Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
Cell ; 184(3): 655-674.e27, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33497611

ABSTRACT

Ras GTPase-activating protein-binding proteins 1 and 2 (G3BP1 and G3BP2, respectively) are widely recognized as core components of stress granules (SGs). We report that G3BPs reside at the cytoplasmic surface of lysosomes. They act in a non-redundant manner to anchor the tuberous sclerosis complex (TSC) protein complex to lysosomes and suppress activation of the metabolic master regulator mechanistic target of rapamycin complex 1 (mTORC1) by amino acids and insulin. Like the TSC complex, G3BP1 deficiency elicits phenotypes related to mTORC1 hyperactivity. In the context of tumors, low G3BP1 levels enhance mTORC1-driven breast cancer cell motility and correlate with adverse outcomes in patients. Furthermore, G3bp1 inhibition in zebrafish disturbs neuronal development and function, leading to white matter heterotopia and neuronal hyperactivity. Thus, G3BPs are not only core components of SGs but also a key element of lysosomal TSC-mTORC1 signaling.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DNA Helicases/metabolism , Lysosomes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Poly-ADP-Ribose Binding Proteins/metabolism , RNA Helicases/metabolism , RNA Recognition Motif Proteins/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction , Tuberous Sclerosis/metabolism , Amino Acid Sequence , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , DNA Helicases/chemistry , Evolution, Molecular , Female , Humans , Insulin/pharmacology , Lysosomal Membrane Proteins/metabolism , Lysosomes/drug effects , Neurons/drug effects , Neurons/metabolism , Phenotype , Poly-ADP-Ribose Binding Proteins/chemistry , RNA Helicases/chemistry , RNA Recognition Motif Proteins/chemistry , Rats, Wistar , Signal Transduction/drug effects , Zebrafish/metabolism
2.
Cell ; 154(4): 859-74, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23953116

ABSTRACT

Mammalian target of rapamycin complex 1 (mTORC1) controls growth and survival in response to metabolic cues. Oxidative stress affects mTORC1 via inhibitory and stimulatory inputs. Whereas downregulation of TSC1-TSC2 activates mTORC1 upon oxidative stress, the molecular mechanism of mTORC1 inhibition remains unknown. Here, we identify astrin as an essential negative mTORC1 regulator in the cellular stress response. Upon stress, astrin inhibits mTORC1 association and recruits the mTORC1 component raptor to stress granules (SGs), thereby preventing mTORC1-hyperactivation-induced apoptosis. In turn, balanced mTORC1 activity enables expression of stress factors. By identifying astrin as a direct molecular link between mTORC1, SG assembly, and the stress response, we establish a unifying model of mTORC1 inhibition and activation upon stress. Importantly, we show that in cancer cells, apoptosis suppression during stress depends on astrin. Being frequently upregulated in tumors, astrin is a potential clinically relevant target to sensitize tumors to apoptosis.


Subject(s)
Apoptosis , Breast Neoplasms/metabolism , Cell Cycle Proteins/metabolism , Multiprotein Complexes/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cytoplasmic Granules/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1 , Oxidative Stress , Regulatory-Associated Protein of mTOR
3.
Cell ; 132(6): 1025-38, 2008 Mar 21.
Article in English | MEDLINE | ID: mdl-18358814

ABSTRACT

Insulin/IGF-1-like signaling (IIS) is central to growth and metabolism and has a conserved role in aging. In C. elegans, reductions in IIS increase stress resistance and longevity, effects that require the IIS-inhibited FOXO protein DAF-16. The C. elegans transcription factor SKN-1 also defends against oxidative stress by mobilizing the conserved phase 2 detoxification response. Here we show that IIS not only opposes DAF-16 but also directly inhibits SKN-1 in parallel. The IIS kinases AKT-1, -2, and SGK-1 phosphorylate SKN-1, and reduced IIS leads to constitutive SKN-1 nuclear accumulation in the intestine and SKN-1 target gene activation. SKN-1 contributes to the increased stress tolerance and longevity resulting from reduced IIS and delays aging when expressed transgenically. Furthermore, SKN-1 that is constitutively active increases life span independently of DAF-16. Our findings indicate that the transcription network regulated by SKN-1 promotes longevity and is an important direct target of IIS.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/physiology , DNA-Binding Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Animals , Gene Regulatory Networks , Insulin/metabolism , Insulin-Like Growth Factor I/metabolism , Intestines , Longevity , Oxidative Stress , Phosphorylation , Receptor, Insulin/metabolism
4.
Bioessays ; 43(12): e2100195, 2021 12.
Article in English | MEDLINE | ID: mdl-34655094

