Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Development ; 150(24)2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38031990

ABSTRACT

Balanced control of stem cell proliferation and differentiation underlines tissue homeostasis. Disruption of tissue homeostasis often results in many diseases. However, how endogenous factors influence the proliferation and differentiation of intestinal stem cells (ISCs) under physiological and pathological conditions remains poorly understood. Here, we find that the evolutionarily conserved endoplasmic reticulum membrane protein complex (EMC) negatively regulates ISC proliferation and intestinal homeostasis. Compromising EMC function in progenitors leads to excessive ISC proliferation and intestinal homeostasis disruption. Mechanistically, the EMC associates with and stabilizes Hippo (Hpo) protein, the key component of the Hpo signaling pathway. In the absence of EMC, Yorkie (Yki) is activated to promote ISC proliferation due to Hpo destruction. The EMC-Hpo-Yki axis also functions in enterocytes to maintain intestinal homeostasis. Importantly, the levels of the EMC are dramatically diminished in tunicamycin-treated animals, leading to Hpo destruction, thereby resulting in intestinal homeostasis disruption due to Yki activation. Thus, our study uncovers the molecular mechanism underlying the action of the EMC in intestinal homeostasis maintenance under physiological and pathological conditions and provides new insight into the pathogenesis of tunicamycin-induced tumorigenesis.


Subject(s)
Drosophila Proteins , Protein Serine-Threonine Kinases , Animals , Protein Serine-Threonine Kinases/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Signal Transduction/physiology , Drosophila Proteins/metabolism , Tunicamycin/metabolism , Trans-Activators/metabolism , Cell Proliferation , Nuclear Proteins/metabolism , Homeostasis , Drosophila melanogaster/metabolism
2.
J Biol Chem ; 299(11): 105354, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37858678

ABSTRACT

O-linked N-acetylglucosamine (O-GlcNAc) glycosylation, a prevalent protein post-translational modification (PTM) that occurs intracellularly, has been shown to crosstalk with phosphorylation and ubiquitination. However, it is unclear whether it interplays with other PTMs. Here we studied its relationship with ADP-ribosylation, which involves decorating target proteins with the ADP-ribose moiety. We discovered that the poly(ADP-ribosyl)ation "eraser", ADP-ribose glycohydrolase (PARG), is O-GlcNAcylated at Ser26, which is in close proximity to its nuclear localization signal. O-GlcNAcylation of PARG promotes nuclear localization and chromatin association. Upon DNA damage, O-GlcNAcylation augments the recruitment of PARG to DNA damage sites and interacting with proliferating cell nuclear antigen (PCNA). In hepatocellular carcinoma (HCC) cells, PARG O-GlcNAcylation enhances the poly(ADP-ribosyl)ation of DNA damage-binding protein 1 (DDB1) and attenuates its auto-ubiquitination, thereby stabilizing DDB1 and allowing it to degrade its downstream targets, such as c-Myc. We further demonstrated that PARG-S26A, the O-GlcNAc-deficient mutant, promoted HCC in mouse xenograft models. Our findings thus reveal that PARG O-GlcNAcylation inhibits HCC, and we propose that O-GlcNAc glycosylation may crosstalk with many other PTMs.


Subject(s)
Carcinoma, Hepatocellular , Glycoside Hydrolases , Liver Neoplasms , Animals , Humans , Mice , Acetylglucosamine , ADP-Ribosylation , Glycoside Hydrolases/genetics , Glycoside Hydrolases/metabolism , Poly Adenosine Diphosphate Ribose/metabolism , Glycosylation , Protein Processing, Post-Translational
3.
J Cell Sci ; 134(3)2021 02 08.
Article in English | MEDLINE | ID: mdl-33468621

ABSTRACT

Haploid male gametes are produced through meiosis during gametogenesis. Whereas the cell biology of mitosis and meiosis is well studied in the nematode Caenorhabditis elegans, comparatively little is known regarding the physical division of primary spermatocytes during meiosis I. Here, we investigated this process using high-resolution time-lapse confocal microscopy and examined the spatiotemporal regulation of contractile ring assembly in C. elegans primary spermatocytes. We found that centralspindlin and RhoA effectors were recruited to the equatorial cortex of dividing primary spermatocytes for contractile ring assembly before segregation of homologous chromosomes. We also observed that perturbations shown to promote centralspindlin oligomerization regulated the cortical recruitment of NMY-2 and impacted the order in which primary spermatocytes along the proximal-distal axis of the gonad enter meiosis I. These results expand our understanding of the cellular division of primary spermatocytes into secondary spermatocytes during meiosis I.This article has an associated First Person interview with the first author of the paper.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Cytokinesis , Male , Meiosis , Spermatocytes
4.
Nat Chem Biol ; 16(11): 1246-1254, 2020 11.
Article in English | MEDLINE | ID: mdl-32807966

ABSTRACT

The diamide insecticide class is one of the top-selling insecticides globally. They are used to control a wide range of pests by targeting their ryanodine receptors (RyRs). Here, we report the highest-resolution cryo-electron microscopy (cryo-EM) structure of RyR1 in the open state, in complex with the anthranilic diamide chlorantraniliprole (CHL). The 3.2-Å local resolution map facilitates unambiguous assignment of the CHL binding site. The molecule induces a conformational change by affecting the S4-S5 linker, triggering channel opening. The binding site is further corroborated by mutagenesis data, which reveal how diamide insecticides are selective to the Lepidoptera group of insects over honeybee or mammalian RyRs. Our data reveal that several pests have developed resistance via two mechanisms, steric hindrance and loss of contact. Our results provide a foundation for the development of highly selective pesticides aimed at overcoming resistance and therapeutic molecules to treat human myopathies.


Subject(s)
Calcium Channel Blockers/metabolism , Diamide/chemistry , Insecticides/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , ortho-Aminobenzoates/metabolism , Amino Acid Sequence , Animals , Bees , Binding Sites , Calcium Channel Blockers/chemistry , Calcium Channel Blockers/pharmacology , Cryoelectron Microscopy , Drug Development , Drug Resistance , Insecticides/chemistry , Insecticides/pharmacology , Lepidoptera , Models, Molecular , Mutagenesis/physiology , Protein Binding , Protein Conformation , Signal Transduction , Substrate Specificity , ortho-Aminobenzoates/chemistry , ortho-Aminobenzoates/pharmacology
5.
Brain Behav Immun ; 94: 196-209, 2021 05.
Article in English | MEDLINE | ID: mdl-33607238

ABSTRACT

Depression is a common mental disorder, and its main environmental risk factor is chronic stress. The activation of mammalian STE20-like kinase 1 (MST1), a key factor involved in the underlying pathophysiology of stress, can trigger synaptic plasticity impairment, neuronal dysfunction and neuroinflammation. However, it is unclear whether down-regulation of MST1 in the hippocampus protects against stress-induced behavioural dysfunctions. In this study, three mouse models were used to assess the role of MST1 in stress. Various behavioural tests, in vivo electrophysiological recordings, Western blotting, Golgi staining and immunofluorescence assay were used. The data showed that the level of phospho-MST1 (T183) was significantly increased in the hippocampus of mice subjected to chronic unpredictable mild stress (CUMS) and that mice with MST1 overexpression showed depression-like behaviours. Importantly, the impairment of cognitive functions and the hippocampal synaptic plasticity induced by CUMS were significantly improved by MST1 knockdown, suggesting that MST1 down-regulation effectively protected against stress-induced behavioural dysfunctions. Moreover, MST1 knockdown suppressed CUMS-induced microglial activation, reduced the abnormal expression of inflammatory cytokines and impeded the activation of p38, implying that the antidepressant-like effects of MST1 knockdown were associated with inhibiting the p38 pathway. These findings suggest that hippocampal MST1 is an essential regulator of stress, which can be an ideal target for the development of antidepressants in the future.


Subject(s)
Depression , Stress, Psychological , Animals , Disease Models, Animal , Down-Regulation , Hippocampus , Mice , Neuronal Plasticity , Stress, Psychological/complications
6.
Int J Mol Sci ; 20(23)2019 Nov 29.
Article in English | MEDLINE | ID: mdl-31795326

ABSTRACT

The Hippo signaling pathway is an evolutionarily conserved regulator that plays important roles in organ size control, homeostasis, and tumorigenesis. As the key effector of the Hippo pathway, Yorkie (Yki) binds to transcription factor Scalloped (Sd) and promotes the expression of target genes, leading to cell proliferation and inhibition of apoptosis. Thus, it is of great significance to understand the regulatory mechanism for Yki protein turnover. Here, we provide evidence that the deubiquitinating enzyme ubiquitin-specific protease 10 (Usp10) binds Yki to counteract Yki ubiquitination and stabilize Yki protein in Drosophila S2 cells. The results in Drosophila wing discs indicate that silence of Usp10 decreases the transcription of target genes of the Hippo pathway by reducing Yki protein. In vivo functional analysis ulteriorly showed that Usp10 upregulates the Yki activity in Drosophila eyes. These findings uncover Usp10 as a novel Hippo pathway modulator and provide a new insight into the regulation of Yki protein stability and activity.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Trans-Activators/metabolism , Animals , Cell Line , Cytoplasm/metabolism , Drosophila Proteins/analysis , Drosophila melanogaster/cytology , Nuclear Proteins/analysis , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Trans-Activators/analysis , Ubiquitin-Specific Proteases , Ubiquitination , YAP-Signaling Proteins
7.
Biochem Biophys Res Commun ; 495(1): 629-633, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29154825

ABSTRACT

The C2H2 type zinc-finger transcription factor Nerfin-1 expresses dominantly in Drosophila nervous system and plays an important role in early axon guidance decisions and preventing neurons dedifferentiation. Recently, increasing reports indicated that INSM1 (homologue to nerfin-1 in mammals) is a useful marker for prognosis of neuroendocrine tumors. The dynamic expression of Nerfin-1 is regulated post-transcriptionally by multiple microRNAs; however, its post-translational regulation is still unclear. Here we showed that the protein turnover of Nerfin-1 is regulated by Slimb, the substrate adaptor of SCFSlimb ubiquitin ligase complex. Mechanistically, Slimb associates with Nerfin-1 and promotes it ubiquitination and degradation in Drosophila S2R+ cells. Furthermore, we determined that the C-terminal half of Nerfin-1 (Nerfin-1CT) is required for its binding to Slimb. Genetic epistasis assays showed that Slimb misexpression antagonizes, while knock-down enhances the activity of Nerfin-1CT in Drosophila eyes. Our data revealed a new link to understand the underlying mechanism for Nerfin-1 turnover in post-translational level, and provided useful insights in animal development and disease treatment by manipulating the activity of Slimb and Nerfin-1.


Subject(s)
Cell Cycle Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Transcription Factors/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/physiology , Animals , Gene Expression Regulation, Developmental/physiology , Metabolic Clearance Rate , SKP Cullin F-Box Protein Ligases/metabolism
8.
PLoS Genet ; 11(10): e1005589, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26474173

ABSTRACT

Interleukin-1 receptor-associated kinases (IRAKs) are crucial mediators of the IL-1R/TLR signaling pathways that regulate the immune and inflammation response in mammals. Recent studies also suggest a critical role of IRAKs in tumor development, though the underlying mechanism remains elusive. Pelle is the sole Drosophila IRAK homolog implicated in the conserved Toll pathway that regulates Dorsal/Ventral patterning, innate immune response, muscle development and axon guidance. Here we report a novel function of pll in modulating apoptotic cell death, which is independent of the Toll pathway. We found that loss of pll results in reduced size in wing tissue, which is caused by a reduction in cell number but not cell size. Depletion of pll up-regulates the transcription of pro-apoptotic genes, and triggers caspase activation and cell death. The transcription factor dFoxO is required for loss-of-pll induced cell death. Furthermore, loss of pll activates dFoxO, promotes its translocation from cytoplasm to nucleus, and up-regulates the transcription of its target gene Thor/4E-BP. Finally, Pll physically interacts with dFoxO and phosphorylates dFoxO directly. This study not only identifies a previously unknown physiological function of pll in cell death, but also shed light on the mechanism of IRAKs in cell survival/death during tumorigenesis.


Subject(s)
Apoptosis/genetics , Drosophila Proteins/genetics , Forkhead Transcription Factors/genetics , Immunity, Innate/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Carcinogenesis/genetics , Drosophila/genetics , Drosophila Proteins/metabolism , Forkhead Transcription Factors/metabolism , Humans , Interleukin-1 Receptor-Associated Kinases/genetics , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , Wings, Animal/growth & development , Wings, Animal/metabolism
9.
Proc Natl Acad Sci U S A ; 112(4): 1065-70, 2015 Jan 27.
Article in English | MEDLINE | ID: mdl-25583514

ABSTRACT

The Hippo and c-Jun N-terminal kinase (JNK) pathway both regulate growth and contribute to tumorigenesis when dysregulated. Whereas the Hippo pathway acts via the transcription coactivator Yki/YAP to regulate target gene expression, JNK signaling, triggered by various modulators including Rho GTPases, activates the transcription factors Jun and Fos. Here, we show that impaired Hippo signaling induces JNK activation through Rho1. Blocking Rho1-JNK signaling suppresses Yki-induced overgrowth in the wing disk, whereas ectopic Rho1 expression promotes tissue growth when apoptosis is prohibited. Furthermore, Yki directly regulates Rho1 transcription via the transcription factor Sd. Thus, our results have identified a novel molecular link between the Hippo and JNK pathways and implicated the essential role of the JNK pathway in Hippo signaling-related tumorigenesis.


Subject(s)
Drosophila Proteins/metabolism , Imaginal Discs/embryology , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/physiology , Protein Serine-Threonine Kinases/metabolism , Wings, Animal/embryology , rho GTP-Binding Proteins/metabolism , Animals , Drosophila Proteins/genetics , Drosophila melanogaster , Imaginal Discs/cytology , Intracellular Signaling Peptides and Proteins/genetics , JNK Mitogen-Activated Protein Kinases/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription, Genetic/physiology , Wings, Animal/cytology , YAP-Signaling Proteins , rho GTP-Binding Proteins/genetics
10.
Biochem Biophys Res Commun ; 482(2): 317-322, 2017 Jan 08.
Article in English | MEDLINE | ID: mdl-27856247

ABSTRACT

SMAD ubiquitination regulatory factors 1 and 2 (Smurf1/2) are members of the HECT domain E3 ligase family which play crucial roles in the regulation of cell cycle progression, planar cell polarity, cancer metastasis and cell apoptosis. We recently showed that the Drosophila homolog dSmurf controls the stability of Warts kinase to regulate the Hippo pathway. In the current study, we found that the F-box protein Slimb controls dSmurf protein level to regulate the Hippo pathway. Slimb physically associates with dSmurf as revealed by co-immunoprecipitation assay in S2 cells. The C-terminal WD40 repeats of Slimb (188-510 amino acid) and the C-terminal HECT domain of dSmurf (723-1061 amino acid) are necessary for their binding. Interaction with Slimb leads to the ubiquitination and degradation of dSmurf, resulting in negative regulation of dSmurf-mediated Yki phosphorylation and activity in the Hippo pathway. Thus our study revealed a new regulatory mechanism of the Hippo pathway which may provide implications for developing tumor treatment.


Subject(s)
Cell Cycle Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/physiology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/physiology , Animals , F-Box Proteins/metabolism , Gene Expression Regulation, Developmental/physiology , Organogenesis/physiology , Ubiquitinated Proteins/metabolism
11.
Yi Chuan ; 39(7): 537-545, 2017 Jul 20.
Article in English | MEDLINE | ID: mdl-28757469

ABSTRACT

The discovery of Hippo signaling pathway is another breakthrough of fly genetics. Similar to the other signaling pathways, Hippo pathway also functions crucially in tremendous physiological and pathological conditions, like organ size control and cancer. There are three main stages of Hippo pathway study: Firstly, identifications of core components by fly genetic screens; secondly, regulations by versatile upstream cues, like cytoskeleton, mechanical tension, and nutrition; thirdly, functions in different biological processes, like cell proliferation regulation, stem cell biology, and immunology. In this review, we summarize the current understanding of Hippo pathway and highlight its regulations and transcriptional complex assembly. We also discuss the potential future directions in Drosophila model system.


Subject(s)
Drosophila Proteins/physiology , Intracellular Signaling Peptides and Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Signal Transduction/physiology , Animals , Drosophila , Transcription, Genetic
12.
Biochem Biophys Res Commun ; 470(1): 29-34, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26718403

ABSTRACT

The LIM-homeodomain (LIM-HD) family member Lmx1a has been successfully used to induce dopaminergic neurons from other cell types, thus showing significant implications in replacement therapies of Parkinson's disease, but the underlying mechanism remains elusive. In this study, we used Drosophila eye as a model system to investigate how forced expression of dLmx1a, the fly homolog of human Lmx1a, alters cell identify. We found that ectopic expression of dLmx1a suppresses the formation of Drosophila eye tissue and identified the LIM and HD as two essential domains. dLmx1a requires and physically binds to Chip, a well-known cofactor of LIM-HD proteins. Chip connects two dLmx1a proteins to form a functional tetrameric complex. In addition, we provide evidence showing that dLmx1a expression results in the suppression of two retina determination gene eyes absent (eya) and string (stg). Taken together, our findings identified Chip as a novel partner of dLmx1a to alter cell differentiation in Drosophila eye through repressing eya and stg expression, and provide an animal model for further understanding the molecular mechanism whereby Lmx1a determines cell fate.


Subject(s)
Drosophila Proteins/metabolism , Eye Proteins/metabolism , Gene Expression Regulation, Developmental/physiology , LIM-Homeodomain Proteins/metabolism , Nuclear Proteins/metabolism , Ocular Physiological Phenomena , Transcription Factors/metabolism , Animals , Drosophila , Protein Structure, Tertiary
13.
Biochem Biophys Res Commun ; 452(3): 808-12, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25201729

ABSTRACT

Cylindromatosis (CYLD), a deubiquitinase and regulator of microtubule dynamics, has important roles in the regulation of inflammation, immune response, apoptosis, mitosis, cell migration and tumorigenesis. Although great progress has been made in the biochemical and cellular functions of CYLD, its role in animal development remains elusive. In this study, we identified Drosophila CYLD (dCYLD) as a negative regulator of the Hippo pathway in vivo. dCYLD associates and colocalizes with Hpo, a core component of the Hippo pathway, in the cytoplasm, and decreases Hpo activity through limiting its phosphorylation at T195. We also showed that dCYLD limits Hippo signal transduction as evidenced by decreasing phosphorylation and thereby increasing activity of Yki, the key downstream effector of the Hippo pathway. These findings uncover dCYLD as a negative regulator of the Hippo pathway and provide new insights into the physiological function of dCYLD in animal development.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/genetics , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Trans-Activators/genetics , Tumor Suppressor Proteins/genetics , Animals , Cell Line , Cytoplasm/metabolism , Deubiquitinating Enzyme CYLD , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Threonine/metabolism , Trans-Activators/metabolism , Transfection , Tumor Suppressor Proteins/metabolism , YAP-Signaling Proteins
14.
Biochem Biophys Res Commun ; 454(1): 167-71, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25450375

ABSTRACT

The Hippo pathway has been implicated in controlling organ size and tumorigenesis and the underlying molecular mechanisms have attracted intensive attentions. In this work, we identified dSmurf as a new regulator of Wts, a core component of the Hippo pathway, in Drosophila. Our data revealed that Wts and dSmurf colocalize to cytoplasm and physically form an immunoprecipitated complex in S2 cells. Sufficient knock-down of dSmurf increases the protein abundance of Wts and thus increases phosphorylation level at S168 of Yki, the key downstream target of Wts in the Hippo pathway. Genetic epistasis assays showed that halving dosage of dSmurf dominantly enhances the phenotype caused by overexpression of Wts and restrains Yki activity in Drosophila eyes. Our works defines a novel role of dSmurf in animal development through modulating Wts turnover and thereby Hippo signal transduction, implying that targeting dSmurf may be a promising therapeutic strategy to manipulate the Hippo pathway in pathological conditions.


Subject(s)
Drosophila Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Animals, Genetically Modified , Cell Line , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Epistasis, Genetic , Eye/growth & development , Eye/metabolism , Gene Knockdown Techniques , Genes, Insect , Male , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Kinases/genetics , Signal Transduction , Trans-Activators/genetics , Trans-Activators/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , YAP-Signaling Proteins
15.
J Genet Genomics ; 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38663479

ABSTRACT

Yorkie (Yki) is a key effector of the Hippo pathway that activates the expression of targets by associating with the transcription factor Scalloped. Various upstream signals, such as cell polarity and mechanical cues, control transcriptional programs by regulating Yki activity. Searching for Yki regulatory factors has far-reaching significance for studying the Hippo pathway in animal development and human diseases. In this study, we identify Calpain-A (CalpA) and Calpain-B (CalpB), two calcium (Ca2+)-dependent modulatory proteases of the calpain family, as critical regulators of Yki in Drosophila that interact with Yki, respectively. Ca2+ induces Yki cleavage in a CalpA/CalpB-dependent manner, and the protease activity of CalpA/CalpB is pivotal for the cleavage. Furthermore, overexpression of CalpA or CalpB in Drosophila partially restores the large wing phenotype caused by Yki overexpression, and F98 of Yki is an important cleavage site by the Ca2+-calpains axis. Our study uncovers a unique mechanism whereby the Ca2+-calpain axis modulates Yki activity through protein cleavage.

16.
Adv Sci (Weinh) ; 11(25): e2400569, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38666385

ABSTRACT

The photoreceptor cilium is vital for maintaining the structure and function of the retina. However, the molecular mechanisms underlying the photoreceptor cilium integrity and retinal homeostasis are largely unknown. Herein, it is shown that kinesin family member 11 (KIF11) localizes at the transition zone (connecting cilium) of the photoreceptor and plays a crucial role in orchestrating the cilium integrity. KIF11 depletion causes malformations of both the photoreceptor ciliary axoneme and membranous discs, resulting in photoreceptor degeneration and the accumulation of drusen-like deposits throughout the retina. Mechanistic studies show that the stability of KIF11 is regulated by an interplay between its UFMylation and ubiquitination; UFMylation of KIF11 at lysine 953 inhibits its ubiquitination by synoviolin 1 and thereby prevents its proteasomal degradation. The lysine 953-to-arginine mutant of KIF11 is more stable than wild-type KIF11 and also more effective in reversing the ciliary and retinal defects induced by KIF11 depletion. These findings identify a critical role for KIF11 UFMylation in the maintenance of photoreceptor cilium integrity and retinal homeostasis.


Subject(s)
Cilia , Homeostasis , Kinesins , Retina , Kinesins/metabolism , Kinesins/genetics , Animals , Mice , Homeostasis/physiology , Cilia/metabolism , Cilia/genetics , Retina/metabolism , Disease Models, Animal , Ubiquitination , Humans , Retinal Degeneration/metabolism , Retinal Degeneration/genetics
17.
Biochem Biophys Res Commun ; 433(4): 538-41, 2013 Apr 19.
Article in English | MEDLINE | ID: mdl-23524264

ABSTRACT

The Hippo (Hpo) pathway is a conserved tumor suppressor pathway that controls organ size through the coordinated regulation of apoptosis and proliferation. Drosophila Salvador (Sav), which limits organ size, is a core component of the Hpo pathway. In this study, Ser-17 was shown to be important for the stability of Sav. Alanine mutation of Ser-17 promoted the proteasomal degradation of Sav. Destabilization and stabilization of the Sav protein mediated by alanine mutation of Ser-17 and by Hpo, respectively, were independent of each other. This implies that the stability of Sav is controlled by two mechanisms, one that is Ser-17-dependent and Hpo-independent, and another that is Ser-17-independent and Hpo-dependent. These dual mechanisms also regulated the human counterpart of Drosophila Sav, WW domain-containing adaptor 45 (WW45). The conservation of this regulation adds to its significance in normal physiology and tumorigenesis.


Subject(s)
Cell Cycle Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila/metabolism , Proteasome Endopeptidase Complex/metabolism , Serine/metabolism , Alanine/genetics , Alanine/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Line, Tumor , Drosophila/genetics , Drosophila Proteins/genetics , Electrophoresis, Polyacrylamide Gel , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Leupeptins/pharmacology , Mutagenesis, Site-Directed , Mutation , Proteasome Endopeptidase Complex/drug effects , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Stability , Proteolysis , Serine/genetics , Species Specificity , Transfection
18.
Proc Natl Acad Sci U S A ; 107(23): 10532-7, 2010 Jun 08.
Article in English | MEDLINE | ID: mdl-20498073

ABSTRACT

The Hippo signaling pathway regulates organ size and tissue homeostasis from Drosophila to mammals. At the core of the Hippo pathway is a kinase cascade extending from the Hippo (Hpo) tumor suppressor to the Yorkie (Yki) oncoprotein. The Hippo kinase cascade, in turn, is regulated by apical membrane-associated proteins such as the FERM domain proteins Merlin and Expanded (Ex), and the WW- and C2-domain protein Kibra. How these apical proteins are themselves regulated remains poorly understood. Here, we identify the transmembrane protein Crumbs (Crb), a determinant of epithelial apical-basal polarity in Drosophila embryos, as an upstream component of the Hippo pathway in imaginal disk growth control. Loss of Crb leads to tissue overgrowth and target gene expression characteristic of defective Hippo signaling. Crb directly binds to Ex through its juxtamembrane FERM-binding motif (FBM). Loss of Crb or mutation of its FBM leads to mislocalization of Ex to basolateral domain of imaginal disk epithelial cells. These results shed light on the mechanism of Ex regulation and provide a molecular link between apical-basal polarity and tissue growth. Furthermore, our studies implicate Crb as a putative cell surface receptor for Hippo signaling by uncovering a transmembrane protein that directly binds to an apical component of the Hippo pathway.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/metabolism , Membrane Proteins/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Animals, Genetically Modified , Cell Polarity , Conserved Sequence , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/growth & development , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Sequence Data , Mutation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
19.
Environ Eng Sci ; 30(4): 179-186, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23633893

ABSTRACT

Microcystis, a genus of potentially harmful cyanobacteria, is known to proliferate in stratified freshwaters due to its capability to change cell density and regulate buoyancy. In this study, a trajectory model was developed to simulate the cell density change and spatial distribution of Microcystis cells with nonuniform colony sizes. Simulations showed that larger colonies migrate to the near-surface water layer during the night to effectively capture irradiation and become heavy enough to sink during daytime. Smaller-sized colonies instead took a longer time to get to the surface. Simulation of the diurnally varying Microcystis population profile matched the observed pattern in the field when the radii of the multisized colonies were in a beta distribution. This modeling approach is able to take into account the history of cells by keeping track of their positions and properties, such as cell density and the sizes of colonies. It also serves as the basis for further developmental modeling of phytoplanktons that are forming colonies and changing buoyancy.

20.
FEBS Lett ; 596(13): 1628-1638, 2022 07.
Article in English | MEDLINE | ID: mdl-35278215

ABSTRACT

Hippo signalling plays key role in tissue growth and homeostasis, and its dysregulation is implicated in various human diseases. Expanded (Ex) is an important upstream activator of Hippo signalling; however, how Ex activates Hippo signalling is still poorly understood. Here, we demonstrate that Ex forms a homodimer via C-terminal interaction, and that the ExC2 region (912-1164 aa) is sufficient and essential for Ex dimerization. Functional analysis shows that ExC2 is required for Ex to promote the phosphorylation and inactivation of Yki in Drosophila cells. Further in vivo analysis shows that ExC2 is important for Ex to control Drosophila tissue growth. Our study, thus, uncovers a novel mechanism whereby Ex homodimerization mediates its full activation to promote Hippo signalling in growth control.


Subject(s)
Drosophila Proteins , Drosophila , Animals , Dimerization , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL