1
|
Schwartz HT, Sternberg PW. A sequencing-based screening method identifies regulators of EGFR signaling from nonviable mutants in Caenorhabditis elegans. Sci Signal 2025; 18:eadp9377. [PMID: 39999212 DOI: 10.1126/scisignal.adp9377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 12/06/2024] [Accepted: 02/03/2025] [Indexed: 02/27/2025]
Abstract
Suppressor screens can identify genetic modifiers of biochemical pathways but generally require that the suppressed mutant be viable and fertile. We developed a screening method that obviated this requirement and enabled the identification of mutations that partially suppressed the early developmental arrest and lethality caused by loss of the epidermal growth factor (EGF) receptor ortholog LET-23 in Caenorhabditis elegans. We chemically mutagenized animals carrying the loss-of-function allele let-23(sy15), recovered let-23(sy15) homozygotes that escaped early developmental arrest but were nevertheless inviable, and sequenced their genomes. Testing of candidate causal mutations identified 11 genes that, when mutated, mitigated the early lethality caused by loss of EGF signaling. These included genes encoding homologs of the small guanosine triphosphatase (GTPase) Ras (let-60), which is a downstream effector of LET-23, and of regulators of the small GTPase Rho, including the homolog of the phosphotyrosine-binding protein TENSIN (tns-1). We also recovered suppressing mutations in genes encoding nuclear proteins that protect against DNA damage, including the homolog of MutS homolog 4 (him-14). Genetic experiments were consistent with the repression of Rho activity or the activation of the DNA damage response compensating for the loss of EGF signaling. This sequencing-based, whole-animal screening method may be adapted to other organisms to enable the identification of mutations for which the phenotype does not allow the recovery of viable animals.
Collapse
Affiliation(s)
- Hillel T Schwartz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Paul W Sternberg
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
2
|
Perez SM, Augustineli HS, Marcello MR. Utilizing C. elegans Spermatogenesis and Fertilization Mutants as a Model for Human Disease. J Dev Biol 2025; 13:4. [PMID: 39982357 PMCID: PMC11843878 DOI: 10.3390/jdb13010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/10/2025] [Accepted: 01/20/2025] [Indexed: 02/22/2025] Open
Abstract
The nematode C. elegans is a proven model for identifying genes involved in human disease, and the study of C. elegans reproduction, specifically spermatogenesis and fertilization, has led to significant contributions to our understanding of cellular function. Approximately 70 genes have been identified in C. elegans that control spermatogenesis and fertilization (spe and fer mutants). This review focuses on eight genes that have human orthologs with known pathogenic phenotypes. Using C. elegans to study these genes has led to critical developments in our understanding of protein domain function and human disease, including understanding the role of OTOF (the ortholog of C. elegans fer-1) in hearing loss, the contribution of the spe-39 ortholog VIPAS39 in vacuolar protein sorting, and the overlapping functions of spe-26 and KLHL10 in spermatogenesis. We discuss the cellular function of both the C. elegans genes and their human orthologs and the impact that C. elegans mutants and human variants have on cellular function and physiology. Utilizing C. elegans to understand the function of the genes reviewed here, and additional understudied and undiscovered genes, represents a unique opportunity to understand the function of variants that could lead to better disease diagnosis and clinical decision making.
Collapse
|
3
|
Spadaro JGA, Wildwater M, Korthout H, Kim HK. Sample Preparation of Caenorhabditis elegans for GC-MS-Based Metabolomics in Toxicity Assessment. Methods Mol Biol 2025; 2895:271-297. [PMID: 39885036 DOI: 10.1007/978-1-0716-4350-1_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
The nematode Caenorhabditis elegans, widely recognized as a model organism due to its ease of breeding and well-characterized genomes, boasts complete digestive, reproductive, and endocrine systems, as well as conserved signaling pathways shared with mammals. It has become an invaluable resource for metabolomics research, particularly in examining responses to chemical or environmental factors and toxicity assessments. In this article, we provide detailed, step-by-step protocols for cultivating C. elegans and conducting metabolomics analyses, specifically focusing on sample preparation for GC-MS analysis in response to toxic compound treatments. We highlight the critical choice of extraction solvent, introducing two representative systems for extracting metabolites from C. elegans.
Collapse
Affiliation(s)
| | | | | | - Hye Kyong Kim
- Fytagoras BV, Leiden, The Netherlands.
- Natural Products Lab, Institute of Biology, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
4
|
Gonzalez HC, Misare KR, Mendenhall TT, Wolf BJ, Mulholland PJ, Gordon KL, Hartman JH. Transgenic expression of human cytochrome P450 2E1 in C. elegans and rat PC-12 cells sensitizes to ethanol-induced locomotor and mitochondrial effects. Biochem Biophys Res Commun 2024; 734:150735. [PMID: 39357336 PMCID: PMC11540131 DOI: 10.1016/j.bbrc.2024.150735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/17/2024] [Accepted: 09/21/2024] [Indexed: 10/04/2024]
Abstract
Chronic alcohol (ethanol) use is increasing in the United States and has been linked to numerous health issues in multiple organ systems including neurological dysfunction and diseases. Ethanol toxicity is mainly driven by the metabolite acetaldehyde, which is generated through three pathways: alcohol dehydrogenase (ADH2), catalase (CAT), and cytochrome P450 2E1 (CYP2E1). ADH2, while the main ethanol clearance pathway in the liver, is not expressed in the mammalian brain, resulting in CAT and CYP2E1 driving local metabolism of ethanol in the central nervous system. CYP2E1 is known to generate reactive metabolites and reactive oxygen species and localizes to the mitochondria (mtCYP2E1) and endoplasmic reticulum (erCYP2E1). We sought to understand the consequences of mtCYP2E1 and erCYP2E1 in the nervous system during acute ethanol exposure. To answer this question, we generated transgenic Caenorhabditis elegans roundworms expressing human CYP2E1 in the mitochondria, endoplasmic reticulum, or both and exposed them to ethanol. We found that at lower concentrations, wild-type and mtCYP2E1-expressing worms had a small but significant inhibition of locomotion, whereas the erCYP2E1-expressing worms showed protection from this inhibition. At higher doses, all strains had reduced locomotion, but the erCYP2E1-expressing worms recovered faster than wild-type controls. CYP2E1 expression, regardless of organellar targeting, reduced mitochondrial respiration in response to ethanol. Similarly, transgenic expression of CYP2E1 in either organelle in PC-12 rat neuronal cell lines sensitized them to ethanol-induced cell death. Together, these findings suggest that subcellular localization of CYP2E1 impacts behavioral effects of ethanol and should be further studied in the mammalian central nervous system.
Collapse
Affiliation(s)
- Hyland C Gonzalez
- Dept of Biochemistry and Molecular Biology, Medical University of South Carolina, 173 Ashley Ave. BSB 501 | MSC 509, Charleston, SC, 29425, USA
| | - Kelly R Misare
- Dept of Biochemistry and Molecular Biology, Medical University of South Carolina, 173 Ashley Ave. BSB 501 | MSC 509, Charleston, SC, 29425, USA
| | - Tsultrim T Mendenhall
- Dept of Biochemistry and Molecular Biology, Medical University of South Carolina, 173 Ashley Ave. BSB 501 | MSC 509, Charleston, SC, 29425, USA
| | - Bethany J Wolf
- Dept of Public Health Sciences, Medical University of South Carolina, 135 Cannon St., Charleston, SC, 29425, USA
| | - Patrick J Mulholland
- Dept of Neuroscience, Charleston Alcohol Research Center, Medical University of South Carolina, 30 Courtenay Drive, Charleston, SC, 29425, USA
| | - Kacy L Gordon
- Dept of Biology, The University of North Carolina at Chapel Hill, 316 Fordham Hall, Chapel Hill, NC, 27599, USA; UNC Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, 450 West Dr, Chapel Hill, NC, 27599, USA
| | - Jessica H Hartman
- Dept of Biochemistry and Molecular Biology, Medical University of South Carolina, 173 Ashley Ave. BSB 501 | MSC 509, Charleston, SC, 29425, USA; Dept of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Ave. BSB 501 | MSC 509, Charleston, SC, 29425, USA.
| |
Collapse
|
5
|
Valera-Alberni M, Yao P, Romero-Sanz S, Lanjuin A, Mair WB. Novel imaging tools to study mitochondrial morphology in Caenorhabditis elegans. Life Sci Alliance 2024; 7:e202402918. [PMID: 39260886 PMCID: PMC11391045 DOI: 10.26508/lsa.202402918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/13/2024] Open
Abstract
Mitochondria exhibit a close interplay between their structure and function. Understanding this intricate relationship requires advanced imaging techniques that can capture the dynamic nature of mitochondria and their impact on cellular processes. However, much of the work on mitochondrial dynamics has been performed in single celled organisms or in vitro cell culture. Here, we introduce novel genetic tools for live imaging of mitochondrial morphology in the nematode Caenorhabditis elegans, addressing a pressing need for advanced techniques in studying organelle dynamics within live intact multicellular organisms. Through a comprehensive analysis, we directly compare our tools with existing methods, demonstrating their advantages for visualizing mitochondrial morphology and contrasting their impact on organismal physiology. We reveal limitations of conventional techniques, whereas showcasing the utility and versatility of our approaches, including endogenous CRISPR tags and ectopic labeling. By providing a guide for selecting the most suitable tools based on experimental goals, our work advances mitochondrial research in C. elegans and enhances the strategic integration of diverse imaging modalities for a holistic understanding of organelle dynamics in living organisms.
Collapse
Affiliation(s)
- Miriam Valera-Alberni
- Department of Molecular Metabolism, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Pallas Yao
- Department of Molecular Metabolism, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Silvia Romero-Sanz
- Department of Molecular Metabolism, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Anne Lanjuin
- Department of Molecular Metabolism, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - William B Mair
- Department of Molecular Metabolism, Harvard TH Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
6
|
Thakor PM, Patel JD, Patel RJ, Chaki SH, Khimani AJ, Vaidya YH, Chauhan AP, Dholakia AB, Patel VC, Patel AJ, Bhavsar NH, Patel HV. Exploring New Schiff Bases: Synthesis, Characterization, and Multifaceted Analysis for Biomedical Applications. ACS OMEGA 2024; 9:35431-35448. [PMID: 39184520 PMCID: PMC11339819 DOI: 10.1021/acsomega.4c02007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/18/2024] [Accepted: 05/08/2024] [Indexed: 08/27/2024]
Abstract
The current work aims to generate novel Schiff bases by reacting substituted aldehydes with amine derivatives catalyzed by a natural acid. The developed compounds underwent diverse physicochemical analyses including liquid chromatography-mass spectrometry, Fourier transform infrared spectroscopy, scanning electron microscopy, 1H- and 13C-nuclear magnetic resonance, and X-ray diffraction. Furthermore, differential thermogravimetric, thermogravimetric, and differential thermal analysis techniques were employed in a nitrogen-free environment to determine kinetic parameters. These data were then used in model-free isoconversional methods (e.g., Friedman, Kissinger-Akahira-Sunose, and Flynn-Wall-Ozawa). The Schiff bases were evaluated for their in vitro and in silico α-amylase inhibitory activity. Schiff base-2 displayed the highest inhibition compared with the reference drug acarbose. In comprehensive MTT assay cytotoxicity investigations, both Schiff bases showed strong anticancer capabilities against the human lung cancer cell line (A549). Moreover, this study demonstrated effectiveness of synthetic compounds in screening Caenorhabditis elegans for anti-Alzheimer's and stress resistance properties. The simplicity of its biology allowed precise evaluation of the effect of compounds on neuronal function and stress response. This research enhances drug discovery efforts for Alzheimer's and stress-related disorders, potentially improving patient outcomes.
Collapse
Affiliation(s)
- Priteshkumar M Thakor
- Department of Chemistry, Shri Alpesh N. Patel Post Graduate Institute of Science and Research, Anand, Gujarat 388001, India
| | - Jatin D Patel
- Department of Chemistry, Shri Alpesh N. Patel Post Graduate Institute of Science and Research, Anand, Gujarat 388001, India
| | - Rajesh J Patel
- Department of Chemistry, Sardar Patel University, Vallabh Vidyanagar, Anand, Gujarat 388001, India
| | - Sunil H Chaki
- Department of Physics, Sardar Patel University, Vallabh Vidyanagar, Anand, Gujarat 388001, India
| | - Ankurkumar J Khimani
- Department of Physics, Shri Alpesh N. Patel Cnce and Research, Anand, Gujarat 388001, India
| | - Yati H Vaidya
- Department of Microbiology, Shri Alpesh N. Patel Post Graduate Institute of Science and Research, Anand, Gujarat 388001, India
| | - Anita P Chauhan
- Department of Biotechnology, Shri Alpesh N. Patel Post Graduate Institute of Science and Research, Anand, Gujarat 388001, India
| | - Amit B Dholakia
- School of Science, Birsa Munda Tribal University, Rajpipla, Gujarat 393145, India
| | - Vishant C Patel
- Department of Chemistry, Shri Alpesh N. Patel Post Graduate Institute of Science and Research, Anand, Gujarat 388001, India
| | - Ankitkumar J Patel
- Department of Chemistry, Shri Alpesh N. Patel Post Graduate Institute of Science and Research, Anand, Gujarat 388001, India
| | - Nirav H Bhavsar
- Department of Microbiology, Shri Alpesh N. Patel Post Graduate Institute of Science and Research, Anand, Gujarat 388001, India
| | - Hiteshkumar V Patel
- Department of Biochemistry, Shri Alpesh N. Patel Post Graduate Institute of Science and Research, Anand, Gujarat 388001, India
| |
Collapse
|
7
|
Wu Y, Zhou D, Hu J. Reconstruction of gene regulatory networks for Caenorhabditis elegans using tree-shaped gene expression data. Brief Bioinform 2024; 25:bbae396. [PMID: 39133097 PMCID: PMC11318059 DOI: 10.1093/bib/bbae396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/11/2024] [Accepted: 08/07/2024] [Indexed: 08/13/2024] Open
Abstract
Constructing gene regulatory networks is a widely adopted approach for investigating gene regulation, offering diverse applications in biology and medicine. A great deal of research focuses on using time series data or single-cell RNA-sequencing data to infer gene regulatory networks. However, such gene expression data lack either cellular or temporal information. Fortunately, the advent of time-lapse confocal laser microscopy enables biologists to obtain tree-shaped gene expression data of Caenorhabditis elegans, achieving both cellular and temporal resolution. Although such tree-shaped data provide abundant knowledge, they pose challenges like non-pairwise time series, laying the inaccuracy of downstream analysis. To address this issue, a comprehensive framework for data integration and a novel Bayesian approach based on Boolean network with time delay are proposed. The pre-screening process and Markov Chain Monte Carlo algorithm are applied to obtain the parameter estimates. Simulation studies show that our method outperforms existing Boolean network inference algorithms. Leveraging the proposed approach, gene regulatory networks for five subtrees are reconstructed based on the real tree-shaped datatsets of Caenorhabditis elegans, where some gene regulatory relationships confirmed in previous genetic studies are recovered. Also, heterogeneity of regulatory relationships in different cell lineage subtrees is detected. Furthermore, the exploration of potential gene regulatory relationships that bear importance in human diseases is undertaken. All source code is available at the GitHub repository https://github.com/edawu11/BBTD.git.
Collapse
Affiliation(s)
- Yida Wu
- School of Mathematical Sciences, Xiamen University, Zengcuo'an West Road, Siming District, Xiamen 361000, China
| | - Da Zhou
- School of Mathematical Sciences, Xiamen University, Zengcuo'an West Road, Siming District, Xiamen 361000, China
| | - Jie Hu
- School of Mathematical Sciences, Xiamen University, Zengcuo'an West Road, Siming District, Xiamen 361000, China
| |
Collapse
|
8
|
Valera-Alberni M, Yao P, Romero-Sanz S, Lanjuin A, Mair WB. Novel Imaging Tools to Study Mitochondrial Dynamics in Caenorhabditis elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.603730. [PMID: 39071403 PMCID: PMC11275731 DOI: 10.1101/2024.07.16.603730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Mitochondria exhibit a close interplay between their structure and function. Understanding this intricate relationship requires advanced imaging techniques that can capture the dynamic nature of mitochondria and their impact on cellular processes. However, much of the work on mitochondrial dynamics has been done in single celled organisms or in vitro cell culture. Here, we introduce novel genetic tools for live imaging of mitochondrial networks in the nematode Caenorhabditis elegans , addressing a pressing need for advanced techniques in studying organelle dynamics within live intact multicellular organisms. Through a comprehensive analysis, we directly compare our tools with existing methods, demonstrating their advantages for visualizing mitochondrial morphology and contrasting their impact on organismal physiology. We reveal limitations of conventional techniques, while showcasing the utility and versatility of our approaches, including endogenous CRISPR tags and ectopic labeling. By providing a guide for selecting the most suitable tools based on experimental goals, our work advances mitochondrial research in C. elegans and enhances the strategic integration of diverse imaging modalities for a holistic understanding of organelle dynamics in living organisms.
Collapse
|
9
|
de Oliveira GV, Soares MV, Cordeiro LM, da Silva AF, Venturini L, Ilha L, Baptista FBO, da Silveira TL, Soares FAA, Iglesias BA. Toxicological assessment of photoactivated tetra-cationic porphyrin molecules under white light exposure in a Caenorhabditis elegans model. Toxicology 2024; 504:153793. [PMID: 38574843 DOI: 10.1016/j.tox.2024.153793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024]
Abstract
Photodynamic therapy (PDT) utilizes the potential of photosensitizing substances to absorb light energy and produce reactive oxygen species. Tetra-cationic porphyrins, which have organic or coordination compounds attached to their periphery, are heterocyclic derivatives with well-described antimicrobial and antitumoral properties. This is due to their ability to produce reactive oxygen species and their photobiological properties in solution. Consequently, these molecules are promising candidates as new and more effective photosensitizers with biomedical, environmental, and other biomedical applications. Prior to human exposure, it is essential to establish the toxicological profile of these molecules using in vivo models. In this study, we used Caenorhabditis elegans, a small free-living nematode, as a model for assessing toxic effects and predicting toxicity in preclinical research. We evaluated the toxic effects of porphyrins (neutral and tetra-cationic) on nematodes under dark/light conditions. Our findings demonstrate that tetra-methylated porphyrins (3TMeP and 4TMeP) at a concentration of 3.3 µg/mL (1.36 and 0.93 µM) exhibit high toxicity (as evidenced by reduced survival, development, and locomotion) under dark conditions. Moreover, photoactivated tetra-methylated porphyrins induce higher ROS levels compared to neutral (3TPyP and 4TPyP), tetra-palladated (3PdTPyP and 4PdTPyP), and tetra-platinated (3PtTPyP and 4PtTPyP) porphyrins, which may be responsible for the observed toxic effects.
Collapse
Affiliation(s)
- Gabriela Vitória de Oliveira
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Marcell Valandro Soares
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Larissa Marafiga Cordeiro
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Aline Franzen da Silva
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Luiza Venturini
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Larissa Ilha
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Fabiane Bicca Obetine Baptista
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Tássia Limana da Silveira
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Félix Alexandre Antunes Soares
- Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil.
| | - Bernardo Almeida Iglesias
- Laboratory of Bioinorganic and Porphyrinic Materials, Department of Chemistry, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil.
| |
Collapse
|
10
|
Errázuriz León R, Araya Salcedo VA, Novoa San Miguel FJ, Llanquinao Tardio CRA, Tobar Briceño AA, Cherubini Fouilloux SF, de Matos Barbosa M, Saldías Barros CA, Waldman WR, Espinosa-Bustos C, Hornos Carneiro MF. Photoaged polystyrene nanoplastics exposure results in reproductive toxicity due to oxidative damage in Caenorhabditis elegans. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 348:123816. [PMID: 38508369 DOI: 10.1016/j.envpol.2024.123816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/15/2024] [Accepted: 03/16/2024] [Indexed: 03/22/2024]
Abstract
The increase of plastic production together with the incipient reuse/recycling system has resulted in massive discards into the environment. This has facilitated the formation of micro- and nanoplastics (MNPs) which poses major risk for environmental health. Although some studies have investigated the effects of pristine MNPs on reproductive health, the effects of weathered MNPs have been poorly investigated. Here we show in Caenorhabditis elegans that exposure to photoaged polystyrene nanoplastics (PSNP-UV) results in worse reproductive performance than pristine PSNP (i.e., embryonic/larval lethality plus a decrease in the brood size, accompanied by a high number of unfertilized eggs), besides it affects size and locomotion behavior. Those effects were potentially generated by reactive products formed during UV-irradiation, since we found higher levels of reactive oxygen species and increased expression of GST-4 in worms exposed to PSNP-UV. Those results are supported by physical-chemical characterization analyses which indicate significant formation of oxidative degradation products from PSNP under UV-C irradiation. Our study also demonstrates that PSNP accumulate predominantly in the gastrointestinal tract of C. elegans (with no accumulation in the gonads), being completely eliminated at 96 h post-exposure. We complemented the toxicological analysis of PSNP/PSNP-UV by showing that the activation of the stress response via DAF-16 is dependent of the nanoplastics accumulation. Our data suggest that exposure to the wild PSNP, i.e., polystyrene nanoplastics more similar to those actually found in the environment, results in more important reprotoxic effects. This is associated with the presence of degradation products formed during UV-C irradiation and their interaction with biological targets.
Collapse
Affiliation(s)
- Rocío Errázuriz León
- Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, 7820436, Chile
| | | | | | | | | | | | - Marcela de Matos Barbosa
- Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto/SP, 14040-901, Brazil
| | | | | | - Christian Espinosa-Bustos
- Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, 7820436, Chile
| | | |
Collapse
|
11
|
Rodríguez-Martín M, Báez-Flores J, Ribes V, Isidoro-García M, Lacal J, Prieto-Matos P. Non-Mammalian Models for Understanding Neurological Defects in RASopathies. Biomedicines 2024; 12:841. [PMID: 38672195 PMCID: PMC11048513 DOI: 10.3390/biomedicines12040841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 03/27/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
RASopathies, a group of neurodevelopmental congenital disorders stemming from mutations in the RAS/MAPK pathway, present a unique opportunity to delve into the intricacies of complex neurological disorders. Afflicting approximately one in a thousand newborns, RASopathies manifest as abnormalities across multiple organ systems, with a pronounced impact on the central and peripheral nervous system. In the pursuit of understanding RASopathies' neurobiology and establishing phenotype-genotype relationships, in vivo non-mammalian models have emerged as indispensable tools. Species such as Danio rerio, Drosophila melanogaster, Caenorhabditis elegans, Xenopus species and Gallus gallus embryos have proven to be invaluable in shedding light on the intricate pathways implicated in RASopathies. Despite some inherent weaknesses, these genetic models offer distinct advantages over traditional rodent models, providing a holistic perspective on complex genetics, multi-organ involvement, and the interplay among various pathway components, offering insights into the pathophysiological aspects of mutations-driven symptoms. This review underscores the value of investigating the genetic basis of RASopathies for unraveling the underlying mechanisms contributing to broader neurological complexities. It also emphasizes the pivotal role of non-mammalian models in serving as a crucial preliminary step for the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Mario Rodríguez-Martín
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca, 37007 Salamanca, Spain; (M.R.-M.); (J.B.-F.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.I.-G.); (P.P.-M.)
| | - Juan Báez-Flores
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca, 37007 Salamanca, Spain; (M.R.-M.); (J.B.-F.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.I.-G.); (P.P.-M.)
| | - Vanessa Ribes
- Institut Jacques Monod, Université Paris Cité, CNRS, F-75013 Paris, France;
| | - María Isidoro-García
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.I.-G.); (P.P.-M.)
- Clinical Biochemistry Department, Hospital Universitario de Salamanca, 37007 Salamanca, Spain
- Clinical Rare Diseases Reference Unit DiERCyL, 37007 Castilla y León, Spain
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| | - Jesus Lacal
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca, 37007 Salamanca, Spain; (M.R.-M.); (J.B.-F.)
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.I.-G.); (P.P.-M.)
| | - Pablo Prieto-Matos
- Institute of Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.I.-G.); (P.P.-M.)
- Clinical Rare Diseases Reference Unit DiERCyL, 37007 Castilla y León, Spain
- Department of Pediatrics, Hospital Universitario de Salamanca, 37007 Salamanca, Spain
- Department of Biomedical and Diagnostics Science, University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
12
|
Shalash R, Levi-Ferber M, Cohen C, Dori A, Brodie C, Henis-Korenblit S. Cross-species modeling of muscular dystrophy in Caenorhabditis elegans using patient-derived extracellular vesicles. Dis Model Mech 2024; 17:dmm050412. [PMID: 38501170 PMCID: PMC11007864 DOI: 10.1242/dmm.050412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 03/04/2024] [Indexed: 03/20/2024] Open
Abstract
Reliable disease models are critical for medicine advancement. Here, we established a versatile human disease model system using patient-derived extracellular vesicles (EVs), which transfer a pathology-inducing cargo from a patient to a recipient naïve model organism. As a proof of principle, we applied EVs from the serum of patients with muscular dystrophy to Caenorhabditis elegans and demonstrated their capability to induce a spectrum of muscle pathologies, including lifespan shortening and robust impairment of muscle organization and function. This demonstrates that patient-derived EVs can deliver disease-relevant pathologies between species and can be exploited for establishing novel and personalized models of human disease. Such models can potentially be used for disease diagnosis, prognosis, analyzing treatment responses, drug screening and identification of the disease-transmitting cargo of patient-derived EVs and their cellular targets. This system complements traditional genetic disease models and enables modeling of multifactorial diseases and of those not yet associated with specific genetic mutations.
Collapse
Affiliation(s)
- Rewayd Shalash
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Mor Levi-Ferber
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Coral Cohen
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
- The Mina and Everard Goodman Faculty of Life Sciences and Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Amir Dori
- Department of Neurology, Sheba Medical Center, Ramat-Gan 52621, Israel
- Faculty of Medicine, Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Chaya Brodie
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
- The Mina and Everard Goodman Faculty of Life Sciences and Institute of Nanotechnology and Advanced Materials (BINA), Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Sivan Henis-Korenblit
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| |
Collapse
|
13
|
Schwartz HT, Tan CH, Peraza J, Raymundo KLT, Sternberg PW. Molecular identification of a peroxidase gene controlling body size in the entomopathogenic nematode Steinernema hermaphroditum. Genetics 2024; 226:iyad209. [PMID: 38078889 PMCID: PMC11491526 DOI: 10.1093/genetics/iyad209] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 12/01/2023] [Indexed: 02/08/2024] Open
Abstract
The entomopathogenic nematode Steinernema hermaphroditum was recently rediscovered and is being developed as a genetically tractable experimental system for the study of previously unexplored biology, including parasitism of its insect hosts and mutualism with its bacterial endosymbiont Xenorhabdus griffiniae. Through whole-genome re-sequencing and genetic mapping we have for the first time molecularly identified the gene responsible for a mutationally defined phenotypic locus in an entomopathogenic nematode. In the process we observed an unexpected mutational spectrum following ethyl methansulfonate mutagenesis in this species. We find that the ortholog of the essential Caenorhabditis elegans peroxidase gene skpo-2 controls body size and shape in S. hermaphroditum. We confirmed this identification by generating additional loss-of-function mutations in the gene using CRISPR-Cas9. We propose that the identification of skpo-2 will accelerate gene targeting in other Steinernema entomopathogenic nematodes used commercially in pest control, as skpo-2 is X-linked and males hemizygous for loss of its function can mate, making skpo-2 an easily recognized and maintained marker for use in co-CRISPR.
Collapse
Affiliation(s)
- Hillel T Schwartz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Chieh-Hsiang Tan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Jackeline Peraza
- Department of Biology, Barnard College of Columbia University, NewYork, NY 10027, USA
| | | | - Paul W Sternberg
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| |
Collapse
|
14
|
Eck RJ, Stair JG, Kraemer BC, Liachko NF. Simple models to understand complex disease: 10 years of progress from Caenorhabditis elegans models of amyotrophic lateral sclerosis and frontotemporal lobar degeneration. Front Neurosci 2024; 17:1300705. [PMID: 38239833 PMCID: PMC10794587 DOI: 10.3389/fnins.2023.1300705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 11/28/2023] [Indexed: 01/22/2024] Open
Abstract
The nematode Caenorhabditis elegans are a powerful model system to study human disease, with numerous experimental advantages including significant genetic and cellular homology to vertebrate animals, a short lifespan, and tractable behavioral, molecular biology and imaging assays. Beginning with the identification of SOD1 as a genetic cause of amyotrophic lateral sclerosis (ALS), C. elegans have contributed to a deeper understanding of the mechanistic underpinnings of this devastating neurodegenerative disease. More recently this work has expanded to encompass models of other types of ALS and the related disease frontotemporal lobar degeneration (FTLD-TDP), including those characterized by mutation or accumulation of the proteins TDP-43, C9orf72, FUS, HnRNPA2B1, ALS2, DCTN1, CHCHD10, ELP3, TUBA4A, CAV1, UBQLN2, ATXN3, TIA1, KIF5A, VAPB, GRN, and RAB38. In this review we summarize these models and the progress and insights from the last ten years of using C. elegans to study the neurodegenerative diseases ALS and FTLD-TDP.
Collapse
Affiliation(s)
- Randall J. Eck
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, United States
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, United States
| | - Jade G. Stair
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| | - Brian C. Kraemer
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, United States
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, United States
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Nicole F. Liachko
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, United States
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| |
Collapse
|
15
|
Kuo-Esser L, Chen R, Lawson K, Kuchinski K, Simmons N, Dominguez M, Scandura T, Vo M, Dasenbrock-Gammon E, Hagan N, Esposito H, Thompson M, Le S, Escorcia W, Wetzel HN. Early-life caffeine exposure induces morphological changes and altered physiology in Caenorhabditiselegans. Biochem Biophys Res Commun 2024; 690:149240. [PMID: 37988878 DOI: 10.1016/j.bbrc.2023.149240] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023]
Abstract
Caffeine, a widely consumed stimulant, is known for its effects on alertness and fatigue reduction by blockade of adenosine receptors. While it holds therapeutic potential, its diverse impacts pose risks, particularly in early development. This study explores the developmental effects of caffeine exposure using Caenorhabditis elegans (C. elegans) as a model organism. We investigated morphological and behavioral changes induced by caffeine exposure at the L1 stage and assessed their impact at the L4 stage, which roughly corresponds to human infancy and adolescence, respectively. Caffeine-exposed worms displayed increased body length, body bends, and pharyngeal pumping rates compared to control worms. These findings indicate heightened food-seeking behavior and greater food intake, leading to the observed morphological changes. While caffeine did not affect other locomotor behaviors, its stimulatory effect on growth and development highlights its significance. This study provides insights into the potential impact of early-life caffeine exposure on long-term health and development, offering a foundation for future research in vertebrates to uncover its implications on metabolism and other metrics of health.
Collapse
Affiliation(s)
- Lance Kuo-Esser
- Biology Department, Xavier University, Cincinnati, OH, 45207, USA
| | - Ramon Chen
- Biology Department, Xavier University, Cincinnati, OH, 45207, USA
| | - Kylie Lawson
- Biology Department, Xavier University, Cincinnati, OH, 45207, USA
| | | | - Nijah Simmons
- Biology Department, Xavier University, Cincinnati, OH, 45207, USA
| | | | - Tommy Scandura
- Biology Department, Xavier University, Cincinnati, OH, 45207, USA
| | - Martin Vo
- Biology Department, Xavier University, Cincinnati, OH, 45207, USA; Lake Erie College of Osteopathic Medicine, Lake Erie, Pennsylvania, 16509, USA
| | - Emma Dasenbrock-Gammon
- Biology Department, Xavier University, Cincinnati, OH, 45207, USA; University of Kentucky College of Medicine, Highland Heights, Kentucky, 41099, USA
| | - Natalie Hagan
- Biology Department, Xavier University, Cincinnati, OH, 45207, USA; University of Kentucky College of Medicine, Highland Heights, Kentucky, 41099, USA
| | - Haley Esposito
- Biology Department, Xavier University, Cincinnati, OH, 45207, USA
| | - Molly Thompson
- Biology Department, Xavier University, Cincinnati, OH, 45207, USA
| | - Steven Le
- Biology Department, Xavier University, Cincinnati, OH, 45207, USA
| | - Wilber Escorcia
- Biology Department, Xavier University, Cincinnati, OH, 45207, USA.
| | - Hanna N Wetzel
- Biology Department, Xavier University, Cincinnati, OH, 45207, USA.
| |
Collapse
|
16
|
Doyle JJ, Parker JA. Genetic Interactions of Progranulin Across the ALS-FTD Spectrum and Beyond. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000304. [PMID: 38188422 PMCID: PMC10767572 DOI: 10.17912/micropub.biology.000304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 08/08/2021] [Accepted: 12/19/2023] [Indexed: 01/09/2024]
Abstract
Progranulin (PGRN) is a growth factor in which mutations are one of the leading causes of frontotemporal dementia (FTD), and has been implicated in an assortment of neurodegenerative diseases. Conversely, higher levels of the protein have shown potential as a general neuronal protective factor. While examining its neuroprotective applications on a broader scale would be unfeasible in mammalian models, we turned to the nematode C. elegans to map the interactions of PGRN across multiple genetic models of neurodegenerative diseases. Our results indicate that while the overexpression of PGRN appears to be protective across all models tested, the loss of PGRN exacerbated the disease phenotypes of all but three of the models tested. Given the ease of genetic analysis in nematodes, we propose this model organism as an efficient tool to build a comprehensive map of PGRN's genetic interactions.
Collapse
Affiliation(s)
- James J. Doyle
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
- Metabolic Disorders and Complications, RI-MUHC, Montreal, McGill, Canada
| | - J Alex Parker
- CRCHUM and Department of Neuroscience, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
17
|
Forte NR, Veasey VN, Christie BJ, Carter A, Hanks MA, Holderfield A, Houston T, Challa AK, Turner AN. Engaging students in a genetics course-based undergraduate research experience utilizing Caenorhabditis elegans in hybrid learning to explore human disease gene variants. JOURNAL OF MICROBIOLOGY & BIOLOGY EDUCATION 2023; 24:e00078-23. [PMID: 38108007 PMCID: PMC10720527 DOI: 10.1128/jmbe.00078-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 10/13/2023] [Indexed: 12/19/2023]
Abstract
Genetic analysis in model systems using bioinformatic approaches provides a rich context for a concrete and conceptual understanding of gene structure and function. With the intent to engage students in research and explore disease biology utilizing the nematode Caenorhabditis elegans model, we developed a semester-long course-based undergraduate research experience (CURE) in a hybrid (online/in-person) learning environment-the gene-editing and evolutionary nematode exploration CURE (GENE-CURE). Using a combination of bioinformatic and molecular genetic tools, students performed structure-function analysis of disease-associated variants of uncertain significance (VUS) in human orthologs. With the aid of a series of workshop-style research sessions, students worked in teams of two to six members to identify a conserved VUS locus across species and design and test a polymerase chain reaction-based assay for targeted editing of a gene in the nematode and downstream genotyping. Research session discussions, responsible conduct of research training, electronic laboratory notebook, project reports, quizzes, and group poster presentations at a research symposium were assessed for mastery of learning objectives and research progress. Self-reflections were collected from students to assess engagement, science identity, and science efficacy. Qualitative analysis of these reflections indicated several gains suggesting that all students found many aspects of the GENE-CURE rewarding (learning process of research, self-confidence in research and science identity, and personal interest) and challenging (iterative research and failure, time management, COVID-19 pandemic, and life issues).
Collapse
Affiliation(s)
- Natalie R. Forte
- Department of Biology, Jacksonville State University, Jacksonville, Alabama, USA
| | - Virginia N. Veasey
- Department of Biology, Jacksonville State University, Jacksonville, Alabama, USA
| | - Bethany J. Christie
- Department of Psychology, Jacksonville State University, Jacksonville, Alabama, USA
| | - Amira Carter
- Department of Biology, Jacksonville State University, Jacksonville, Alabama, USA
| | - Marli A. Hanks
- Department of Secondary Education, Jacksonville State University, Jacksonville, Alabama, USA
| | - Alan Holderfield
- Department of Secondary Education, Jacksonville State University, Jacksonville, Alabama, USA
| | - Taylor Houston
- Department of Secondary Education, Jacksonville State University, Jacksonville, Alabama, USA
| | - Anil K. Challa
- Department of Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Department of Life Sciences, Shiv Nadar University, Delhi, National Capital Region, India
| | - Ashley N. Turner
- Department of Biology, Jacksonville State University, Jacksonville, Alabama, USA
| |
Collapse
|
18
|
Banerjee S, Bajire SK, Mithun HK, Shastry RP. 3-(Bromoacetyl) coumarin is a potential therapeutic agent against neonatal sepsis-associated Pseudomonas extremorientalis. Arch Microbiol 2023; 205:312. [PMID: 37603073 DOI: 10.1007/s00203-023-03653-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/24/2023] [Accepted: 08/08/2023] [Indexed: 08/22/2023]
Abstract
Neonatal sepsis is a severe bacterial infection that can lead to life-threatening complications in newborns. Pseudomonas extremorientalis is a Gram-negative bacterium and these Gram-negative organisms have been identified as a major cause of neonatal sepsis. The virulence factors produced by this bacterium play a crucial role in its pathogenicity. Therefore, targeting these virulence factors could be a potential strategy to treat neonatal sepsis caused by P. extremorientalis. In this study, we investigated the efficacy of 3-(bromoacetyl) coumarin (3-BC) in reducing the virulence factors of P. extremorientalis strains isolated from neonatal sepsis. Our results showed that 3-BC effectively reduced the production of various virulence factors, including protease, elastase, siderophore, and exopolysaccharide in these strains. Furthermore, at a concentration of 125 µg/ml, 3-BC also inhibited the biofilm formation ability of these strains in combination with ciprofloxacin. It was discovered that 3-BC was functionally effective in protecting C. elegans against bacterial infection. Moreover, the in vitro and in vivo outcomes were strongly correlated with docking studies of various activator proteins. Overall, our findings suggest that 3-BC could be a potential therapeutic agent for the treatment of neonatal sepsis caused by P. extremorientalis. Further studies are needed to explore the mechanism of action of 3-BC and its potential use in clinical settings.
Collapse
Affiliation(s)
- Shukla Banerjee
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangaluru, 575018, India
| | - Sukesh Kumar Bajire
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangaluru, 575018, India
| | - H K Mithun
- Department of Pediatrics, Yenepoya Medical College Hospital, Yenepoya (Deemed to be University), Deralakatte, Mangaluru, 575018, India
| | - Rajesh P Shastry
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangaluru, 575018, India.
| |
Collapse
|
19
|
Anilkumar A, Batra A, Talukder S, Sharma R. Microfluidics based bioimaging with cost-efficient fabrication of multi-level micrometer-sized trenches. BIOMICROFLUIDICS 2023; 17:034103. [PMID: 37334275 PMCID: PMC10275646 DOI: 10.1063/5.0151868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 05/31/2023] [Indexed: 06/20/2023]
Abstract
Microfluidic devices, through their vast applicability as tools for miniaturized experimental setups, have become indispensable for cutting edge research and diagnostics. However, the high operational cost and the requirement of sophisticated equipment and clean room facility for the fabrication of these devices make their use unfeasible for many research laboratories in resource limited settings. Therefore, with the aim of increasing accessibility, in this article, we report a novel, cost-effective micro-fabrication technique for fabricating multi-layer microfluidic devices using only common wet-lab facilities, thereby significantly lowering the cost. Our proposed process-flow-design eliminates the need for a mastermold, does not require any sophisticated lithography tools, and can be executed successfully outside a clean room. In this work, we also optimized the critical steps (such as spin coating and wet etching) of our fabrication process and validated the process flow and the device by trapping and imaging Caenorhabditis elegans. The fabricated devices are effective in conducting lifetime assays and flushing out larvae, which are, in general, manually picked from Petri dishes or separated using sieves. Our technique is not only cost effective but also scalable, as it can be used to fabricate devices with multiple layers of confinements ranging from 0.6 to more than 50 μ m, thus enabling the study of unicellular and multicellular organisms. This technique, therefore, has the potential to be adopted widely by many research laboratories for a variety of applications.
Collapse
Affiliation(s)
- Anand Anilkumar
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Bhopal 462066, Madhya Pradesh, India
| | - Abhilasha Batra
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Bhopal 462066, Madhya Pradesh, India
| | - Santanu Talukder
- Department of Electrical Engineering and Computer Science, Indian Institute of Science Education and Research (IISER), Bhopal 462066, Madhya Pradesh, India
| | - Rati Sharma
- Department of Chemistry, Indian Institute of Science Education and Research (IISER), Bhopal 462066, Madhya Pradesh, India
| |
Collapse
|
20
|
Migliori ML, Goya ME, Lamberti ML, Silva F, Rota R, Bénard C, Golombek DA. Caenorhabditis elegans as a Promising Model Organism in Chronobiology. J Biol Rhythms 2023; 38:131-147. [PMID: 36680418 DOI: 10.1177/07487304221143483] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Circadian rhythms represent an adaptive feature, ubiquitously found in nature, which grants living beings the ability to anticipate daily variations in their environment. They have been found in a multitude of organisms, ranging from bacteria to fungi, plants, and animals. Circadian rhythms are generated by endogenous clocks that can be entrained daily by environmental cycles such as light and temperature. The molecular machinery of circadian clocks includes a transcriptional-translational feedback loop that takes approximately 24 h to complete. Drosophila melanogaster has been a model organism of choice to understand the molecular basis of circadian clocks. However, alternative animal models are also being adopted, each offering their respective experimental advantages. The nematode Caenorhabditis elegans provides an excellent model for genetics and neuro-behavioral studies, which thanks to its ease of use and manipulation, as well as availability of genetic data and mutant strains, is currently used as a novel model for circadian research. Here, we aim to evaluate C. elegans as a model for chronobiological studies, focusing on its strengths and weaknesses while reviewing the available literature. Possible zeitgebers (including light and temperature) are also discussed. Determining the molecular bases and the neural circuitry involved in the central pacemaker of the C. elegans' clock will contribute to the understanding of its circadian system, becoming a novel model organism for the study of diseases due to alterations of the circadian cycle.
Collapse
Affiliation(s)
- María Laura Migliori
- Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Argentina
| | - María Eugenia Goya
- European Institute for the Biology of Aging, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Francisco Silva
- Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Argentina
| | - Rosana Rota
- Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Argentina
| | - Claire Bénard
- Department of Biological Sciences, CERMO-FC Research Center, Universite du Québec à Montréal, Montreál, QC, Canada
| | - Diego Andrés Golombek
- Departamento de Ciencia y Tecnología, Universidad Nacional de Quilmes, Bernal, Argentina
- Universidad de San Andrés, Victoria, Argentina
| |
Collapse
|
21
|
Mele S, Martelli F, Lin J, Kanca O, Christodoulou J, Bellen HJ, Piper MDW, Johnson TK. Drosophila as a diet discovery tool for treating amino acid disorders. Trends Endocrinol Metab 2023; 34:85-105. [PMID: 36567227 DOI: 10.1016/j.tem.2022.12.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 12/06/2022] [Indexed: 12/24/2022]
Abstract
Amino acid disorders (AADs) are a large group of rare inherited conditions that collectively impact one in 6500 live births, often resulting in rapid neurological decline and death during infancy. For several AADs, including phenylketonuria, dietary modification prevents physiological deterioration and ameliorates symptoms. Despite this remarkable potential for treatment success, dietary therapy for most AADs remains largely unexplored. Although animal models have provided novel insights into AAD mechanisms, few have been used for therapeutic diet discovery. Here, we find that of all the animal models, Drosophila is particularly well suited for nutrigenomic disease modelling, having amino acid pathways conserved with humans, exceptional genetic tractability, and the unique availability of a synthetic customisable diet.
Collapse
Affiliation(s)
- Sarah Mele
- School of Biological Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - Felipe Martelli
- School of Biological Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - Jiayi Lin
- School of Biological Sciences, Monash University, Clayton, Victoria 3800, Australia
| | - Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Duncan Neurological Research Institute of Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - John Christodoulou
- Murdoch Children's Research Institute, Parkville, Australia; Department of Paediatrics, University of Melbourne, Melbourne, Victoria 3052, Australia
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Duncan Neurological Research Institute of Texas Children's Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Matthew D W Piper
- School of Biological Sciences, Monash University, Clayton, Victoria 3800, Australia.
| | - Travis K Johnson
- School of Biological Sciences, Monash University, Clayton, Victoria 3800, Australia.
| |
Collapse
|
22
|
Davies M, Jurynec MJ, Gomez-Alvarado F, Hu D, Feeley SE, Allen-Brady K, Tashjian RZ, Feeley BT. Current cellular and molecular biology techniques for the orthopedic surgeon-scientist. J Shoulder Elbow Surg 2023; 32:e11-e22. [PMID: 35988889 DOI: 10.1016/j.jse.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 06/27/2022] [Accepted: 07/07/2022] [Indexed: 02/01/2023]
Affiliation(s)
- Michael Davies
- Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Michael J Jurynec
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA
| | - Francisco Gomez-Alvarado
- Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Daniel Hu
- Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Sonali E Feeley
- Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Kristina Allen-Brady
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Robert Z Tashjian
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA.
| | - Brian T Feeley
- Department of Orthopedic Surgery, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
23
|
Peng H, Bai H, Pan Y, Li J, Pei Z, Liao Y, Wu C, Li C, Tao L, Zhong S, Ma C, Chen Z, Li X, Gong Y, Wang L, Li F. Immunological pathogenesis of Bovine E. coli infection in a model of C. elegans. BMC Microbiol 2022; 22:311. [PMID: 36539715 PMCID: PMC9764636 DOI: 10.1186/s12866-022-02733-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 09/06/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Cattle industry is critical for China's livestock industry, whereas E. coli infection and relevant diseases could lead huge economic loss. Traditional mammalian models would be costly, time consuming and complicated to study pathological changes of bovine E. coli. There is an urgent need for a simple but efficient animal model to quantitatively evaluate the pathological changes of bovine-derived E. coli in vivo. Caenorhabditis elegans (C. elegans) has a broad host range of diverse E. coli strains with advantages, including a short life cycle, a simple structure, a transparent body which is easily visualized, a well-studied genetic map, an intrinsic immune system which is conservable with more complicated mammalians. RESULTS Here, we considered that O126 was the dominant serotype, and a total of 19 virulence factors were identified from 41 common E. coli virulence factors. Different E. coli strains with diverse pathogenicity strengths were tested in C. elegans in E. coli with higher pathogenicity (EC3/10), Nsy-1, Sek-1 and Pmk-1 of the p38 MAPK signaling pathway cascade and the expression of the antimicrobial peptides Abf-3 and Clec-60 were significantly up-regulated comparing with other groups. E. coli with lower pathogenicity (EC5/13) only activated the expression of Nsy-1 and Sek-1 genes in the p38 MAPK signaling pathway, Additionally, both groups of E. coli strains caused significant upregulation of the antimicrobial peptide Spp-1. CONCLUSION Thirteen E. coli strains showed diverse pathogenicity in nematodes and the detection rate of virulence factors did not corresponding to the virulence in nematodes, indicating complex pathogenicity mechanisms. We approved that C. elegans is a fast and convenient detection model for pathogenic bacteria virulence examinations.
Collapse
Affiliation(s)
- Hao Peng
- grid.418337.aGuangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001 China
| | - Huili Bai
- grid.418337.aGuangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001 China
| | - Yan Pan
- Guangxi Agricultural Vocational University, Nanning, China
| | - Jun Li
- grid.418337.aGuangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001 China
| | - Zhe Pei
- grid.254250.40000 0001 2264 7145The City College of New York, New York, USA
| | - Yuying Liao
- grid.418337.aGuangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001 China
| | - Cuilan Wu
- grid.418337.aGuangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001 China
| | - Changting Li
- grid.418337.aGuangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001 China
| | - Li Tao
- grid.418337.aGuangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001 China
| | - Shuhong Zhong
- grid.418337.aGuangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001 China
| | - Chunxia Ma
- grid.418337.aGuangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001 China
| | - Zhongwei Chen
- grid.418337.aGuangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001 China
| | - Xiaoning Li
- grid.418337.aGuangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001 China
| | - Yu Gong
- Animal Science and Technology Station of Guizhou, Guiyang, China
| | - Leping Wang
- grid.418337.aGuangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001 China
| | - Fengsheng Li
- grid.418337.aGuangxi Key Laboratory of Veterinary Biotechnology, Guangxi Veterinary Research Institute, Nanning, 530001 China
| |
Collapse
|
24
|
Sridhar N, Fajrial AK, Doser RL, Hoerndli FJ, Ding X. Surface acoustic wave microfluidics for repetitive and reversible temporary immobilization of C. elegans. LAB ON A CHIP 2022; 22:4882-4893. [PMID: 36377422 PMCID: PMC10091851 DOI: 10.1039/d2lc00737a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Caenorhabditis elegans is an important genetic model for neuroscience studies, used for analyses of how genes control connectivity, neuronal function, and behavior. To date, however, most studies of neuronal function in C. elegans are incapable of obtaining microscopy imaging with subcellular resolution and behavior analysis in the same set of animals. This constraint stems from the immobilization requirement for high-resolution imaging that is incompatible with behavioral analysis using conventional immobilization techniques. Here, we present a novel microfluidic device that uses surface acoustic waves (SAW) as a non-contact method to temporarily immobilize worms for a short period (30 seconds). We optimize the SAW based protocol for rapid switching between free-swimming and immobilized states, facilitating non-invasive analysis of swimming behavior as well as high-resolution synaptic imaging in the same animal. We find that the coupling of heat and acoustic pressure play a key role in the immobilization process. We introduce a proof-of-concept longitudinal study, illustrating that the device enables repeated imaging of fluorescently tagged synaptic receptors in command interneurons and analysis of swimming behavior in the same animals for three days. This longitudinal approach provides the first correlative analysis of synaptic glutamatergic receptors and swimming behavior in aging animals. We anticipate that this device will enable further longitudinal analysis of animal motility and subcellular morphological changes during development and aging in C. elegans.
Collapse
Affiliation(s)
- Nakul Sridhar
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado, USA.
| | - Apresio Kefin Fajrial
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado, USA.
| | - Rachel L Doser
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA.
| | - Frederic J Hoerndli
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA.
| | - Xiaoyun Ding
- Paul M. Rady Department of Mechanical Engineering, University of Colorado Boulder, Boulder, Colorado, USA.
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, Colorado, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, Colorado, USA
| |
Collapse
|
25
|
Lim SYM, Alshagga M, Kong C, Alshawsh MA, Alshehade SA, Pan Y. CYP35 family in Caenorhabditis elegans biological processes: fatty acid synthesis, xenobiotic metabolism, and stress responses. Arch Toxicol 2022; 96:3163-3174. [PMID: 36175686 DOI: 10.1007/s00204-022-03382-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/14/2022] [Indexed: 01/08/2023]
Abstract
With more than 80 cytochrome P450 (CYP) encoding genes found in the nematode Caenorhabditis elegans (C. elegans), the cyp35 genes are one of the important genes involved in many biological processes such as fatty acid synthesis and storage, xenobiotic stress response, dauer and eggshell formation, and xenobiotic metabolism. The C. elegans CYP35 subfamily consisted of A, B, C, and D, which have the closest homolog to human CYP2 family. C. elegans homologs could answer part of the hunt for human disease genes. This review aims to provide an overview of CYP35 in C. elegans and their human homologs, to explore the roles of CYP35 in various C. elegans biological processes, and how the genes of cyp35 upregulation or downregulation are influenced by biological processes, upon exposure to xenobiotics or changes in diet and environment. The C. elegans CYP35 gene expression could be upregulated by heavy metals, pesticides, anti-parasitic and anti-chemotherapeutic agents, polycyclic aromatic hydrocarbons (PAHs), nanoparticles, drugs, and organic chemical compounds. Among the cyp35 genes, cyp-35A2 is involved in most of the C. elegans biological processes regulation. Further venture of cyp35 genes, the closest homolog of CYP2 which is the largest family of human CYPs, may have the power to locate cyps gene targets, discovery of novel therapeutic strategies, and possibly a successful medical regime to combat obesity, cancers, and cyps gene-related diseases.
Collapse
Affiliation(s)
- Sharoen Yu Ming Lim
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, 43500, Semenyih, Malaysia.
| | - Mustafa Alshagga
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, 43500, Semenyih, Malaysia
| | - Cin Kong
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, 43500, Semenyih, Malaysia
| | | | | | - Yan Pan
- Division of Biomedical Sciences, School of Pharmacy, University of Nottingham Malaysia, 43500, Semenyih, Malaysia
| |
Collapse
|
26
|
Abdullahi M, Li X, Abdallah MAE, Stubbings W, Yan N, Barnard M, Guo LH, Colbourne JK, Orsini L. Daphnia as a Sentinel Species for Environmental Health Protection: A Perspective on Biomonitoring and Bioremediation of Chemical Pollution. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:14237-14248. [PMID: 36169655 PMCID: PMC9583619 DOI: 10.1021/acs.est.2c01799] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Indexed: 05/14/2023]
Abstract
Despite available technology and the knowledge that chemical pollution damages human and ecosystem health, chemical pollution remains rampant, ineffectively monitored, rarely prevented, and only occasionally mitigated. We present a framework that helps address current major challenges in the monitoring and assessment of chemical pollution by broadening the use of the sentinel species Daphnia as a diagnostic agent of water pollution. And where prevention has failed, we propose the application of Daphnia as a bioremediation agent to help reduce hazards from chemical mixtures in the environment. By applying "omics" technologies to Daphnia exposed to real-world ambient chemical mixtures, we show improvements at detecting bioactive components of chemical mixtures, determining the potential effects of untested chemicals within mixtures, and identifying targets of toxicity. We also show that using Daphnia strains that naturally adapted to chemical pollution as removal agents of ambient chemical mixtures can sustainably improve environmental health protection. Expanding the use of Daphnia beyond its current applications in regulatory toxicology has the potential to improve both the assessment and the remediation of environmental pollution.
Collapse
Affiliation(s)
- Muhammad Abdullahi
- Environmental
Genomics Group, School of Biosciences, the
University of Birmingham, Birmingham B15 2TT, U.K.
| | - Xiaojing Li
- Environmental
Genomics Group, School of Biosciences, the
University of Birmingham, Birmingham B15 2TT, U.K.
| | | | - William Stubbings
- School
of Geography, Earth and Environmental Sciences, the University of Birmingham, Birmingham B15 2TT, U.K.
| | - Norman Yan
- Department
of Biology, York University, and Friends of the Muskoka Watershed, Bracebridge, Ontario P1L 1T7, Canada
| | - Marianne Barnard
- Environmental
Genomics Group, School of Biosciences, the
University of Birmingham, Birmingham B15 2TT, U.K.
| | - Liang-Hong Guo
- Institute
of Environmental and Health Sciences, China
Jiliang University, 258 Xueyuan Street, Hangzhou, Zhejiang 310018, People’s Republic of China
| | - John K. Colbourne
- Environmental
Genomics Group, School of Biosciences, the
University of Birmingham, Birmingham B15 2TT, U.K.
| | - Luisa Orsini
- Environmental
Genomics Group, School of Biosciences, the
University of Birmingham, Birmingham B15 2TT, U.K.
- The
Alan Turing Institute, British Library, 96 Euston Road, London NW1 2DB, U.K.
| |
Collapse
|
27
|
van Es-Remers M, Spadaro JA, Poppelaars E, Kim HK, van Haaster M, de Wit M, ILiopoulou E, Wildwater M, Korthout H. C. elegans as a test system to study relevant compounds that contribute to the specific health-related effects of different cannabis varieties. J Cannabis Res 2022; 4:53. [PMID: 36184617 PMCID: PMC9528106 DOI: 10.1186/s42238-022-00162-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 09/21/2022] [Indexed: 11/05/2022] Open
Abstract
Background The medicinal effects of cannabis varieties on the market cannot be explained solely by the presence of the major cannabinoids Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD). Evidence for putative entourage effects caused by other compounds present in cannabis is hard to obtain due to the subjective nature of patient experience data. Caenorhabditis elegans (C. elegans) is an objective test system to identify cannabis compounds involved in claimed health and entourage effects. Methods From a medicinal cannabis breeding program by MariPharm BV, the Netherlands a set of 12 varieties were selected both THC rich varieties as well as CBD rich varieties. A consecutive extraction process was applied resulting in a non-polar (cannabinoid-rich) and polar (cannabinoid-poor) extract of each variety. The test model C. elegans was exposed to these extracts in a broad set of bioassays for appetite control, body oscillation, motility, and nervous system function. Results Exposing C. elegans to extracts with a high concentration of cannabinoids (> 1 μg/mL) reduces the life span of C. elegans dramatically. Exposing the nematodes to the low-cannabinoid (< 0.005 μg/mL) polar extracts, however, resulted in significant effects with respect to appetite control, body oscillation, motility, and nervous system-related functions in a dose-dependent and variety-dependent manner. Discussion C. elegans is a small, transparent organism with a complete nervous system, behavior and is due to its genetic robustness and short life cycle highly suitable to unravel entourage effects of Cannabis compounds. Although C. elegans lacks an obvious CB1 and CB2 receptor it has orthologs of Serotonin and Vanilloid receptor which are also involved in (endo)cannabinoid signaling. Conclusion By using C. elegans, we were able to objectively distinguish different effects of different varieties despite the cannabinoid content. C. elegans seems a useful test system for studying entourage effects, for targeted medicinal cannabis breeding programs and product development. Supplementary Information The online version contains supplementary material available at 10.1186/s42238-022-00162-9.
Collapse
Affiliation(s)
| | | | - Eefje Poppelaars
- Vivaltes B.V., Bunnik, Regulierenring 9, 3981 LA Bunnik the Netherlands
| | - Hye Kyong Kim
- Fytagoras B.V., Sylviusweg 72, 2333 BE Leiden, the Netherlands
| | - Marieke van Haaster
- Maripham B.V., Nieuw-Mathenesserstraat 33, 3029 AV Rotterdam, the Netherlands
| | - Marcel de Wit
- Maripham B.V., Nieuw-Mathenesserstraat 33, 3029 AV Rotterdam, the Netherlands
| | - Eva ILiopoulou
- Vivaltes B.V., Bunnik, Regulierenring 9, 3981 LA Bunnik the Netherlands
| | | | - Henrie Korthout
- Fytagoras B.V., Sylviusweg 72, 2333 BE Leiden, the Netherlands
| |
Collapse
|
28
|
Hooper FW, Morrow J, Rodriguez J, Webb C. Teaching the Applications of CRISPR/Cas9: Using the African Turquoise Killifish as a Novel Model of Aging and Age-Related Diseases. JOURNAL OF UNDERGRADUATE NEUROSCIENCE EDUCATION : JUNE : A PUBLICATION OF FUN, FACULTY FOR UNDERGRADUATE NEUROSCIENCE 2022; 20:R5-R8. [PMID: 39036719 PMCID: PMC11256378 DOI: 10.59390/xzql5300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 02/08/2024]
Abstract
The development of genome editing technologies, including the novel CRISPR/Cas9 technique, has advanced scientific research concerning the contribution of genetics to disease through the creation of new model organisms. The subject of this review is a 2015 study done by Harel et al. from the journal Cell. This study is a prime example of using CRISPR/Cas9 to create a new model organism to accurately model the effects of aging and age-related diseases on a short-lived vertebrate. This study found that the African turquoise killifish is a reliable model to study the physiological process of aging due to its compressed lifespan. In addition, it provides a genotype-to-phenotype platform to study genes related to the hallmarks of aging and age-related diseases. This paper demonstrates this by showing that killifish deficient in the protein subunit of telomerase display telomerase-related pathologies faster than other established vertebrate models. From a teaching perspective, this paper could be used as a resource for educators to teach students about new technologies emerging in the field of neuroscience and the importance of model organisms. Specifically, for upper-level undergraduate students, this paper could serve as a real-world example of how scientific techniques such as CRISPR/Cas9 could be used to answer scientific questions. Further, it shows how these techniques could bring forward new model organisms better suited to answer the scientific questions being asked. Learning these techniques and being open minded to new approaches will be advantageous to students' future careers in science.
Collapse
Affiliation(s)
- Frances W Hooper
- Department of Psychology and Neuroscience, University of St Andrews, St Andrews, UK KY16 9JP
| | - Jonathan Morrow
- Department of Psychology and Neuroscience, University of St Andrews, St Andrews, UK KY16 9JP
| | - Jasmine Rodriguez
- Department of Psychology and Neuroscience, University of St Andrews, St Andrews, UK KY16 9JP
| | - Claire Webb
- Department of Psychology and Neuroscience, University of St Andrews, St Andrews, UK KY16 9JP
| |
Collapse
|
29
|
Cheng KC, Burdine RD, Dickinson ME, Ekker SC, Lin AY, Lloyd KCK, Lutz CM, MacRae CA, Morrison JH, O'Connor DH, Postlethwait JH, Rogers CD, Sanchez S, Simpson JH, Talbot WS, Wallace DC, Weimer JM, Bellen HJ. Promoting validation and cross-phylogenetic integration in model organism research. Dis Model Mech 2022; 15:dmm049600. [PMID: 36125045 PMCID: PMC9531892 DOI: 10.1242/dmm.049600] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Model organism (MO) research provides a basic understanding of biology and disease due to the evolutionary conservation of the molecular and cellular language of life. MOs have been used to identify and understand the function of orthologous genes, proteins, cells and tissues involved in biological processes, to develop and evaluate techniques and methods, and to perform whole-organism-based chemical screens to test drug efficacy and toxicity. However, a growing richness of datasets and the rising power of computation raise an important question: How do we maximize the value of MOs? In-depth discussions in over 50 virtual presentations organized by the National Institutes of Health across more than 10 weeks yielded important suggestions for improving the rigor, validation, reproducibility and translatability of MO research. The effort clarified challenges and opportunities for developing and integrating tools and resources. Maintenance of critical existing infrastructure and the implementation of suggested improvements will play important roles in maintaining productivity and facilitating the validation of animal models of human biology and disease.
Collapse
Affiliation(s)
- Keith C. Cheng
- Department of Pathology, Penn State College of Medicine, Hershey, PA 17033, USA
- Institute for Computational and Data Sciences, Pennsylvania State University, Park, PA 16802, USA
| | - Rebecca D. Burdine
- Department of Molecular Biology, Princeton University, Princeton, NJ 08540, USA
| | - Mary E. Dickinson
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77007, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77007, USA
| | - Stephen C. Ekker
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55906, USA
| | - Alex Y. Lin
- Department of Pathology, Penn State College of Medicine, Hershey, PA 17033, USA
| | - K. C. Kent Lloyd
- Mouse Biology Program, School of Medicinel, University of California Davis, Davis, CA 95618, USA
- Department of Surgery, School of Medicine, University of California Davis, Davis, CA 95618, USA
| | - Cathleen M. Lutz
- The Jackson Laboratory, Genetic Resource Science, Bar Harbor, ME 04609, USA
| | - Calum A. MacRae
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, 360 Longwood Avenue, Boston, MA 02215, USA
| | - John H. Morrison
- California National Primate Research Center, University of California Davis, Davis, CA 95616, USA
- Department of Neurology, University of California Davis, Davis, CA 95616, USA
| | - David H. O'Connor
- Department of Pathology and Laboratory Medicine, University ofWisconsin-Madison, Madison, WI 53711, USA
| | | | - Crystal D. Rogers
- School of Veterinary Medicine, University of California Davis, Davis, CA 95616, USA
| | - Susan Sanchez
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA
| | - Julie H. Simpson
- Department of Molecular, Cell and Developmental Biology, University of California, Santa Barbara, CA 93117, USA
| | - William S. Talbot
- Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA
| | - Douglas C. Wallace
- Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Jill M. Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD 57104, USA
| | - Hugo J. Bellen
- Department of Molecular and Human Genetics, Neurological Research Institute (TCH), Baylor College of Medicine, Houston, TX 77007, USA
| |
Collapse
|
30
|
Weinkove D, Zavagno G. Applying C. elegans to the Industrial Drug Discovery Process to Slow Aging. FRONTIERS IN AGING 2022; 2:740582. [PMID: 35821999 PMCID: PMC9261450 DOI: 10.3389/fragi.2021.740582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/29/2021] [Indexed: 01/29/2023]
Abstract
The increase in our molecular understanding of the biology of aging, coupled with a recent surge in investment, has led to the formation of several companies developing pharmaceuticals to slow aging. Research using the tiny nematode worm Caenorhabditis elegans was the first to show that mutations in single genes can extend lifespan, and subsequent research has shown that this model organism is uniquely suited to testing interventions to slow aging. Yet, with a few notable exceptions, C. elegans is not in the standard toolkit of longevity companies. Here we discuss the paths to overcome the barriers to using C. elegans in industrial drug discovery. We address the predictive power of C. elegans for human aging, how C. elegans research can be applied to specific challenges in the typical drug discovery pipeline, and how standardised and quantitative assays will help C. elegans fulfil its potential in the biotech and pharmaceutical industry. We argue that correct application of this model and its knowledge base will significantly accelerate progress to slow human aging.
Collapse
Affiliation(s)
- David Weinkove
- Department of Biosciences, Durham University, Durham, United Kingdom.,Magnitude Biosciences Ltd., NETpark Plexus, Sedgefield, United Kingdom
| | - Giulia Zavagno
- Department of Biosciences, Durham University, Durham, United Kingdom.,Magnitude Biosciences Ltd., NETpark Plexus, Sedgefield, United Kingdom
| |
Collapse
|
31
|
The great small organisms of developmental genetics: Caenorhabditis elegans and Drosophila melanogaster. Dev Biol 2022; 485:93-122. [PMID: 35247454 PMCID: PMC9092520 DOI: 10.1016/j.ydbio.2022.02.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 12/30/2022]
Abstract
Experimental embryologists working at the turn of the 19th century suggested fundamental mechanisms of development, such as localized cytoplasmic determinants and tissue induction. However, the molecular basis underlying these processes proved intractable for a long time, despite concerted efforts in many developmental systems to isolate factors with a biological role. That road block was overcome by combining developmental biology with genetics. This powerful approach used unbiased genome-wide screens to isolate mutants with developmental defects and to thereby identify genes encoding key determinants and regulatory pathways that govern development. Two small invertebrates were the pioneers: the fruit fly Drosophila melanogaster and the nematode Caenorhabditis elegans. Their modes of development differ in many ways, but the two together led the way to unraveling the molecular mechanisms of many fundamental developmental processes. The discovery of the grand homologies between key players in development throughout the animal kingdom underscored the usefulness of studying these small invertebrate models for animal development and even human disease. We describe developmental genetics in Drosophila and C. elegans up to the rise of genomics at the beginning of the 21st Century. Finally, we discuss themes that emerge from the histories of such distinct organisms and prospects of this approach for the future.
Collapse
|
32
|
Mello DF, Bergemann CM, Fisher K, Chitrakar R, Bijwadia SR, Wang Y, Caldwell A, Baugh LR, Meyer JN. Rotenone Modulates Caenorhabditis elegans Immunometabolism and Pathogen Susceptibility. Front Immunol 2022; 13:840272. [PMID: 35273616 PMCID: PMC8902048 DOI: 10.3389/fimmu.2022.840272] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/31/2022] [Indexed: 11/21/2022] Open
Abstract
Mitochondria are central players in host immunometabolism as they function not only as metabolic hubs but also as signaling platforms regulating innate immunity. Environmental exposures to mitochondrial toxicants occur widely and are increasingly frequent. Exposures to these mitotoxicants may pose a serious threat to organismal health and the onset of diseases by disrupting immunometabolic pathways. In this study, we investigated whether the Complex I inhibitor rotenone could alter C. elegans immunometabolism and disease susceptibility. C. elegans embryos were exposed to rotenone (0.5 µM) or DMSO (0.125%) until they reached the L4 larval stage. Inhibition of mitochondrial respiration by rotenone and disruption of mitochondrial metabolism were evidenced by rotenone-induced detrimental effects on mitochondrial efficiency and nematode growth and development. Next, through transcriptomic analysis, we investigated if this specific but mild mitochondrial stress that we detected would lead to the modulation of immunometabolic pathways. We found 179 differentially expressed genes (DEG), which were mostly involved in detoxification, energy metabolism, and pathogen defense. Interestingly, among the down-regulated DEG, most of the known genes were involved in immune defense, and most of these were identified as commonly upregulated during P. aeruginosa infection. Furthermore, rotenone increased susceptibility to the pathogen Pseudomonas aeruginosa (PA14). However, it increased resistance to Salmonella enterica (SL1344). To shed light on potential mechanisms related to these divergent effects on pathogen resistance, we assessed the activation of the mitochondrial unfolded protein response (UPRmt), a well-known immunometabolic pathway in C. elegans which links mitochondria and immunity and provides resistance to pathogen infection. The UPRmt pathway was activated in rotenone-treated nematodes further exposed for 24 h to the pathogenic bacteria P. aeruginosa and S. enterica or the common bacterial food source Escherichia coli (OP50). However, P. aeruginosa alone suppressed UPRmt activation and rotenone treatment rescued its activation only to the level of DMSO-exposed nematodes fed with E. coli. Module-weighted annotation bioinformatics analysis was also consistent with UPRmt activation in rotenone-exposed nematodes consistent with the UPR being involved in the increased resistance to S. enterica. Together, our results demonstrate that the mitotoxicant rotenone can disrupt C. elegans immunometabolism in ways likely protective against some pathogen species but sensitizing against others.
Collapse
Affiliation(s)
- Danielle F. Mello
- Nicholas School of the Environment, Duke University, Durham, NC, United States
| | | | - Kinsey Fisher
- Department of Biology, Duke University, Durham, NC, United States
| | - Rojin Chitrakar
- Department of Biology, Duke University, Durham, NC, United States
| | - Shefali R. Bijwadia
- Nicholas School of the Environment, Duke University, Durham, NC, United States
| | - Yang Wang
- Nicholas School of the Environment, Duke University, Durham, NC, United States
| | - Alexis Caldwell
- Nicholas School of the Environment, Duke University, Durham, NC, United States
| | - Larry Ryan Baugh
- Department of Biology, Duke University, Durham, NC, United States
- Center for Genomic and Computational Biology, Duke University, Durham, NC, United States
| | - Joel N. Meyer
- Nicholas School of the Environment, Duke University, Durham, NC, United States
| |
Collapse
|
33
|
Wang Y, Sun Y, Wang X, Wang Y, Liao L, Zhang Y, Fang B, Fu Y. Novel antioxidant peptides from Yak bones collagen enhanced the capacities of antiaging and antioxidant in Caenorhabditis elegans. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.104933] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
34
|
Tang X, Zhao Y, Liu Y, Liu Y, Liu Y, Niu F, Fang F. 3,6'-disinapoyl sucrose attenuates Aβ 1-42 - induced neurotoxicity in Caenorhabditis elegans by enhancing antioxidation and regulating autophagy. J Cell Mol Med 2022; 26:1024-1033. [PMID: 35044105 PMCID: PMC8831957 DOI: 10.1111/jcmm.17153] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 02/06/2023] Open
Abstract
The aggregation of β‐amyloid (Aβ) has the neurotoxicity, which is thought to play critical role in the pathogenesis of Alzheimer's disease (AD). Inhibiting Aβ deposition and neurotoxicity has been considered as an important strategy for AD treatment. 3,6'‐Disinapoyl sucrose (DISS), one of the oligosaccharide esters derived from traditional Chinese medicine Polygalae Radix, possesses antioxidative activity, neuroprotective effect and anti‐depressive activity. This study was to explore whether DISS could attenuate the pathological changes of Aβ1‐42 transgenic Caenorhabditis elegans (C. elegans). The results showed that DISS (5 and 50 μM) treatment significantly prolonged the life span, increased the number of egg‐laying, reduced paralysis rate, decreased the levels of lipofuscin and ROS and attenuated Aβ deposition in Aβ1‐42 transgenic C. elegans. Gene analysis showed that DISS could up‐regulate the mRNA expression of sod‐3, gst‐4, daf‐16, bec‐1 and lgg‐1, while down‐regulate the mRNA expression of daf‐2 and daf‐15 in Aβ1‐42 transgenic C. elegans. These results suggested that DISS has the protective effect against Aβ1‐42‐induced pathological damages and prolongs the life span of C. elegans, which may be related to the reduction of Aβ deposition and neurotoxicity by regulating expression of genes related to antioxidation and autophagy.
Collapse
Affiliation(s)
- Xiaoli Tang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yuming Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yanan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Liu
- State Key Lab for Structural Chemistry of Unstable and Stable Species, Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Yue Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Fenxi Niu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Fang Fang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
35
|
Nieto-Torres JL, Hansen M. Macroautophagy and aging: The impact of cellular recycling on health and longevity. Mol Aspects Med 2021; 82:101020. [PMID: 34507801 PMCID: PMC8671213 DOI: 10.1016/j.mam.2021.101020] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/11/2021] [Accepted: 08/31/2021] [Indexed: 02/08/2023]
Abstract
Aging is associated with many deleterious changes at the cellular level, including the accumulation of potentially toxic components that can have devastating effects on health. A key protective mechanism to this end is the cellular recycling process called autophagy. During autophagy, damaged or surplus cellular components are delivered to acidic vesicles called lysosomes, that secure degradation and recycling of the components. Numerous links between autophagy and aging exist. Autophagy declines with age, and increasing evidence suggests that this reduction plays important roles in both physiological aging and the development of age-associated disorders. Studies in pharmacologically and genetically manipulated model organisms indicate that defects in autophagy promote age-related diseases, and conversely, that enhancement of autophagy has beneficial effects on both healthspan and lifespan. Here, we review our current understanding of the role of autophagy in different physiological processes and their molecular links with aging and age-related diseases. We also highlight some recent advances in the field that could accelerate the development of autophagy-based therapeutic interventions.
Collapse
Affiliation(s)
- Jose L Nieto-Torres
- Sanford Burnham Prebys Medical Discovery Institute. Program of Development, Aging, and Regeneration, La Jolla, CA, USA
| | - Malene Hansen
- Sanford Burnham Prebys Medical Discovery Institute. Program of Development, Aging, and Regeneration, La Jolla, CA, USA.
| |
Collapse
|
36
|
Behrouzi M, Youssef K, Rezai P, Tabatabaei N. Low-cost optofluidic add-on enables rapid selective plane illumination microscopy of C. elegans with a conventional wide-field microscope. JOURNAL OF BIOMEDICAL OPTICS 2021; 26:JBO-210275RR. [PMID: 34894114 PMCID: PMC8664272 DOI: 10.1117/1.jbo.26.12.126501] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/22/2021] [Indexed: 06/14/2023]
Abstract
SIGNIFICANCE Selective plane illumination microscopy (SPIM) is an emerging fluorescent imaging technique suitable for noninvasive volumetric imaging of C. elegans. These promising microscopy systems, however, are scarce in academic and research institutions due to their high cost and technical complexities. Simple and low-cost solutions that enable conversion of commonplace wide-field microscopes to rapid SPIM platforms promote widespread adoption of SPIM by biologist for studying neuronal expressions of C. elegans. AIM We sought to develop a simple and low-cost optofluidic add-on device that enables rapid and immobilization-free volumetric SPIM imaging of C. elegans with conventional fluorescent microscopes. APPROACH A polydimethylsiloxane (PDMS)-based device with integrated optical and fluidic elements was developed as a low-cost and miniaturized SPIM add-on for the conventional wide-field microscope. The developed optofluidic chip contained an integrated PDMS cylindrical lens for on-chip generation of the light-sheet across a microchannel. Cross-sectional SPIM images of C. elegans were continuously acquired by the native objective of microscope as worms flowed in an L-shape microchannel and through the light sheet. RESULTS On-chip SPIM imaging of C. elegans strains demonstrated possibility of visualizing the entire neuronal system in few seconds at single-neuron resolution, with high contrast and without worm immobilization. Volumetric visualization of neuronal system from the acquired cross-sectional two-dimensional images is also demonstrated, enabling the standard microscope to acquire three-dimensional fluorescent images of C. elegans. The full-width at half-maximum width of the point spread function was measured as 1.1 and 2.4 μm in the lateral and axial directions, respectively. CONCLUSION The developed low-cost optofluidic device is capable of continuous SPIM imaging of C. elegans model organism with a conventional fluorescent microscope, at high speed, and with single neuron resolution.
Collapse
Affiliation(s)
- Mehran Behrouzi
- York University, Lassonde School of Engineering, Department of Mechanical Engineering, Toronto, Canada
| | - Khaled Youssef
- York University, Lassonde School of Engineering, Department of Mechanical Engineering, Toronto, Canada
| | - Pouya Rezai
- York University, Lassonde School of Engineering, Department of Mechanical Engineering, Toronto, Canada
| | - Nima Tabatabaei
- York University, Lassonde School of Engineering, Department of Mechanical Engineering, Toronto, Canada
| |
Collapse
|
37
|
Lehrbach NJ. NGLY1: Insights from C. elegans. J Biochem 2021; 171:145-152. [PMID: 34697631 DOI: 10.1093/jb/mvab112] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/01/2021] [Indexed: 01/31/2023] Open
Abstract
Peptide:N-glycanase is an evolutionarily conserved deglycosylating enzyme that catalyzes the removal of N-linked glycans from cytosolic glycoproteins. Recessive mutations that inactivate this enzyme cause NGLY1 deficiency, a multisystemic disorder with symptoms including developmental delay and defects in cognition and motor control. Developing treatments for NGLY1 deficiency will require an understanding of how failure to deglycosylate NGLY1 substrates perturbs cellular and organismal function. In this review, I highlight insights into peptide:N-glycanase biology gained by studies in the highly tractable genetic model animal C. elegans. I focus on the recent discovery of SKN-1A/Nrf1, an N-glycosylated transcription factor, as a peptide:N-glycanase substrate critical for regulation of the proteasome. I describe the elaborate post-translational mechanism that culminates in activation of SKN-1A/Nrf1 via NGLY1-dependent 'sequence editing' and discuss the implications of these findings for our understanding of NGLY1 deficiency.
Collapse
|
38
|
Abstract
A variety of effector proteins contribute to host defense in Caenorhabditis elegans. However, beyond lytic enzymes and antimicrobial peptides and proteins, little is known about the exact function of these infection-related effectors. This study set out to identify pathogen-dependent cytokine-like molecules, focusing on C-type lectin domain-containing proteins (CLECs). In total, 38 CLECs that are differentially regulated in response to bacterial infections have been previously identified by microarray and transcriptome sequencing (RNA-seq) analyses in C. elegans. We successfully cloned 18 of these 38 CLECs and chose to focus on CLEC-47 because, among these 18 cloned CLECs, it was the smallest protein and was recombinantly expressed at the highest levels in prokaryotic cells examined by SDS-PAGE. Quantitative real-time PCR (qRT-PCR/qPCR) showed that the expression of clec-47 was induced by a variety of Gram-positive bacterial pathogens, including Enterococcus faecium, Staphylococcus aureus, and Cutibacterium acnes, but was suppressed by the Gram-negative bacteria Klebsiella pneumoniae and Pseudomonas aeruginosa. By expressing CLEC-47 in HEK 293 cells, we showed that CLEC-47 is released into the culture media, which the Golgi apparatus inhibitors (brefeldin A [BFA] and GolgiStop) could block. Purified recombinant CLEC-47 (maltose binding protein [MBP]–CLEC-47–His) did not display antimicrobial activity against ESKAPE pathogen isolates but bound directly to murine macrophage J774A.1 cells. Recombinant CLEC-47 attracted and recruited J774A.1 cells in a chemotaxis assay. In addition, qPCR studies and enzyme-linked immunosorbent assays (ELISAs) showed that CLEC-47 activates J774A.1 cells in a dose- and time-dependent manner to express the proinflammatory cytokines tumor necrosis factor alpha (TNF-α), interleukin-1β (IL-1β), IL-6, and Macrophage Inflammatory Protein 2 (MIP-2). Moreover, C. elegans, fed with CLEC-47-expressing Escherichia coli, demonstrated enhanced expression of several antimicrobial proteins (CNC-1, CNC-2, CPR-1, and CPR-2) as well as the detoxification protein MTL-1. These data suggest that CLEC-47 functions as a novel cytokine-like signaling molecule and exemplify how the study of infection-related effectors in C. elegans can help elucidate the evolution of immune responses.
Collapse
|
39
|
Salem JB, Nkambeu B, Arvanitis DN, Beaudry F. Resiniferatoxin Hampers the Nocifensive Response of Caenorhabditis elegans to Noxious Heat, and Pathway Analysis Revealed that the Wnt Signaling Pathway is Involved. Neurochem Res 2021; 47:622-633. [PMID: 34694534 DOI: 10.1007/s11064-021-03471-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 10/13/2021] [Accepted: 10/18/2021] [Indexed: 10/20/2022]
Abstract
Resiniferatoxin (RTX) is a metabolite extracted from Euphorbia resinifera. RTX is a potent capsaicin analog with specific biological activities resulting from its agonist activity with the transient receptor potential channel vanilloid subfamily member 1 (TRPV1). RTX has been examined as a pain reliever, and more recently, investigated for its ability to desensitize cardiac sensory fibers expressing TRPV1 to improve chronic heart failure (CHF) outcomes using validated animal models. Caenorhabditis elegans (C. elegans) expresses orthologs of vanilloid receptors activated by capsaicin, producing antinociceptive effects. Thus, we used C. elegans to characterize the antinociceptive properties and performed proteomic profiling to uncover specific signaling networks. After exposure to RTX, wild-type (N2) and mutant C. elegans were placed on petri dishes divided into quadrants for heat stimulation. The thermal avoidance index was used to phenotype each tested C. elegans experimental group. The data revealed for the first time that RTX can hamper the nocifensive response of C. elegans to noxious heat (32 - 35 °C). The effect was reversed 6 h after RTX exposure. Additionally, we identified the RTX target, the C. elegans transient receptor potential channel OCR-3. The proteomics and pathway enrichment analysis results suggest that Wnt signaling is triggered by the agonistic effects of RTX on C. elegans vanilloid receptors.
Collapse
Affiliation(s)
- Jennifer Ben Salem
- Groupe de Recherche en Pharmacologie Animal du Québec (GREPAQ), Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, Canada.,Centre de recherche sur le cerveau et l'apprentissage (CIRCA), Université de Montréal, Montréal, Québec, Canada.,Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR1297, Université de Toulouse, Toulouse, France
| | - Bruno Nkambeu
- Groupe de Recherche en Pharmacologie Animal du Québec (GREPAQ), Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, Canada.,Centre de recherche sur le cerveau et l'apprentissage (CIRCA), Université de Montréal, Montréal, Québec, Canada
| | - Dina N Arvanitis
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM UMR1297, Université de Toulouse, Toulouse, France
| | - Francis Beaudry
- Groupe de Recherche en Pharmacologie Animal du Québec (GREPAQ), Département de Biomédecine Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, Canada. .,Centre de recherche sur le cerveau et l'apprentissage (CIRCA), Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
40
|
Gaur AV, Agarwal R. Risperidone induced alterations in feeding and locomotion behavior of Caenorhabditis elegans. Curr Res Toxicol 2021; 2:367-374. [PMID: 34806037 PMCID: PMC8585583 DOI: 10.1016/j.crtox.2021.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/14/2021] [Accepted: 10/14/2021] [Indexed: 11/19/2022] Open
Abstract
Antipsychotic drugs (APDs) are prescribed for the treatment of psychiatric illness. However, these drugs can also contribute to several developmental and behavioral disorders. Contemporary studies to evaluate the toxic effects of numerous atypical antipsychotics are reported to cause behavioral alteration at variable doses in mammals and nematodes. Risperidone, the second most prescribed drug in India, requires more exploration of its adverse effects on humans. Here, we explore effects on feeding behavior and locomotion patterns due to risperidone exposure in C. elegans model. The study targets to work out the toxic effects of risperidone exposure on feeding and locomotion behavior in addition to the expected pharmacological effects. N2 wild type strain was exposed in liquid culture assay for 2, 4, 6, 8, 10, and 12 hours with fixed 50 µM concentration. Feeding behavior was depleted due to inhibition in pharyngeal pumping varying from 11.05% - 45.67% in a time-dependent manner. Results of locomotion assay also show time-varying increase in reversals (4.9%-34.03%) and omega bends (26.23%-62.17%) with reduction in turn counts (29.07%- 42.2%) and peristaltic speed (31.38%-42.22%) amongst exposed groups as to control. The present work shows behavioral alterations due to risperidone exposure (50 µM) in C. elegans is in a time-dependent manner. The study concludes that risperidone exposure in C. elegans produces toxic effects with time, possibly caused by antagonizing other receptors apart from serotonin (5-H2T) and dopamine (D2) adding to its expected pharmacological effects.
Collapse
Key Words
- 5-H2T
- 5-HT, 5-hydroxytryptamine
- ADF, Amphid Neuron
- APDs, Antipsychotic drugs
- Antipsychotic drugs
- Behavioral alteration
- C, Control Group
- C-0h, Control Group at 0 h
- C-10h, Control Group at 10 h
- C-12h, Control Group at 12 h. E-2h, Exposure Group at 2 h
- C-2h, Control Group at 2 h
- C-4h, Control Group at 4 h
- C-6h, Control Group at 6 h
- C-8h, Control Group at 8 h
- C. elegans
- C. elegans, Caenorhabditis elegans
- D2
- D2, Dopamine Receptor 2
- E, Exposed Group
- E-10h, Exposure Group at ten
- E-12h, Exposure Group at 12 h
- E-4h, Exposure Group at 4 h
- E-6h, Exposure Group at 6 h
- E-8h, Exposure Group at 8 h
- E. coli, Escherichia coli BOD-Biochemical Oxygen Demand
- GPR, G coupled Protein Receptor
- HSN, Hermaphrodite Specific Neuron
- Min., Minutes
- N2 Wild type
- NSM, Neurosecretory Motor Neuron
- Peristaltic speed
- Pharyngeal pumping
- Reversals
- Risperidone
- SD, Standard Deviation
- SDA, Serotonin Dopamine Antagonist
- Turn counts
- omega bends
Collapse
Affiliation(s)
- Aaditya Vikram Gaur
- Laboratory of Analytical & Molecular Toxicology (Forensic Chemistry & Toxicology Laboratory), School of Forensic Science, National Forensic Sciences University, Sector 09, Gandhinagar 382007, Gujarat, India
- Forensic Science Laboratory, Kirumampakkam, Puducherry 607402, India
| | - Rakhi Agarwal
- Laboratory of Analytical & Molecular Toxicology (Forensic Chemistry & Toxicology Laboratory), School of Forensic Science, National Forensic Sciences University, Sector 09, Gandhinagar 382007, Gujarat, India
| |
Collapse
|
41
|
Pham TD. Recurrence eigenvalues of movements from brain signals. Brain Inform 2021; 8:22. [PMID: 34652546 PMCID: PMC8521564 DOI: 10.1186/s40708-021-00143-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/21/2021] [Indexed: 12/02/2022] Open
Abstract
The ability to characterize muscle activities or skilled movements controlled by signals from neurons in the motor cortex of the brain has many useful implications, ranging from biomedical perspectives to brain–computer interfaces. This paper presents the method of recurrence eigenvalues for differentiating moving patterns in non-mammalian and human models. The non-mammalian models of Caenorhabditis elegans have been studied for gaining insights into behavioral genetics and discovery of human disease genes. Systematic probing of the movement of these worms is known to be useful for these purposes. Study of dynamics of normal and mutant worms is important in behavioral genetic and neuroscience. However, methods for quantifying complexity of worm movement using time series are still not well explored. Neurodegenerative diseases adversely affect gait and mobility. There is a need to accurately quantify gait dynamics of these diseases and differentiate them from the healthy control to better understand their pathophysiology that may lead to more effective therapeutic interventions. This paper attempts to explore the potential application of the method for determining the largest eigenvalues of convolutional fuzzy recurrence plots of time series for measuring the complexity of moving patterns of Caenorhabditis elegans and neurodegenerative disease subjects. Results obtained from analyses demonstrate that the largest recurrence eigenvalues can differentiate phenotypes of behavioral dynamics between wild type and mutant strains of Caenorhabditis elegans; and walking patterns among healthy control subjects and patients with Parkinson’s disease, Huntington’s disease, or amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Tuan D Pham
- Center for Artificial Intelligence, Prince Mohammad Bin Fahd University, Khobar, Saudi Arabia.
| |
Collapse
|
42
|
Wang J, Cao H. Zebrafish and Medaka: Important Animal Models for Human Neurodegenerative Diseases. Int J Mol Sci 2021; 22:10766. [PMID: 34639106 PMCID: PMC8509648 DOI: 10.3390/ijms221910766] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023] Open
Abstract
Animal models of human neurodegenerative disease have been investigated for several decades. In recent years, zebrafish (Danio rerio) and medaka (Oryzias latipes) have become popular in pathogenic and therapeutic studies about human neurodegenerative diseases due to their small size, the optical clarity of embryos, their fast development, and their suitability to large-scale therapeutic screening. Following the emergence of a new generation of molecular biological technologies such as reverse and forward genetics, morpholino, transgenesis, and gene knockout, many human neurodegenerative disease models, such as Parkinson's, Huntington's, and Alzheimer's, were constructed in zebrafish and medaka. These studies proved that zebrafish and medaka genes are functionally conserved in relation to their human homologues, so they exhibit similar neurodegenerative phenotypes to human beings. Therefore, fish are a suitable model for the investigation of pathologic mechanisms of neurodegenerative diseases and for the large-scale screening of drugs for potential therapy. In this review, we summarize the studies in modelling human neurodegenerative diseases in zebrafish and medaka in recent years.
Collapse
Affiliation(s)
- Jing Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Donghu South Road 7#, Wuhan 430072, China;
- College of Advanced Agriculture Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hong Cao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Donghu South Road 7#, Wuhan 430072, China;
- College of Advanced Agriculture Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
43
|
Wellenberg A, Brinkmann V, Bornhorst J, Ventura N, Honnen S, Fritz G. Cisplatin-induced neurotoxicity involves the disruption of serotonergic neurotransmission. Pharmacol Res 2021; 174:105921. [PMID: 34601079 DOI: 10.1016/j.phrs.2021.105921] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/23/2021] [Accepted: 09/28/2021] [Indexed: 11/18/2022]
Abstract
Neurotoxicity is a frequent side effect of cisplatin (CisPt)-based anticancer therapy whose pathophysiology is largely vague. Here, we exploited C. elegans as a 3R-compliant in vivo model to elucidate molecular mechanisms contributing to CisPt-induced neuronal dysfunction. To this end, we monitored the impact of CisPt on various sensory functions as well as pharyngeal neurotransmission by recording electropharyngeograms (EPGs). CisPt neither affected food and odor sensation nor mechano-sensation, which involve dopaminergic and glutaminergic neurotransmission. However, CisPt reduced serotonin-regulated pharyngeal pumping activity independent of changes in the morphology of related neurons. CisPt-mediated alterations in EPGs were fully rescued by addition of serotonin (5-HT) (≤ 2 mM). Moreover, the CisPt-induced pharyngeal injury was prevented by co-incubation with the clinically approved serotonin re-uptake inhibitory drug duloxetine. A protective effect of 5-HT was also observed with respect to CisPt-mediated impairment of another 5-HT-dependent process, the egg laying activity. Importantly, CisPt-induced apoptosis in the gonad and learning disability were not influenced by 5-HT. Using different C. elegans mutants we found that CisPt-mediated (neuro)toxicity is independent of serotonin biosynthesis and re-uptake and likely involves serotonin-receptor subtype 7 (SER-7)-related functions. In conclusion, by measuring EPGs as a surrogate parameter of neuronal dysfunction, we provide first evidence that CisPt-induced neurotoxicity in C. elegans involves 5-HT-dependent neurotransmission and SER-7-mediated signaling mechanisms and can be prevented by the clinically approved antidepressant duloxetine. The data highlight the particular suitability of C. elegans as a 3R-conform in vivo model in molecular (neuro)toxicology and, moreover, for the pre-clinical identification of neuroprotective candidate drugs.
Collapse
Affiliation(s)
- Anna Wellenberg
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, D-40225 Düsseldorf, Germany
| | - Vanessa Brinkmann
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, D-40225 Düsseldorf, Germany
| | - Julia Bornhorst
- Faculty of Mathematics and Natural Sciences, Food Chemistry, University of Wuppertal, D-42119 Wuppertal, Germany
| | - Natascia Ventura
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty, Heinrich Heine University and Leibniz Research Institute for Environmental Medicine (IUF), D-40225 Düsseldorf, Germany
| | - Sebastian Honnen
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, D-40225 Düsseldorf, Germany.
| | - Gerhard Fritz
- Institute of Toxicology, Medical Faculty, Heinrich Heine University, D-40225 Düsseldorf, Germany.
| |
Collapse
|
44
|
Zheng F, Aschner M, Li H. Evaluations of Environmental Pollutant-Induced Mitochondrial Toxicity Using Caenorhabditis elegans as a Model System. Methods Mol Biol 2021; 2326:33-46. [PMID: 34097259 DOI: 10.1007/978-1-0716-1514-0_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Environmental pollutants inevitably exert adverse effects on humans and other species. Quick identification and in-depth characterization of the pollutants are requisite objectives for clinicians and environmental health scientists. The nematode Caenorhabditis elegans has been utilized as a model organism for toxicity evaluation of environmental pollutants, due to its transparency, short lifespan, entire genome sequencing, and economical characteristics. However, few researchers have systematically addressed mitochondrial toxicity in response to toxicants, despite the critical role mitochondria play in energy production and respiration, as well as the generation of reactive oxygen species. Mitochondria are vulnerable to environmental pollutants, and their dysfunction contributes to cellular damage and toxicity in plethora of diseases. Here, we describe methods in step-by-step for mitochondrial toxicity evaluation in response to pollutants, including exposure of C. elegans to toxicants, mitochondrial ROS detection, mitochondrial morphology analysis, mitochondrial function analysis, such as ATP production and oxygen consumption, and gene expression studies, with the application of corresponding genetically modified strains.
Collapse
Affiliation(s)
- Fuli Zheng
- The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, China.,Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Huangyuan Li
- The Key Laboratory of Environment and Health, School of Public Health, Fujian Medical University, Fuzhou, China. .,Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
45
|
Rzepnikowska W, Kochański A. Models for IGHMBP2-associated diseases: an overview and a roadmap for the future. Neuromuscul Disord 2021; 31:1266-1278. [PMID: 34785121 DOI: 10.1016/j.nmd.2021.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/16/2021] [Accepted: 08/06/2021] [Indexed: 12/13/2022]
Abstract
Models are practical tools with which to establish the basic aspects of a diseases. They allow systematic research into the significance of mutations, of cellular and molecular pathomechanisms, of therapeutic options and of functions of diseases associated proteins. Thus, disease models are an integral part of the study of enigmatic proteins such as immunoglobulin mu-binding protein 2 (IGHMBP2). IGHMBP2 has been well defined as a helicase, however there is little known about its role in cellular processes. Notably, it is unclear why changes in such an abundant protein lead to specific neuronal disorders including spinal muscular atrophy with respiratory distress type 1 (SMARD1) and Charcot-Marie-Tooth type 2S (CMT2S). SMARD1 is caused by a loss of motor neurons in the spinal cord that results in muscle atrophy and is accompanied by rapid respiratory failure. In contrast, CMT2S manifests as a severe neuropathy, but typically without critical breathing problems. Here, we present the clinical manifestation of IGHMBP2 mutations, function of protein and models that may be used for the study of IGHMBP2-associated disorders. We highlight the strengths and weaknesses of specific models and discuss the orthologs of IGHMBP2 that are found in different systems with regard to their similarity to human IGHMBP2.
Collapse
Affiliation(s)
- Weronika Rzepnikowska
- Neuromuscular Unit, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw 02-106, Poland.
| | - Andrzej Kochański
- Neuromuscular Unit, Mossakowski Medical Research Institute Polish Academy of Sciences, Warsaw 02-106, Poland
| |
Collapse
|
46
|
Long NP, Kim HM. Distinct metabolic alterations in different Caenorhabditis elegans mitochondrial mutants. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1179:122863. [PMID: 34325308 DOI: 10.1016/j.jchromb.2021.122863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 10/20/2022]
Abstract
Mitochondria play an essential role in various biochemical processes that maintain cellular homeostasis. Minor defects in the mitochondrial genome can lead to aversive behavioral responses in an organism. Nevertheless, little is known about the impact of mitochondrial mutations on the metabolome of Caenorhabditis elegans (C. elegans). In this study, an untargeted metabolomics approach was employed to elucidate the metabolic aberrant caused by mitochondrial DNA mutations in C. elegans. Specifically, three mutant strains of C. elegans, including clk-1, mev-1, and phb-2, were adopted to study corresponding metabolic signatures. Adult worms were collected, and metabolites were extracted and analyzed by gas chromatography-mass spectrometry. Uni- and multivariate analyses were performed to elucidate perturbed metabolism between wildtype worms and mutant strains, and metabolic differences among the mutants. The tricarboxylic acid cycle intermediates, amino acids, and sugars were significantly affected in the mitochondrial mutants. Overall, each mitochondrial DNA mutation exhibited a different pattern of metabolic alterations. The shift of metabolome appeared to be associated with the lifespan of C. elegans. In particular, clk-1 and mev-1 strains, which had the opposite phenotypes of lifespan, had apparently different metabolomes. Our findings set light on the metabolic consequences of mitochondrial genetic variants, which may help better understand mitochondrial disease mechanisms.
Collapse
Affiliation(s)
- Nguyen Phuoc Long
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyung Min Kim
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea.
| |
Collapse
|
47
|
Braems E, Tziortzouda P, Van Den Bosch L. Exploring the alternative: Fish, flies and worms as preclinical models for ALS. Neurosci Lett 2021; 759:136041. [PMID: 34118308 DOI: 10.1016/j.neulet.2021.136041] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 04/15/2021] [Accepted: 06/01/2021] [Indexed: 12/22/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is an incurable neurodegenerative disorder characterized by the loss of upper and lower motor neurons. In general, patients succumb to respiratory insufficiency due to respiratory muscle weakness. Despite many promising therapeutic strategies primarily identified in rodent models, patient trials remain rather unsuccessful. There is a clear need for alternative approaches, which could provide directions towards the justified use of rodents and which increase the likelihood to identify new promising clinical candidates. In the last decades, the use of fast genetic approaches and the development of high-throughput screening platforms in the nematode Caenorhabditis elegans, in the fruit fly (Drosophila melanogaster) and in zebrafish (Danio rerio) have contributed to new insights into ALS pathomechanisms, disease modifiers and therapeutic targets. In this mini-review, we provide an overview of these alternative small animal studies, modeling the most common ALS genes and discuss the most recent preclinical discoveries. We conclude that small animal models will not replace rodent models, yet they clearly represent an important asset for preclinical studies.
Collapse
Affiliation(s)
- Elke Braems
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Paraskevi Tziortzouda
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Ludo Van Den Bosch
- KU Leuven - University of Leuven, Department of Neurosciences, Experimental Neurology, and Leuven Brain Institute (LBI), Leuven, Belgium; VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium.
| |
Collapse
|
48
|
Yadav J, El Hassani M, Sodhi J, Lauschke VM, Hartman JH, Russell LE. Recent developments in in vitro and in vivo models for improved translation of preclinical pharmacokinetics and pharmacodynamics data. Drug Metab Rev 2021; 53:207-233. [PMID: 33989099 DOI: 10.1080/03602532.2021.1922435] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Improved pharmacokinetics/pharmacodynamics (PK/PD) prediction in the early stages of drug development is essential to inform lead optimization strategies and reduce attrition rates. Recently, there have been significant advancements in the development of new in vitro and in vivo strategies to better characterize pharmacokinetic properties and efficacy of drug leads. Herein, we review advances in experimental and mathematical models for clearance predictions, advancements in developing novel tools to capture slowly metabolized drugs, in vivo model developments to capture human etiology for supporting drug development, limitations and gaps in these efforts, and a perspective on the future in the field.
Collapse
Affiliation(s)
- Jaydeep Yadav
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Inc., Boston, MA, USA
| | | | - Jasleen Sodhi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jessica H Hartman
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | | |
Collapse
|
49
|
Shahzad U, Taccone MS, Kumar SA, Okura H, Krumholtz S, Ishida J, Mine C, Gouveia K, Edgar J, Smith C, Hayes M, Huang X, Derry WB, Taylor MD, Rutka JT. Modeling human brain tumors in flies, worms, and zebrafish: From proof of principle to novel therapeutic targets. Neuro Oncol 2021; 23:718-731. [PMID: 33378446 DOI: 10.1093/neuonc/noaa306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
For decades, cell biologists and cancer researchers have taken advantage of non-murine species to increase our understanding of the molecular processes that drive normal cell and tissue development, and when perturbed, cause cancer. The advent of whole-genome sequencing has revealed the high genetic homology of these organisms to humans. Seminal studies in non-murine organisms such as Drosophila melanogaster, Caenorhabditis elegans, and Danio rerio identified many of the signaling pathways involved in cancer. Studies in these organisms offer distinct advantages over mammalian cell or murine systems. Compared to murine models, these three species have shorter lifespans, are less resource intense, and are amenable to high-throughput drug and RNA interference screening to test a myriad of promising drugs against novel targets. In this review, we introduce species-specific breeding strategies, highlight the advantages of modeling brain tumors in each non-mammalian species, and underscore the successes attributed to scientific investigation using these models. We conclude with an optimistic proposal that discoveries in the fields of cancer research, and in particular neuro-oncology, may be expedited using these powerful screening tools and strategies.
Collapse
Affiliation(s)
- Uswa Shahzad
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Canada.,Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada
| | - Michael S Taccone
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Sachin A Kumar
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Hidehiro Okura
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada
| | - Stacey Krumholtz
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada
| | - Joji Ishida
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada
| | - Coco Mine
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada
| | - Kyle Gouveia
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada
| | - Julia Edgar
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada
| | - Christian Smith
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada
| | - Madeline Hayes
- Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Canada
| | - Xi Huang
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada.,Developmental and Stem Cell Biology, Hospital for Sick Children, Toronto, Canada
| | - W Brent Derry
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Michael D Taylor
- Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada
| | - James T Rutka
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, Canada.,Arthur and Sonia Labatt Brain Tumor Research Center, Hospital for Sick Children, Toronto, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada
| |
Collapse
|
50
|
Garcia-Sanchez JA, Ewbank JJ, Visvikis O. Ubiquitin-related processes and innate immunity in C. elegans. Cell Mol Life Sci 2021; 78:4305-4333. [PMID: 33630111 PMCID: PMC11072174 DOI: 10.1007/s00018-021-03787-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/18/2021] [Accepted: 02/03/2021] [Indexed: 02/06/2023]
Abstract
Innate immunity is an evolutionary ancient defence strategy that serves to eliminate infectious agents while maintaining host health. It involves a complex network of sensors, signaling proteins and immune effectors that detect the danger, then relay and execute the immune programme. Post-translational modifications relying on conserved ubiquitin and ubiquitin-like proteins are an integral part of the system. Studies using invertebrate models of infection, such as the nematode Caenorhabditis elegans, have greatly contributed to our understanding of how ubiquitin-related processes act in immune sensing, regulate immune signaling pathways, and participate to host defence responses. This review highlights the interest of working with a genetically tractable model organism and illustrates how C. elegans has been used to identify ubiquitin-dependent immune mechanisms, discover novel ubiquitin-based resistance strategies that mediate pathogen clearance, and unravel the role of ubiquitin-related processes in tolerance, preserving host fitness during pathogen attack. Special emphasis is placed on processes that are conserved in mammals.
Collapse
Affiliation(s)
- Juan A Garcia-Sanchez
- INSERM, C3M, Côte D'Azur University, Nice, France
- INSERM, CNRS, CIML, