Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.385
Filter
1.
J Environ Sci (China) ; 147: 244-258, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39003044

ABSTRACT

4-Nitrophenol (4-NP), as a toxic and refractory pollutant, has generated significant concern due to its adverse effects. However, the potential toxic effects and mechanism remained unclear. In this study, the reproduction, development, locomotion and reactive oxygen species (ROS) production of Caenorhabditis elegans were investigated to evaluate the 4-NP toxicity. We used metabolomics to assess the potential damage mechanisms. The role of metabolites in mediating the relationship between 4-NP and phenotypes was examined by correlation and mediation analysis. 4-NP (8 ng/L and 8 µg/L) caused significant reduction of brood size, ovulation rate, total germ cells numbers, head thrashes and body bends, and an increase in ROS. However, the oosperm numbers in uterus, body length and body width were decreased in 8 µg/L. Moreover, 36 differential metabolites were enriched in the significant metabolic pathways, including lysine biosynthesis, ß-alanine metabolism, tryptophan metabolism, pentose phosphate pathway, pentose and glucuronate interconversions, amino sugar and nucleotide sugar metabolism, starch and sucrose metabolism, galactose metabolism, propanoate metabolism, glycerolipid metabolism, and estrogen signaling pathway. The mechanism of 4-NP toxicity was that oxidative stress caused by the perturbation of amino acid, which had effects on energy metabolism through disturbing carbohydrate and lipid metabolism, and finally affected the estrogen signaling pathway to exert toxic effects. Moreover, correlation and mediation analysis showed glycerol-3P, glucosamine-6P, glucosamine-1P, UDP-galactose, L-aspartic acid, and uracil were potential markers for the reproduction and glucose-1,6P2 for developmental toxicity. The results provided insight into the pathways involved in the toxic effects caused by 4-NP and developed potential biomarkers to evaluate 4-NP toxicity.


Subject(s)
Caenorhabditis elegans , Estrogens , Nitrophenols , Reproduction , Signal Transduction , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/physiology , Reproduction/drug effects , Signal Transduction/drug effects , Nitrophenols/toxicity , Estrogens/toxicity , Reactive Oxygen Species/metabolism , Oxidative Stress/drug effects
2.
Nat Commun ; 15(1): 6594, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39097618

ABSTRACT

Neurons coordinate inter-tissue protein homeostasis to systemically manage cytotoxic stress. In response to neuronal mitochondrial stress, specific neuronal signals coordinate the systemic mitochondrial unfolded protein response (UPRmt) to promote organismal survival. Yet, whether chemical neurotransmitters are sufficient to control the UPRmt in physiological conditions is not well understood. Here, we show that gamma-aminobutyric acid (GABA) inhibits, and acetylcholine (ACh) promotes the UPRmt in the Caenorhabditis elegans intestine. GABA controls the UPRmt by regulating extra-synaptic ACh release through metabotropic GABAB receptors GBB-1/2. We find that elevated ACh levels in animals that are GABA-deficient or lack ACh-degradative enzymes induce the UPRmt through ACR-11, an intestinal nicotinic α7 receptor. This neuro-intestinal circuit is critical for non-autonomously regulating organismal survival of oxidative stress. These findings establish chemical neurotransmission as a crucial regulatory layer for nervous system control of systemic protein homeostasis and stress responses.


Subject(s)
Acetylcholine , Caenorhabditis elegans Proteins , Caenorhabditis elegans , Mitochondria , Oxidative Stress , Signal Transduction , Unfolded Protein Response , gamma-Aminobutyric Acid , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Acetylcholine/metabolism , Mitochondria/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , gamma-Aminobutyric Acid/metabolism , Intestines/physiology , Stress, Physiological , Neurons/metabolism , Receptors, GABA-B/metabolism , Receptors, GABA-B/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism , alpha7 Nicotinic Acetylcholine Receptor/genetics , Synaptic Transmission/physiology
3.
Curr Biol ; 34(14): R693-R696, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39043144

ABSTRACT

Cells experience dynamic internal and external forces during animal development. Two new studies reveal critical and unexpected roles for cytoskeletal regulators and nuclear positioning in maintaining the physical integrity of migrating leader cells during Caenorhabditis elegans organogenesis.


Subject(s)
Caenorhabditis elegans , Cell Movement , Organogenesis , Animals , Caenorhabditis elegans/physiology , Cell Movement/physiology , Organogenesis/physiology , Cytoskeleton/physiology , Cytoskeleton/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics
4.
Curr Biol ; 34(14): R682-R684, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39043140

ABSTRACT

A new analysis of cytokinetic furrow ingression in the Caenorhabditis elegans zygote at high spatiotemporal resolution demonstrates that, rather than being a process of steady, spatially uniform constriction, furrow ingression is modulated by complex contractile oscillations that move around the furrow, possibly in the form of propagating waves.


Subject(s)
Actomyosin , Caenorhabditis elegans , Animals , Caenorhabditis elegans/physiology , Actomyosin/metabolism , Cytokinesis/physiology , Zygote/physiology , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics
5.
Sci Rep ; 14(1): 16881, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39043768

ABSTRACT

Securing complete control of complex systems comprised of tens of thousands of interconnected nodes holds immense significance across various fields, spanning from cell biology and brain science to human-engineered systems. However, depending on specific functional requirements, it can be more practical and efficient to focus on a pre-defined subset of nodes for control, a concept known as target control. While some methods have been proposed to find the smallest driver node set for target control, they either rely on heuristic approaches based on k-walk theory, lacking a guarantee of optimal solutions, or they are overly complex and challenging to implement in real-world networks. To address this challenge, we introduce a simple and elegant algorithm, inspired by the path cover problem, which efficiently identifies the nodes required to control a target node set within polynomial time. To practically apply the algorithm in real-world systems, we have selected several networks in which a specific set of nodes with functional significance can be designated as a target control set. The analysed systems include the complete connectome of the nematode worm C. elegans, the recently disclosed connectome of the Drosophila larval brain, as well as dozens of genome-wide metabolic networks spanning major plant lineages. The target control analysis shed light on distinctions between neural systems in nematode worms and larval brain insects, particularly concerning the number of nodes necessary to regulate specific functional systems. Furthermore, our analysis uncovers evolutionary trends within plant lineages, notably when examining the proportion of nodes required to control functional pathways.


Subject(s)
Algorithms , Caenorhabditis elegans , Connectome , Animals , Caenorhabditis elegans/physiology , Metabolic Networks and Pathways , Drosophila/physiology , Brain/physiology , Humans
6.
Mar Pollut Bull ; 205: 116668, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38972217

ABSTRACT

Two concentrations (6.25 and 1.25 mg/L) were used for two Parkinson's disease medications, Benserazide, and Trihexyphenidyl, to test their effects on the meiobenthic nematofauna. It is predicted that these highly hydrosoluble drugs will end up in marine environments. The results showed that both medications when added alone, induced (i) important changes in the numbers and (ii) taxonomic composition. The impact of Benserazide and Trihexyphenidyl was also reflected in the (iii) functional traits of nematofauna, with the most affected categories following exposure being the trophic group 1B, the clavate tails, the circular amphids, the c-p2 life history, and the body length of 1-2 mm. These results were supported by the molecular interactions of the studied drugs with both GLD-3 and SDP proteins of Caenorhabditis elegans. (iv) The mixtures of both drugs did not show any changes in the nematode communities, suggesting that no synergistic or antagonistic interactions exist between them.


Subject(s)
Antiparkinson Agents , Benserazide , Caenorhabditis elegans , Trihexyphenidyl , Animals , Antiparkinson Agents/pharmacology , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/physiology , Water Pollutants, Chemical , Nematoda/drug effects
7.
Sci Total Environ ; 946: 174418, 2024 Oct 10.
Article in English | MEDLINE | ID: mdl-38960162

ABSTRACT

Micro-nano plastics have been reported as important carriers of polycyclic aromatic hydrocarbons (PAHs) for long-distance migration in the environment. However, the combined toxicity from long-term chronic exposure beyond the vehicle-release mechanism remains elusive. In this study, we investigated the synergistic action of Benzo[a]pyrene (BaP) and Polystyrene nanoparticles (PS) in Caenorhabditis elegans (C. elegans) as a combined exposure model with environmental concentrations. We found that the combined exposure to BaP and PS, as opposed to single exposures at low concentrations, significantly shortened the lifespan of C. elegans, leading to the occurrence of multiple senescence phenotypes. Multi-omics data indicated that the combined exposure to BaP and PS is associated with the disruption of glutathione homeostasis. Consequently, the accumulated reactive oxygen species (ROS) cannot be effectively cleared, which is highly correlated with mitochondrial dysfunction. Moreover, the increase in ROS promoted lipid peroxidation in C. elegans and downregulated Ferritin-1 (Ftn-1), resulting in ferroptosis and ultimately accelerating the aging process of C. elegans. Collectively, our study provides a new perspective to explain the long-term compound toxicity caused by BaP and PS at real-world exposure concentrations.


Subject(s)
Benzo(a)pyrene , Caenorhabditis elegans , Ferroptosis , Mitochondria , Reactive Oxygen Species , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/physiology , Benzo(a)pyrene/toxicity , Mitochondria/drug effects , Ferroptosis/drug effects , Reactive Oxygen Species/metabolism , Nanoparticles/toxicity , Microplastics/toxicity , Aging
8.
Nat Commun ; 15(1): 5793, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987256

ABSTRACT

Temperature is a critical environmental cue that controls the development and lifespan of many animal species; however, mechanisms underlying low-temperature adaptation are poorly understood. Here, we describe cold-inducible diapause (CID), another type of diapause induced by low temperatures in Caenorhabditis elegans. A premature stop codon in heat shock factor 1 (hsf-1) triggers entry into CID at 9 °C, whereas wild-type animals enter CID at 4 °C. Furthermore, both wild-type and hsf-1(sy441) mutant animals undergoing CID can survive for weeks, and resume growth at 20 °C. Using epistasis analysis, we demonstrate that neural signalling pathways, namely tyraminergic and neuromedin U signalling, regulate entry into CID of the hsf-1 mutant. Overexpression of anti-ageing genes, such as hsf-1, XBP1/xbp-1, FOXO/daf-16, Nrf2/skn-1, and TFEB/hlh-30, also inhibits CID entry of the hsf-1 mutant. Based on these findings, we hypothesise that regulators of the hsf-1 mutant CID may impact longevity, and successfully isolate 16 long-lived mutants among 49 non-CID mutants via genetic screening. Furthermore, we demonstrate that the nonsense mutation of MED23/sur-2 prevents CID entry of the hsf-1(sy441) mutant and extends lifespan. Thus, CID is a powerful model to investigate neural networks involving cold acclimation and to explore new ageing mechanisms.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Cold Temperature , DNA-Binding Proteins , Diapause , Longevity , Transcription Factors , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Diapause/genetics , Diapause/physiology , Longevity/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mutation , Signal Transduction , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Codon, Nonsense/genetics , Neuropeptides/metabolism , Neuropeptides/genetics , Carrier Proteins , Basic Helix-Loop-Helix Transcription Factors
9.
Int J Mol Sci ; 25(13)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38999947

ABSTRACT

Alcohol tolerance is a neuroadaptive response that leads to a reduction in the effects of alcohol caused by previous exposure. Tolerance plays a critical role in the development of alcohol use disorder (AUD) because it leads to the escalation of drinking and dependence. Understanding the molecular mechanisms underlying alcohol tolerance is therefore important for the development of effective therapeutics and for understanding addiction in general. This review explores the molecular basis of alcohol tolerance in invertebrate models, Drosophila and C. elegans, focusing on synaptic transmission. Both organisms exhibit biphasic responses to ethanol and develop tolerance similar to that of mammals. Furthermore, the availability of several genetic tools makes them a great candidate to study the molecular basis of ethanol response. Studies in invertebrate models show that tolerance involves conserved changes in the neurotransmitter systems, ion channels, and synaptic proteins. These neuroadaptive changes lead to a change in neuronal excitability, most likely to compensate for the enhanced inhibition by ethanol.


Subject(s)
Caenorhabditis elegans , Ethanol , Neuronal Plasticity , Synaptic Transmission , Animals , Neuronal Plasticity/drug effects , Ethanol/pharmacology , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/physiology , Caenorhabditis elegans/metabolism , Synaptic Transmission/drug effects , Drug Tolerance , Synapses/metabolism , Synapses/drug effects , Synapses/physiology , Alcoholism/metabolism , Drosophila/physiology , Humans , Invertebrates/physiology
10.
Curr Biol ; 34(14): 3201-3214.e5, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38991614

ABSTRACT

The actomyosin cortex is an active material that generates force to drive shape changes via cytoskeletal remodeling. Cytokinesis is the essential cell division event during which a cortical actomyosin ring closes to separate two daughter cells. Our active gel theory predicted that actomyosin systems controlled by a biochemical oscillator and experiencing mechanical strain would exhibit complex spatiotemporal behavior. To test whether active materials in vivo exhibit spatiotemporally complex kinetics, we imaged the C. elegans embryo with unprecedented temporal resolution and discovered that sections of the cytokinetic cortex undergo periodic phases of acceleration and deceleration. Contractile oscillations exhibited a range of periodicities, including those much longer periods than the timescale of RhoA pulses, which was shorter in cytokinesis than in any other biological context. Modifying mechanical feedback in vivo or in silico revealed that the period of contractile oscillation is prolonged as a function of the intensity of mechanical feedback. Fast local ring ingression occurs where speed oscillations have long periods, likely due to increased local stresses and, therefore, mechanical feedback. Fast ingression also occurs where material turnover is high, in vivo and in silico. We propose that downstream of initiation by pulsed RhoA activity, mechanical feedback, including but not limited to material advection, extends the timescale of contractility beyond that of biochemical input and, therefore, makes it robust to fluctuations in activation. Circumferential propagation of contractility likely allows for sustained contractility despite cytoskeletal remodeling necessary to recover from compaction. Thus, like biochemical feedback, mechanical feedback affords active materials responsiveness and robustness.


Subject(s)
Actomyosin , Caenorhabditis elegans , Cytokinesis , Cytokinesis/physiology , Animals , Caenorhabditis elegans/physiology , Actomyosin/metabolism , Biomechanical Phenomena , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Feedback, Physiological , rhoA GTP-Binding Protein/metabolism , Embryo, Nonmammalian/physiology
11.
Front Biosci (Landmark Ed) ; 29(7): 264, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39082355

ABSTRACT

BACKGROUND: The ability to maintain muscle function decreases with age and loss of proteostatic function. Diet, drugs, and genetic interventions that restrict nutrients or nutrient signaling help preserve long-term muscle function and slow age-related decline. Previously, it was shown that attenuating protein synthesis downstream of the mechanistic target of rapamycin (mTOR) gradually increases expression of heat shock response (HSR) genes in a manner that correlates with increased resilience to protein unfolding stress. Here, we investigate the role of specific tissues in mediating the cytoprotective effects of low translation. METHODS: This study uses genetic tools (transgenic Caenorhabditis elegans (C. elegans), RNA interference and gene expression analysis) as well as physiological assays (survival and paralysis assays) in order to better understand how specific tissues contribute to adaptive changes involving cellular cross-talk that enhance proteostasis under low translation conditions. RESULTS: We use the C. elegans system to show that lowering translation in neurons or the germline increases heat shock gene expression and survival under conditions of heat stress. In addition, we find that low translation in these tissues protects motility in a body muscle-specific model of proteotoxicity that results in paralysis. Low translation in neurons or germline also results in increased expression of certain muscle regulatory and structural genes, reversing reduced expression normally observed with aging in C. elegans. Enhanced resilience to protein unfolding stress requires neuronal expression of cbp-1. CONCLUSIONS: Low translation in either neurons or the germline orchestrate protective adaptation in other tissues, including body muscle.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Heat-Shock Response , Protein Biosynthesis , Proteostasis , TOR Serine-Threonine Kinases , Animals , Animals, Genetically Modified , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Heat-Shock Response/genetics , Neurons/metabolism , RNA Interference , TOR Serine-Threonine Kinases/metabolism , TOR Serine-Threonine Kinases/genetics
12.
Proc Natl Acad Sci U S A ; 121(32): e2318805121, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39083417

ABSTRACT

How do we capture the breadth of behavior in animal movement, from rapid body twitches to aging? Using high-resolution videos of the nematode worm Caenorhabditis elegans, we show that a single dynamics connects posture-scale fluctuations with trajectory diffusion and longer-lived behavioral states. We take short posture sequences as an instantaneous behavioral measure, fixing the sequence length for maximal prediction. Within the space of posture sequences, we construct a fine-scale, maximum entropy partition so that transitions among microstates define a high-fidelity Markov model, which we also use as a means of principled coarse-graining. We translate these dynamics into movement using resistive force theory, capturing the statistical properties of foraging trajectories. Predictive across scales, we leverage the longest-lived eigenvectors of the inferred Markov chain to perform a top-down subdivision of the worm's foraging behavior, revealing both "runs-and-pirouettes" as well as previously uncharacterized finer-scale behaviors. We use our model to investigate the relevance of these fine-scale behaviors for foraging success, recovering a trade-off between local and global search strategies.


Subject(s)
Behavior, Animal , Caenorhabditis elegans , Markov Chains , Animals , Caenorhabditis elegans/physiology , Behavior, Animal/physiology , Models, Biological , Movement/physiology
13.
PLoS Biol ; 22(7): e3002729, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39024405

ABSTRACT

Sensory neurons specialize in detecting and signaling the presence of diverse environmental stimuli. Neuronal injury or disease may undermine such signaling, diminishing the availability of crucial information. Can animals distinguish between a stimulus not being present and the inability to sense that stimulus in the first place? To address this question, we studied Caenorhabditis elegans nematode worms that lack gentle body touch sensation due to genetic mechanoreceptor dysfunction. We previously showed that worms can compensate for the loss of touch by enhancing their sense of smell, via an FLP-20 neuropeptide pathway. Here, we find that touch-deficient worms exhibit, in addition to sensory compensation, also cautious-like behavior, as if preemptively avoiding potential undetectable hazards. Intriguingly, these behavioral adjustments are abolished when the touch neurons are removed, suggesting that touch neurons are required for signaling the unavailability of touch information, in addition to their conventional role of signaling touch stimulation. Furthermore, we found that the ASE taste neurons, which similarly to the touch neurons, express the FLP-20 neuropeptide, exhibit altered FLP-20 expression levels in a touch-dependent manner, thus cooperating with the touch circuit. These results imply a novel form of neuronal signaling that enables C. elegans to distinguish between lack of touch stimulation and loss of touch sensation, producing adaptive behavioral adjustments that could overcome the inability to detect potential threats.


Subject(s)
Behavior, Animal , Caenorhabditis elegans Proteins , Caenorhabditis elegans , Mechanoreceptors , Touch , Animals , Caenorhabditis elegans/physiology , Mechanoreceptors/physiology , Mechanoreceptors/metabolism , Touch/physiology , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Behavior, Animal/physiology , Neuropeptides/metabolism , Neuropeptides/genetics , Mechanotransduction, Cellular/physiology , Smell/physiology , Sensory Receptor Cells/physiology , Sensory Receptor Cells/metabolism
14.
Curr Biol ; 34(14): 3133-3151.e10, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-38964319

ABSTRACT

The sense of touch is conferred by the conjoint function of somatosensory neurons and skin cells. These cells meet across a gap filled by a basal lamina, an ancient structure found in metazoans. Using Caenorhabditis elegans, we investigate the composition and ultrastructure of the extracellular matrix at the epidermis and touch receptor neuron (TRN) interface. We show that membrane-matrix complexes containing laminin, nidogen, and the MEC-4 mechano-electrical transduction channel reside at this interface and are central to proper touch sensation. Interestingly, the dimensions and spacing of these complexes correspond with the discontinuous beam-like extracellular matrix structures observed in serial-section transmission electron micrographs. These complexes fail to coalesce in touch-insensitive extracellular matrix mutants and in dissociated neurons. Loss of nidogen reduces the density of mechanoreceptor complexes and the amplitude of the touch-evoked currents they carry. Thus, neuron-epithelium cell interfaces are instrumental in mechanosensory complex assembly and function. Unlike the basal lamina ensheathing the pharynx and body wall muscle, nidogen recruitment to the puncta along TRNs is not dependent upon laminin binding. MEC-4, but not laminin or nidogen, is destabilized by point mutations in the C-terminal Kunitz domain of the extracellular matrix component, MEC-1. These findings imply that somatosensory neurons secrete proteins that actively repurpose the basal lamina to generate special-purpose mechanosensory complexes responsible for vibrotactile sensing.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Mechanoreceptors , Mechanotransduction, Cellular , Animals , Caenorhabditis elegans/physiology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Mechanoreceptors/metabolism , Mechanoreceptors/physiology , Mechanotransduction, Cellular/physiology , Touch/physiology , Basement Membrane/metabolism , Basement Membrane/physiology , Extracellular Matrix/metabolism , Laminin/metabolism , Membrane Glycoproteins , Membrane Proteins
15.
J Food Sci ; 89(8): 5101-5112, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39030756

ABSTRACT

Macauba (Acrocomia aculeata) is a Brazilian palm tree whose oil in the pulp is rich in oleic acid and carotenoids. However, its physiological function remains unknown. This study aimed to investigate the effects of macauba pulp oil (MPO) on the metabolic link between lipid metabolism and lifespan using Caenorhabditis elegans (C. elegans). C. elegans were treated with 5.0 mg/mL of MPO for analyzing triglyceride and glycerol accumulation, fatty acid profile, gene expression of lipid and oxidative metabolism proteins under cold (4°C) stress conditions, and lifespan analysis under stress conditions such as cold (4°C), heat (37°C), and oxidative (paraquat) stress. MPO significantly suppressed fat accumulation and increased glycerol (a lipolysis index) and the lifespan of C. elegans at low temperature (4°C). This was accompanied by decreased mRNA levels of the genes involved in lipogenesis (spb-1 and pod-2) and increased levels of the genes involved in fatty acid ß-oxidation (acs-2 and nhr-49) and fat mobilization genes (hosl-1 and aak-2). Additionally, MPO treatment modulated fatty acid pools in C. elegans at low temperatures in that MPO treatment decreased saturated fatty acid levels and shifted the fatty acid profile to long-chain fatty acids. Moreover, the effect of MPO on fat accumulation at low temperatures was abolished in fat-7 mutants, whereas both fat-1 and fat-7 contribute, at least in part, to MPO-elevated survival of C. elegans under cold conditions. PRACTICAL APPLICATION: The results obtained in the present study may contribute to the understanding of the health benefits of consuming macauba pulp oil and consequently stimulate economic growth and the industrial application of this new type of oil, which may result in the creation of new jobs and increased value of small producers.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Cold Temperature , Lipid Metabolism , Longevity , Animals , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/physiology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/genetics , Longevity/drug effects , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Lipid Metabolism/drug effects , Plant Oils/pharmacology , Arecaceae/chemistry , Fatty Acids/metabolism , Triglycerides/metabolism , Glycerol/metabolism , Glycerol/pharmacology , Oxidative Stress/drug effects , Palm Oil/pharmacology
16.
Cell Mol Life Sci ; 81(1): 252, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38849591

ABSTRACT

Animals have evolved to seek, select, and exploit food sources in their environment. Collectively termed foraging, these ubiquitous behaviors are necessary for animal survival. As a foundation for understanding foraging, behavioral ecologists established early theoretical and mathematical frameworks which have been subsequently refined and supported by field and laboratory studies of foraging animals. These simple models sought to explain how animals decide which strategies to employ when locating food, what food items to consume, and when to explore the environment for new food sources. These foraging decisions involve integration of prior experience with multimodal sensory information about the animal's current environment and internal state. We suggest that the nematode Caenorhabditis elegans is well-suited for a high-resolution analysis of complex goal-oriented behaviors such as foraging. We focus our discussion on behavioral studies highlighting C. elegans foraging on bacteria and summarize what is known about the underlying neuronal and molecular pathways. Broadly, we suggest that this simple model system can provide a mechanistic understanding of decision-making and present additional avenues for advancing our understanding of complex behavioral processes.


Subject(s)
Caenorhabditis elegans , Decision Making , Feeding Behavior , Neurons , Animals , Caenorhabditis elegans/physiology , Decision Making/physiology , Feeding Behavior/physiology , Neurons/physiology , Models, Biological
17.
Elife ; 122024 Jun 10.
Article in English | MEDLINE | ID: mdl-38857169

ABSTRACT

Understanding how different neuronal types connect and communicate is critical to interpreting brain function and behavior. However, it has remained a formidable challenge to decipher the genetic underpinnings that dictate the specific connections formed between neuronal types. To address this, we propose a novel bilinear modeling approach that leverages the architecture similar to that of recommendation systems. Our model transforms the gene expressions of presynaptic and postsynaptic neuronal types, obtained from single-cell transcriptomics, into a covariance matrix. The objective is to construct this covariance matrix that closely mirrors a connectivity matrix, derived from connectomic data, reflecting the known anatomical connections between these neuronal types. When tested on a dataset of Caenorhabditis elegans, our model achieved a performance comparable to, if slightly better than, the previously proposed spatial connectome model (SCM) in reconstructing electrical synaptic connectivity based on gene expressions. Through a comparative analysis, our model not only captured all genetic interactions identified by the SCM but also inferred additional ones. Applied to a mouse retinal neuronal dataset, the bilinear model successfully recapitulated recognized connectivity motifs between bipolar cells and retinal ganglion cells, and provided interpretable insights into genetic interactions shaping the connectivity. Specifically, it identified unique genetic signatures associated with different connectivity motifs, including genes important to cell-cell adhesion and synapse formation, highlighting their role in orchestrating specific synaptic connections between these neurons. Our work establishes an innovative computational strategy for decoding the genetic programming of neuronal type connectivity. It not only sets a new benchmark for single-cell transcriptomic analysis of synaptic connections but also paves the way for mechanistic studies of neural circuit assembly and genetic manipulation of circuit wiring.


Subject(s)
Caenorhabditis elegans , Connectome , Neurons , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Mice , Neurons/physiology , Single-Cell Analysis , Models, Neurological
18.
Sci Adv ; 10(25): eadn0014, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38905346

ABSTRACT

The central nervous system coordinates peripheral cellular stress responses, including the unfolded protein response of the mitochondria (UPRMT); however, the contexts for which this regulatory capability evolved are unknown. UPRMT is up-regulated upon pathogenic infection and in metabolic flux, and the olfactory nervous system has been shown to regulate pathogen resistance and peripheral metabolic activity. Therefore, we asked whether the olfactory nervous system in Caenorhabditis elegans controls the UPRMT cell nonautonomously. We found that silencing a single inhibitory olfactory neuron pair, AWC, led to robust induction of UPRMT and reduction of oxidative phosphorylation dependent on serotonin signaling and parkin-mediated mitophagy. Further, AWC ablation confers resistance to the pathogenic bacteria Pseudomonas aeruginosa partially dependent on the UPRMT transcription factor atfs-1 and fully dependent on mitophagy machinery. These data illustrate a role for the olfactory nervous system in regulating whole-organism mitochondrial dynamics, perhaps in preparation for postprandial metabolic stress or pathogenic infection.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Mitochondria , Mitophagy , Smell , Animals , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/physiology , Mitochondria/metabolism , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , Smell/physiology , Unfolded Protein Response , Pseudomonas aeruginosa/physiology , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Oxidative Phosphorylation , Signal Transduction , Serotonin/metabolism , Transcription Factors
19.
Sci Adv ; 10(24): eadk9481, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38865452

ABSTRACT

The molecular mechanisms underlying diversity in animal behavior are not well understood. A major experimental challenge is determining the contribution of genetic variants that affect neuronal gene expression to differences in behavioral traits. In Caenorhabditis elegans, the neuroendocrine transforming growth factor-ß ligand, DAF-7, regulates diverse behavioral responses to bacterial food and pathogens. The dynamic neuron-specific expression of daf-7 is modulated by environmental and endogenous bacteria-derived cues. Here, we investigated natural variation in the expression of daf-7 from the ASJ pair of chemosensory neurons. We identified common genetic variants in gap-2, encoding a Ras guanosine triphosphatase (GTPase)-activating protein homologous to mammalian synaptic Ras GTPase-activating protein, which modify daf-7 expression cell nonautonomously and promote exploratory foraging behavior in a partially DAF-7-dependent manner. Our data connect natural variation in neuron-specific gene expression to differences in behavior and suggest that genetic variation in neuroendocrine signaling pathways mediating host-microbe interactions may give rise to diversity in animal behavior.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Genetic Variation , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Gene Expression Regulation , Neurosecretory Systems/metabolism , Feeding Behavior , Behavior, Animal/physiology , Neurons/metabolism , Signal Transduction , Transforming Growth Factor beta
20.
Curr Biol ; 34(12): 2756-2763.e2, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38838665

ABSTRACT

Extracellular vesicles (EVs) are submicron membranous structures and key mediators of intercellular communication.1,2 Recent research has highlighted roles for cilia-derived EVs in signal transduction, underscoring their importance as bioactive extracellular organelles containing conserved ciliary signaling proteins.3,4 Members of the transient receptor potential (TRP) channel polycystin-2 (PKD-2) family are found in ciliary EVs of the green algae Chlamydomonas and the nematode Caenorhabditis elegans5,6 and in EVs in the mouse embryonic node and isolated from human urine.7,8 In C. elegans, PKD-2 is expressed in male-specific EV-releasing sensory neurons, which extend ciliary tips to ciliary pore and directly release EVs into the environment.6,9 Males release EVs in a mechanically stimulated manner, regulate EV cargo content in response to mating partners, and deposit PKD-2::GFP-labeled EVs on the vulval cuticle of hermaphrodites during mating.9,10 Combined, our findings suggest that ciliary EV release is a dynamic process. Herein, we identify mechanisms controlling dynamic EV shedding using time-lapse imaging. Cilia can sustain the release of PKD-2-labeled EVs for 2 h. This extended release doesn't require neuronal transmission. Instead, ciliary intrinsic mechanisms regulate PKD-2 ciliary membrane replenishment and dynamic EV release. The kinesin-3 motor kinesin-like protein 6 (KLP-6) is necessary for initial and extended EV release, while the transition zone protein NPHP-4 is required only for sustained EV release. The dynamic replenishment of PKD-2 at the ciliary tip is key to sustained EV release. Our study provides a comprehensive portrait of real-time ciliary EV release and mechanisms supporting cilia as proficient EV release platforms.


Subject(s)
Caenorhabditis elegans Proteins , Caenorhabditis elegans , Cilia , Extracellular Vesicles , Sensory Receptor Cells , TRPP Cation Channels , Animals , Cilia/metabolism , Cilia/physiology , Extracellular Vesicles/metabolism , Extracellular Vesicles/physiology , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/physiology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/physiology , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans Proteins/genetics , TRPP Cation Channels/metabolism , TRPP Cation Channels/genetics , Male
SELECTION OF CITATIONS
SEARCH DETAIL