Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Nucleic Acids Res ; 52(15): 9247-9266, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-38943346

ABSTRACT

Classification of introns, which is crucial to understanding their evolution and splicing, has historically been binary and has resulted in the naming of major and minor introns that are spliced by their namesake spliceosome. However, a broad range of intron consensus sequences exist, leading us to here reclassify introns as minor, minor-like, hybrid, major-like, major and non-canonical introns in 263 species across six eukaryotic supergroups. Through intron orthology analysis, we discovered that minor-like introns are a transitory node for intron conversion across evolution. Despite close resemblance of their consensus sequences to minor introns, these introns possess an AG dinucleotide at the -1 and -2 position of the 5' splice site, a salient feature of major introns. Through combined analysis of CoLa-seq, CLIP-seq for major and minor spliceosome components, and RNAseq from samples in which the minor spliceosome is inhibited we found that minor-like introns are also an intermediate class from a splicing mechanism perspective. Importantly, this analysis has provided insight into the sequence elements that have evolved to make minor-like introns amenable to recognition by both minor and major spliceosome components. We hope that this revised intron classification provides a new framework to study intron evolution and splicing.


Subject(s)
Evolution, Molecular , Introns , RNA Splicing , Spliceosomes , Introns/genetics , Spliceosomes/genetics , Humans , RNA Splice Sites , Animals , Consensus Sequence , Eukaryota/genetics , Eukaryota/classification , Base Sequence
2.
Development ; 149(20)2022 10 15.
Article in English | MEDLINE | ID: mdl-35997536

ABSTRACT

Complex organ development depends on single lumen formation and its expansion during tubulogenesis. This can be achieved by correct mitotic spindle orientation during cell division, combined with luminal fluid filling that generates hydrostatic pressure. Using a human 3D cell culture model, we have identified two regulators of these processes. We find that pleckstrin homology leucine-rich repeat protein phosphatase (PHLPP) 2 regulates mitotic spindle orientation, and thereby midbody positioning and maintenance of a single lumen. Silencing the sole PHLPP family phosphatase in Drosophila melanogaster, phlpp, resulted in defective spindle orientation in Drosophila neuroblasts. Importantly, cystic fibrosis transmembrane conductance regulator (CFTR) is the main channel regulating fluid transport in this system, stimulated by phosphorylation by protein kinase A and inhibited by the AMP-activated protein kinase AMPK. During lumen expansion, CFTR remains open through the action of PHLPP1, which stops activated AMPK from inhibiting ion transport through CFTR. In the absence of PHLPP1, the restraint on AMPK activity is lost and this tips the balance in the favour of channel closing, resulting in the lack of lumen expansion and accumulation of mucus.


Subject(s)
AMP-Activated Protein Kinases , Cystic Fibrosis Transmembrane Conductance Regulator , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/metabolism , Phosphoric Monoester Hydrolases/metabolism , Phosphorylation
3.
Nature ; 560(7717): 253-257, 2018 08.
Article in English | MEDLINE | ID: mdl-30069049

ABSTRACT

Acetylation of histones by lysine acetyltransferases (KATs) is essential for chromatin organization and function1. Among the genes coding for the MYST family of KATs (KAT5-KAT8) are the oncogenes KAT6A (also known as MOZ) and KAT6B (also known as MORF and QKF)2,3. KAT6A has essential roles in normal haematopoietic stem cells4-6 and is the target of recurrent chromosomal translocations, causing acute myeloid leukaemia7,8. Similarly, chromosomal translocations in KAT6B have been identified in diverse cancers8. KAT6A suppresses cellular senescence through the regulation of suppressors of the CDKN2A locus9,10, a function that requires its KAT activity10. Loss of one allele of KAT6A extends the median survival of mice with MYC-induced lymphoma from 105 to 413 days11. These findings suggest that inhibition of KAT6A and KAT6B may provide a therapeutic benefit in cancer. Here we present highly potent, selective inhibitors of KAT6A and KAT6B, denoted WM-8014 and WM-1119. Biochemical and structural studies demonstrate that these compounds are reversible competitors of acetyl coenzyme A and inhibit MYST-catalysed histone acetylation. WM-8014 and WM-1119 induce cell cycle exit and cellular senescence without causing DNA damage. Senescence is INK4A/ARF-dependent and is accompanied by changes in gene expression that are typical of loss of KAT6A function. WM-8014 potentiates oncogene-induced senescence in vitro and in a zebrafish model of hepatocellular carcinoma. WM-1119, which has increased bioavailability, arrests the progression of lymphoma in mice. We anticipate that this class of inhibitors will help to accelerate the development of therapeutics that target gene transcription regulated by histone acetylation.


Subject(s)
Benzenesulfonates/pharmacology , Cellular Senescence/drug effects , Histone Acetyltransferases/antagonists & inhibitors , Hydrazines/pharmacology , Lymphoma/drug therapy , Lymphoma/pathology , Sulfonamides/pharmacology , Acetylation/drug effects , Animals , Benzenesulfonates/therapeutic use , Cell Proliferation/drug effects , Cells, Cultured , Drug Development , Fibroblasts , Gene Expression Regulation, Neoplastic/drug effects , Histone Acetyltransferases/deficiency , Histone Acetyltransferases/genetics , Histones/chemistry , Histones/metabolism , Hydrazines/therapeutic use , Lymphoma/enzymology , Lymphoma/genetics , Lysine/chemistry , Lysine/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Sulfonamides/therapeutic use
4.
Nucleic Acids Res ; 49(6): 3524-3545, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33660780

ABSTRACT

Vertebrate genomes contain major (>99.5%) and minor (<0.5%) introns that are spliced by the major and minor spliceosomes, respectively. Major intron splicing follows the exon-definition model, whereby major spliceosome components first assemble across exons. However, since most genes with minor introns predominately consist of major introns, formation of exon-definition complexes in these genes would require interaction between the major and minor spliceosomes. Here, we report that minor spliceosome protein U11-59K binds to the major spliceosome U2AF complex, thereby supporting a model in which the minor spliceosome interacts with the major spliceosome across an exon to regulate the splicing of minor introns. Inhibition of minor spliceosome snRNAs and U11-59K disrupted exon-bridging interactions, leading to exon skipping by the major spliceosome. The resulting aberrant isoforms contained a premature stop codon, yet were not subjected to nonsense-mediated decay, but rather bound to polysomes. Importantly, we detected elevated levels of these alternatively spliced transcripts in individuals with minor spliceosome-related diseases such as Roifman syndrome, Lowry-Wood syndrome and early-onset cerebellar ataxia. In all, we report that the minor spliceosome informs splicing by the major spliceosome through exon-definition interactions and show that minor spliceosome inhibition results in aberrant alternative splicing in disease.


Subject(s)
Alternative Splicing , Exons , Introns , Spliceosomes/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Cardiomyopathies/genetics , Cells, Cultured , Cerebellar Ataxia/genetics , Growth Disorders/genetics , Humans , Intellectual Disability/genetics , Mental Retardation, X-Linked/genetics , Mice , Microcephaly/genetics , Nonsense Mediated mRNA Decay , Osteochondrodysplasias/genetics , Polyribosomes/metabolism , Primary Immunodeficiency Diseases/genetics , RNA, Small Nuclear/antagonists & inhibitors , Retinal Diseases/genetics , Transcription Factors/metabolism
5.
RNA ; 24(12): 1856-1870, 2018 12.
Article in English | MEDLINE | ID: mdl-30254136

ABSTRACT

Splicing is an essential step in eukaryotic gene expression. While the majority of introns is excised by the U2-dependent, or major class, spliceosome, the appropriate expression of a very small subset of genes depends on U12-dependent, or minor class, splicing. The U11/U12 65K protein (hereafter 65K), encoded by RNPC3, is one of seven proteins that are unique to the U12-dependent spliceosome, and previous studies including our own have established that it plays a role in plant and vertebrate development. To pinpoint the impact of 65K loss during mammalian development and in adulthood, we generated germline and conditional Rnpc3-deficient mice. Homozygous Rnpc3-/- embryos died prior to blastocyst implantation, whereas Rnpc3+/- mice were born at the expected frequency, achieved sexual maturity, and exhibited a completely normal lifespan. Systemic recombination of conditional Rnpc3 alleles in adult (Rnpc3lox/lox ) mice caused rapid weight loss, leukopenia, and degeneration of the epithelial lining of the entire gastrointestinal tract, the latter due to increased cell death and a reduction in cell proliferation. Accompanying this, we observed a loss of both 65K and the pro-proliferative phospho-ERK1/2 proteins from the stem/progenitor cells at the base of intestinal crypts. RT-PCR analysis of RNA extracted from purified preparations of intestinal epithelial cells with recombined Rnpc3lox alleles revealed increased frequency of U12-type intron retention in all transcripts tested. Our study, using a novel conditional mouse model of Rnpc3 deficiency, establishes that U12-dependent splicing is not only important during development but is indispensable throughout life.


Subject(s)
RNA Splicing/genetics , RNA-Binding Proteins/genetics , Ribonucleoproteins, Small Nuclear/genetics , Alleles , Animals , Gastrointestinal Tract/metabolism , Humans , Introns/genetics , Mice , RNA, Small Nuclear/chemical synthesis , RNA, Small Nuclear/genetics , RNA-Binding Proteins/chemistry , Ribonucleoproteins, Small Nuclear/chemistry , Ribonucleoproteins, Small Nuclear/deficiency , Spliceosomes/chemistry , Spliceosomes/genetics
6.
Proc Natl Acad Sci U S A ; 111(8): 3062-7, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24516132

ABSTRACT

Minor class or U12-type splicing is a highly conserved process required to remove a minute fraction of introns from human pre-mRNAs. Defects in this splicing pathway have recently been linked to human disease, including a severe developmental disorder encompassing brain and skeletal abnormalities known as Taybi-Linder syndrome or microcephalic osteodysplastic primordial dwarfism 1, and a hereditary intestinal polyposis condition, Peutz-Jeghers syndrome. Although a key mechanism for regulating gene expression, the impact of impaired U12-type splicing on the transcriptome is unknown. Here, we describe a unique zebrafish mutant, caliban (clbn), with arrested development of the digestive organs caused by an ethylnitrosourea-induced recessive lethal point mutation in the rnpc3 [RNA-binding region (RNP1, RRM) containing 3] gene. rnpc3 encodes the zebrafish ortholog of human RNPC3, also known as the U11/U12 di-snRNP 65-kDa protein, a unique component of the U12-type spliceosome. The biochemical impact of the mutation in clbn is the formation of aberrant U11- and U12-containing small nuclear ribonucleoproteins that impair the efficiency of U12-type splicing. Using RNA sequencing and microarrays, we show that multiple genes involved in various steps of mRNA processing, including transcription, splicing, and nuclear export are disrupted in clbn, either through intron retention or differential gene expression. Thus, clbn provides a useful and specific model of aberrant U12-type splicing in vivo. Analysis of its transcriptome reveals efficient mRNA processing as a critical process for the growth and proliferation of cells during vertebrate development.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Protein Conformation , RNA Splicing/physiology , RNA, Small Nuclear/chemistry , RNA-Binding Proteins/genetics , Spliceosomes/metabolism , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Base Sequence , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , Intestines/abnormalities , Liver/abnormalities , Microarray Analysis , Molecular Sequence Data , Pancreas/abnormalities , Point Mutation/genetics , RNA Splicing/genetics , RNA-Binding Proteins/metabolism , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Spliceosomes/genetics , Zebrafish/growth & development , Zebrafish Proteins/metabolism
7.
Adv Exp Med Biol ; 916: 411-37, 2016.
Article in English | MEDLINE | ID: mdl-27165364

ABSTRACT

Colorectal cancer, encompassing colon and rectal cancer, arises from the epithelial lining of the large bowel. It is most prevalent in Westernised societies and is increasing in frequency as the world becomes more industrialised. Unfortunately, metastatic colorectal cancer is not cured by chemotherapy and the annual number of deaths caused by colorectal cancer, currently 700,000, is expected to rise. Our understanding of the contribution that genetic mutations make to colorectal cancer, although incomplete, is reasonably well advanced. However, it has only recently become widely appreciated that in addition to the ongoing accumulation of genetic mutations, chronic inflammation also plays a critical role in the initiation and progression of this disease. While a robust and tractable genetic model of colorectal cancer in zebrafish, suitable for pre-clinical studies, is not yet available, the identification of genes required for the rapid proliferation of zebrafish intestinal epithelial cells during development has highlighted a number of essential genes that could be targeted to disable colorectal cancer cells. Moreover, appreciation of the utility of zebrafish to study intestinal inflammation is on the rise. In particular, zebrafish provide unique opportunities to investigate the impact of genetic and environmental factors on the integrity of intestinal epithelial barrier function. With currently available tools, the interplay between epigenetic regulators, intestinal injury, microbiota composition and innate immune cell mobilisation can be analysed in exquisite detail. This provides excellent opportunities to define critical events that could potentially be targeted therapeutically. Further into the future, the use of zebrafish larvae as hosts for xenografts of human colorectal cancer tissue, while still in its infancy, holds great promise that zebrafish could one day provide a practical, preclinical personalized medicine platform for the rapid assessment of the metastatic potential and drug-sensitivity of patient-derived cancers.


Subject(s)
Colorectal Neoplasms/pathology , Disease Models, Animal , Animals , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/therapy , Prevalence , Zebrafish
8.
PLoS Genet ; 9(2): e1003279, 2013.
Article in English | MEDLINE | ID: mdl-23408911

ABSTRACT

Ribosome biogenesis underpins cell growth and division. Disruptions in ribosome biogenesis and translation initiation are deleterious to development and underlie a spectrum of diseases known collectively as ribosomopathies. Here, we describe a novel zebrafish mutant, titania (tti(s450)), which harbours a recessive lethal mutation in pwp2h, a gene encoding a protein component of the small subunit processome. The biochemical impacts of this lesion are decreased production of mature 18S rRNA molecules, activation of Tp53, and impaired ribosome biogenesis. In tti(s450), the growth of the endodermal organs, eyes, brain, and craniofacial structures is severely arrested and autophagy is up-regulated, allowing intestinal epithelial cells to evade cell death. Inhibiting autophagy in tti(s450) larvae markedly reduces their lifespan. Somewhat surprisingly, autophagy induction in tti(s450) larvae is independent of the state of the Tor pathway and proceeds unabated in Tp53-mutant larvae. These data demonstrate that autophagy is a survival mechanism invoked in response to ribosomal stress. This response may be of relevance to therapeutic strategies aimed at killing cancer cells by targeting ribosome biogenesis. In certain contexts, these treatments may promote autophagy and contribute to cancer cells evading cell death.


Subject(s)
Autophagy/genetics , Cell Cycle Proteins , Ribosomes , TOR Serine-Threonine Kinases , Tumor Suppressor Protein p53 , Zebrafish Proteins , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Survival , Genes, Lethal/genetics , Mutation , Protein Biosynthesis/genetics , RNA, Ribosomal, 18S/genetics , RNA, Ribosomal, 18S/metabolism , Ribosomes/genetics , Ribosomes/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/genetics , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
9.
Development ; 139(3): 525-36, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22223680

ABSTRACT

The isthmic organiser located at the midbrain-hindbrain boundary (MHB) is the crucial developmental signalling centre responsible for patterning mesencephalic and metencephalic regions of the vertebrate brain. Formation and maintenance of the MHB is characterised by a hierarchical program of gene expression initiated by fibroblast growth factor 8 (Fgf8), coupled with cellular morphogenesis, culminating in the formation of the tectal-isthmo-cerebellar structures. Here, we show in zebrafish that one orthologue of the transcription factor grainy head-like 2 (Grhl2), zebrafish grhl2b plays a central role in both MHB maintenance and folding by regulating two distinct, non-linear pathways. Loss of grhl2b expression induces neural apoptosis and extinction of MHB markers, which are rescued by re-expression of engrailed 2a (eng2a), an evolutionarily conserved target of the Grhl family. Co-injection of sub-phenotypic doses of grhl2b and eng2a morpholinos reproduces the apoptosis and MHB marker loss, but fails to substantially disrupt formation of the isthmic constriction. By contrast, a novel direct grhl2b target, spec1, identified by phylogenetic analysis and confirmed by ChIP, functionally cooperates with grhl2b to induce MHB morphogenesis, but plays no role in apoptosis or maintenance of MHB markers. Collectively, these data show that MHB maintenance and morphogenesis are dissociable events regulated by grhl2b through diverse transcriptional targets.


Subject(s)
Carrier Proteins/metabolism , Mesencephalon/growth & development , Morphogenesis , Rhombencephalon/growth & development , Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Zebrafish/growth & development , Animals , Apoptosis , Carrier Proteins/genetics , Gene Expression Regulation, Developmental/genetics , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Mesencephalon/metabolism , Morpholinos/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Phylogeny , Rhombencephalon/metabolism , Signal Transduction , Transcription Factors/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
10.
Biochem J ; 458(2): 251-8, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24320611

ABSTRACT

PIK3CA, the gene encoding the p110α catalytic subunit of PI3K (phosphoinositide 3-kinase), is mutated in approximately 20% of sporadic CRCs (colorectal cancers), but the role of these mutations in the pathogenesis of CRC remains unclear. In the present study we used a novel mouse model to investigate the role of the Pik3caH1047R mutation, the most common PIK3CA mutation in CRC, during the development and progression of intestinal cancer. Our results demonstrate that Pik3caH1047R, when expressed at physiological levels, is insufficient to initiate intestinal tumorigenesis; however, in the context of Apc (adenomatous polyposis coli) loss, which is observed in 80% of CRCs and by itself results in benign intestinal adenomas, the Pik3caH1047R mutation promotes the development of highly aggressive and invasive adenocarcinomas in both the small and large intestines. The results of the present study show that an activating Pik3ca mutation can act in tandem with Apc loss to drive the progression of gastrointestinal cancer and thus this disease may be susceptible to therapeutic targeting using PI3K pathway inhibitors.


Subject(s)
Adenocarcinoma/genetics , Adenomatous Polyposis Coli Protein/deficiency , Gene Expression Regulation, Neoplastic , Intestinal Neoplasms/genetics , Mutation/genetics , Neoplasm Invasiveness , Phosphatidylinositol 3-Kinases/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenomatous Polyposis Coli Protein/genetics , Animals , Class I Phosphatidylinositol 3-Kinases , Disease Progression , Female , Gene Knock-In Techniques , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Phosphatidylinositol 3-Kinases/biosynthesis
11.
Development ; 137(13): 2107-15, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20504962

ABSTRACT

It is currently unclear how intrinsic and extrinsic mechanisms cooperate to control the progression from self-renewing to neurogenic divisions in retinal precursor cells. Here, we use the zebrafish flotte lotte (flo) mutant, which carries a mutation in the elys (ahctf1) gene, to study the relationship between cell cycle progression and neuronal differentiation by investigating how proliferating progenitor cells transition towards differentiation in a retinal stem cell niche termed the ciliary marginal zone (CMZ). In zebrafish embryos without Elys, CMZ cells retain the capacity to proliferate but lose the ability to enter their final neurogenic divisions to differentiate as neurons. However, mosaic retinae composed of wild-type and flo cells show that despite inherent cell cycle defects, flo mutant cells progress from proliferation to differentiation when in the vicinity of wild-type retinal neurons. We propose that the differentiated retinal environment limits the proliferation of precursors emerging from the CMZ in a manner that explains the spatial organisation of cells in the CMZ and ensures that proliferative retinal progenitors are driven towards differentiation.


Subject(s)
Neurogenesis , Nuclear Pore Complex Proteins/metabolism , Retina/cytology , Stem Cells/cytology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Apoptosis , Feedback , Nuclear Pore Complex Proteins/genetics , Organ Size , Retina/metabolism , Zebrafish Proteins/genetics
12.
PLoS Genet ; 6(1): e1000816, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20084116

ABSTRACT

Contributions of null and hypomorphic alleles of Apc in mice produce both developmental and pathophysiological phenotypes. To ascribe the resulting genotype-to-phenotype relationship unambiguously to the Wnt/beta-catenin pathway, we challenged the allele combinations by genetically restricting intracellular beta-catenin expression in the corresponding compound mutant mice. Subsequent evaluation of the extent of resulting Tcf4-reporter activity in mouse embryo fibroblasts enabled genetic measurement of Wnt/beta-catenin signaling in the form of an allelic series of mouse mutants. Different permissive Wnt signaling thresholds appear to be required for the embryonic development of head structures, adult intestinal polyposis, hepatocellular carcinomas, liver zonation, and the development of natural killer cells. Furthermore, we identify a homozygous Apc allele combination with Wnt/beta-catenin signaling capacity similar to that in the germline of the Apc(min) mice, where somatic Apc loss-of-heterozygosity triggers intestinal polyposis, to distinguish whether co-morbidities in Apc(min) mice arise independently of intestinal tumorigenesis. Together, the present genotype-phenotype analysis suggests tissue-specific response levels for the Wnt/beta-catenin pathway that regulate both physiological and pathophysiological conditions.


Subject(s)
Mice/genetics , Mice/metabolism , Signal Transduction , beta Catenin/metabolism , Adenomatous Polyposis Coli Protein/genetics , Adenomatous Polyposis Coli Protein/metabolism , Animals , Cells, Cultured , Embryo, Mammalian , Female , Fibroblasts/metabolism , Intestinal Mucosa/metabolism , Intestines/embryology , Intestines/growth & development , Liver/embryology , Liver/growth & development , Liver/metabolism , Male , Mice/embryology , Mice/growth & development , Mice, Inbred C57BL , Mice, Knockout , Wnt Proteins , Wnt3 Protein , beta Catenin/genetics
13.
Elife ; 122023 01 17.
Article in English | MEDLINE | ID: mdl-36648336

ABSTRACT

The nucleoporin (NUP) ELYS, encoded by AHCTF1, is a large multifunctional protein with essential roles in nuclear pore assembly and mitosis. Using both larval and adult zebrafish models of hepatocellular carcinoma (HCC), in which the expression of an inducible mutant kras transgene (krasG12V) drives hepatocyte-specific hyperplasia and liver enlargement, we show that reducing ahctf1 gene dosage by 50% markedly decreases liver volume, while non-hyperplastic tissues are unaffected. We demonstrate that in the context of cancer, ahctf1 heterozygosity impairs nuclear pore formation, mitotic spindle assembly, and chromosome segregation, leading to DNA damage and activation of a Tp53-dependent transcriptional programme that induces cell death and cell cycle arrest. Heterozygous expression of both ahctf1 and ranbp2 (encoding a second nucleoporin), or treatment of heterozygous ahctf1 larvae with the nucleocytoplasmic transport inhibitor, Selinexor, completely blocks krasG12V-driven hepatocyte hyperplasia. Gene expression analysis of patient samples in the liver hepatocellular carcinoma (LIHC) dataset in The Cancer Genome Atlas shows that high expression of one or more of the transcripts encoding the 10 components of the NUP107-160 subcomplex, which includes AHCTF1, is positively correlated with worse overall survival. These results provide a strong and feasible rationale for the development of novel cancer therapeutics that target ELYS function and suggest potential avenues for effective combinatorial treatments.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Zebrafish/genetics , Zebrafish/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Hyperplasia , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
14.
Nat Commun ; 13(1): 2073, 2022 04 19.
Article in English | MEDLINE | ID: mdl-35440107

ABSTRACT

Modulation of protein abundance using tag-Targeted Protein Degrader (tTPD) systems targeting FKBP12F36V (dTAGs) or HaloTag7 (HaloPROTACs) are powerful approaches for preclinical target validation. Interchanging tags and tag-targeting degraders is important to achieve efficient substrate degradation, yet limited degrader/tag pairs are available and side-by-side comparisons have not been performed. To expand the tTPD repertoire we developed catalytic NanoLuc-targeting PROTACs (NanoTACs) to hijack the CRL4CRBN complex and degrade NanoLuc tagged substrates, enabling rapid luminescence-based degradation screening. To benchmark NanoTACs against existing tTPD systems we use an interchangeable reporter system to comparatively test optimal degrader/tag pairs. Overall, we find the dTAG system exhibits superior degradation. To align tag-induced degradation with physiology we demonstrate that NanoTACs limit MLKL-driven necroptosis. In this work we extend the tTPD platform to include NanoTACs adding flexibility to tTPD studies, and benchmark each tTPD system to highlight the importance of comparing each system against each substrate.


Subject(s)
Benchmarking , Tacrolimus Binding Protein 1A , Luciferases , Proteolysis , Tacrolimus Binding Protein 1A/genetics
15.
Nat Med ; 8(10): 1089-97, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12219085

ABSTRACT

The intracellular signaling mechanisms that specify tissue-specific responses to the interleukin-6 (IL-6) family of cytokines are not well understood. Here, we evaluated the functions of the two major signaling pathways, the signal transducers and activators of transcription 1 and 3 (STAT1/3) and the Src-homology tyrosine phosphatase 2 (SHP2)-Ras-ERK, emanating from the common signal transducer, gp130, in the gastrointestinal tract. Gp130(757F) mice, with a 'knock-in' mutation abrogating SHP2-Ras-ERK signaling, developed gastric adenomas by three months of age. In contrast, mice harboring the reciprocal mutation ablating STAT1/3 signaling (gp130(Delta STAT)), or deficient in IL-6-mediated gp130 signaling (IL-6(-/-) mice), showed impaired colonic mucosal wound healing. These gastrointestinal phenotypes are highly similar to the phenotypes exhibited by mice deficient in trefoil factor 1 (pS2/TFF1) and intestinal trefoil factor (ITF)/TFF3, respectively, and corresponded closely with the capacity of the two pathways to stimulate transcription of the genes encoding pS2/TFF1 and ITF/TFF3. We propose a model whereby mucosal wound healing depends solely on activation of STAT1/3, whereas gastric hyperplasia ensues when the coordinated activation of the STAT1/3 and SHP2-Ras-ERK pathways is disrupted.


Subject(s)
Antigens, CD/metabolism , DNA-Binding Proteins/metabolism , Digestive System Physiological Phenomena , Growth Substances/genetics , Homeostasis , Membrane Glycoproteins/metabolism , Mucins , Muscle Proteins , Neuropeptides , Peptides/genetics , Protein Tyrosine Phosphatases/metabolism , Trans-Activators/metabolism , Adenoma/metabolism , Adenoma/pathology , Animals , Antigens, CD/genetics , Cell Line , Colon/metabolism , Colon/pathology , Cytokine Receptor gp130 , Gastric Mucosa/metabolism , Gene Expression Regulation , Growth Substances/metabolism , Interleukin-11/metabolism , Interleukin-6/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intracellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinases/metabolism , Peptides/metabolism , Protein Phosphatase 2 , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Proteins/genetics , Proteins/metabolism , STAT1 Transcription Factor , Signal Transduction/physiology , Spleen/cytology , Spleen/metabolism , Stomach/cytology , Stomach/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transcriptional Activation , Trefoil Factor-2 , Trefoil Factor-3 , Wound Healing
16.
Nat Cell Biol ; 23(11): 1136-1147, 2021 11.
Article in English | MEDLINE | ID: mdl-34750583

ABSTRACT

The development of a functional vasculature requires the coordinated control of cell fate, lineage differentiation and network growth. Cellular proliferation is spatiotemporally regulated in developing vessels, but how this is orchestrated in different lineages is unknown. Here, using a zebrafish genetic screen for lymphatic-deficient mutants, we uncover a mutant for the RNA helicase Ddx21. Ddx21 cell-autonomously regulates lymphatic vessel development. An established regulator of ribosomal RNA synthesis and ribosome biogenesis, Ddx21 is enriched in sprouting venous endothelial cells in response to Vegfc-Flt4 signalling. Ddx21 function is essential for Vegfc-Flt4-driven endothelial cell proliferation. In the absence of Ddx21, endothelial cells show reduced ribosome biogenesis, p53 and p21 upregulation and cell cycle arrest that blocks lymphangiogenesis. Thus, Ddx21 coordinates the lymphatic endothelial cell response to Vegfc-Flt4 signalling by balancing ribosome biogenesis and p53 function. This mechanism may be targetable in diseases of excessive lymphangiogenesis such as cancer metastasis or lymphatic malformation.


Subject(s)
Cell Proliferation , DEAD-box RNA Helicases/metabolism , Endothelial Cells/enzymology , Lymphangiogenesis , Lymphatic Vessels/enzymology , RNA, Ribosomal/biosynthesis , Ribosomes/metabolism , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor C/metabolism , Zebrafish Proteins/metabolism , Animals , Animals, Genetically Modified , Cell Cycle Checkpoints , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DEAD-box RNA Helicases/genetics , Gene Expression Regulation, Developmental , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Lymphatic Vessels/embryology , RNA, Ribosomal/genetics , Ribosomes/genetics , Signal Transduction , Tumor Suppressor Protein p53/genetics , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
17.
J Am Chem Soc ; 132(45): 15881-3, 2010 Nov 17.
Article in English | MEDLINE | ID: mdl-20977221

ABSTRACT

Targeted delivery of drugs to specific cells allows a high therapeutic dose to be delivered to the target site with minimal harmful side effects. Combining targeting molecules with nanoengineered drug carriers, such as polymer capsules, micelles and polymersomes, has significant potential to improve the therapeutic delivery and index of a range of drugs. We present a general approach for functionalization of low-fouling, nanoengineered polymer capsules with antibodies using click chemistry. We demonstrate that antibody (Ab)-functionalized capsules specifically bind to colorectal cancer cells even when the target cells constitute less than 0.1% of the total cell population. This precise targeting offers promise for drug delivery applications.


Subject(s)
Antibodies/chemistry , Click Chemistry , Nanocapsules/chemistry , Neoplasms/drug therapy , Polymers/chemistry , Antibodies/administration & dosage , Azides/chemistry , Cell Line, Tumor , Flow Cytometry , Humans , Microscopy, Fluorescence , Nanocapsules/administration & dosage , Neoplasms/immunology , Polymers/administration & dosage , Polymers/chemical synthesis
18.
Gastroenterology ; 137(3): 902-13, 913.e1-11, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19454286

ABSTRACT

BACKGROUND & AIMS: Aberrant DNA methylation is a common early event in neoplasia, but it is unclear how this relates to dysregulation of DNA (cytosine-5) methyltransferases (Dnmts). Here we use knock-in transgenic mice to investigate the consequences of intestinal epithelium-specific overexpression of de novo Dnmt3a. METHODS: A novel gene targeting strategy, based on the intestinal epithelium-specific, uniform expression of the A33 glycoprotein, is employed to restrict Dnmt3a overexpression in homozygous A33(Dnmt3a) mutant mice. RESULTS: A33(Dnmt3a) mice infrequently develop spontaneous intestinal polyps. However, when genetically challenged, tumor multiplicity in A33(Dnmt3a);Apc(Min) compound mice is 3-fold higher than in Apc(Min) mice. Although we observe a requirement for spontaneous loss of heterozygosity of the adenomatous polyposis coli (Apc) gene to trigger tumorigenesis in Apc(Min) mice, lesions in A33(Dnmt3a);Apc(Min) mice frequently retain the wild-type Apc allele. However, epithelia from normal mucosa and polyps of A33(Dnmt3a);Apc(Min) mice show hypermethylation-mediated transcriptional silencing of the Wnt antagonists Sfrp5, and to a lesser extent, Sfrp1 and increased nuclear beta-catenin alongside activation of the Wnt-target gene Axin2/Conductin. Conversely, enforced Sfrp5 expression suppresses canonical Wnt-signaling more effectively in wild-type than in Apc(Min) cells. CONCLUSIONS: Aberrant activation of the canonical Wnt pathway, either by mono-allelic Apc loss or transcriptional silencing of Sfrp5 is largely insufficient to promote polyposis, but epistatic interactions between these genetic and epigenetic events enables initiation and promotion of disease. This mechanism is likely to play a role in human colorectal cancer, because we also show that elevated DNMT3A expression coincides with repressed SFRP5 and enhanced AXIN2/CONDUCTIN expression in paired patient biopsies.


Subject(s)
Adenomatous Polyposis Coli/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , Genes, APC , Intestinal Mucosa/metabolism , Signal Transduction , Wnt Proteins/metabolism , Adenomatous Polyposis Coli/genetics , Animals , Cells, Cultured , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Gene Expression Regulation, Neoplastic , Gene Knock-In Techniques , Gene Silencing , Loss of Heterozygosity , Membrane Glycoproteins/genetics , Mice , Mice, Mutant Strains , Mice, Transgenic
19.
Gastroenterology ; 136(3): 902-11, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19073184

ABSTRACT

BACKGROUND & AIMS: Zebrafish mutants generated by ethylnitrosourea-mutagenesis provide a powerful tool for dissecting the genetic regulation of developmental processes, including organogenesis. One zebrafish mutant, "flotte lotte" (flo), displays striking defects in intestinal, liver, pancreas, and eye formation at 78 hours postfertilization (hpf). In this study, we sought to identify the underlying mutated gene in flo and link the genetic lesion to its phenotype. METHODS: Positional cloning was employed to map the flo mutation. Subcellular characterization of flo embryos was achieved using histology, immunocytochemistry, bromodeoxyuridine incorporation analysis, and confocal and electron microscopy. RESULTS: The molecular lesion in flo is a nonsense mutation in the elys (embryonic large molecule derived from yolk sac) gene, which encodes a severely truncated protein lacking the Elys C-terminal AT-hook DNA binding domain. Recently, the human ELYS protein has been shown to play a critical, and hitherto unsuspected, role in nuclear pore assembly. Although elys messenger RNA (mRNA) is expressed broadly during early zebrafish development, widespread early defects in flo are circumvented by the persistence of maternally expressed elys mRNA until 24 hpf. From 72 hpf, elys mRNA expression is restricted to proliferating tissues, including the intestinal epithelium, pancreas, liver, and eye. Cells in these tissues display disrupted nuclear pore formation; ultimately, intestinal epithelial cells undergo apoptosis. CONCLUSIONS: Our results demonstrate that Elys regulates digestive organ formation.


Subject(s)
Apoptosis/physiology , Intestinal Mucosa/abnormalities , Intestinal Mucosa/physiology , Nuclear Pore Complex Proteins/genetics , Nuclear Pore/pathology , Zebrafish Proteins/genetics , Animals , Cell Differentiation/physiology , Cell Lineage/physiology , Enteric Nervous System/abnormalities , Enteric Nervous System/pathology , Enteric Nervous System/physiology , Eye Abnormalities/pathology , Eye Abnormalities/physiopathology , Gene Expression Regulation, Developmental , Intestinal Mucosa/pathology , Intestines/abnormalities , Intestines/pathology , Intestines/physiology , Liver/abnormalities , Liver/pathology , Liver/physiology , Microscopy, Electron , Nuclear Pore/physiology , Nuclear Pore/ultrastructure , Nuclear Pore Complex Proteins/metabolism , Pancreas/abnormalities , Pancreas/pathology , Pancreas/physiology , Phenotype , Zebrafish , Zebrafish Proteins/metabolism
20.
Dev Biol ; 314(1): 12-22, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18154948

ABSTRACT

The homeobox transcription factor Mtx2 is essential for epiboly, the first morphogenetic movement of gastrulation in zebrafish. Morpholino knockdown of Mtx2 results in stalling of epiboly and lysis due to yolk rupture. However, the mechanism of Mtx2 action is unknown. The role of mtx2 is surprising as most mix/bix family genes are thought to have roles in mesendoderm specification. Using a transgenic sox17-promoter driven EGFP line, we show that Mtx2 is not required for endoderm specification but is required for correct morphogenetic movements of endoderm and axial mesoderm. During normal zebrafish development, mtx2 is expressed at both the blastoderm margin and in the zebrafish equivalent of visceral endoderm, the extra-embryonic yolk syncytial layer (YSL). We show that formation of the YSL is not Mtx2 dependent, but that Mtx2 directs spatial arrangement of YSL nuclei. Furthermore, we demonstrate that Mtx2 knockdown results in loss of the YSL F-actin ring, a microfilament structure previously shown to be necessary for epiboly progression. In summary, we propose that Mtx2 acts within the YSL to regulate morphogenetic movements of both embryonic and extra-embryonic tissues, independently of cell fate specification.


Subject(s)
Actin Cytoskeleton/physiology , Actins/physiology , Membrane Proteins/physiology , Zebrafish Proteins/physiology , Zebrafish/physiology , Animals , Cell Movement/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Embryo, Nonmammalian , Gastrulation/physiology , High Mobility Group Proteins/genetics , High Mobility Group Proteins/physiology , Promoter Regions, Genetic , SOXF Transcription Factors , Transcription Factors/genetics , Transcription Factors/physiology , Zebrafish/embryology , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL