted database of Vietnamese genetic variants, their allele frequency distribution, and their associated diseases presents a valuable resource for future genetic studies.


Subject(s)
Alleles , Asian People/genetics , Gene Frequency , Genetic Testing , Genotype , Noninvasive Prenatal Testing , Female , Genome-Wide Association Study , Humans , Phenotype , Polymorphism, Single Nucleotide , Pregnancy , Vietnam
5.
Blood ; 135(12): 934-947, 2020 03 19.
Article in English | MEDLINE | ID: mdl-31972002

ABSTRACT

Adult T-cell leukemia/lymphoma (ATL) is a highly aggressive hematological malignancy derived from mature CD4+ T-lymphocytes. Here, we demonstrate the transcriptional regulatory network driven by 2 oncogenic transcription factors, IRF4 and NF-κB, in ATL cells. Gene expression profiling of primary ATL samples demonstrated that the IRF4 gene was more highly expressed in ATL cells than in normal T cells. Chromatin immunoprecipitation sequencing analysis revealed that IRF4-bound regions were more frequently found in super-enhancers than in typical enhancers. NF-κB was found to co-occupy IRF4-bound regulatory elements and formed a coherent feed-forward loop to coordinately regulate genes involved in T-cell functions and development. Importantly, IRF4 and NF-κB regulated several cancer genes associated with super-enhancers in ATL cells, including MYC, CCR4, and BIRC3. Genetic inhibition of BIRC3 induced growth inhibition in ATL cells, implicating its role as a critical effector molecule downstream of the IRF4-NF-κB transcriptional network.


Subject(s)
Interferon Regulatory Factors/metabolism , Leukemia-Lymphoma, Adult T-Cell/etiology , Leukemia-Lymphoma, Adult T-Cell/metabolism , NF-kappa B/metabolism , Signal Transduction , Apoptosis/genetics , Baculoviral IAP Repeat-Containing 3 Protein/metabolism , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Computational Biology , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Models, Biological , RNA, Small Interfering/genetics , Receptors, CCR4/metabolism
6.
Nat Commun ; 10(1): 5622, 2019 12 09.
Article in English | MEDLINE | ID: mdl-31819055

ABSTRACT

A heritable polymorphism within regulatory sequences of the LMO1 gene is associated with its elevated expression and increased susceptibility to develop neuroblastoma, but the oncogenic pathways downstream of the LMO1 transcriptional co-regulatory protein are unknown. Our ChIP-seq and RNA-seq analyses reveal that a key gene directly regulated by LMO1 and MYCN is ASCL1, which encodes a basic helix-loop-helix transcription factor. Regulatory elements controlling ASCL1 expression are bound by LMO1, MYCN and the transcription factors GATA3, HAND2, PHOX2B, TBX2 and ISL1-all members of the adrenergic (ADRN) neuroblastoma core regulatory circuitry (CRC). ASCL1 is required for neuroblastoma cell growth and arrest of differentiation. ASCL1 and LMO1 directly regulate the expression of CRC genes, indicating that ASCL1 is a member and LMO1 is a coregulator of the ADRN neuroblastoma CRC.


Subject(s)
Adrenergic Agents/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA-Binding Proteins/metabolism , Gene Regulatory Networks , LIM Domain Proteins/metabolism , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/metabolism , Transcription Factors/metabolism , Cell Differentiation , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Neuroblastoma/genetics , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Survival Analysis
7.
Blood ; 134(3): 239-251, 2019 07 18.
Article in English | MEDLINE | ID: mdl-31076442

ABSTRACT

The oncogenic transcription factor TAL1 regulates the transcriptional program in T-ALL. ARID5B is one of the critical downstream targets of TAL1, which further activates the oncogenic regulatory circuit in T-ALL cells. Here, we elucidated the molecular functions of the noncoding RNA, ARID5B-inducing enhancer associated long noncoding RNA (ARIEL), in T-ALL pathogenesis. We demonstrated that ARIEL is specifically activated in TAL1 + T-ALL cases, and its expression is associated with ARID5B enhancer activity. ARIEL recruits mediator proteins to the ARID5B enhancer, promotes enhancer-promoter interactions, and activates the expression of ARID5B, thereby positively regulating the TAL1-induced transcriptional program and the MYC oncogene. The TAL1 complex coordinately regulates the expression of ARIEL Knockdown of ARIEL inhibits cell growth and survival of T-ALL cells in culture and blocks disease progression in a murine xenograft model. Our results indicate that ARIEL plays an oncogenic role as an enhancer RNA in T-ALL.


