Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Science ; 381(6662): eabn4180, 2023 09 08.
Article in English | MEDLINE | ID: mdl-37676964

ABSTRACT

Despite substantial advances in targeting mutant KRAS, tumor resistance to KRAS inhibitors (KRASi) remains a major barrier to progress. Here, we report proteostasis reprogramming as a key convergence point of multiple KRASi-resistance mechanisms. Inactivation of oncogenic KRAS down-regulated both the heat shock response and the inositol-requiring enzyme 1α (IRE1α) branch of the unfolded protein response, causing severe proteostasis disturbances. However, IRE1α was selectively reactivated in an ER stress-independent manner in acquired KRASi-resistant tumors, restoring proteostasis. Oncogenic KRAS promoted IRE1α protein stability through extracellular signal-regulated kinase (ERK)-dependent phosphorylation of IRE1α, leading to IRE1α disassociation from 3-hydroxy-3-methylglutaryl reductase degradation (HRD1) E3-ligase. In KRASi-resistant tumors, both reactivated ERK and hyperactivated AKT restored IRE1α phosphorylation and stability. Suppression of IRE1α overcame resistance to KRASi. This study reveals a druggable mechanism that leads to proteostasis reprogramming and facilitates KRASi resistance.


Subject(s)
Antineoplastic Agents , Drug Resistance, Neoplasm , Endoribonucleases , Enzyme Inhibitors , Extracellular Signal-Regulated MAP Kinases , Heat Shock Transcription Factors , Neoplasms , Proteostasis , Proto-Oncogene Proteins p21(ras) , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Protein Serine-Threonine Kinases , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Enzyme Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Heat Shock Transcription Factors/metabolism
2.
Sci Adv ; 9(31): eadf3984, 2023 08 04.
Article in English | MEDLINE | ID: mdl-37540752

ABSTRACT

The glioblastoma (GBM) stem cell-like cells (GSCs) are critical for tumorigenesis/therapeutic resistance of GBM. Mounting evidence supports tumor-promoting function of long noncoding RNAs (lncRNAs), but their role in GSCs remains poorly understood. By combining CRISPRi screen with orthogonal multiomics approaches, we identified a lncRNA DARS1-AS1-controlled posttranscriptional circuitry that promoted the malignant properties of GBM cells/GSCs. Depleting DARS1-AS1 inhibited the proliferation of GBM cells/GSCs and self-renewal of GSCs, prolonging survival in orthotopic GBM models. DARS1-AS1 depletion also impaired the homologous recombination (HR)-mediated double-strand break (DSB) repair and enhanced the radiosensitivity of GBM cells/GSCs. Mechanistically, DARS1-AS1 interacted with YBX1 to promote target mRNA binding and stabilization, forming a mixed transcriptional/posttranscriptional feed-forward loop to up-regulate expression of the key regulators of G1-S transition, including E2F1 and CCND1. DARS1-AS1/YBX1 also stabilized the mRNA of FOXM1, a master transcription factor regulating GSC self-renewal and DSB repair. Our findings suggest DARS1-AS1/YBX1 axis as a potential therapeutic target for sensitizing GBM to radiation/HR deficiency-targeted therapy.


Subject(s)
Brain Neoplasms , Glioblastoma , RNA, Long Noncoding , Humans , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Carcinogenesis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Multiomics , RNA, Long Noncoding/genetics , Y-Box-Binding Protein 1/genetics , Y-Box-Binding Protein 1/metabolism
3.
J Clin Invest ; 133(5)2023 03 01.
Article in English | MEDLINE | ID: mdl-36856111

ABSTRACT

Emerging evidence suggests that cryptic translation within long noncoding RNAs (lncRNAs) may produce novel proteins with important developmental/physiological functions. However, the role of this cryptic translation in complex diseases (e.g., cancer) remains elusive. Here, we applied an integrative strategy combining ribosome profiling and CRISPR/Cas9 screening with large-scale analysis of molecular/clinical data for breast cancer (BC) and identified estrogen receptor α-positive (ER+) BC dependency on the cryptic ORFs encoded by lncRNA genes that were upregulated in luminal tumors. We confirmed the in vivo tumor-promoting function of an unannotated protein, GATA3-interacting cryptic protein (GT3-INCP) encoded by LINC00992, the expression of which was associated with poor prognosis in luminal tumors. GTE-INCP was upregulated by estrogen/ER and regulated estrogen-dependent cell growth. Mechanistically, GT3-INCP interacted with GATA3, a master transcription factor key to mammary gland development/BC cell proliferation, and coregulated a gene expression program that involved many BC susceptibility/risk genes and impacted estrogen response/cell proliferation. GT3-INCP/GATA3 bound to common cis regulatory elements and upregulated the expression of the tumor-promoting and estrogen-regulated BC susceptibility/risk genes MYB and PDZK1. Our study indicates that cryptic lncRNA-encoded proteins can be an important integrated component of the master transcriptional regulatory network driving aberrant transcription in cancer, and suggests that the "hidden" lncRNA-encoded proteome might be a new space for therapeutic target discovery.


Subject(s)
Breast Neoplasms , RNA, Long Noncoding , Humans , Female , RNA, Long Noncoding/genetics , Open Reading Frames , CRISPR-Cas Systems , Breast Neoplasms/genetics , Estrogens
4.
Genome Biol ; 22(1): 240, 2021 08 23.
Article in English | MEDLINE | ID: mdl-34425866

ABSTRACT

BACKGROUND: The human genome encodes over 14,000 pseudogenes that are evolutionary relics of protein-coding genes and commonly considered as nonfunctional. Emerging evidence suggests that some pseudogenes may exert important functions. However, to what extent human pseudogenes are functionally relevant remains unclear. There has been no large-scale characterization of pseudogene function because of technical challenges, including high sequence similarity between pseudogene and parent genes, and poor annotation of transcription start sites. RESULTS: To overcome these technical obstacles, we develop an integrated computational pipeline to design the first genome-wide library of CRISPR interference (CRISPRi) single-guide RNAs (sgRNAs) that target human pseudogene promoter-proximal regions. We perform the first pseudogene-focused CRISPRi screen in luminal A breast cancer cells and reveal approximately 70 pseudogenes that affect breast cancer cell fitness. Among the top hits, we identify a cancer-testis unitary pseudogene, MGAT4EP, that is predominantly localized in the nucleus and interacts with FOXA1, a key regulator in luminal A breast cancer. By enhancing the promoter binding of FOXA1, MGAT4EP upregulates the expression of oncogenic transcription factor FOXM1. Integrative analyses of multi-omic data from the Cancer Genome Atlas (TCGA) reveal many unitary pseudogenes whose expressions are significantly dysregulated and/or associated with overall/relapse-free survival of patients in diverse cancer types. CONCLUSIONS: Our study represents the first large-scale study characterizing pseudogene function. Our findings suggest the importance of nuclear function of unitary pseudogenes and underscore their underappreciated roles in human diseases. The functional genomic resources developed here will greatly facilitate the study of human pseudogene function.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Pseudogenes/genetics , Breast Neoplasms/genetics , Cell Nucleus/genetics , Cell Proliferation , Computational Biology , Forkhead Box Protein M1/metabolism , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , MCF-7 Cells , Promoter Regions, Genetic/genetics , Protein Binding , RNA, Guide, Kinetoplastida/genetics , Reproducibility of Results , Up-Regulation/genetics
5.
Elife ; 102021 07 09.
Article in English | MEDLINE | ID: mdl-34240701

ABSTRACT

Signals from the pre-T cell receptor and Notch coordinately instruct ß-selection of CD4-CD8-double negative (DN) thymocytes to generate αß T cells in the thymus. However, how these signals ensure a high-fidelity proteome and safeguard the clonal diversification of the pre-selection TCR repertoire given the considerable translational activity imposed by ß-selection is largely unknown. Here, we identify the endoplasmic reticulum (ER)-associated degradation (ERAD) machinery as a critical proteostasis checkpoint during ß-selection. Expression of the SEL1L-HRD1 complex, the most conserved branch of ERAD, is directly regulated by the transcriptional activity of the Notch intracellular domain. Deletion of Sel1l impaired DN3 to DN4 thymocyte transition and severely impaired mouse αß T cell development. Mechanistically, Sel1l deficiency induced unresolved ER stress that triggered thymocyte apoptosis through the PERK pathway. Accordingly, genetically inactivating PERK rescued T cell development from Sel1l-deficient thymocytes. In contrast, IRE1α/XBP1 pathway was induced as a compensatory adaptation to alleviate Sel1l-deficiency-induced ER stress. Dual loss of Sel1l and Xbp1 markedly exacerbated the thymic defect. Our study reveals a critical developmental signal controlled proteostasis mechanism that enforces T cell development to ensure a healthy adaptive immunity.


Subject(s)
Endoplasmic Reticulum-Associated Degradation/drug effects , Receptors, Notch/metabolism , Thymocytes/metabolism , Animals , Endoplasmic Reticulum Stress , Endoribonucleases/metabolism , Female , Inflammation , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/metabolism , Proteostasis , Ubiquitin-Protein Ligases/metabolism , X-Box Binding Protein 1/metabolism
6.
Cell Discov ; 6: 43, 2020.
Article in English | MEDLINE | ID: mdl-32637151

ABSTRACT

Genetic robustness is an important characteristic to tolerate genetic or nongenetic perturbations and ensure phenotypic stability. Morphogens, a type of evolutionarily conserved diffusible molecules, govern tissue patterns in a direction-dependent or concentration-dependent manner by differentially regulating downstream gene expression. However, whether the morphogen-directed gene regulatory network possesses genetic robustness remains elusive. In the present study, we collected 4217 morphogen-responsive genes along A-P axis of Drosophila wing discs from the RNA-seq data, and clustered them into 12 modules. By applying mathematical model to the measured data, we constructed a gene modular network (GMN) to decipher the module regulatory interactions and robustness in morphogen-directed development. The computational analyses on asymptotical dynamics of this GMN demonstrated that this morphogen-directed GMN is robust to tolerate a majority of genetic perturbations, which has been further validated by biological experiments. Furthermore, besides the genetic alterations, we further demonstrated that this morphogen-directed GMN can well tolerate nongenetic perturbations (Hh production changes) via computational analyses and experimental validation. Therefore, these findings clearly indicate that the morphogen-directed GMN is robust in response to perturbations and is important for Drosophila to ensure the proper tissue patterning in wing disc.

7.
J Vis Exp ; (159)2020 05 18.
Article in English | MEDLINE | ID: mdl-32478757

ABSTRACT

Preclinical models that faithfully recapitulate tumor heterogeneity and therapeutic response are critical for translational breast cancer research. Immortalized cell lines are easy to grow and genetically modify to study molecular mechanisms, yet the selective pressure from cell culture often leads to genetic and epigenetic alterations over time. Patient-derived xenograft (PDX) models faithfully recapitulate the heterogeneity and drug response of human breast tumors. PDX models exhibit a relatively short latency after orthotopic transplantation that facilitates the investigation of breast tumor biology and drug response. The transplantable genetically engineered mouse models allow the study of breast tumor immunity. The current protocol describes the method to orthotopically transplant breast tumor fragments into the mammary fat pad followed by drug treatments. These preclinical models provide valuable approaches to investigate breast tumor biology, drug response, biomarker discovery and mechanisms of drug resistance.


Subject(s)
Breast Neoplasms/surgery , Transplants/surgery , Animals , Breast Neoplasms/pathology , Female , Humans , Mice
8.
Nucleic Acids Res ; 47(21): 11114-11131, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31586391

ABSTRACT

Establishment and subsequent maintenance of distinct chromatin domains during embryonic stem cell (ESC) differentiation are crucial for lineage specification and cell fate determination. Here we show that the histone chaperone Chromatin Assembly Factor 1 (CAF-1), which is recruited to DNA replication forks through its interaction with proliferating cell nuclear antigen (PCNA) for nucleosome assembly, participates in the establishment of H3K27me3-mediated silencing during differentiation. Deletion of CAF-1 p150 subunit impairs the silencing of many genes including Oct4, Sox2 and Nanog as well as the establishment of H3K27me3 at these gene promoters during ESC differentiation. Mutations of PCNA residues involved in recruiting CAF-1 to the chromatin also result in defects in differentiation in vitro and impair early embryonic development as p150 deletion. Together, these results reveal that the CAF-1-PCNA nucleosome assembly pathway plays an important role in the establishment of H3K27me3-mediated silencing during cell fate determination.


Subject(s)
Cell Differentiation/genetics , Chromatin Assembly Factor-1/physiology , Embryonic Stem Cells/physiology , Heterochromatin/metabolism , Pluripotent Stem Cells/physiology , Animals , Cells, Cultured , Chromatin Assembly Factor-1/genetics , Chromatin Assembly Factor-1/metabolism , Chromatin Assembly and Disassembly/genetics , DNA Replication/genetics , Gene Silencing/physiology , Histone Methyltransferases/metabolism , Histones/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Proliferating Cell Nuclear Antigen/metabolism
10.
J Clin Invest ; 128(4): 1283-1299, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29480818

ABSTRACT

The unfolded protein response (UPR) is a cellular homeostatic mechanism that is activated in many human cancers and plays pivotal roles in tumor progression and therapy resistance. However, the molecular mechanisms for UPR activation and regulation in cancer cells remain elusive. Here, we show that oncogenic MYC regulates the inositol-requiring enzyme 1 (IRE1)/X-box binding protein 1 (XBP1) branch of the UPR in breast cancer via multiple mechanisms. We found that MYC directly controls IRE1 transcription by binding to its promoter and enhancer. Furthermore, MYC forms a transcriptional complex with XBP1, a target of IRE1, and enhances its transcriptional activity. Importantly, we demonstrate that XBP1 is a synthetic lethal partner of MYC. Silencing of XBP1 selectively blocked the growth of MYC-hyperactivated cells. Pharmacological inhibition of IRE1 RNase activity with small molecule inhibitor 8866 selectively restrained the MYC-overexpressing tumor growth in vivo in a cohort of preclinical patient-derived xenograft models and genetically engineered mouse models. Strikingly, 8866 substantially enhanced the efficacy of docetaxel chemotherapy, resulting in rapid regression of MYC-overexpressing tumors. Collectively, these data establish the synthetic lethal interaction of the IRE1/XBP1 pathway with MYC hyperactivation and provide a potential therapy for MYC-driven human breast cancers.


Subject(s)
Breast Neoplasms/drug therapy , Docetaxel/pharmacology , Drug Delivery Systems , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Response Elements , Signal Transduction/drug effects , X-Box Binding Protein 1/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Endoribonucleases/genetics , Female , Humans , Mice , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-myc/genetics , Saccharomyces cerevisiae , Signal Transduction/genetics , Unfolded Protein Response/drug effects , Unfolded Protein Response/genetics , X-Box Binding Protein 1/genetics , Xenograft Model Antitumor Assays
11.
J Mol Cell Biol ; 10(5): 437-447, 2018 10 01.
Article in English | MEDLINE | ID: mdl-29432547

ABSTRACT

The Hedgehog (Hh) signaling pathway plays important roles in both embryonic development and adult tissue homeostasis. Such biological functions are mediated by the transcription factor Cubitus interruptus (Ci). Yet the transcriptional regulation of the effector Ci itself is poorly investigated. Through an RNAi-based genetic screen, we identified that female sterile (1) homeotic (Fsh), a transcription co-activator, directly activates Ci transcription. Biochemistry assays demonstrated physical interactions among Fsh, Sex combs extra (Sce), and Polycomb (Pc). Functional assays further showed that both Pc and Sce are required for Ci expression, which is not likely mediated by the derepression of Engrailed (En), a repressor of Ci, in Pc or Sce mutant cells. Finally, we provide evidence showing that Pc/Sce facilitates the binding of Fsh at Ci locus and that the physical interaction between Fsh and Pc is essential for Fsh-mediated Ci transcription. Taken together, we not only uncover that Ci is transcriptionally regulated by Fsh-Pc-Sce complex but also provide evidence for the coordination between Fsh and PcG proteins in transcriptional regulation.


Subject(s)
DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Polycomb Repressive Complex 1/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Animals, Genetically Modified , DNA-Binding Proteins/metabolism , Drosophila melanogaster/genetics , Epigenesis, Genetic , Gene Expression Regulation , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutation , Polycomb Repressive Complex 1/genetics , Transcription, Genetic , Wings, Animal/growth & development
12.
EMBO Rep ; 18(11): 1922-1934, 2017 11.
Article in English | MEDLINE | ID: mdl-28887318

ABSTRACT

The Hh pathway controls many morphogenetic processes in metazoans and plays important roles in numerous pathologies and in cancer. Hh signaling is mediated by the activity of the Gli/Ci family of transcription factors. Several studies in Drosophila have shown that ubiquitination by the ubiquitin E3 ligases Slimb and Rdx(Hib) plays a crucial role in controlling Ci stability dependent on the levels of Hh signals. If Hh levels are low, Slimb adds K11- and K48-linked poly-ubiquitin chains on Ci resulting in partial degradation. Ubiquitin E2 enzymes are pivotal in determining the topologies of ubiquitin chains. However, which E2 enzymes participate in the selective ubiquitination-degradation of Ci remains elusive. Here, we find that the E2 enzyme UbcD1 negatively regulates Hh signaling activity in Drosophila wing disks. Genetic and biochemical analyses in wing disks and in cultured cells reveal that UbcD1 directly controls Ci stability. Interestingly, UbcD1 is found to be selectively involved in Slimb-mediated Ci degradation. Finally, we show that the homologs of UbcD1 play a conserved role in modulating Hh signaling in vertebrates.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Hedgehog Proteins/genetics , Protein Processing, Post-Translational , Ubiquitin-Conjugating Enzymes/genetics , Zebrafish/genetics , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Conserved Sequence , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Embryo, Nonmammalian , Hedgehog Proteins/metabolism , Imaginal Discs/growth & development , Imaginal Discs/metabolism , Larva/genetics , Larva/growth & development , Larva/metabolism , Patched-2 Receptor/genetics , Patched-2 Receptor/metabolism , Polyubiquitin/genetics , Polyubiquitin/metabolism , Protein Stability , Proteolysis , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
13.
Sci Rep ; 7(1): 5101, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28698559

ABSTRACT

Hox genes play a fundamental role in regulating animal development. However, less is known about their functions on homeostasis maintenance in adult stem cells. Here, we report that the repression of an important axial Hox gene, Abdominal-B (Abd-B), in cyst stem cells (CySCs) is essential for the homeostasis and cell identity maintenance in the adult Drosophila testis. Derepression of Abd-B in CySCs disrupts the proper self-renewal of both germline stem cells (GSCs) and CySCs, and leads to an excessive expansion of early stage somatic cells, which originate from both lineages. We further demonstrate that canonical Polycomb (Pc) and functional pathway of Polycomb group (PcG) proteins are responsible for maintaining the germline cell identity non-autonomously via repressing Abd-B in CySCs in the adult Drosophila testis.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila/cytology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Polycomb Repressive Complex 1/metabolism , Testis/cytology , Animals , Cell Lineage , Cells, Cultured , Drosophila/genetics , Drosophila/metabolism , Histones/metabolism , Male , Polycomb Repressive Complex 1/genetics , Polycomb-Group Proteins/metabolism , Spermatozoa/cytology , Spermatozoa/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Testis/metabolism
15.
Cell Res ; 26(5): 529-42, 2016 05.
Article in English | MEDLINE | ID: mdl-27002220

ABSTRACT

The highly conserved polycomb group (PcG) proteins maintain heritable transcription repression of the genes essential for development from fly to mammals. However, sporadic reports imply a potential role of PcGs in positive regulation of gene transcription, although systematic investigation of such function and the underlying mechanism has rarely been reported. Here, we report a Pc-mediated, H3K27me3-dependent positive transcriptional regulation of Senseless (Sens), a key transcription factor required for development. Mechanistic studies show that Pc regulates Sens expression by promoting H4K20me1 at the Sens locus. Further bioinformatic analysis at genome-wide level indicates that the existence of H4K20me1 acts as a selective mark for positive transcriptional regulation by Pc/H3K27me3. Both the intensities and specific patterns of Pc and H3K27me3 are important for the fates of target gene transcription. Moreover, binding of transcription factor Broad (Br), which physically interacts with Pc and positively regulates the transcription of Sens, is observed in Pc(+)H3K27me3(+)H4K20me1(+) genes, but not in Pc(+)H3K27me3(+)H4K20me1(-) genes. Taken together, our study reveals that, coupling with the transcription factor Br, Pc positively regulates transcription of Pc(+)H3K27me3(+)H4K20me1(+) genes in developing Drosophila wing disc.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Gene Expression Regulation, Developmental , Histones/metabolism , Lysine/metabolism , Polycomb Repressive Complex 1/metabolism , Animals , Base Sequence , Biomarkers/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/embryology , Methylation , Nuclear Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Transcription, Genetic , Wings, Animal/metabolism
16.
Development ; 143(10): 1655-62, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27013244

ABSTRACT

SUMO (Small ubiquitin-related modifier) modification (SUMOylation) is a highly dynamic post-translational modification (PTM) that plays important roles in tissue development and disease progression. However, its function in adult stem cell maintenance is largely unknown. Here, we report the function of SUMOylation in somatic cyst stem cell (CySC) self-renewal in adult Drosophila testis. The SUMO pathway cell-autonomously regulates CySC maintenance. Reduction of SUMOylation promotes premature differentiation of CySCs and impedes the proliferation of CySCs, which leads to a reduction in the number of CySCs. Consistent with this, CySC clones carrying a mutation of the SUMO-conjugating enzyme are rapidly lost. Furthermore, inhibition of the SUMO pathway phenocopies disruption of the Hedgehog (Hh) pathway, and can block the proliferation of CySCs induced by Hh activation. Importantly, the SUMO pathway directly regulates the SUMOylation of Hh pathway transcription factor Cubitus interruptus (Ci), which is required for promoting CySC proliferation. Thus, we conclude that SUMO directly targets the Hh pathway and regulates CySC maintenance in adult Drosophila testis.


Subject(s)
Aging/physiology , Drosophila Proteins/metabolism , Hedgehog Proteins/metabolism , Signal Transduction , Small Ubiquitin-Related Modifier Proteins/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Testis/cytology , Animals , Cell Differentiation , Cell Proliferation , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Epistasis, Genetic , Male , Sumoylation , Testis/metabolism
17.
Methods Mol Biol ; 1322: 61-70, 2015.
Article in English | MEDLINE | ID: mdl-26179039

ABSTRACT

Protein-protein interactions and signal-induced protein conformational changes are fundamental molecular events that are considered as essential in modern life sciences. Among various techniques developed to study such phenomena, fluorescence resonance energy transfer (FRET) is a widely used method with many advantages in detecting these molecular events. Here, we describe the application of FRET in the mechanistic investigation of cell signal transduction, taking the example of the Hh signaling pathway, which plays a critical role in embryonic development and tissue homeostasis. A number of general guidelines as well as some key notes have been summarized as a protocol for reader's reference.


Subject(s)
Fluorescence Resonance Energy Transfer , Hedgehog Proteins/chemistry , Hedgehog Proteins/metabolism , Protein Conformation , Protein Interaction Mapping , Signal Transduction , Animals , Cell Line , Drosophila , Fluorescence Resonance Energy Transfer/methods , Gene Expression , Hedgehog Proteins/genetics , Mice , Molecular Imaging , Wings, Animal/metabolism
18.
Cell Discov ; 1: 15006, 2015.
Article in English | MEDLINE | ID: mdl-27462407

ABSTRACT

Intestinal homeostasis is maintained by intestinal stem cells (ISCs) and their progenies. A complex autonomic nervous system spreads over posterior intestine. However, whether and how neurons regulate posterior intestinal homeostasis is largely unknown. Here we report that neurons regulate Drosophila posterior intestinal homeostasis. Specifically, downregulation of neuronal Hedgehog (Hh) signaling inhibits the differentiation of ISCs toward enterocytes (ECs), whereas upregulated neuronal Hh signaling promotes such process. We demonstrate that, among multiple sources of Hh ligand, those secreted by ECs induces similar phenotypes as does neuronal Hh. In addition, intestinal JAK/STAT signaling responds to activated neuronal Hh signaling, suggesting that JAK/STAT signaling acts downstream of neuronal Hh signaling in intestine. Collectively, our results indicate that neuronal Hh signaling is essential for the determination of ISC fate.

19.
J Biol Chem ; 289(32): 22333-41, 2014 Aug 08.
Article in English | MEDLINE | ID: mdl-24962581

ABSTRACT

The hedgehog (Hh) signaling pathway plays a very important role in metazoan development by controlling pattern formation. Drosophila imaginal discs are subdivided into anterior and posterior compartments that derive from adjacent cell populations. The anterior/posterior (A/P) boundaries, which are critical to maintaining the position of organizers, are established by a complex mechanism involving Hh signaling. Here, we uncover the regulation of ptc in the Hh signaling pathway by two subunits of mediator complex, Kto and Skd, which can also regulate boundary location. Collectively, we provide further evidence that Kto-Skd affects the A/P-axial development of the whole wing disc. Kto can interact with Cubitus interruptus (Ci), bind to the Ci-binding region on ptc promoter, which are both regulated by Hh signals to down-regulate ptc expression.


Subject(s)
DNA-Binding Proteins/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Eye Proteins/metabolism , Hedgehog Proteins/metabolism , Receptors, Cell Surface/genetics , Transcription Factors/metabolism , Animals , Animals, Genetically Modified , Body Patterning , Drosophila melanogaster/genetics , Eye Proteins/genetics , Gene Expression Regulation, Developmental , Genes, Insect , Hedgehog Proteins/genetics , Multiprotein Complexes/metabolism , Mutation , Promoter Regions, Genetic , Signal Transduction , Wings, Animal/growth & development , Wings, Animal/metabolism
20.
PLoS Biol ; 11(11): e1001721, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24302888

ABSTRACT

Hedgehog signaling plays conserved roles in controlling embryonic development; its dysregulation has been implicated in many human diseases including cancers. Hedgehog signaling has an unusual reception system consisting of two transmembrane proteins, Patched receptor and Smoothened signal transducer. Although activation of Smoothened and its downstream signal transduction have been intensively studied, less is known about how Patched receptor is regulated, and particularly how this regulation contributes to appropriate Hedgehog signal transduction. Here we identified a novel role of Smurf E3 ligase in regulating Hedgehog signaling by controlling Patched ubiquitination and turnover. Moreover, we showed that Smurf-mediated Patched ubiquitination depends on Smo activity in wing discs. Mechanistically, we found that Smo interacts with Smurf and promotes it to mediate Patched ubiquitination by targeting the K1261 site in Ptc. The further mathematic modeling analysis reveals that a bidirectional control of activation of Smo involving Smurf and Patched is important for signal-receiving cells to precisely interpret external signals, thereby maintaining Hedgehog signaling reliability. Finally, our data revealed an evolutionarily conserved role of Smurf proteins in controlling Hh signaling by targeting Ptc during development.


Subject(s)
Drosophila Proteins/metabolism , Drosophila Proteins/physiology , Drosophila melanogaster/enzymology , Hedgehog Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Line , Enzyme Activation , Protein Structure, Tertiary , Proteolysis , Signal Transduction , Smoothened Receptor , Two-Hybrid System Techniques , Ubiquitination , Wings, Animal/enzymology , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...