ABSTRACT

In sexually reproducing organisms maintenance of germ stem cell immortality is fundamental for transmitting genetic material to future generations. While previous research has mainly considered intrinsic regulatory mechanisms in the germline, our recent study has found a direct contribution of somatic cells in preserving germline immortality via the somatically expressed endoribonuclease ENDU-2 in Caenorhabditis elegans. We have identified ENDU-2 as a secreted protein that can be taken up by the germline. Here, we discuss how ENDU-2 might uncouple its RNA-binding and RNA-cleavage activities to control gene expression via either an endoribonuclease dependent or an independent way. We also speculate on a possible functional conservation of its mammalian homologs in mediating cell-cell communication as well as its potential significance in understanding human pathogenesis such as cancer development.


Subject(s)
Caenorhabditis elegans Proteins , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Endoribonucleases/genetics , Germ Cells , Humans , Stem Cells
5.
PLoS Genet ; 15(7): e1008099, 2019 07.
Article in English | MEDLINE | ID: mdl-31339880

ABSTRACT

The length of cilia is controlled by a poorly understood mechanism that involves members of the conserved RCK kinase group, and among them, the LF4/MOK kinases. The multiciliated protist model, Tetrahymena, carries two types of cilia (oral and locomotory) and the length of the locomotory cilia is dependent on their position with the cell. In Tetrahymena, loss of an LF4/MOK ortholog, LF4A, lengthened the locomotory cilia, but also reduced their number. Without LF4A, cilia assembled faster and showed signs of increased intraflagellar transport (IFT). Consistently, overproduced LF4A shortened cilia and downregulated IFT. GFP-tagged LF4A, expressed in the native locus and imaged by total internal reflection microscopy, was enriched at the basal bodies and distributed along the shafts of cilia. Within cilia, most LF4A-GFP particles were immobile and a few either diffused or moved by IFT. We suggest that the distribution of LF4/MOK along the cilium delivers a uniform dose of inhibition to IFT trains that travel from the base to the tip. In a longer cilium, the IFT machinery may experience a higher cumulative dose of inhibition by LF4/MOK. Thus, LF4/MOK activity could be a readout of cilium length that helps to balance the rate of IFT-driven assembly with the rate of disassembly at steady state. We used a forward genetic screen to identify a CDK-related kinase, CDKR1, whose loss-of-function suppressed the shortening of cilia caused by overexpression of LF4A, by reducing its kinase activity. Loss of CDKR1 alone lengthened both the locomotory and oral cilia. CDKR1 resembles other known ciliary CDK-related kinases: LF2 of Chlamydomonas, mammalian CCRK and DYF-18 of C. elegans, in lacking the cyclin-binding motif and acting upstream of RCKs. The new genetic tools we developed here for Tetrahymena have potential for further dissection of the principles of cilia length regulation in multiciliated cells.


Subject(s)
Cilia/metabolism , Cyclin-Dependent Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Tetrahymena/cytology , Gene Expression Regulation , Locomotion , Protozoan Proteins/metabolism , Tetrahymena/metabolism , Tetrahymena/physiology
6.
PLoS Genet ; 13(5): e1006801, 2017 May.
Article in English | MEDLINE | ID: mdl-28549065

ABSTRACT

Activation of the FOXO transcription factor DAF-16 by reduced insulin/IGF signaling (IIS) is considered to be beneficial in C. elegans due to its ability to extend lifespan and to enhance stress resistance. In the germline, cell-autonomous DAF-16 activity prevents stem cell proliferation, thus acting tumor-suppressive. In contrast, hypodermal DAF-16 causes a tumorous germline phenotype characterized by hyperproliferation of the germline stem cells and rupture of the adjacent basement membrane. Here we show that cross-talk between DAF-16 and the transforming growth factor ß (TGFß)/bone morphogenic protein (BMP) signaling pathway causes germline hyperplasia and results in disruption of the basement membrane. In addition to activating MADM/NRBP/hpo-11 gene alone, DAF-16 also directly interacts with both R-SMAD proteins SMA-2 and SMA-3 in the nucleus to regulate the expression of mTORC1 pathway. Knocking-down of BMP genes or each of the four target genes in the hypodermis was sufficient to inhibit germline proliferation, indicating a cell-non-autonomously controlled regulation of stem cell proliferation by somatic tissues. We propose the existence of two antagonistic DAF-16/FOXO functions, a cell-proliferative somatic and an anti-proliferative germline activity. Whereas germline hyperplasia under reduced IIS is inhibited by DAF-16 cell-autonomously, activation of somatic DAF-16 in the presence of active IIS promotes germline proliferation and eventually induces tumor-like germline growth. In summary, our results suggest a novel pathway crosstalk of DAF-16 and TGF-ß/BMP that can modulate mTORC1 at the transcriptional level to cause stem-cell hyperproliferation. Such cell-type specific differences may help explaining why human FOXO activity is considered to be tumor-suppressive in most contexts, but may become oncogenic, e.g. in chronic and acute myeloid leukemia.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Carcinogenesis/genetics , Forkhead Transcription Factors/metabolism , Multiprotein Complexes/metabolism , Stem Cells/metabolism , TOR Serine-Threonine Kinases/metabolism , Transforming Growth Factor beta/metabolism , Animals , Caenorhabditis elegans/cytology , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Cell Membrane/metabolism , Cell Proliferation , Epidermal Cells , Epidermis/metabolism , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Mechanistic Target of Rapamycin Complex 1 , Protein Binding , Signal Transduction , Stem Cells/physiology
7.
PLoS Genet ; 12(10): e1006377, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27716778

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pgen.1003346.].

8.
BMC Biol ; 16(1): 36, 2018 03 29.
Article in English | MEDLINE | ID: mdl-29598825

ABSTRACT

BACKGROUND: Advances in automated image-based microscopy platforms coupled with high-throughput liquid workflows have facilitated the design of large-scale screens utilising multicellular model organisms such as Caenorhabditis elegans to identify genetic interactions, therapeutic drugs or disease modifiers. However, the analysis of essential genes has lagged behind because lethal or sterile mutations pose a bottleneck for high-throughput approaches, and a systematic way to analyse genetic interactions of essential genes in multicellular organisms has been lacking. RESULTS: In C. elegans, non-conditional lethal mutations can be maintained in heterozygosity using chromosome balancers, commonly expressing green fluorescent protein (GFP) in the pharynx. However, gene expression or function is typically monitored by the use of fluorescent reporters marked with the same fluorophore, presenting a challenge to sort worm populations of interest, particularly at early larval stages. Here, we develop a sorting strategy capable of selecting homozygous mutants carrying a GFP stress reporter from GFP-balanced animals at the second larval stage. Because sorting is not completely error-free, we develop an automated high-throughput image analysis protocol that identifies and discards animals carrying the chromosome balancer. We demonstrate the experimental usefulness of combining sorting of homozygous lethal mutants and automated image analysis in a functional genomic RNA interference (RNAi) screen for genes that genetically interact with mitochondrial prohibitin (PHB). Lack of PHB results in embryonic lethality, while homozygous PHB deletion mutants develop into sterile adults due to maternal contribution and strongly induce the mitochondrial unfolded protein response (UPRmt). In a chromosome-wide RNAi screen for C. elegans genes having human orthologues, we uncover both known and new PHB genetic interactors affecting the UPRmt and growth. CONCLUSIONS: The method presented here allows the study of balanced lethal mutations in a high-throughput manner. It can be easily adapted depending on the user's requirements and should serve as a useful resource for the C. elegans community for probing new biological aspects of essential nematode genes as well as the generation of more comprehensive genetic networks.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Flow Cytometry/methods , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Mitochondria/metabolism , Mutation , Prohibitins
9.
PLoS Genet ; 9(3): e1003346, 2013.
Article in English | MEDLINE | ID: mdl-23516373

ABSTRACT

Disturbance of cellular functions results in the activation of stress-signaling pathways that aim at restoring homeostasis. We performed a genome-wide screen to identify components of the signal transduction of the mitochondrial unfolded protein response (UPR(mt)) to a nuclear chaperone promoter. We used the ROS generating complex I inhibitor paraquat to induce the UPR(mt), and we employed RNAi exposure post-embryonically to allow testing genes whose knockdown results in embryonic lethality. We identified 54 novel regulators of the ROS-induced UPR(mt). Activation of the UPR(mt), but not of other stress-signaling pathways, failed when homeostasis of basic cellular mechanisms such as translation and protein transport were impaired. These mechanisms are monitored by a recently discovered surveillance system that interprets interruption of these processes as pathogen attack and depends on signaling through the JNK-like MAP-kinase KGB-1. Mutation of kgb-1 abrogated the inhibition of ROS-induced UPR(mt), suggesting that surveillance-activated defenses specifically inhibit the UPR(mt) but do not compromise activation of the heat shock response, the UPR of the endoplasmic reticulum, or the SKN-1/Nrf2 mediated response to cytosolic stress. In addition, we identified PIFK-1, the orthologue of the Drosophila PI 4-kinase four wheel drive (FWD), and found that it is the only known factor so far that is essential for the unfolded protein responses of both mitochondria and endoplasmic reticulum. This suggests that both UPRs may share a common membrane associated mechanism.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , JNK Mitogen-Activated Protein Kinases , Mitochondria , Reactive Oxygen Species , ATP-Binding Cassette Transporters/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Nucleus/genetics , Cell Nucleus/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Chaperones , Paraquat/pharmacology , Phosphorylation , Protein Folding , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/toxicity , Signal Transduction/drug effects , Transcription Factors/metabolism , Unfolded Protein Response/drug effects
10.
PLoS Genet ; 8(8): e1002836, 2012.
Article in English | MEDLINE | ID: mdl-22916022

ABSTRACT

In Caenorhabditis elegans (C. elegans), the promotion of longevity by the transcription factor DAF-16 requires reduced insulin/IGF receptor (IIR) signaling or the ablation of the germline, although the reason for the negative impact of germ cells is unknown. FOXO/DAF-16 activity inhibits germline proliferation in both daf-2 mutants and gld-1 tumors. In contrast to its function as a germline tumor suppressor, we now provide evidence that somatic DAF-16 in the presence of IIR signaling can also result in tumorigenic activity, which counteracts robust lifespan extension. In contrast to the cell-autonomous IIR signaling, which is required for larval germline proliferation, activation of DAF-16 in the hypodermis results in hyperplasia of the germline and disruption of the surrounding basement membrane. SHC-1 adaptor protein and AKT-1 kinase antagonize, whereas AKT-2 and SGK-1 kinases promote, this cell-nonautonomous DAF-16 function. Our data suggest that a functional balance of DAF-16 activities in different tissues determines longevity and reveals a novel, cell-nonautonomous role of FOXO/DAF-16 to affect stem cells.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Cell Transformation, Neoplastic/metabolism , Longevity/genetics , Signal Transduction/genetics , Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Forkhead Transcription Factors , Gene Expression Regulation, Developmental , Germ Cells/cytology , Germ Cells/metabolism , Humans , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Somatomedin/genetics , Receptors, Somatomedin/metabolism , Shc Signaling Adaptor Proteins/genetics , Shc Signaling Adaptor Proteins/metabolism , Stem Cells/cytology , Transcription Factors/genetics
11.
Hum Mol Genet ; 21(16): 3587-603, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22611162

ABSTRACT

Increased Tau protein amyloidogenicity has been causatively implicated in several neurodegenerative diseases, collectively called tauopathies. In pathological conditions, Tau becomes hyperphosphorylated and forms intracellular aggregates. The deletion of K280, which is a mutation that commonly appears in patients with frontotemporal dementia with Parkinsonism linked to chromosome 17, enhances Tau aggregation propensity (pro-aggregation). In contrast, introduction of the I277P and I308P mutations prevents ß-sheet formation and subsequent aggregation (anti-aggregation). In this study, we created a tauopathy model by expressing pro- or anti-aggregant Tau species in the nervous system of Caenorhabditis elegans. Animals expressing the highly amyloidogenic Tau species showed accelerated Tau aggregation and pathology manifested by severely impaired motility and evident neuronal dysfunction. In addition, we observed that the axonal transport of mitochondria was perturbed in these animals. Control animals expressing the anti-aggregant combination had rather mild phenotype. We subsequently tested several Tau aggregation inhibitor compounds and observed a mitigation of Tau proteotoxicity. In particular, a novel compound that crosses the blood-brain barrier of mammals proved effective in ameliorating the motility as well as delaying the accumulation of neuronal defects. Our study establishes a new C. elegans model of Tau aggregation-mediated toxicity and supports the emerging notion that inhibiting the nucleation of Tau aggregation can be neuroprotective.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Neurons/pathology , Tauopathies/etiology , tau Proteins/metabolism , Animals , Animals, Genetically Modified , Axonal Transport , Blood-Brain Barrier/drug effects , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Disease Models, Animal , Humans , Hydrazines/pharmacology , Methylene Blue/pharmacology , Mitochondria/metabolism , Motor Neurons/metabolism , Motor Neurons/pathology , Mutation , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , Phenotype , Phosphorylation , Protein Structure, Tertiary , Sensory Receptor Cells/metabolism , Tauopathies/pathology , Thiazoles/pharmacology , Vesicle-Associated Membrane Protein 1/metabolism , tau Proteins/antagonists & inhibitors , tau Proteins/genetics
12.
MicroPubl Biol ; 20232023.
Article in English | MEDLINE | ID: mdl-37799207

ABSTRACT

Maintaining iron levels is crucial for health, but iron overload has been associated with tumorigenesis. Therefore, critical enzymes involved in iron homeostasis are under tight, typically posttranslational control. In C. elegans , the mTORC2 and insulin/IGF-1 activated kinase SGK-1 is induced upon exogenous iron overload to couple iron storage and fat accumulation. Here we show that, already at physiological iron conditions, sgk-1 loss-of-function increases intracellular iron levels that may impair lifespan. Reducing iron levels by diminishing cellular or mitochondrial iron import is sufficient to extend the short lifespan of sgk-1 loss-of-function animals. Our results indicate another regulatory level of sgk-1 in iron homeostasis via negative feedback regulation on iron transporters.

13.
Cells ; 12(10)2023 05 10.
Article in English | MEDLINE | ID: mdl-37408188

ABSTRACT

Temperature changes and periods of detrimental cold occur frequently for many organisms in their natural habitats. Homeothermic animals have evolved metabolic adaptation strategies to increase mitochondrial-based energy expenditure and heat production, largely relying on fat as a fuel source. Alternatively, certain species are able to repress their metabolism during cold periods and enter a state of decreased physiological activity known as torpor. By contrast, poikilotherms, which are unable to maintain their internal temperature, predominantly increase membrane fluidity to diminish cold-related damage from low-temperature stress. However, alterations of molecular pathways and the regulation of lipid-metabolic reprogramming during cold exposure are poorly understood. Here, we review organismal responses that adjust fat metabolism during detrimental cold stress. Cold-related changes in membranes are detected by membrane-bound sensors, which signal to downstream transcriptional effectors, including nuclear hormone receptors of the PPAR (peroxisome proliferator-activated receptor) subfamily. PPARs control lipid metabolic processes, such as fatty acid desaturation, lipid catabolism and mitochondrial-based thermogenesis. Elucidating the underlying molecular mechanisms of cold adaptation may improve beneficial therapeutic cold treatments and could have important implications for medical applications of hypothermia in humans. This includes treatment strategies for hemorrhagic shock, stroke, obesity and cancer.


Subject(s)
Adaptation, Physiological , Cold Temperature , Cold-Shock Response , Lipid Metabolism , Peroxisome Proliferator-Activated Receptors , Thermogenesis , Torpor , Torpor/physiology , Animals , Peroxisome Proliferator-Activated Receptors/metabolism , Fatty Acids/metabolism , Cold-Shock Response/physiology , Membrane Fluidity , Mitochondria/metabolism
14.
Nat Commun ; 14(1): 4176, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37443152

ABSTRACT

Transient stress experiences not only trigger acute stress responses, but can also have long-lasting effects on cellular functions. In Caenorhabditis elegans, a brief exposure to heat shock during early adulthood extends lifespan and improves stress resistance, a phenomenon known as heat hormesis. Here, we investigated the prolonged effect of hormetic heat stress on the transcriptome of worms and found that the canonical heat shock response is followed by a profound transcriptional reprogramming in the post-stress period. This reprogramming relies on the endoribonuclease ENDU-2 but not the heat shock factor 1. ENDU-2 co-localizes with chromatin and interacts with RNA polymerase II, enabling specific regulation of transcription after the stress period. Failure to activate the post-stress response does not affect the resistance of animals to heat shock but eliminates the beneficial effects of hormetic heat stress. In summary, our work discovers that the RNA-binding protein ENDU-2 mediates the long-term impacts of transient heat stress via reprogramming transcriptome after stress exposure.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Transcriptome , Hormesis/physiology , Heat-Shock Response/genetics , Longevity/physiology
15.
Hum Mol Genet ; 19(14): 2804-16, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-20442138

ABSTRACT

The tyrosine kinase receptor RET51 is expressed in distinct families of neurons where it promotes different functions. FKBP52 is an immunophilin with neuroprotective effects on different kinds of neurons. In this paper, we demonstrate that RET51 activation by both glial cell line-derived neurotrophic factor (GDNF) and NGF triggers the formation of RET51/FKBP52 complex. The substitution of the tyrosine 905 of RET51, a key residue phosphorylated by both GDNF and NGF, disrupts the RET51/FKBP52 complex. NGF and GDNF have a functional role in dopaminergic (DA) neurons where RET51 and FKBP52 are expressed with a yet undefined function. To clarify if RET51/FKBP52 complex should exert its function in DA neurons, we used an indirect approach by screening the genes encoding for RET51 and FKBP52 in a group of 30 Parkinson's disease patients. The degeneration of DA neurons is the main feature of PD, which is associated to a complex multifactorial aetiology combining environmental, age-related and genetic factors. We found a compound heterozygous carrying two mutations in RET and FKBP52 that are sufficient to disrupt the RET51/FKBP52 complex, indicating its potential role in PD.


Subject(s)
Parkinson Disease/etiology , Proto-Oncogene Proteins c-ret/metabolism , Proto-Oncogene Proteins c-ret/physiology , Tacrolimus Binding Proteins/metabolism , Tacrolimus Binding Proteins/physiology , Adult , Cell Line , Female , Genetic Association Studies , Genetic Testing , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Humans , Male , Middle Aged , Models, Biological , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Multiprotein Complexes/physiology , Nerve Growth Factor/pharmacology , Parkinson Disease/genetics , Parkinson Disease/metabolism , Phosphorylation , Protein Binding/drug effects , Protein Interaction Domains and Motifs/genetics , Protein Interaction Mapping , Protein Kinases/metabolism , Proto-Oncogene Proteins c-ret/chemistry , Proto-Oncogene Proteins c-ret/genetics , Tacrolimus Binding Proteins/chemistry , Tacrolimus Binding Proteins/genetics , Young Adult
16.
Hum Mol Genet ; 19(12): 2347-59, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20233749

ABSTRACT

Mutations of the immunoglobulin superfamily proteins nephrin and Neph1 lead to congenital nephrotic syndrome in humans or mice. Neph proteins are three closely related molecules that are evolutionarily conserved and mediate cell recognition. Their importance for morphogenetic processes including the formation of the kidney filtration barrier in vertebrates and synaptogenesis in Caenorhabditis elegans has recently been uncovered. However, the individual morphogenetic function of mammalian Neph1-3 isoforms remained elusive. We demonstrate now that the Neph/nephrin family proteins can form cell-cell adhesion modules across species. Expression of all three mammalian Neph isoforms partially rescued mutant C. elegans lacking their Neph homolog syg-1 and restored synapse formation, suggesting a functional redundancy between the three isoforms. Strikingly, the rescue of defective synaptic connectivity was prevented by deletion of the highly conserved cytoplasmic PSD95/Dlg/ZO-1-binding motif of SYG-1/Neph proteins, indicating the critical role of this intracellular signaling motif for SYG-1/Neph-dependent morphogenetic events. To determine the significance of Neph isoform redundancy for vertebrate kidney development, we analyzed the expression pattern and the functional role of Neph proteins in zebrafish. In situ hybridizations identified zNeph1 and zNeph2 as glomerular proteins. Morpholino knockdown of either zNeph1 or zNeph2 resulted in loss of slit diaphragms and leakiness of the glomerular filtration barrier. This is the first report utilizing C. elegans to study mammalian Neph/nephrin protein function and to demonstrate a functional overlap of Neph1-3 proteins. Furthermore, we identify Neph2 as a novel critical regulator of glomerular function, indicating that both Neph1 and Neph2 are required for glomerular maintenance and development.


Subject(s)
Immunoglobulins/physiology , Kidney/growth & development , Membrane Proteins/physiology , Neurons/physiology , Amino Acid Sequence , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/classification , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/physiology , Cell Adhesion , HeLa Cells , Humans , Immunoglobulins/classification , Immunoglobulins/genetics , Membrane Proteins/classification , Membrane Proteins/genetics , Mice , Models, Animal , Morphogenesis/genetics , PDZ Domains , Phylogeny
17.
Hepatology ; 53(3): 875-84, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21374660

ABSTRACT

UNLABELLED: Early detection of malignant biliary tract diseases, especially cholangiocarcinoma (CC) in patients with primary sclerosing cholangitis (PSC), is very difficult and often comes too late to give the patient a therapeutic benefit. We hypothesize that bile proteomic analysis distinguishes CC from nonmalignant lesions. We used capillary electrophoresis mass spectrometry (CE-MS) to identify disease-specific peptide patterns in patients with choledocholithiasis (n = 16), PSC (n = 18), and CC (n = 16) in a training set. A model for differentiation of choledocholithiasis from PSC and CC (PSC/CC model) and another model distinguishing CC from PSC (CC model) were subsequently validated in independent cohorts (choledocholithiasis [n = 14], PSC [n = 18] and CC [n = 25]). Peptides were characterized by sequencing. Application of the PSC/CC model in the independent test cohort resulted in correct exclusion of 12/14 bile samples from patients with choledocholithiasis and identification of 40/43 patients with PSC or CC (86% specificity, 93% sensitivity). The corresponding receiver operating characteristic (ROC) analysis revealed an area under the curve (AUC) of 0.93 (95% confidence interval [CI]: 0.82-0.98, P = 0.0001). The CC model succeeded in an accurate detection of 14/18 bile samples from patients with PSC and 21/25 samples with CC (78% specificity, 84% sensitivity) in the independent cohort, resulting in an AUC value of 0.87 (95% CI: 0.73-0.95, P = 0.0001) in ROC analysis. Eight out of 10 samples of patients with CC complicating PSC were identified. CONCLUSION: Bile proteomic analysis discriminates benign conditions from CC accurately. This method may become a diagnostic tool in future as it offers a new possibility to diagnose malignant bile duct disease and thus enables efficient therapy particularly in patients with PSC.


Subject(s)
Bile/chemistry , Cholangiocarcinoma/diagnosis , Choledocholithiasis/diagnosis , Proteome/analysis , Adult , Aged , Aged, 80 and over , Cholangitis, Sclerosing/diagnosis , Electrophoresis, Capillary , Female , Gene Expression Profiling , Humans , Male , Mass Spectrometry , Middle Aged
18.
Nucleic Acids Res ; 38(6): e91, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20047968

ABSTRACT

Systematic tandem-affinity-purification (TAP) of protein complexes was tremendously successful in yeast and has changed the general concept of how we understand protein function in eukaryotic cells. The transfer of this method to other model organisms has been difficult and may require specific adaptations. We were especially interested to establish a cell-type-specific TAP system for Caenorhabditis elegans, a model animal well suited to high-throughput analysis, proteomics and systems biology. By combining the high-affinity interaction between in vivo biotinylated target-proteins and streptavidin with the usage of a newly identified epitope of the publicly shared SB1 monoclonal antibody we created a novel in vivo fluorescent tag, the SnAvi-Tag. We show the versatile application of the SnAvi-Tag in Escherichia coli, vertebrate cells and in C. elegans for tandem affinity purification of protein complexes, western blotting and also for the in vivo sub-cellular localization of labelled proteins.


Subject(s)
Multiprotein Complexes/isolation & purification , Recombinant Fusion Proteins/chemistry , Animals , Antibodies, Monoclonal/immunology , Caenorhabditis elegans Proteins/isolation & purification , Caenorhabditis elegans Proteins/metabolism , Cell Line , Epitopes/chemistry , Escherichia coli/genetics , Fluorescent Dyes , Green Fluorescent Proteins/genetics , Humans , Recombinant Fusion Proteins/analysis , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/metabolism , Vesicle-Associated Membrane Protein 1/chemistry , Vesicle-Associated Membrane Protein 1/immunology
19.
Nat Cell Biol ; 6(12): 1173-9, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15558032

ABSTRACT

Redundant gene function frequently hampers investigations of the physiological roles of mammalian proteins. This is the case for Eps8, a receptor tyrosine kinase (RTK) substrate that participates in the activation of the Rac-specific guanine nucleotide-exchange function of Sos1 (refs 2-5), thereby regulating actin remodelling by RTKs. EPS8-knockout mice, however, exhibit no evident phenotype, owing to the redundant function of three other EPS8-related genes. Here we show that in the nematode Caenorhabditis elegans, only one orthologue of the EPS8 gene exists, which gives rise to two alternatively spliced isoforms, EPS-8A and EPS-8B, differing at their carboxyl termini. In the nematode, eps-8 is essential for embryonic development. Furthermore, EPS-8A, but not EPS-8B, is specifically required for proper apical morphogenesis in the intestinal cells. This latter phenotype could be precisely correlated with a previously unknown actin barbed-end-capping activity, which is present in the C terminus of the EPS-8A isoform. Therefore, nematode genetics allowed not only the unmasking of distinct EPS-8-linked phenotypes, but also the definition of a novel function for this molecule in actin dynamics.


Subject(s)
Actins/biosynthesis , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/embryology , Carrier Proteins/metabolism , Intestines/embryology , Organogenesis/physiology , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/ultrastructure , Adaptor Proteins, Signal Transducing , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/isolation & purification , Carrier Proteins/genetics , Carrier Proteins/isolation & purification , Cytoskeletal Proteins , DNA, Complementary/analysis , DNA, Complementary/genetics , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Intestinal Mucosa/metabolism , Intestines/ultrastructure , Microscopy, Electron, Transmission , Microvilli/metabolism , Microvilli/ultrastructure , Molecular Sequence Data , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary/genetics , Proteins/genetics , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid
20.
Nat Commun ; 12(1): 1262, 2021 02 24.
Article in English | MEDLINE | ID: mdl-33627668

ABSTRACT

Multicellular organisms coordinate tissue specific responses to environmental information via both cell-autonomous and non-autonomous mechanisms. In addition to secreted ligands, recent reports implicated release of small RNAs in regulating gene expression across tissue boundaries. Here, we show that the conserved poly-U specific endoribonuclease ENDU-2 in C. elegans is secreted from the soma and taken-up by the germline to ensure germline immortality at elevated temperature. ENDU-2 binds to mature mRNAs and negatively regulates mRNA abundance both in the soma and the germline. While ENDU-2 promotes RNA decay in the soma directly via its endoribonuclease activity, ENDU-2 prevents misexpression of soma-specific genes in the germline and preserves germline immortality independent of its RNA-cleavage activity. In summary, our results suggest that the secreted RNase ENDU-2 regulates gene expression across tissue boundaries in response to temperature alterations and contributes to maintenance of stem cell immortality, probably via retaining a stem cell specific program of gene expression.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/genetics , Endoribonucleases/metabolism , Animals , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Endoribonucleases/genetics , Germ-Line Mutation/genetics , RNA Stability/genetics , RNA Stability/physiology , RNA, Messenger/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL