Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Mol Cancer Ther ; 20(10): 1757-1768, 2021 10.
Article in English | MEDLINE | ID: mdl-34330842

ABSTRACT

The MAPK signaling pathway is commonly upregulated in human cancers. As the primary downstream effector of the MAPK pathway, ERK is an attractive therapeutic target for the treatment of MAPK-activated cancers and for overcoming resistance to upstream inhibition. ASTX029 is a highly potent and selective dual-mechanism ERK inhibitor, discovered using fragment-based drug design. Because of its distinctive ERK-binding mode, ASTX029 inhibits both ERK catalytic activity and the phosphorylation of ERK itself by MEK, despite not directly inhibiting MEK activity. This dual mechanism was demonstrated in cell-free systems, as well as cell lines and xenograft tumor tissue, where the phosphorylation of both ERK and its substrate, ribosomal S6 kinase (RSK), were modulated on treatment with ASTX029. Markers of sensitivity were highlighted in a large cell panel, where ASTX029 preferentially inhibited the proliferation of MAPK-activated cell lines, including those with BRAF or RAS mutations. In vivo, significant antitumor activity was observed in MAPK-activated tumor xenograft models following oral treatment. ASTX029 also demonstrated activity in both in vitro and in vivo models of acquired resistance to MAPK pathway inhibitors. Overall, these findings highlight the therapeutic potential of a dual-mechanism ERK inhibitor such as ASTX029 for the treatment of MAPK-activated cancers, including those which have acquired resistance to inhibitors of upstream components of the MAPK pathway. ASTX029 is currently being evaluated in a first in human phase I-II clinical trial in patients with advanced solid tumors (NCT03520075).


Subject(s)
Colonic Neoplasms/drug therapy , Drug Resistance, Neoplasm , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Indoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Animals , Apoptosis , Cell Cycle , Cell Movement , Cell Proliferation , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
BMC Dev Biol ; 20(1): 4, 2020 02 26.
Article in English | MEDLINE | ID: mdl-32098630

ABSTRACT

BACKGROUND: Pancreatic organoid systems have recently been described for the in vitro culture of pancreatic ductal cells from mouse and human. Mouse pancreatic organoids exhibit unlimited expansion potential, while previously reported human pancreas organoid (hPO) cultures do not expand efficiently long-term in a chemically defined, serum-free medium. We sought to generate a 3D culture system for long-term expansion of human pancreas ductal cells as hPOs to serve as the basis for studies of human pancreas ductal epithelium, exocrine pancreatic diseases and the development of a genomically stable replacement cell therapy for diabetes mellitus. RESULTS: Our chemically defined, serum-free, human pancreas organoid culture medium supports the generation and expansion of hPOs with high efficiency from both fresh and cryopreserved primary tissue. hPOs can be expanded from a single cell, enabling their genetic manipulation and generation of clonal cultures. hPOs expanded for months in vitro maintain their ductal morphology, biomarker expression and chromosomal integrity. Xenografts of hPOs survive long-term in vivo when transplanted into the pancreas of immunodeficient mice. Notably, mouse orthotopic transplants show no signs of tumorigenicity. Crucially, our medium also supports the establishment and expansion of hPOs in a chemically defined, modifiable and scalable, biomimetic hydrogel. CONCLUSIONS: hPOs can be expanded long-term, from both fresh and cryopreserved human pancreas tissue in a chemically defined, serum-free medium with no detectable tumorigenicity. hPOs can be clonally expanded, genetically manipulated and are amenable to culture in a chemically defined hydrogel. hPOs therefore represent an abundant source of pancreas ductal cells that retain the characteristics of the tissue-of-origin, which opens up avenues for modelling diseases of the ductal epithelium and increasing understanding of human pancreas exocrine biology as well as for potentially producing insulin-secreting cells for the treatment of diabetes.


Subject(s)
Organoids/cytology , Pancreas/cytology , Cell Differentiation/physiology , Cells, Cultured , Female , Flow Cytometry , Genomic Instability/physiology , Humans , In Vitro Techniques , Lentivirus/genetics , Male , Organ Culture Techniques , Organoids/metabolism , Pancreas/metabolism , Reverse Transcriptase Polymerase Chain Reaction
3.
Development ; 146(12)2019 06 12.
Article in English | MEDLINE | ID: mdl-31142540

ABSTRACT

During mouse embryogenesis, progenitors within the liver known as hepatoblasts give rise to adult hepatocytes and cholangiocytes. Hepatoblasts, which are specified at E8.5-E9.0, have been regarded as a homogeneous progenitor population that initiate differentiation from E13.5. Recently, scRNA-seq analysis has identified sub-populations of transcriptionally distinct hepatoblasts at E11.5. Here, we show that hepatoblasts are not only transcriptionally but also functionally heterogeneous, and that a subpopulation of E9.5-E10.0 hepatoblasts exhibit a previously unidentified early commitment to cholangiocyte fate. Importantly, we also identify a subpopulation constituting 2% of E9.5-E10.0 hepatoblasts that express the adult stem cell marker Lgr5, and generate both hepatocyte and cholangiocyte progeny that persist for the lifespan of the mouse. Combining lineage tracing and scRNA-seq, we show that Lgr5 marks E9.5-E10.0 bipotent liver progenitors residing at the apex of a hepatoblast hierarchy. Furthermore, isolated Lgr5+ hepatoblasts can be clonally expanded in vitro into embryonic liver organoids, which can commit to either hepatocyte or cholangiocyte fates. Our study demonstrates functional heterogeneity within E9.5 hepatoblasts and identifies Lgr5 as a marker for a subpopulation of bipotent liver progenitors.


Subject(s)
Gene Expression Regulation, Developmental , Hepatocytes/cytology , Liver/embryology , Receptors, G-Protein-Coupled/metabolism , Alleles , Animals , Base Sequence , Cell Count , Cell Culture Techniques , Cell Differentiation , Cell Lineage , Cells, Cultured , Embryonic Development , Epithelial Cells/cytology , Female , Hepatocytes/metabolism , Homeostasis , Male , Mice , Microscopy, Confocal , Stem Cells/cytology
4.
Dev Cell ; 46(3): 360-375.e5, 2018 08 06.
Article in English | MEDLINE | ID: mdl-30057275

ABSTRACT

Pancreas development involves a coordinated process in which an early phase of cell segregation is followed by a longer phase of lineage restriction, expansion, and tissue remodeling. By combining clonal tracing and whole-mount reconstruction with proliferation kinetics and single-cell transcriptional profiling, we define the functional basis of pancreas morphogenesis. We show that the large-scale organization of mouse pancreas can be traced to the activity of self-renewing precursors positioned at the termini of growing ducts, which act collectively to drive serial rounds of stochastic ductal bifurcation balanced by termination. During this phase of branching morphogenesis, multipotent precursors become progressively fate-restricted, giving rise to self-renewing acinar-committed precursors that are conveyed with growing ducts, as well as ductal progenitors that expand the trailing ducts and give rise to delaminating endocrine cells. These findings define quantitatively how the functional behavior and lineage progression of precursor pools determine the large-scale patterning of pancreatic sub-compartments.


Subject(s)
Cell Lineage , Endocrine Cells/metabolism , Gene Expression Regulation, Developmental/genetics , Organogenesis/physiology , Pancreas/growth & development , Acinar Cells/metabolism , Animals , Cell Differentiation/physiology , Cell Lineage/physiology , Cell Proliferation/physiology , Morphogenesis/physiology , Stem Cells/metabolism
5.
Nat Phys ; 14(5): 469-474, 2018 May.
Article in English | MEDLINE | ID: mdl-29736183

ABSTRACT

The emergence of complex organs is driven by the coordinated proliferation, migration and differentiation of precursor cells. The fate behaviour of these cells is reflected in the time evolution their progeny, termed clones, which serve as a key experimental observable. In adult tissues, where cell dynamics is constrained by the condition of homeostasis, clonal tracing studies based on transgenic animal models have advanced our understanding of cell fate behaviour and its dysregulation in disease (1, 2). But what can be learned from clonal dynamics in development, where the spatial cohesiveness of clones is impaired by tissue deformations during tissue growth? Drawing on the results of clonal tracing studies, we show that, despite the complexity of organ development, clonal dynamics may converge to a critical state characterized by universal scaling behaviour of clone sizes. By mapping clonal dynamics onto a generalization of the classical theory of aerosols, we elucidate the origin and range of scaling behaviours and show how the identification of universal scaling dependences may allow lineage-specific information to be distilled from experiments. Our study shows the emergence of core concepts of statistical physics in an unexpected context, identifying cellular systems as a laboratory to study non-equilibrium statistical physics.

6.
Nat Protoc ; 11(9): 1724-43, 2016 09.
Article in English | MEDLINE | ID: mdl-27560176

ABSTRACT

Adult somatic tissues have proven difficult to expand in vitro, largely because of the complexity of recreating appropriate environmental signals in culture. We have overcome this problem recently and developed culture conditions for adult stem cells that allow the long-term expansion of adult primary tissues from small intestine, stomach, liver and pancreas into self-assembling 3D structures that we have termed 'organoids'. We provide a detailed protocol that describes how to grow adult mouse and human liver and pancreas organoids, from cell isolation and long-term expansion to genetic manipulation in vitro. Liver and pancreas cells grow in a gel-based extracellular matrix (ECM) and a defined medium. The cells can self-organize into organoids that self-renew in vitro while retaining their tissue-of-origin commitment, genetic stability and potential to differentiate into functional cells in vitro (hepatocytes) and in vivo (hepatocytes and endocrine cells). Genetic modification of these organoids opens up avenues for the manipulation of adult stem cells in vitro, which could facilitate the study of human biology and allow gene correction for regenerative medicine purposes. The complete protocol takes 1-4 weeks to generate self-renewing 3D organoids and to perform genetic manipulation experiments. Personnel with basic scientific training can conduct this protocol.


Subject(s)
Adult Stem Cells/cytology , Cell Culture Techniques/methods , Cell Self Renewal , Genetic Engineering/methods , Liver/cytology , Organoids/cytology , Pancreas/cytology , Adult Stem Cells/metabolism , Animals , Female , Humans , Male , Mice , Organoids/metabolism
7.
Dev Biol ; 420(2): 251-261, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27364469

ABSTRACT

The liver and pancreas are critical organs maintaining whole body metabolism. Historically, the expansion of adult-derived cells from these organs in vitro has proven challenging and this in turn has hampered studies of liver and pancreas stem cell biology, as well as being a roadblock to disease modelling and cell replacement therapies for pathologies in these organs. Recently, defined culture conditions have been described which allow the in vitro culture and manipulation of adult-derived liver and pancreatic material. Here we review these systems and assess their physiological relevance, as well as their potential utility in biomedicine.


Subject(s)
Liver/growth & development , Organoids/growth & development , Pancreas/growth & development , Animals , Cell- and Tissue-Based Therapy , Genetic Therapy , Humans , Liver/cytology , Liver/embryology , Liver Regeneration , Models, Biological , Organ Culture Techniques/methods , Organogenesis , Organoids/cytology , Organoids/embryology , Pancreas/cytology , Pancreas/embryology , Signal Transduction , Stem Cells/cytology
8.
Sci Rep ; 6: 23208, 2016 Mar 16.
Article in English | MEDLINE | ID: mdl-26980066

ABSTRACT

The Hippo/YAP pathway serves as a major integrator of cell surface-mediated signals and regulates key processes during development and tumorigenesis. The neural crest is an embryonic tissue known to respond to multiple environmental cues in order to acquire appropriate cell fate and migration properties. Using multiple in vitro models of human neural development (pluripotent stem cell-derived neural stem cells; LUHMES, NTERA2 and SH-SY5Y cell lines), we investigated the role of Hippo/YAP signaling in neural differentiation and neural crest development. We report that the activity of YAP promotes an early neural crest phenotype and migration, and provide the first evidence for an interaction between Hippo/YAP and retinoic acid signaling in this system.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Neural Crest/cytology , Neural Stem Cells/physiology , Neurogenesis , Phosphoproteins/physiology , Antigens, CD/metabolism , Cell Line, Tumor , Cell Movement , Cell Nucleus/metabolism , Humans , Signal Transduction , Transcription Factors , Tretinoin/pharmacology , YAP-Signaling Proteins
9.
Open Biol ; 5(12): 150209, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26701933

ABSTRACT

To address the long-known relationship between supernumerary centrosomes and cancer, we have generated a transgenic mouse that permits inducible expression of the master regulator of centriole duplication, Polo-like-kinase-4 (Plk4). Over-expression of Plk4 from this transgene advances the onset of tumour formation that occurs in the absence of the tumour suppressor p53. Plk4 over-expression also leads to hyperproliferation of cells in the pancreas and skin that is enhanced in a p53 null background. Pancreatic islets become enlarged following Plk4 over-expression as a result of equal expansion of α- and ß-cells, which exhibit centrosome amplification. Mice overexpressing Plk4 develop grey hair due to a loss of differentiated melanocytes and bald patches of skin associated with a thickening of the epidermis. This reflects an increase in proliferating cells expressing keratin 5 in the basal epidermal layer and the expansion of these cells into suprabasal layers. Such cells also express keratin 6, a marker for hyperplasia. This is paralleled by a decreased expression of later differentiation markers, involucrin, filaggrin and loricrin. Proliferating cells showed an increase in centrosome number and a loss of primary cilia, events that were mirrored in primary cultures of keratinocytes established from these animals. We discuss how repeated duplication of centrioles appears to prevent the formation of basal bodies leading to loss of primary cilia, disruption of signalling and thereby aberrant differentiation of cells within the epidermis. The absence of p53 permits cells with increased centrosomes to continue dividing, thus setting up a neoplastic state of error prone mitoses, a prerequisite for cancer development.


Subject(s)
Centrosome/metabolism , Cilia/metabolism , Hyperplasia/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Proliferation , Cells, Cultured , Centrioles/metabolism , Filaggrin Proteins , Intermediate Filament Proteins/metabolism , Islets of Langerhans/metabolism , Membrane Proteins/metabolism , Mice , Protein Precursors/metabolism , Protein Serine-Threonine Kinases/genetics
10.
J Clin Invest ; 124(12): 5099-102, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25401467

ABSTRACT

The liver is capable of full regeneration following several types and rounds of injury, ranging from hepatectomy to toxin-mediated damage. The source of this regenerative capacity has long been a hotly debated topic. The damage response that occurs when hepatocyte proliferation is impaired is thought to be mediated by oval/dedifferentiated progenitor cells, which replenish the hepatocyte and ductal compartments of the liver. Recently, reports have questioned whether these oval/progenitor cells truly serve as the facultative stem cell of the liver following toxin-mediated damage. In this issue of the JCI, Kordes and colleagues use lineage tracing to follow transplanted rat hepatic stellate cells, a resident liver mesenchymal cell population, in hosts that have suffered liver damage. Transplanted stellate cells repopulated the damaged rat liver by contributing to the oval cell response. These data establish yet another cell type of mesenchymal origin as the progenitor for the oval/ductular response in the rat. The lack of uniformity between different damage models, the extent of the injury to the liver parenchyma, and potential species-specific differences might be at the core of the discrepancy between different studies. Taken together, these data imply a considerable degree of plasticity in the liver, whereby several cell types can contribute to regeneration.


Subject(s)
Cell Differentiation , Hepatic Stellate Cells , Liver Regeneration , Liver/metabolism , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Animals
11.
BMC Biochem ; 15: 24, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25374254

ABSTRACT

BACKGROUND: Neuronal differentiation is largely under the control of basic Helix-Loop-Helix (bHLH) proneural transcription factors that play key roles during development of the embryonic nervous system. In addition to well-characterised regulation of their expression, increasing evidence is emerging for additional post-translational regulation of proneural protein activity. Of particular interest is the bHLH proneural factor Neurogenin2 (Ngn2), which orchestrates progression from neural progenitor to differentiated neuron in several regions of the central nervous system. Previous studies have demonstrated a key role for cell cycle-dependent multi-site phosphorylation of Ngn2 protein at Serine-Proline (SP) sites for regulation of its neuronal differentiation activity, although the potential structural and functional consequences of phosphorylation at different regions of the protein are unclear. RESULTS: Here we characterise the role of phosphorylation of specific regions of Ngn2 on the stability of Ngn2 protein and on its neuronal differentiation activity in vivo in the developing embryo, demonstrating clearly that the location of SP sites is less important than the number of SP sites available for control of Ngn2 activity in vivo. We also provide structural evidence that Ngn2 contains large, intrinsically disordered regions that undergo phosphorylation by cyclin-dependent kinases (cdks). CONCLUSIONS: Phosphorylation of Ngn2 occurs in both the N- and C-terminal regions, either side of the conserved basic Helix-Loop-Helix domain. While these phosphorylation events do not change the intrinsic stability of Ngn2, phosphorylation on multiple sites acts to limit its ability to drive neuronal differentiation in vivo. Phosphorylated regions of Ngn2 are predicted to be intrinsically disordered and cdk-dependent phosphorylation of these intrinsically disordered regions contributes to Ngn2 regulation.


Subject(s)
Nerve Tissue Proteins/metabolism , Protein Processing, Post-Translational , Xenopus Proteins/metabolism , Animals , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/metabolism , Nerve Tissue Proteins/chemistry , Neurogenesis , Phosphorylation , Protein Structure, Tertiary , Xenopus Proteins/chemistry , Xenopus laevis
12.
Nat Commun ; 5: 4028, 2014 Jun 06.
Article in English | MEDLINE | ID: mdl-24905578

ABSTRACT

Mutations in the acid ß-glucocerebrosidase (GBA1) gene, responsible for the lysosomal storage disorder Gaucher's disease (GD), are the strongest genetic risk factor for Parkinson's disease (PD) known to date. Here we generate induced pluripotent stem cells from subjects with GD and PD harbouring GBA1 mutations, and differentiate them into midbrain dopaminergic neurons followed by enrichment using fluorescence-activated cell sorting. Neurons show a reduction in glucocerebrosidase activity and protein levels, increase in glucosylceramide and α-synuclein levels as well as autophagic and lysosomal defects. Quantitative proteomic profiling reveals an increase of the neuronal calcium-binding protein 2 (NECAB2) in diseased neurons. Mutant neurons show a dysregulation of calcium homeostasis and increased vulnerability to stress responses involving elevation of cytosolic calcium. Importantly, correction of the mutations rescues such pathological phenotypes. These findings provide evidence for a link between GBA1 mutations and complex changes in the autophagic/lysosomal system and intracellular calcium homeostasis, which underlie vulnerability to neurodegeneration.


Subject(s)
Autophagy , Calcium/metabolism , Glucosylceramidase/genetics , Homeostasis , Induced Pluripotent Stem Cells/pathology , Neurons/pathology , Parkinson Disease/pathology , Cell Differentiation , Glycoside Hydrolases/metabolism , Humans , Induced Pluripotent Stem Cells/enzymology , Neurons/enzymology , Parkinson Disease/immunology , Parkinson Disease/metabolism
13.
PLoS One ; 8(6): e68519, 2013.
Article in English | MEDLINE | ID: mdl-23826393

ABSTRACT

Surface molecule profiles undergo dynamic changes in physiology and pathology, serve as markers of cellular state and phenotype and can be exploited for cell selection strategies and diagnostics. The isolation of well-defined cell subsets is needed for in vivo and in vitro applications in stem cell biology. In this technical report, we present an approach for defining a subset of interest in a mixed cell population by flow cytometric detection of intracellular antigens. We have developed a fully validated protocol that enables the co-detection of cluster of differentiation (CD) surface antigens on fixed, permeabilized neural cell populations defined by intracellular staining. Determining the degree of co-expression of surface marker candidates with intracellular target population markers (nestin, MAP2, doublecortin, TUJ1) on neuroblastoma cell lines (SH-SY5Y, BE(2)-M17) yielded a combinatorial CD49f(-)/CD200(high) surface marker panel. Its application in fluorescence-activated cell sorting (FACS) generated enriched neuronal cultures from differentiated cell suspensions derived from human induced pluripotent stem cells. Our data underlines the feasibility of using the described co-labeling protocol and co-expression analysis for quantitative assays in mammalian neurobiology and for screening approaches to identify much needed surface markers in stem cell biology.


Subject(s)
Antigens, CD/metabolism , Cell Differentiation , Cell Membrane/metabolism , Flow Cytometry/methods , Intracellular Space/metabolism , Neurons/cytology , Neurons/metabolism , Biomarkers/metabolism , Cell Line , Cell Separation , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Reproducibility of Results
14.
Int J Dev Biol ; 55(3): 249-60, 2011.
Article in English | MEDLINE | ID: mdl-21710433

ABSTRACT

The developing embryo is patterned by a complex set of signals and interactions resulting in changes in cell division, cell fate determination and differentiation. An increasing body of evidence points to the role of the ubiquitin proteasome system (UPS) and ubiquitin-mediated protein degradation as a major mechanism of protein regulation, crucial for control of developmental processes. The specific and irreversible signal generated by protein degradation can function as an integrator of cell signaling events, coupled with other post-translational protein modifications, but also as a master switch for differentiation in its own right. The UPS also displays more subtle mechanisms of regulating signaling than decreasing protein levels, such as proteolytic processing and altering subcellular localization. In particular, the SCF E3 ligase family plays pivotal roles in regulating diverse developmental events in varied species. This review will focus on the role played by SCF E3 ligases in cell fate determination and differentiation.


Subject(s)
Cell Lineage , SKP Cullin F-Box Protein Ligases/metabolism , Animals , Cell Differentiation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Humans , Signal Transduction
15.
Neural Dev ; 5: 1, 2010 Jan 04.
Article in English | MEDLINE | ID: mdl-20047651

ABSTRACT

BACKGROUND: The neural crest is a unique population of cells that arise in the vertebrate ectoderm at the neural plate border after which they migrate extensively throughout the embryo, giving rise to a wide range of derivatives. A number of proteins involved in neural crest development have dynamic expression patterns, and it is becoming clear that ubiquitin-mediated protein degradation is partly responsible for this. RESULTS: Here we demonstrate a novel role for the F-box protein Cdc4/Fbxw7 in neural crest development. Two isoforms of Xenopus laevis Cdc4 were identified, and designated xCdc4alpha and xCdc4beta. These are highly conserved with vertebrate Cdc4 orthologs, and the Xenopus proteins are functionally equivalent in terms of their ability to degrade Cyclin E, an established vertebrate Cdc4 target. Blocking xCdc4 function specifically inhibited neural crest development at an early stage, prior to expression of c-Myc, Snail2 and Snail. CONCLUSIONS: We demonstrate that Cdc4, an ubiquitin E3 ligase subunit previously identified as targeting primarily cell cycle regulators for proteolysis, has additional roles in control of formation of the neural crest. Hence, we identify Cdc4 as a protein with separable but complementary functions in control of cell proliferation and differentiation.


Subject(s)
F-Box Proteins/metabolism , Neural Crest/embryology , Neural Crest/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Animals , Blotting, Western , Cell Differentiation/genetics , Cell Proliferation , Cyclin E/metabolism , DNA, Complementary , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Gene Expression Regulation, Developmental , In Situ Hybridization , Microinjections , Plasmids/genetics , Point Mutation , Polymerase Chain Reaction , Protein Isoforms/metabolism , RNA, Messenger , Sequence Deletion , Ubiquitin/genetics , Xenopus Proteins/genetics , Xenopus laevis/genetics , Xenopus laevis/metabolism
16.
J Biol Chem ; 284(23): 15458-68, 2009 Jun 05.
Article in English | MEDLINE | ID: mdl-19336407

ABSTRACT

Polyubiquitylation targets multiple proteins for degradation by the proteasome. Typically, the first ubiquitin is linked to lysine residues in the substrate for degradation via an isopeptide bond, although rarely ubiquitin linkage to the N-terminal residue has also been observed. We have recently shown that Neurogenin (NGN), a basic helix-loop-helix transcription factor that plays a central role in regulating neuronal differentiation, is degraded by ubiquitin-mediated proteolysis. We have taken a biochemical and mutagenesis approach to investigate sites of ubiquitylation of NGN, initially using extracts of eggs from the frog Xenopus laevis as a source of ubiquitylation and degradation components. NGN can be targeted for destruction by ubiquitylation via lysines or the N terminus. However, we see that a modified NGN, where canonical lysine ubiquitylation and N-terminally linked ubiquitylation are prevented, is nevertheless ubiquitylated and degraded by the proteasome. We show that polyubiquitin chains covalently attach to non-canonical cysteine residues in NGN, and these non-canonical linkages alone are capable of targeting NGN protein for destruction. Importantly, canonical and non-canonical ubiquitylation occurs simultaneously in the native protein and may differ in importance for driving degradation in interphase and mitosis. We conclude that native NGN is ubiquitylated on multiple canonical and non-canonical sites by cellular ubiquitin ligases, and all types of linkage can contribute to protein turnover.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Embryo, Nonmammalian/physiology , Nerve Tissue Proteins/metabolism , Ubiquitin/metabolism , Acetylation , Animals , Female , Lysine/metabolism , Methionine/metabolism , Mitosis/physiology , Ovum/cytology , Ovum/physiology , Substrate Specificity , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL
...