Subject(s)
Carcinogenesis/genetics , Gene Expression Regulation, Leukemic , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , RNA, Long Noncoding/genetics , Transcription, Genetic , Animals , Binding Sites , Cell Line, Tumor , Cell Proliferation , Cell Survival/genetics , Chromatin Immunoprecipitation Sequencing , DNA-Binding Proteins/metabolism , Disease Models, Animal , Disease Progression , Enhancer Elements, Genetic , Gene Knockdown Techniques , Gene Targeting , Heterografts , Humans , Mice , Models, Biological , Multiprotein Complexes , Oncogenes , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Promoter Regions, Genetic , Protein Binding , T-Cell Acute Lymphocytic Leukemia Protein 1/metabolism , Transcription Factors/metabolism
8.
Cell ; 173(5): 1204-1216.e26, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29628141

ABSTRACT

Pseudouridylation (Ψ) is the most abundant and widespread type of RNA epigenetic modification in living organisms; however, the biological role of Ψ remains poorly understood. Here, we show that a Ψ-driven posttranscriptional program steers translation control to impact stem cell commitment during early embryogenesis. Mechanistically, the Ψ "writer" PUS7 modifies and activates a novel network of tRNA-derived small fragments (tRFs) targeting the translation initiation complex. PUS7 inactivation in embryonic stem cells impairs tRF-mediated translation regulation, leading to increased protein biosynthesis and defective germ layer specification. Remarkably, dysregulation of this posttranscriptional regulatory circuitry impairs hematopoietic stem cell commitment and is common to aggressive subtypes of human myelodysplastic syndromes. Our findings unveil a critical function of Ψ in directing translation control in stem cells with important implications for development and disease.


Subject(s)
Intramolecular Transferases/metabolism , Protein Biosynthesis , Pseudouridine/metabolism , RNA, Transfer/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Cycle Proteins , Cell Differentiation , Eukaryotic Initiation Factors/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Intramolecular Transferases/antagonists & inhibitors , Intramolecular Transferases/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Myelodysplastic Syndromes/pathology , Nucleic Acid Conformation , Phosphoproteins/metabolism , Poly(A)-Binding Protein I/antagonists & inhibitors , Poly(A)-Binding Protein I/genetics , Poly(A)-Binding Protein I/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Stem Cell Niche
9.
Leukemia ; 32(10): 2138-2151, 2018 10.
Article in English | MEDLINE | ID: mdl-29654272

ABSTRACT

TAL1/SCL is one of the most prevalent oncogenes in T-cell acute lymphoblastic leukemia (T-ALL). TAL1 and its regulatory partners (GATA3, RUNX1, and MYB) positively regulate each other and coordinately regulate the expression of their downstream target genes in T-ALL cells. However, long non-coding RNAs (lncRNAs) regulated by these factors are largely unknown. Here we established a bioinformatics pipeline and analyzed RNA-seq datasets with deep coverage to identify lncRNAs regulated by TAL1 in T-ALL cells. Our analysis predicted 57 putative lncRNAs that are activated by TAL1. Many of these transcripts were regulated by GATA3, RUNX1, and MYB in a coordinated manner. We identified two novel transcripts that were activated in multiple T-ALL cell samples but were downregulated in normal thymocytes. One transcript near the ARID5B gene locus was specifically expressed in TAL1-positive T-ALL cases. The other transcript located between the FAM49A and MYCN gene locus was also expressed in normal hematopoietic stem cells and T-cell progenitor cells. In addition, we identified a subset of lncRNAs that were negatively regulated by TAL1 and positively regulated by E-proteins in T-ALL cells. This included a known lncRNA (lnc-OAZ3-2:7) located near the RORC gene, which was expressed in normal thymocytes but repressed in TAL1-positive T-ALL cells.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , RNA, Long Noncoding/genetics , T-Cell Acute Lymphocytic Leukemia Protein 1/genetics , Animals , Cell Line, Tumor , Core Binding Factor Alpha 2 Subunit/genetics , DNA-Binding Proteins/genetics , GATA3 Transcription Factor/genetics , Gene Expression Regulation, Leukemic/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , N-Myc Proto-Oncogene Protein/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Thymocytes/physiology
10.
Blood ; 130(21): 2326-2338, 2017 11 23.
Article in English | MEDLINE | ID: mdl-28978570

ABSTRACT

A number of studies have recently demonstrated that super-enhancers, which are large cluster of enhancers typically marked by a high level of acetylation of histone H3 lysine 27 and mediator bindings, are frequently associated with genes that control and define cell identity during normal development. Super-enhancers are also often enriched at cancer genes in various malignancies. The identification of such enhancers would pinpoint critical factors that directly contribute to pathogenesis. In this study, we performed enhancer profiling using primary leukemia samples from adult T-cell leukemia/lymphoma (ATL), which is a genetically heterogeneous intractable cancer. Super-enhancers were enriched at genes involved in the T-cell activation pathway, including IL2RA/CD25, CD30, and FYN, in both ATL and normal mature T cells, which reflected the origin of the leukemic cells. Super-enhancers were found at several known cancer gene loci, including CCR4, PIK3R1, and TP73, in multiple ATL samples, but not in normal mature T cells, which implicated those genes in ATL pathogenesis. A small-molecule CDK7 inhibitor, THZ1, efficiently inhibited cell growth, induced apoptosis, and downregulated the expression of super-enhancer-associated genes in ATL cells. Furthermore, enhancer profiling combined with gene expression analysis identified a previously uncharacterized gene, TIAM2, that was associated with super-enhancers in all ATL samples, but not in normal T cells. Knockdown of TIAM2 induced apoptosis in ATL cell lines, whereas overexpression of this gene promoted cell growth. Our study provides a novel strategy for identifying critical cancer genes.


Subject(s)
Enhancer Elements, Genetic/genetics , Gene Expression Profiling , Genes, Neoplasm , Leukemia-Lymphoma, Adult T-Cell/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Gene Expression Regulation, Leukemic/drug effects , Genetic Association Studies , Humans , Leukemia-Lymphoma, Adult T-Cell/drug therapy , Leukemia-Lymphoma, Adult T-Cell/immunology , Leukemia-Lymphoma, Adult T-Cell/pathology , Lymphocyte Activation/genetics , Phenylenediamines/pharmacology , Phenylenediamines/therapeutic use , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , RNA Polymerase II/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Cyclin-Dependent Kinase-Activating Kinase
11.
Adv Exp Med Biol ; 962: 139-147, 2017.
Article in English | MEDLINE | ID: mdl-28299656

ABSTRACT

Enhancers are regulatory elements in genomic DNA that contain specific sequence motifs that are bound by DNA-binding transcription factors. The activity of enhancers is tightly regulated in an integrated and combinatorial manner, thus yielding complex patterns of transcription in different tissues. Identifying enhancers is crucial to understanding the physiological and pathogenic roles of their target genes. The RUNX1 intronic enhancer, eR1, acts in cis to regulate RUNX1 gene expression in hematopoietic stem cells (HSCs) and hemogenic endothelial cells. RUNX1 and other hematopoietic transcription factors TAL1/SCL, GATA2, PU.1, LMO2 and LDB1 bind at this region. Interestingly, recent studies have revealed that this region is involved in a large cluster of enhancers termed a super-enhancer. The RUNX1 super-enhancer is observed in normal HSCs and T-cell acute lymphoblastic leukemia cells. In this review, we describe the discovery of eR1 and its roles in normal development and leukemogenesis, as well as its potential applications in stem cell research.


Subject(s)
Core Binding Factor Alpha 2 Subunit/metabolism , Hematopoiesis/physiology , Leukemia/metabolism , Leukemia/pathology , Animals , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelial Cells/physiology , Gene Expression/physiology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Hematopoietic Stem Cells/physiology , Humans , Transcription Factors/metabolism
12.
Genes Dev ; 31(23-24): 2343-2360, 2017 12 01.
Article in English | MEDLINE | ID: mdl-29326336

ABSTRACT

The oncogenic transcription factor TAL1/SCL induces an aberrant transcriptional program in T-cell acute lymphoblastic leukemia (T-ALL) cells. However, the critical factors that are directly activated by TAL1 and contribute to T-ALL pathogenesis are largely unknown. Here, we identified AT-rich interactive domain 5B (ARID5B) as a collaborating oncogenic factor involved in the transcriptional program in T-ALL. ARID5B expression is down-regulated at the double-negative 2-4 stages in normal thymocytes, while it is induced by the TAL1 complex in human T-ALL cells. The enhancer located 135 kb upstream of the ARID5B gene locus is activated under a superenhancer in T-ALL cells but not in normal T cells. Notably, ARID5B-bound regions are associated predominantly with active transcription. ARID5B and TAL1 frequently co-occupy target genes and coordinately control their expression. ARID5B positively regulates the expression of TAL1 and its regulatory partners. ARID5B also activates the expression of the oncogene MYC Importantly, ARID5B is required for the survival and growth of T-ALL cells, and forced expression of ARID5B in immature thymocytes results in thymus retention, differentiation arrest, radioresistance, and tumor formation in zebrafish. Our results indicate that ARID5B reinforces the oncogenic transcriptional program by positively regulating the TAL1-induced regulatory circuit and MYC in T-ALL, thereby contributing to T-cell leukemogenesis.


Subject(s)
Carcinogenesis/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , T-Cell Acute Lymphocytic Leukemia Protein 1/metabolism , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Cell Survival/genetics , DNA-Binding Proteins/genetics , Enhancer Elements, Genetic/genetics , Gene Expression Profiling , Genes, myc/genetics , HEK293 Cells , Humans , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Protein Binding , Protein Domains/genetics , Thymocytes/metabolism , Thymus Gland/growth & development , Transcription Factors/genetics , Transcriptional Activation/genetics , Zebrafish
13.
Genome Biol Evol ; 8(11): 3323-3339, 2016 12 14.
Article in English | MEDLINE | ID: mdl-27797949

ABSTRACT

While mechanisms to detoxify plant produced, anti-herbivore compounds have been associated with plant host use by herbivores, less is known about the role of chemosensory perception in their life histories. This is especially true for generalists, including chelicerate herbivores that evolved herbivory independently from the more studied insect lineages. To shed light on chemosensory perception in a generalist herbivore, we characterized the chemosensory receptors (CRs) of the chelicerate two-spotted spider mite, Tetranychus urticae, an extreme generalist. Strikingly, T. urticae has more CRs than reported in any other arthropod to date. Including pseudogenes, 689 gustatory receptors were identified, as were 136 degenerin/Epithelial Na+ Channels (ENaCs) that have also been implicated as CRs in insects. The genomic distribution of T. urticae gustatory receptors indicates recurring bursts of lineage-specific proliferations, with the extent of receptor clusters reminiscent of those observed in the CR-rich genomes of vertebrates or C. elegans Although pseudogenization of many gustatory receptors within clusters suggests relaxed selection, a subset of receptors is expressed. Consistent with functions as CRs, the genomic distribution and expression of ENaCs in lineage-specific T. urticae expansions mirrors that observed for gustatory receptors. The expansion of ENaCs in T. urticae to > 3-fold that reported in other animals was unexpected, raising the possibility that ENaCs in T. urticae have been co-opted to fulfill a major role performed by unrelated CRs in other animals. More broadly, our findings suggest an elaborate role for chemosensory perception in generalist herbivores that are of key ecological and agricultural importance.


Subject(s)
Acari/genetics , Epithelial Sodium Channels/genetics , Evolution, Molecular , Insect Proteins/genetics , Receptors, Cell Surface/genetics , Taste , Acari/metabolism , Acari/physiology , Animals , Epithelial Sodium Channels/metabolism , Herbivory/genetics , Insect Proteins/metabolism , Multigene Family , Receptors, Cell Surface/metabolism
14.
Nature ; 479(7374): 487-92, 2011 Nov 23.
Article in English | MEDLINE | ID: mdl-22113690

ABSTRACT

The spider mite Tetranychus urticae is a cosmopolitan agricultural pest with an extensive host plant range and an extreme record of pesticide resistance. Here we present the completely sequenced and annotated spider mite genome, representing the first complete chelicerate genome. At 90 megabases T. urticae has the smallest sequenced arthropod genome. Compared with other arthropods, the spider mite genome shows unique changes in the hormonal environment and organization of the Hox complex, and also reveals evolutionary innovation of silk production. We find strong signatures of polyphagy and detoxification in gene families associated with feeding on different hosts and in new gene families acquired by lateral gene transfer. Deep transcriptome analysis of mites feeding on different plants shows how this pest responds to a changing host environment. The T. urticae genome thus offers new insights into arthropod evolution and plant-herbivore interactions, and provides unique opportunities for developing novel plant protection strategies.


Subject(s)
Adaptation, Physiological/genetics , Genome/genetics , Herbivory/genetics , Tetranychidae/genetics , Tetranychidae/physiology , Adaptation, Physiological/physiology , Animals , Ecdysterone/analogs & derivatives , Ecdysterone/genetics , Evolution, Molecular , Fibroins/genetics , Gene Expression Regulation , Gene Transfer, Horizontal/genetics , Genes, Homeobox/genetics , Genomics , Herbivory/physiology , Molecular Sequence Data , Molting/genetics , Multigene Family/genetics , Nanostructures/chemistry , Plants/parasitology , Silk/biosynthesis , Silk/chemistry , Transcriptome/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...