Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Br J Clin Pharmacol ; 89(2): 660-671, 2023 02.
Article in English | MEDLINE | ID: mdl-35998099

ABSTRACT

AIMS: High-dose methotrexate (HDMTX) is an essential part of the treatment of several adult and paediatric malignancies. Despite meticulous supportive care during HDMTX administration, severe toxicities, including acute kidney injury (AKI), may occur contributing to patient morbidity. Population pharmacokinetics provide a powerful tool to predict time to clear HDMTX and adjust subsequent doses. We sought to develop and validate pharmacokinetic models for HDMTX in adults with diverse malignancies and to relate systemic exposure with the occurrence of severe toxicity. METHODS: Anonymized, de-identified data were provided from 101 US oncology practices that participate in the Guardian Research Network, a non-profit clinical research consortium. Modelled variables included clinical, laboratory, demographic and pharmacological data. Population pharmacokinetic analysis was performed by means of nonlinear mixed effects modelling using MonolixSuite. RESULTS: A total of 693 HDMTX courses from 243 adults were analysed, of which 62 courses (8.8%) were associated with stage 2/3 acute kidney injury (43 stage 2, 19 stage 3). A three-compartment model adequately fitted the data. Time-dependent serum creatinine, baseline serum albumin and allometrically scaled bodyweight were clinically significant covariates related to methotrexate clearance. External evaluation confirmed a satisfactory predictive performance of the model in adults receiving HDMTX. Dose-normalized methotrexate concentration at 24 and 48 hours correlated with AKI incidence. CONCLUSION: We developed a population pharmacometric model that considers weight, albumin and time-dependent creatinine that can be used to guide supportive care in adult patients with delayed HDMTX elimination.


Subject(s)
Acute Kidney Injury , Neoplasms , Child , Humans , Adult , Methotrexate , Antimetabolites, Antineoplastic , Neoplasms/drug therapy , Acute Kidney Injury/chemically induced , Acute Kidney Injury/epidemiology , Seizures/drug therapy
2.
EMBO J ; 35(12): 1254-75, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27220849

ABSTRACT

Membrane-less organelles in cells are large, dynamic protein/protein or protein/RNA assemblies that have been reported in some cases to have liquid droplet properties. However, the molecular interactions underlying the recruitment of components are not well understood. Herein, we study how the ability to form higher-order assemblies influences the recruitment of the speckle-type POZ protein (SPOP) to nuclear speckles. SPOP, a cullin-3-RING ubiquitin ligase (CRL3) substrate adaptor, self-associates into higher-order oligomers; that is, the number of monomers in an oligomer is broadly distributed and can be large. While wild-type SPOP localizes to liquid nuclear speckles, self-association-deficient SPOP mutants have a diffuse distribution in the nucleus. SPOP oligomerizes through its BTB and BACK domains. We show that BTB-mediated SPOP dimers form linear oligomers via BACK domain dimerization, and we determine the concentration-dependent populations of the resulting oligomeric species. Higher-order oligomerization of SPOP stimulates CRL3(SPOP) ubiquitination efficiency for its physiological substrate Gli3, suggesting that nuclear speckles are hotspots of ubiquitination. Dynamic, higher-order protein self-association may be a general mechanism to concentrate functional components in membrane-less cellular bodies.


Subject(s)
Cell Nucleus/metabolism , Macromolecular Substances/metabolism , Nuclear Proteins/metabolism , Protein Multimerization , Repressor Proteins/metabolism , Humans , Kruppel-Like Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Protein Binding , Protein Domains , Ubiquitination , Zinc Finger Protein Gli3
3.
Development ; 144(19): 3612-3624, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28827391

ABSTRACT

During development, extracellular cues guiding cell fate determination are provided by morphogens. One mechanism by which morphogens are proposed to traverse extracellular space is by traveling along specialized filopodia called cytonemes. These cellular highways extend between signal-producing and -receiving cells to enable direct morphogen delivery. Although genetic studies support cytoneme involvement in morphogen transport, mechanistic insight into how they are regulated is limited owing to technical challenges associated with performing cell biological analysis of the delicate filopodial structures. Here, we introduce a fixation method whereby cultured cell cytonemes can be preserved for imaging studies, allowing investigation of cytoneme regulation using standard cell biological techniques. Using this method, we examined Hedgehog-containing cytonemes and identified a role for the Hedgehog deployment protein Dispatched in cytoneme stabilization. We demonstrate that Hedgehog and Dispatched colocalize in cytonemes, and that cholesterol-modified Hedgehog acts through Dispatched to increase cytoneme occurrence. Live imaging suggests that this occurs through Dispatched-mediated slowing of cytoneme retraction rates. Dispatched-induced cytoneme modulation was recapitulated in wing imaginal discs of transgenic Drosophila, providing evidence that cultured cell cytoneme analysis is predictive of in vivo functionality.


Subject(s)
Drosophila Proteins/metabolism , Pseudopodia/metabolism , Tissue Fixation/methods , Animals , Biological Transport , Cells, Cultured , Cholesterol/metabolism , Drosophila melanogaster/metabolism , Mice , NIH 3T3 Cells , Reproducibility of Results
4.
PLoS Genet ; 11(8): e1005473, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26291458

ABSTRACT

The G protein-coupled receptor (GPCR) Smoothened (Smo) is the requisite signal transducer of the evolutionarily conserved Hedgehog (Hh) pathway. Although aspects of Smo signaling are conserved from Drosophila to vertebrates, significant differences have evolved. These include changes in its active sub-cellular localization, and the ability of vertebrate Smo to induce distinct G protein-dependent and independent signals in response to ligand. Whereas the canonical Smo signal to Gli transcriptional effectors occurs in a G protein-independent manner, its non-canonical signal employs Gαi. Whether vertebrate Smo can selectively bias its signal between these routes is not yet known. N-linked glycosylation is a post-translational modification that can influence GPCR trafficking, ligand responsiveness and signal output. Smo proteins in Drosophila and vertebrate systems harbor N-linked glycans, but their role in Smo signaling has not been established. Herein, we present a comprehensive analysis of Drosophila and murine Smo glycosylation that supports a functional divergence in the contribution of N-linked glycans to signaling. Of the seven predicted glycan acceptor sites in Drosophila Smo, one is essential. Loss of N-glycosylation at this site disrupted Smo trafficking and attenuated its signaling capability. In stark contrast, we found that all four predicted N-glycosylation sites on murine Smo were dispensable for proper trafficking, agonist binding and canonical signal induction. However, the under-glycosylated protein was compromised in its ability to induce a non-canonical signal through Gαi, providing for the first time evidence that Smo can bias its signal and that a post-translational modification can impact this process. As such, we postulate a profound shift in N-glycan function from affecting Smo ER exit in flies to influencing its signal output in mice.


Subject(s)
Drosophila Proteins/metabolism , Protein Processing, Post-Translational , Receptors, G-Protein-Coupled/metabolism , Amino Acid Sequence , Animals , Conserved Sequence , Drosophila melanogaster , Glycosylation , HEK293 Cells , Humans , Mice , Molecular Sequence Data , NIH 3T3 Cells , Protein Binding , Protein Transport , Signal Transduction , Smoothened Receptor , Species Specificity
5.
Dev Biol ; 409(1): 84-94, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26481064

ABSTRACT

During development, the evolutionarily conserved Hedgehog (Hh) morphogen provides instructional cues that influence cell fate, cell affinity and tissue morphogenesis. To do so, the Hh signaling cascade must coordinate its activity with other morphogenetic signals. This can occur through engagement of or response to effectors that do not typically function as core Hh pathway components. Given the ability of small G proteins of the Ras family to impact cell survival, differentiation, growth and adhesion, we wanted to determine whether Hh and Ras signaling might intersect during development. We performed genetic modifier tests in Drosophila to examine the ability of select Ras family members to influence Hh signal output, and identified Rap1 as a positive modulator of Hh pathway activity. Our results suggest that Rap1 is activated to its GTP-bound form in response to Hh ligand, and that the GTPase exchange factor C3G likely contributes to this activation. The Rap1 effector Canoe (Cno) also impacts Hh signal output, suggesting that a C3G-Rap1-Cno axis intersects the Hh pathway during tissue morphogenesis.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Hedgehog Proteins/metabolism , Signal Transduction , Telomere-Binding Proteins/metabolism , Animals , Genes, Suppressor , Guanosine Triphosphate/metabolism , Shelterin Complex
6.
Development ; 139(3): 612-21, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22223683

ABSTRACT

The Hedgehog (Hh) signaling pathway plays an instructional role during development, and is frequently activated in cancer. Ligand-induced pathway activation requires signaling by the transmembrane protein Smoothened (Smo), a member of the G-protein-coupled receptor (GPCR) superfamily. The extracellular (EC) loops of canonical GPCRs harbor cysteine residues that engage in disulfide bonds, affecting active and inactive signaling states through regulating receptor conformation, dimerization and/or ligand binding. Although a functional importance for cysteines localized to the N-terminal extracellular cysteine-rich domain has been described, a functional role for a set of conserved cysteines in the EC loops of Smo has not yet been established. In this study, we mutated each of the conserved EC cysteines, and tested for effects on Hh signal transduction. Cysteine mutagenesis reveals that previously uncharacterized functional roles exist for Smo EC1 and EC2. We provide in vitro and in vivo evidence that EC1 cysteine mutation induces significant Hh-independent Smo signaling, triggering a level of pathway activation similar to that of a maximal Hh response in Drosophila and mammalian systems. Furthermore, we show that a single amino acid change in EC2 attenuates Hh-induced Smo signaling, whereas deletion of the central region of EC2 renders Smo fully active, suggesting that the conformation of EC2 is crucial for regulated Smo activity. Taken together, these findings are consistent with loop cysteines engaging in disulfide bonds that facilitate a Smo conformation that is silent in the absence of Hh, but can transition to a fully active state in response to ligand.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Hedgehog Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Amino Acid Sequence , Animals , Cysteine/chemistry , Cysteine/genetics , Cysteine/metabolism , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Male , Molecular Sequence Data , Mutagenesis , Protein Conformation , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Smoothened Receptor , Wings, Animal/metabolism
7.
Elife ; 72018 01 23.
Article in English | MEDLINE | ID: mdl-29359685

ABSTRACT

Hedgehog ligands activate an evolutionarily conserved signaling pathway that provides instructional cues during tissue morphogenesis, and when corrupted, contributes to developmental disorders and cancer. The transmembrane protein Dispatched is an essential component of the machinery that deploys Hedgehog family ligands from producing cells, and is absolutely required for signaling to long-range targets. Despite this crucial role, regulatory mechanisms controlling Dispatched activity remain largely undefined. Herein, we reveal vertebrate Dispatched is activated by proprotein convertase-mediated cleavage at a conserved processing site in its first extracellular loop. Dispatched processing occurs at the cell surface to instruct its membrane re-localization in polarized epithelial cells. Cleavage site mutation alters Dispatched membrane trafficking and reduces ligand release, leading to compromised pathway activity in vivo. As such, convertase-mediated cleavage is required for Dispatched maturation and functional competency in Hedgehog ligand-producing cells.


Subject(s)
Furin/metabolism , Hedgehog Proteins/metabolism , Membrane Proteins/metabolism , Proprotein Convertases/metabolism , Proteolysis , Animals , Cell Line , Mice
8.
Data Brief ; 7: 485-9, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27014736

ABSTRACT

This data article includes supporting information for the research article entitled "The Small GTPase Rap1 is a Modulator of Hedgehog Signaling" [1]. Drosophila wing phenotypes induced by expression of a dominant negative Smoothened (Smo) mutant were cataloged into five distinct classes. Class distributions observed following expression of dominant negative Smo in control and sensitized backgrounds were quantified to serve as references for strength of phenotypic modification. Shifts in class distribution of Hedgehog (Hh) wing phenotypes resulting from introduction of loss-of-function alleles of select Ras family G protein genes and the Hh pathway regulators Fused and Suppressor of Fused are shown.

9.
Trends Pharmacol Sci ; 37(1): 62-72, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26432668

ABSTRACT

The G protein-coupled receptor (GPCR) Smoothened (Smo) is the signal transducer of the developmentally and therapeutically relevant Hedgehog (Hh) pathway. Although recent structural analyses have advanced our understanding of Smo biology, several questions remain. Chief among them are the identity of its natural ligand, the regulatory processes controlling its activation, and the mechanisms by which it signals to downstream effectors. In this review, we discuss recent discoveries from multiple model systems that have set the stage for solving these mysteries. We focus on the roles of distinct Smo functional domains, post-translational modifications, and trafficking, and conclude by discussing their contributions to signal output.


Subject(s)
Receptors, G-Protein-Coupled/metabolism , Animals , Drosophila Proteins/metabolism , Hedgehog Proteins/metabolism , Humans , Protein Processing, Post-Translational , Signal Transduction , Smoothened Receptor
10.
Genetics ; 196(1): 91-105, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24172131

ABSTRACT

The Drosophila melanogaster genome has been extensively characterized, but there remains a pressing need to associate gene products with phenotypes, subcellular localizations, and interaction partners. A multifunctional, Minos transposon-based protein trapping system called Hostile takeover (Hto) was developed to facilitate in vivo analyses of endogenous genes, including live imaging, purification of protein complexes, and mutagenesis. The Hto transposon features a UAS enhancer with a basal promoter, followed by an artificial exon 1 and a standard 5' splice site. Upon GAL4 induction, exon 1 can splice to the next exon downstream in the flanking genomic DNA, belonging to a random target gene. Exon 1 encodes a dual tag (FLAG epitope and mCherry red fluorescent protein), which becomes fused to the target protein. Hto was mobilized throughout the genome and then activated by eye-specific GAL4; an F1 screen for abnormal eye phenotypes was used to identify inserts that express disruptive fusion proteins. Approximately 1.7% of new inserts cause eye phenotypes. Of the first 23 verified target genes, 21 can be described as regulators of cell biology and development. Most are transcription factor genes, including AP-2, CG17181, cut, klu, mamo, Sox102F, and sv. Other target genes [l(1)G0232, nuf, pum, and Syt4] make cytoplasmic proteins, and these lines produce diverse fluorescence localization patterns. Hto permits the expression of stable carboxy-terminal subfragments of proteins, which are rarely tested in conventional genetic screens. Some of these may disrupt specific cell pathways, as exemplified by truncated forms of Mastermind and Nuf.


Subject(s)
Chromosome Mapping/methods , DNA Transposable Elements/genetics , Drosophila melanogaster/genetics , Transposases/genetics , Animals , Animals, Genetically Modified , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental , Genes, Reporter , Promoter Regions, Genetic , Transcription Factors/genetics
11.
Mol Cell Biol ; 33(12): 2375-87, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23572559

ABSTRACT

The Hedgehog signaling pathway, an essential regulator of developmental patterning, has been implicated in playing causative and survival roles in a range of human cancers. The signal-transducing component of the pathway, Smoothened, has revealed itself to be an efficacious therapeutic target in combating oncogenic signaling. However, therapeutic challenges remain in cases where tumors acquire resistance to Smoothened antagonists, and also in cases where signaling is driven by active Smoothened mutants that exhibit reduced sensitivity to these compounds. We previously demonstrated that active Smoothened mutants are subjected to prolonged endoplasmic reticulum (ER) retention, likely due to their mutations triggering conformation shifts that are detected by ER quality control. We attempted to exploit this biology and demonstrate that deregulated Hedgehog signaling driven by active Smoothened mutants is specifically attenuated by ER stressors that induce the unfolded protein response (UPR). Upon UPR induction, active Smoothened mutants are targeted by ER-associated degradation, resulting in attenuation of inappropriate pathway activity. Accordingly, we found that the UPR agonist thapsigargin attenuated mutant Smoothened-induced phenotypes in vivo in Drosophila melanogaster. Wild-type Smoothened and physiological Hedgehog patterning were not affected, suggesting that UPR modulation may provide a novel therapeutic window to be evaluated for targeting active Smoothened mutants in disease.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum/physiology , Hedgehog Proteins/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Unfolded Protein Response/physiology , 3T3 Cells , Animals , Antineoplastic Agents/pharmacology , Benzoquinones/pharmacology , Boronic Acids/pharmacology , Bortezomib , Carcinoma, Basal Cell/metabolism , Cell Line , DNA-Binding Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Enzyme Inhibitors/pharmacology , Green Fluorescent Proteins/genetics , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Heterocyclic Compounds, 4 or More Rings/pharmacology , Lactams, Macrocyclic/pharmacology , Medulloblastoma/metabolism , Mice , Mutant Proteins/metabolism , Pyrazines/pharmacology , RNA Interference , RNA, Small Interfering , Smoothened Receptor , Thapsigargin/pharmacology , Ubiquitin-Protein Ligases/genetics
12.
Nat Commun ; 4: 2965, 2013.
Article in English | MEDLINE | ID: mdl-24351982

ABSTRACT

Smoothened (Smo) is a member of the Frizzled (FzD) class of G-protein-coupled receptors (GPCRs), and functions as the key transducer in the Hedgehog (Hh) signalling pathway. Smo has an extracellular cysteine-rich domain (CRD), indispensable for its function and downstream Hh signalling. Despite its essential role, the functional contribution of the CRD to Smo signalling has not been clearly elucidated. However, given that the FzD CRD binds to the endogenous Wnt ligand, it has been proposed that the Smo CRD may bind its own endogenous ligand. Here we present the NMR solution structure of the Drosophila Smo CRD, and describe interactions between the glucocorticoid budesonide (Bud) and the Smo CRDs from both Drosophila and human. Our results highlight a function of the Smo CRD, demonstrating its role in binding to small-molecule modulators.


Subject(s)
Drosophila Proteins/metabolism , Hedgehog Proteins/metabolism , Protein Structure, Tertiary , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Budesonide/metabolism , Drosophila melanogaster , Frizzled Receptors/metabolism , Glucocorticoids/metabolism , Humans , Ligands , Models, Molecular , Molecular Sequence Data , Protein Binding , Sequence Homology, Amino Acid , Smoothened Receptor , Species Specificity
13.
Curr Biol ; 22(5): 389-96, 2012 Mar 06.
Article in English | MEDLINE | ID: mdl-22305752

ABSTRACT

The conserved Hippo signaling pathway acts in growth control and is fundamental to animal development and oncogenesis. Hippo signaling has also been implicated in adult midgut homeostasis in Drosophila. Regulated divisions of intestinal stem cells (ISCs), giving rise to an ISC and an enteroblast (EB) that differentiates into an enterocyte (EC) or an enteroendocrine (EE) cell, enable rapid tissue turnover in response to intestinal stress. The damage-related increase in ISC proliferation requires deactivation of the Hippo pathway and consequential activation of the transcriptional coactivator Yorkie (Yki) in both ECs and ISCs. Here, we identify Pez, an evolutionarily conserved FERM domain protein containing a protein tyrosine phosphatase (PTP) domain, as a novel binding partner of the upstream Hippo signaling component Kibra. Pez function--but not its PTP domain--is essential for Hippo pathway activity specifically in the fly midgut epithelium. Thus, Pez displays a tissue-specific requirement and functions as a negative upstream regulator of Yki in the regulation of ISC proliferation.


Subject(s)
Cytoskeletal Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Tyrosine Phosphatases/metabolism , Stem Cells/metabolism , Animals , Cell Differentiation , Cell Proliferation , Drosophila/physiology , ErbB Receptors/metabolism , Intestinal Mucosa/metabolism , Intestines/cytology , Janus Kinases/metabolism , Nuclear Proteins/metabolism , Signal Transduction , Trans-Activators/metabolism , Tumor Suppressor Proteins/metabolism , YAP-Signaling Proteins
14.
Cancer Cell ; 22(5): 683-97, 2012 Nov 13.
Article in English | MEDLINE | ID: mdl-23153540

ABSTRACT

To define the mutation spectrum in non-Down syndrome acute megakaryoblastic leukemia (non-DS-AMKL), we performed transcriptome sequencing on diagnostic blasts from 14 pediatric patients and validated our findings in a recurrency/validation cohort consisting of 34 pediatric and 28 adult AMKL samples. Our analysis identified a cryptic chromosome 16 inversion (inv(16)(p13.3q24.3)) in 27% of pediatric cases, which encodes a CBFA2T3-GLIS2 fusion protein. Expression of CBFA2T3-GLIS2 in Drosophila and murine hematopoietic cells induced bone morphogenic protein (BMP) signaling and resulted in a marked increase in the self-renewal capacity of hematopoietic progenitors. These data suggest that expression of CBFA2T3-GLIS2 directly contributes to leukemogenesis.


Subject(s)
Kruppel-Like Transcription Factors/genetics , Leukemia, Megakaryoblastic, Acute/genetics , Oncogene Proteins, Fusion/genetics , Repressor Proteins/genetics , Tumor Suppressor Proteins/genetics , Animals , Bone Morphogenetic Proteins/metabolism , Child , Chromosome Inversion , Chromosomes, Human, Pair 16 , Drosophila/genetics , Drosophila/growth & development , Gene Expression Profiling , Humans , Leukemia, Megakaryoblastic, Acute/classification , Leukemia, Megakaryoblastic, Acute/diagnosis , Mice , Oncogene Proteins, Fusion/metabolism , Oncogene Proteins, Fusion/physiology , Prognosis , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/physiology , Sequence Analysis, RNA , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL