Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Article in English | MEDLINE | ID: mdl-30142450

ABSTRACT

Glyphosate-based formulation is used as non-selective and post-emergent herbicides in urban and rural activities. In view of its recurring applications in agricultural producing countries, the increase of glyphosate concentration in the environment stresses the need to test the adverse effects on non-target organisms and assess the risk of its use. This paper analyzes the toxicological and oxidative stress and modulatory effects of a glyphosate commercial formulation (glyphosate F) on the nematode Caenorhabditis elegans. We detected ROS production and enhancement of oxidative stress response in glyphosate F-treated nematodes. Particularly, we found an increased ctl-1 catalase gene expression of a catalase specific activity. In addition, we showed that glyphosate F treatment activated the FOXO transcription factor DAF-16, a critical target of the insulin/IGF-1 signaling pathway, which modulates the transcription of a broad range of genes involved in stress resistance, reproductive development, dauer formation, and longevity. In summary, the exposure of glyphosate F induces an oxidative imbalance in C. elegans that leads to the DAF-16 activation and consequently to the expression of genes that boost the antioxidant defense system. In this regard, clt-1 gene and catalase activity proved to be excellent biomarkers to develop more sensitive protocols to assess the environmental risk of glyphosate use.


Subject(s)
Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans/drug effects , Forkhead Transcription Factors/agonists , Gene Expression Regulation, Developmental/drug effects , Glycine/analogs & derivatives , Herbicides/toxicity , Models, Biological , Oxidative Stress/drug effects , Animals , Animals, Genetically Modified , Biomarkers/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Catalase/chemistry , Catalase/genetics , Catalase/metabolism , Dose-Response Relationship, Drug , Environmental Biomarkers/drug effects , Environmental Monitoring/methods , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Glycine/toxicity , Hormesis , Larva/drug effects , Larva/genetics , Larva/growth & development , Larva/metabolism , Mutation , Pesticide Residues/toxicity , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , Soil Pollutants/toxicity , Toxicity Tests, Acute/methods , Glyphosate
2.
PLoS One ; 13(5): e0196870, 2018.
Article in English | MEDLINE | ID: mdl-29723292

ABSTRACT

The bionematicidal effect of a synthetic volatile mixture (SVM) of four volatile organic compounds (VOCs) emitted by the endophytic fungus Daldinia cf. concentrica against the devastating plant-parasitic root-knot nematode Meloidogyne javanica has been recently demonstrated in both in vitro and greenhouse experiments. However, the mode of action governing the observed irreversible paralysis of J2 larvae upon exposure to SVM is unknown. To unravel the mechanism underlying the anthelmintic and nematicidal activities, we used the tractable model worm Caenorhabditis elegans. C. elegans was also susceptible to both the fungal VOCs and SVM. Among compounds comprising SVM, 3-methyl-1-butanol, (±)-2-methyl-1-butanol, and 4-heptanone showed significant nematicidal activity toward L1, L4 and young adult stages. Egg hatching was only negatively affected by 4-heptanone. To determine the mechanism underlying this activity, we examined the response of C. elegans mutants for glutamate-gated chloride channel and acetylcholine transporter, targets of the nematicidal drugs ivermectin and aldicarb, respectively, to 4-heptanone and SVM. These aldicarb- and ivermectin-resistant mutants retained susceptibility upon exposure to 4-heptanone and SVM. Next, we used C. elegans TJ356 strain zIs356 (daf-16::GFP+rol-6), LD1 ldIs7 [skn-1B/C::GFP + pRF4(rol-6(su1006))], LD1171 ldIs3 [gcs-1p::gfp; rol-6(su1006))], CL2166 dvIs19 (gst-4p::GFP) and CF1553 muIs84 (sod-3p::GFP+rol-6), which have mutations in genes regulating multiple stress responses. Following exposure of L4 larvae to 4-heptanone or SVM, there was clear nuclear translocation of DAF-16::GFP, and SKN-1::GFP indicating that their susceptibility involves DAF-16 and SKN1 regulation. Application of 4-heptanone, but not SVM, induced increased expression of, gcs-1::GFP and gst-4::GFP compared to controls. In contrast, application of 4-heptanone or SVM to the sod-3::GFP line elicited a significant decline in overall fluorescence intensity compared to controls, indicating SOD-3 downregulation and therefore overall reduction in cellular redox machinery. Our data indicate that the mode of action of SVM and 4-heptanone from D. cf. concentrica differs from that of currently available nematicides, potentially offering new solutions for nematode management.


Subject(s)
Anthelmintics/pharmacology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/drug effects , Forkhead Transcription Factors/genetics , Larva/drug effects , Volatile Organic Compounds/pharmacology , Xylariales/chemistry , Aldicarb/pharmacology , Animals , Anthelmintics/isolation & purification , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans Proteins/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Forkhead Transcription Factors/agonists , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Ivermectin/pharmacology , Ketones/chemistry , Ketones/pharmacology , Larva/genetics , Larva/growth & development , Larva/metabolism , Pentanols/chemistry , Pentanols/pharmacology , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Volatile Organic Compounds/isolation & purification , Xylariales/metabolism , Zygote/drug effects , Zygote/growth & development , Zygote/metabolism
3.
J Inorg Biochem ; 181: 104-110, 2018 04.
Article in English | MEDLINE | ID: mdl-29150325

ABSTRACT

Ilex paraguariensis, yerba mate is a native plant from the southern region of Brazil. Studies showed that yerba mate has an antioxidant potential, which could help to reduce the risk of developing neurodegenerative diseases, as Alzheimer's Disease (AD). It's known that I. paraguariensis grows in acid soils with aluminium (Al), which is bioavailable in these soils. Al has a neurotoxic potential related with the progression of neurological disorders. This study aim was to evaluate the potential of I. paraguariensis in the etiology of AD using strains of Caenorhabditis elegans and the concentration of Al and antioxidants in the yerba mate extract. The results of the I. paraguariensis infusions made at 65°C and at 75° C show that there was no significant difference between both temperatures when preparing the tea infusion in relation to the presence of Al, methylxanthines, phenolic compounds and flavonoids. Additionally, in the case of Al, there was no difference between the extracts prepared at both temperatures. The behavioral parameters of C. elegans were altered after a long-term exposure to both factors: I. paraguariensis extract and Al. Through the antioxidant levels results along with the Al content on the Acetylcholinesterase (AChE) activity it is possible to observe that the acute and chronic exposure to Al and I. paraguariensis leaves extract are very similar to wild-type worms. Moreover, we can observe that the results in both the transgenic strains long-term exposed to I. paraguariensis leaves extract and to the Al concentrations presented an increase in the AChE activity.


Subject(s)
Aluminum/toxicity , Alzheimer Disease/etiology , Disease Models, Animal , Food Contamination , Ilex paraguariensis/chemistry , Soil Pollutants/toxicity , Teas, Herbal/adverse effects , Acetylcholinesterase/chemistry , Acetylcholinesterase/metabolism , Aluminum/analysis , Alzheimer Disease/prevention & control , Animals , Animals, Genetically Modified , Antioxidants/analysis , Antioxidants/therapeutic use , Behavior, Animal/drug effects , Brazil , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans Proteins/metabolism , Flavonoids/analysis , Flavonoids/therapeutic use , Ilex paraguariensis/growth & development , Neurotoxicity Syndromes/physiopathology , Phenols/analysis , Phenols/therapeutic use , Plant Leaves/chemistry , Plant Leaves/growth & development , Soil Pollutants/analysis , Teas, Herbal/analysis , Toxicity Tests, Acute , Toxicity Tests, Chronic , Xanthines/analysis , Xanthines/chemistry , Xanthines/therapeutic use
4.
Br J Pharmacol ; 174(9): 781-795, 2017 05.
Article in English | MEDLINE | ID: mdl-28146602

ABSTRACT

BACKGROUND AND PURPOSE: Ionotropic GABA receptors are evolutionarily conserved proteins that mediate cellular and network inhibition in both vertebrates and invertebrates. A unique class of excitatory GABA receptors has been identified in several nematode species. Despite well-characterized functions in Caenorhabditis elegans, little is known about the pharmacology of the excitatory GABA receptors EXP-1 and LGC-35. Using a panel of compounds that differentially activate and modulate ionotropic GABA receptors, we investigated the agonist binding site and allosteric modulation of EXP-1 and LGC-35. EXPERIMENTAL APPROACH: We used two-electrode voltage clamp recordings to characterize the pharmacological profile of EXP-1 and LGC-35 receptors expressed in Xenopus laevis oocytes. KEY RESULTS: The pharmacology of EXP-1 and LGC-35 is different from that of GABAA and GABAA -ρ receptors. Both nematode receptors are resistant to the competitive orthosteric antagonist bicuculline and to classical ionotropic receptor pore blockers. The GABAA -ρ specific antagonist, TPMPA, was the only compound tested that potently inhibited EXP-1 and LGC-35. Neurosteroids have minimal effects on GABA-induced currents, but ethanol selectively potentiates LGC-35. CONCLUSIONS AND IMPLICATIONS: The pharmacological properties of EXP-1 and LGC-35 more closely resemble the ionotropic GABAA -ρ family. However, EXP-1 and LGC-35 exhibit a unique profile that differs from vertebrate GABAA and GABAA -ρ receptors, insect GABA receptors and nematode GABA receptors. As a pair, EXP-1 and LGC-35 may be utilized to further understand the differential molecular mechanisms of agonist, antagonist and allosteric modulation at ionotropic GABA receptors and may aid in the design of new and more specific anthelmintics that target GABA neurotransmission.


Subject(s)
Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans Proteins/metabolism , Cysteine Loop Ligand-Gated Ion Channel Receptors/agonists , Cysteine Loop Ligand-Gated Ion Channel Receptors/metabolism , Receptors, GABA/metabolism , Animals , Binding Sites/physiology , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Cysteine Loop Ligand-Gated Ion Channel Receptors/genetics , Dose-Response Relationship, Drug , Female , GABA Agonists/metabolism , GABA Agonists/pharmacology , Receptors, GABA/genetics , Xenopus laevis
5.
J Diabetes Complications ; 31(2): 304-310, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27776915

ABSTRACT

BACKGROUND: Glucose derived metabolism generates reactive metabolites affecting the neuronal system and lifespan in C. elegans. Here, the role of the insulin homologue ins-7 and its downstream effectors in the generation of high glucose induced neuronal damage and shortening of lifespan was studied. RESULTS: In C. elegans high glucose conditions induced the expression of the insulin homologue ins-7. Abrogating ins-7 under high glucose conditions in non-neuronal cells decreased reactive oxygen species (ROS)-formation and accumulation of methylglyoxal derived advanced glycation endproducts (AGEs), prevented structural neuronal damage and normalised head motility and lifespan. The restoration of lifespan by decreased ins-7 expression was dependent on the concerted action of sod-3 and glod-4 coding for the homologues of iron-manganese superoxide dismutase and glyoxalase 1, respectively. CONCLUSIONS: Under high glucose conditions mitochondria-mediated oxidative stress and glycation are downstream targets of ins-7. This impairs the neuronal system and longevity via a non-neuronal/neuronal crosstalk by affecting sod-3 and glod-4, thus giving further insight into the pathophysiology of diabetic complications.


Subject(s)
Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Gene Expression Regulation, Developmental , Glucose/poisoning , Lactoylglutathione Lyase/metabolism , Oxidative Stress , Peptide Hormones/antagonists & inhibitors , Superoxide Dismutase/metabolism , Animals , Behavior, Animal , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/growth & development , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans Proteins/genetics , Feedback, Physiological , Gene Knockdown Techniques , Gene Knockout Techniques , Glycation End Products, Advanced/metabolism , Lactoylglutathione Lyase/antagonists & inhibitors , Lactoylglutathione Lyase/genetics , Longevity , Mutation , Neuroprotection , Osmolar Concentration , Peptide Hormones/agonists , Peptide Hormones/genetics , Peptide Hormones/metabolism , RNA Interference , Reactive Oxygen Species/metabolism , Superoxide Dismutase/antagonists & inhibitors , Superoxide Dismutase/genetics , Survival Analysis
6.
Nat Commun ; 7: 13135, 2016 10 12.
Article in English | MEDLINE | ID: mdl-27731314

ABSTRACT

TRPV ion channels are directly activated by sensory stimuli and participate in thermo-, mechano- and chemo-sensation. They are also hypothesized to respond to endogenous agonists that would modulate sensory responses. Here, we show that the nicotinamide (NAM) form of vitamin B3 is an agonist of a Caenorhabditis elegans TRPV channel. Using heterologous expression in Xenopus oocytes, we demonstrate that NAM is a soluble agonist for a channel consisting of the well-studied OSM-9 TRPV subunit and relatively uncharacterized OCR-4 TRPV subunit as well as the orthologous Drosophila Nan-Iav TRPV channel, and we examine stoichiometry of subunit assembly. Finally, we show that behaviours mediated by these C. elegans and Drosophila channels are responsive to NAM, suggesting conservation of activity of this soluble endogenous metabolite on TRPV activity. Our results in combination with the role of NAM in NAD+ metabolism suggest an intriguing link between metabolic regulation and TRPV channel activity.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Nerve Tissue Proteins/genetics , Niacinamide/pharmacology , Protein Subunits/genetics , TRPV Cation Channels/genetics , Animals , Animals, Genetically Modified , Behavior, Animal/drug effects , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans Proteins/metabolism , Conserved Sequence , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Expression , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/metabolism , Niacinamide/metabolism , Oocytes/cytology , Oocytes/drug effects , Oocytes/metabolism , Patch-Clamp Techniques , Protein Subunits/agonists , Protein Subunits/metabolism , Sensation/drug effects , Sensation/physiology , TRPV Cation Channels/agonists , TRPV Cation Channels/metabolism , Xenopus laevis
7.
J Neurosci ; 36(20): 5498-508, 2016 05 18.
Article in English | MEDLINE | ID: mdl-27194330

ABSTRACT

UNLABELLED: The ability to detect noxious stimuli, process the nociceptive signal, and elicit an appropriate behavioral response is essential for survival. In Caenorhabditis elegans, opioid receptor agonists, such as morphine, mimic serotonin, and suppress the overall withdrawal from noxious stimuli through a pathway requiring the opioid-like receptor, NPR-17. This serotonin- or morphine-dependent modulation can be rescued in npr-17-null animals by the expression of npr-17 or a human κ opioid receptor in the two ASI sensory neurons, with ASI opioid signaling selectively inhibiting ASI neuropeptide release. Serotonergic modulation requires peptides encoded by both nlp-3 and nlp-24, and either nlp-3 or nlp-24 overexpression mimics morphine and suppresses withdrawal. Peptides encoded by nlp-3 act differentially, with only NLP-3.3 mimicking morphine, whereas other nlp-3 peptides antagonize NLP-3.3 modulation. Together, these results demonstrate that opiates modulate nociception in Caenorhabditis elegans through a complex monoaminergic/peptidergic cascade, and suggest that this model may be useful for dissecting opiate signaling in mammals. SIGNIFICANCE STATEMENT: Opiates are used extensively to treat chronic pain. In Caenorhabditis elegans, opioid receptor agonists suppress the overall withdrawal from noxious chemical stimuli through a pathway requiring an opioid-like receptor and two distinct neuropeptide-encoding genes, with individual peptides from the same gene functioning antagonistically to modulate nociception. Endogenous opioid signaling functions as part of a complex, monoaminergic/peptidergic signaling cascade and appears to selectively inhibit neuropeptide release, mediated by a α-adrenergic-like receptor, from two sensory neurons. Importantly, receptor null animals can be rescued by the expression of the human κ opioid receptor, and injection of human opioid receptor ligands mimics exogenous opiates, highlighting the utility of this model for dissecting opiate signaling in mammals.


Subject(s)
Biogenic Monoamines/metabolism , Caenorhabditis elegans/metabolism , Neuropeptides/metabolism , Nociception , Opiate Alkaloids/pharmacology , Receptors, Opioid/metabolism , Animals , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Receptors, Opioid/agonists , Receptors, Opioid/genetics , Sensory Receptor Cells/drug effects , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Signal Transduction
8.
Mol Cells ; 39(2): 163-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26743903

ABSTRACT

Caffeine has both positive and negative effects on physiological functions in a dose-dependent manner. C. elegans has been used as an animal model to investigate the effects of caffeine on development. Caffeine treatment at a high dose (30 mM) showed detrimental effects and caused early larval arrest. We performed a comparative proteomic analysis to investigate the mode of action of high-dose caffeine treatment in C. elegans and found that the stress response proteins, heat shock protein (HSP)-4 (endoplasmic reticulum [ER] chaperone), HSP-6 (mitochondrial chaperone), and HSP-16 (cytosolic chaperone), were induced and their expression was regulated at the transcriptional level. These findings suggest that high-dose caffeine intake causes a strong stress response and activates all three stress-response pathways in the worms, including the ER-, mitochondrial-, and cytosolic pathways. RNA interference of each hsp gene or in triple combination retarded growth. In addition, caffeine treatment stimulated a food-avoidance behavior (aversion phenotype), which was enhanced by RNAi depletion of the hsp-4 gene. Therefore, up-regulation of hsp genes after caffeine treatment appeared to be the major responses to alleviate stress and protect against developmental arrest.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/drug effects , Caffeine/pharmacology , Gene Expression Regulation, Developmental/drug effects , Heat-Shock Proteins/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/metabolism , Dose-Response Relationship, Drug , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Heat-Shock Proteins/agonists , Heat-Shock Proteins/antagonists & inhibitors , Heat-Shock Proteins/metabolism , Larva/drug effects , Larva/genetics , Larva/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Stress, Physiological , Transcription, Genetic
9.
Biosci Biotechnol Biochem ; 79(10): 1676-83, 2015.
Article in English | MEDLINE | ID: mdl-26027643

ABSTRACT

Echinacoside (ECH), a natural polyphenolic compound, has been reported to possess important pharmacological activities. However, very little is known about whether or how ECH affects longevity in vivo. We have examined the effects of ECH on the life span and stress tolerance in Caenorhabditis elegans. Our studies demonstrate that the life span of wild-type worms could be extended in the presence of ECH. Furthermore, ECH was found to increase tolerance of worms to heat shock and oxidative stress, while not exerting any influence on pharyngeal pumping rate and progeny production. Our mechanistic studies indicate that supplementation of ECH increases the transcript level of daf-16. ECH treatment also modulates the nuclear localization and transcriptional activities of daf-16, thus fine tunes the expression of daf-16 target genes to promote longevity and increases stress response in C. elegans. Overall, this work reveals the longevity effect of ECH and elucidates the underpinning mechanisms.


Subject(s)
Antioxidants/pharmacology , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans/drug effects , Forkhead Transcription Factors/agonists , Glycosides/pharmacology , Longevity/drug effects , Protective Agents/pharmacology , RNA, Messenger/agonists , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Dose-Response Relationship, Drug , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Heat-Shock Response/drug effects , Hot Temperature , Larva/drug effects , Larva/genetics , Larva/growth & development , Larva/metabolism , Longevity/genetics , Molecular Structure , Oxidative Stress/drug effects , Paraquat/antagonists & inhibitors , Paraquat/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproduction , Transcription, Genetic
10.
PLoS Pathog ; 11(4): e1004794, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25928899

ABSTRACT

Monoamines, such as 5-HT and tyramine (TA), paralyze both free-living and parasitic nematodes when applied exogenously and serotonergic agonists have been used to clear Haemonchus contortus infections in vivo. Since nematode cell lines are not available and animal screening options are limited, we have developed a screening platform to identify monoamine receptor agonists. Key receptors were expressed heterologously in chimeric, genetically-engineered Caenorhabditis elegans, at sites likely to yield robust phenotypes upon agonist stimulation. This approach potentially preserves the unique pharmacologies of the receptors, while including nematode-specific accessory proteins and the nematode cuticle. Importantly, the sensitivity of monoamine-dependent paralysis could be increased dramatically by hypotonic incubation or the use of bus mutants with increased cuticular permeabilities. We have demonstrated that the monoamine-dependent inhibition of key interneurons, cholinergic motor neurons or body wall muscle inhibited locomotion and caused paralysis. Specifically, 5-HT paralyzed C. elegans 5-HT receptor null animals expressing either nematode, insect or human orthologues of a key Gαo-coupled 5-HT1-like receptor in the cholinergic motor neurons. Importantly, 8-OH-DPAT and PAPP, 5-HT receptor agonists, differentially paralyzed the transgenic animals, with 8-OH-DPAT paralyzing mutant animals expressing the human receptor at concentrations well below those affecting its C. elegans or insect orthologues. Similarly, 5-HT and TA paralyzed C. elegans 5-HT or TA receptor null animals, respectively, expressing either C. elegans or H. contortus 5-HT or TA-gated Cl- channels in either C. elegans cholinergic motor neurons or body wall muscles. Together, these data suggest that this heterologous, ectopic expression screening approach will be useful for the identification of agonists for key monoamine receptors from parasites and could have broad application for the identification of ligands for a host of potential anthelmintic targets.


Subject(s)
Animals, Genetically Modified/metabolism , Anthelmintics/pharmacology , Behavior, Animal/drug effects , Caenorhabditis elegans/drug effects , Chloride Channel Agonists/pharmacology , Drug Discovery/methods , Serotonin 5-HT1 Receptor Agonists/pharmacology , Animals , Animals, Genetically Modified/genetics , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Drosophila Proteins/agonists , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , GTP-Binding Protein alpha Subunits/chemistry , GTP-Binding Protein alpha Subunits/genetics , GTP-Binding Protein alpha Subunits/metabolism , Haemonchus , Helminth Proteins/agonists , Helminth Proteins/genetics , Helminth Proteins/metabolism , Humans , Hypotonic Solutions/toxicity , Interneurons/drug effects , Interneurons/metabolism , Motor Activity/drug effects , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, Biogenic Amine/agonists , Receptors, Biogenic Amine/genetics , Receptors, Biogenic Amine/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
11.
Mar Drugs ; 13(4): 2250-66, 2015 Apr 14.
Article in English | MEDLINE | ID: mdl-25874922

ABSTRACT

Parkinson's disease (PD) is the second most common neurodegenerative disorder in the elderly people, currently with no cure. Its mechanisms are not well understood, thus studies targeting cause-directed therapy or prevention are needed. This study uses the transgenic Caenorhabditis elegans PD model. We demonstrated that dietary supplementation of the worms with an extract from the cultivated red seaweed Chondrus crispus decreased the accumulation of α-synulein and protected the worms from the neuronal toxin-, 6-OHDA, induced dopaminergic neurodegeneration. These effects were associated with a corrected slowness of movement. We also showed that the enhancement of oxidative stress tolerance and an up-regulation of the stress response genes, sod-3 and skn-1, may have served as the molecular mechanism for the C. crispus-extract-mediated protection against PD pathology. Altogether, apart from its potential as a functional food, the tested red seaweed, C. crispus, might find promising pharmaceutical applications for the development of potential novel anti-neurodegenerative drugs for humans.


Subject(s)
Chondrus/chemistry , Dietary Supplements , Neuroprotective Agents/therapeutic use , Parkinson Disease/diet therapy , Plant Extracts/therapeutic use , Seaweed/chemistry , alpha-Synuclein/antagonists & inhibitors , Animals , Animals, Genetically Modified , Aquaculture , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Behavior, Animal/drug effects , Caenorhabditis elegans , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Chondrus/growth & development , Disease Models, Animal , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Movement/drug effects , Neuroprotective Agents/administration & dosage , Neurotoxins/antagonists & inhibitors , Neurotoxins/toxicity , Oxidative Stress/drug effects , Parkinson Disease/metabolism , Parkinson Disease/pathology , Plant Extracts/administration & dosage , Recombinant Fusion Proteins/metabolism , Seaweed/growth & development , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
12.
PLoS One ; 9(12): e113893, 2014.
Article in English | MEDLINE | ID: mdl-25474681

ABSTRACT

The C. elegans pharyngeal neuron M4 is a multi-functional cell that acts as a cholinergic motor neuron to stimulate peristaltic pharyngeal muscle contraction and as a neuroendocrine cell secreting neuropeptides and growth factors to affect other cells both inside and outside the pharynx. The conserved transcription factors ZAG-1 and CEH-28 are co-expressed in M4 through most of development, and here we examine how these factors contribute to M4 differentiation. We find ZAG-1 functions upstream of CEH-28 in a branched pathway to activate expression of different sets of M4 differentiation markers. CEH-28 activates expression of the growth factor genes dbl-1 and egl-17, and the neuropeptide genes flp-5 and flp-2, while ZAG-1 activates expression of the serotonin receptor ser-7, as well as expression of ceh-28 and its downstream targets. Other markers of M4 differentiation are expressed normally in both zag-1 and ceh-28 mutants, including the neuropeptide gene flp-21 and the acetylcholine biosynthetic gene unc-17. Unlike ceh-28 mutants, zag-1 mutants completely lack peristaltic muscle contractions resulting from broader defects in M4 differentiation. Despite these defects, neither ZAG-1 nor CEH-28 are terminal selectors of the M4 phenotype, and we suggest they function in a hierarchy to regulate different aspects of M4 differentiation.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/cytology , Cell Differentiation , Homeodomain Proteins/metabolism , Neurons/cytology , Repressor Proteins/metabolism , Animals , Arecoline/pharmacology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans Proteins/genetics , Feeding Behavior/drug effects , Homeodomain Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Larva/drug effects , Larva/metabolism , Mutation , Neurons/metabolism , Neuropeptides/metabolism , Peristalsis/drug effects , Promoter Regions, Genetic , Repressor Proteins/genetics , Serotonin/pharmacology , Serotonin Receptor Agonists/pharmacology , Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Vesicular Acetylcholine Transport Proteins/metabolism
13.
Biochem J ; 461(3): 509-20, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24854345

ABSTRACT

Lysosomes act as terminal degradation organelles to hydrolyse macromolecules derived from both the extracellular space and the cytoplasm. In Caenorhabditis elegans fasting induces the lysosomal compartment to expand. However, the molecular and cellular mechanisms for this stress response remain largely unclear. In the present study, we find that short-term fasting leads to increased accumulation of polar lipids in lysosomes. The fasting response is co-ordinately regulated by EGL-4, the C. elegans PKG (protein kinase G) orthologue, and nuclear hormone receptor NHR-49. Further results demonstrate that EGL-4 acts in sensory neurons to enhance lysosomal lipid accumulation through inhibiting the DAF-3/SMAD pathway, whereas NHR-49 acts in intestine to inhibit lipids accumulation via activation of IPLA-2 (intracellular membrane-associated calcium-independent phospholipase A2) in cytoplasm and other hydrolases in lysosomes. Remarkably, the lysosomal lipid accumulation is independent of autophagy and RAB-7-mediated endocytosis. Taken together, our results reveal a new mechanism for lysosomal lipid metabolism during the stress response, which may provide new clues for investigations of lysosome function in energy homoeostasis.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/physiology , Cyclic GMP-Dependent Protein Kinases/metabolism , Lipid Metabolism , Lysosomes/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Stress, Physiological , Animals , Animals, Genetically Modified , Caenorhabditis elegans/enzymology , Caenorhabditis elegans/genetics , Caenorhabditis elegans/ultrastructure , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Cyclic GMP-Dependent Protein Kinases/genetics , Enzyme Activation , Fasting/adverse effects , Hydrolases/chemistry , Hydrolases/genetics , Hydrolases/metabolism , Kinetics , Lysosomes/ultrastructure , Mutation , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phospholipases A2, Calcium-Independent/chemistry , Phospholipases A2, Calcium-Independent/genetics , Phospholipases A2, Calcium-Independent/metabolism , RNA Interference , Receptors, Cytoplasmic and Nuclear/genetics , Sensory Receptor Cells/enzymology , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/ultrastructure , Smad Proteins/antagonists & inhibitors , Smad Proteins/genetics , Smad Proteins/metabolism , Up-Regulation
14.
Neurochem Res ; 39(4): 777-84, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24604007

ABSTRACT

Many neurotransmitter receptors are known to interact with a variety of intracellular proteins that modulate signaling processes. In an effort to understand the molecular mechanism by which acetylcholine (ACh) signaling is modulated, we searched for proteins that interact with GAR-3, the Caenorhabditis elegans homolog of muscarinic ACh receptors. We isolated two proteins, VIG-1 and FRM-1, in a yeast two-hybrid screen of a C. elegans cDNA library using the third intracellular (i3) loop of GAR-3 as bait. To test whether these proteins regulate ACh signaling, we utilized Chinese hamster ovary (CHO) cells stably expressing GAR-3 (GAR-3/CHO cells). Previously we have shown that the cholinergic agonist carbachol stimulates extracellular signal-regulated kinases 1 and 2 (ERK1/2) activation in an atropine-sensitive manner in this cell line. When VIG-1 was transiently expressed in GAR-3/CHO cells, carbachol-stimulated ERK1/2 activation was substantially reduced. In contrast, transient expression of FRM-1 significantly enhanced carbachol-stimulated ERK1/2 activation. Neither VIG-1 nor FRM-1 expression appeared to alter the affinity between GAR-3 and carbachol. In support of this notion, expression of these proteins did not affect GAR-3-mediated phospholipase C activation. To verify the modulation of ERK1/2 activity by VIG-1 and FRM-1, we used an i3 loop deletion mutant of GAR-3 (termed GAR-3Δi3). Carbachol treatment evoked robust ERK1/2 activation in CHO cells stably expressing the deletion mutant (GAR-3Δi3/CHO cells). However, transient expression of either VIG-1 or FRM-1 had little effect on carbachol-stimulated ERK1/2 activation in GAR-3Δi3/CHO cells. Taken together, these results indicate that VIG-1 and FRM-1 regulate GAR-3-mediated ERK1/2 activation by interacting with the i3 loop of GAR-3.


Subject(s)
Caenorhabditis elegans Proteins/biosynthesis , Caenorhabditis elegans Proteins/physiology , Carbachol/pharmacology , MAP Kinase Signaling System/physiology , Receptors, Muscarinic/biosynthesis , Ribonucleoproteins/physiology , Animals , CHO Cells , Caenorhabditis elegans , Caenorhabditis elegans Proteins/agonists , Cholinergic Agonists/pharmacology , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/physiology , Gene Expression Regulation , MAP Kinase Signaling System/drug effects
15.
J Neurosci ; 34(6): 2155-9, 2014 Feb 05.
Article in English | MEDLINE | ID: mdl-24501356

ABSTRACT

Propofol is an intravenous general anesthetic that alters neuronal excitability by modulating agonist responses of pentameric ligand-gated ion channels (pLGICs). Evidence suggests that propofol enhancement of anion-selective pLGICs is mediated by a binding site between adjacent subunits, whereas propofol inhibition of cation-selective pLGICs occurs via a binding site contained within helices M1-M4 of individual subunits. We considered this idea by testing propofol modulation of homomeric human glycine receptors (GlyRs) and nematode glutamate-gated chloride channels (GluCls) recombinantly expressed in Xenopus laevis oocytes with electrophysiology. The Haemonchus contortus AVR-14B GluCl was inhibited by propofol with an IC50 value of 252 ± 48 µM, providing the first example of propofol inhibition of an anion-selective pLGIC. Remarkably, inhibition was converted to enhancement by a single I18'S substitution in the channel-forming M2 helix (EC50 = 979 ± 88 µM). When a previously identified site between adjacent subunits was disrupted by the M3 G329I substitution, both propofol inhibition and enhancement of GluCls were severely impaired (IC50 and EC50 values could not be calculated). Similarly, when the equivalent positions were examined in GlyRs, the M2 S18'I substitution significantly altered the maximum level of enhancement by propofol, and the M3 A288I substitution abolished propofol enhancement. These data are not consistent with separate binding sites for the opposing effects of propofol. Instead, these data suggest that propofol enhancement and inhibition are mediated by binding to a single site in anion-selective pLGICs, and the modulatory effect on channel gating depends on the M2 18' residue.


Subject(s)
Anesthetics, Intravenous/metabolism , Ligand-Gated Ion Channels/chemistry , Ligand-Gated Ion Channels/metabolism , Propofol/metabolism , Anesthetics, Intravenous/pharmacology , Animals , Binding Sites/physiology , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , Crystallization , Female , Humans , Ligand-Gated Ion Channels/agonists , Propofol/pharmacology , Protein Structure, Secondary , Protein Structure, Tertiary , Xenopus laevis
16.
PLoS One ; 9(1): e85305, 2014.
Article in English | MEDLINE | ID: mdl-24416384

ABSTRACT

BACKGROUND: Parkinson's disease (PD) is the second most common degenerative disorder of the central nervous system that impairs motor skills and cognitive function. To date, the disease has no effective therapies. The identification of new drugs that provide benefit in arresting the decline seen in PD patients is the focus of much recent study. However, the lengthy time frame for the progression of neurodegeneration in PD increases both the time and cost of examining potential therapeutic compounds in mammalian models. An alternative is to first evaluate the efficacy of compounds in Caenorhabditis elegans models, which reduces examination time from months to days. n-Butylidenephthalide is the naturally-occurring component derived from the chloroform extract of Angelica sinensis. It has been shown to have anti-tumor and anti-inflammatory properties, but no reports have yet described the effects of n-butylidenephthalide on PD. The aim of this study was to assess the potential for n-butylidenephthalide to improve PD in C. elegans models. METHODOLOGY/PRINCIPAL FINDINGS: In the current study, we employed a pharmacological strain that expresses green fluorescent protein specifically in dopaminergic neurons (BZ555) and a transgenic strain that expresses human α-synuclein in muscle cells (OW13) to investigate the antiparkinsonian activities of n-butylidenephthalide. Our results demonstrate that in PD animal models, n-butylidenephthalide significantly attenuates dopaminergic neuron degeneration induced by 6-hydroxydopamine; reduces α-synuclein accumulation; recovers lipid content, food-sensing behavior, and dopamine levels; and prolongs life-span of 6-hydroxydopamine treatment, thus revealing its potential as a possible antiparkinsonian drug. n-Butylidenephthalide may exert its effects by blocking egl-1 expression to inhibit apoptosis pathways and by raising rpn-6 expression to enhance the activity of proteasomes. CONCLUSIONS/SIGNIFICANCE: n-Butylidenephthalide may be one of the effective neuroprotective agents for PD.


Subject(s)
Angelica sinensis/chemistry , Antiparkinson Agents/pharmacology , Caenorhabditis elegans/drug effects , Dopaminergic Neurons/drug effects , Phthalic Anhydrides/pharmacology , alpha-Synuclein/antagonists & inhibitors , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Drugs, Chinese Herbal/chemistry , Gene Expression Regulation , Humans , Longevity/drug effects , Muscle Cells/drug effects , Muscle Cells/metabolism , Muscle Cells/pathology , Oxidopamine/pharmacology , Parkinson Disease/drug therapy , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/pathology , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transgenes , alpha-Synuclein/biosynthesis , alpha-Synuclein/genetics
17.
Oncogene ; 33(29): 3878-85, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-23995781

ABSTRACT

Insulin and insulin-like growth factor-1 signaling have fundamental roles in energy metabolism, growth and development. Recent research suggests hyperactive insulin receptor (IR) and hyperinsulinemia are cancer risk factors. However, the mechanisms that account for the link between the hyperactive insulin signaling and cancer risk are not well understood. Here we show that an insulin-like signaling inhibits the DAF-18/(phosphatase and tensin homolog) PTEN tumour suppressor in Caenorhabditis elegans and that this regulation is conserved in human breast cancer cells. We show that inhibiting the IR increases PTEN protein levels, while increasing insulin signaling decreases PTEN protein levels. Our results show that the kinase region of IRß subunit physically binds to PTEN and phosphorylates on Y27 and Y174. Our genetic results also show that DAF-2/IR negatively regulates DAF-18/PTEN during C. elegans axon guidance. As PTEN is an important tumour suppressor, our results therefore suggest a possible mechanism for increased cancer risk observed in hyperinsulinemia and hyperactive IR individuals.


Subject(s)
Insulin/metabolism , PTEN Phosphohydrolase/metabolism , Receptor, Insulin/metabolism , Animals , Animals, Genetically Modified , Axons/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Cell Line , Gene Expression , Humans , Insulin/pharmacology , Insulins , Intestinal Mucosa/metabolism , Models, Biological , Mutation , Neurons/metabolism , PTEN Phosphohydrolase/genetics , Protein Binding , Receptor, Insulin/agonists , Receptor, Insulin/genetics
18.
Curr Aging Sci ; 6(3): 220-4, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24304198

ABSTRACT

Vitamin D is an essential micronutrient, necessary for human health. To determine if Caenorhabditis elegans (C. elegans) could function as an effective model to study the mechanisms of action of vitamin D, we asked if vitamin D3 affects C. elegans lifespan. Multiple factors positively impact lifespan in this system including dietary restriction and vitamin E. In addition, the C. elegans DAF-12 nuclear hormone receptor is homologous to the vitamin D receptor in humans and is therefore a candidate for a functional vitamin D receptor. It was hypothesized that vitamin D3 supplementation would increase the lifespan of C. elegans in a DAF-12-dependent manner. Dose-response curves were completed, and results indicate that exposure to 1,000 µg/ml vitamin D3 significantly increased the lifespan of wild-type worms by up to 39% (p<0.001). The daf-12 mutants exposed to 1,000 µg/ml vitamin D3 lived significantly longer than daf-12 controls exposed to 0 µg/ml (p<0.001), but among worms exposed to 1,000 µg/ml vitamin D3, wild type lived significantly longer than daf-12 (p<0.01). The data suggest that vitamin D3 can interact with multiple receptors, possibly implicating the NHR family of nuclear hormone receptors related to DAF-12. This research is the first to our knowledge to utilize C. elegans as a model to study the impact of vitamin D3 on longevity and supports the use of this model system to increase our understanding of vitamin D function at the cellular level, its role in cellular health, and its potential medicinal utility in humans.


Subject(s)
Aging/metabolism , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans/drug effects , Cholecalciferol/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , Aging/genetics , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Dose-Response Relationship, Drug , Genotype , Longevity , Models, Animal , Mutation , Phenotype , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Time Factors
19.
Science ; 338(6106): 540-3, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-23112335

ABSTRACT

Many biological functions are conserved, but the extent to which conservation applies to integrative behaviors is unknown. Vasopressin and oxytocin neuropeptides are strongly implicated in mammalian reproductive and social behaviors, yet rodent loss-of-function mutants have relatively subtle behavioral defects. Here we identify an oxytocin/vasopressin-like signaling system in Caenorhabditis elegans, consisting of a peptide and two receptors that are expressed in sexually dimorphic patterns. Males lacking the peptide or its receptors perform poorly in reproductive behaviors, including mate search, mate recognition, and mating, but other sensorimotor behaviors are intact. Quantitative analysis indicates that mating motor patterns are fragmented and inefficient in mutants, suggesting that oxytocin/vasopressin peptides increase the coherence of mating behaviors. These results indicate that conserved molecules coordinate diverse behavioral motifs in reproductive behavior.


Subject(s)
Biological Evolution , Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Neuropeptides/physiology , Oxytocin/physiology , Receptors, G-Protein-Coupled/physiology , Sexual Behavior, Animal/physiology , Vasopressins/physiology , Amino Acid Sequence , Animals , CHO Cells , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/pharmacology , Cricetinae , Humans , Male , Neuropeptides/chemistry , Neuropeptides/genetics , Neuropeptides/pharmacology , Oxytocin/chemistry , Oxytocin/genetics , Oxytocin/pharmacology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Reproduction , Vasopressins/chemistry , Vasopressins/genetics , Vasopressins/pharmacology
20.
Science ; 338(6106): 543-5, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-23112336

ABSTRACT

Vasopressin- and oxytocin-related neuropeptides are key regulators of animal physiology, including water balance and reproduction. Although these neuropeptides also modulate social behavior and cognition in mammals, the mechanism for influencing behavioral plasticity and the evolutionary origin of these effects are not well understood. Here, we present a functional vasopressin- and oxytocin-like signaling system in the nematode Caenorhabditis elegans. Through activation of its receptor NTR-1, a vasopressin/oxytocin-related neuropeptide, designated nematocin, facilitates the experience-driven modulation of salt chemotaxis, a type of gustatory associative learning in C. elegans. Our study suggests that vasopressin and oxytocin neuropeptides have ancient roles in modulating sensory processing in neural circuits that underlie behavioral plasticity.


Subject(s)
Caenorhabditis elegans Proteins/physiology , Caenorhabditis elegans/physiology , Learning/physiology , Neuropeptides/physiology , Oxytocin/physiology , Receptors, G-Protein-Coupled/physiology , Taste/physiology , Vasopressins/physiology , Amino Acid Sequence , Animals , Biological Evolution , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/agonists , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/pharmacology , Learning/drug effects , Male , Molecular Sequence Data , Neuropeptides/chemistry , Neuropeptides/genetics , Neuropeptides/pharmacology , Oxytocin/chemistry , Oxytocin/genetics , Oxytocin/pharmacology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Taste/drug effects , Vasopressins/chemistry , Vasopressins/genetics , Vasopressins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL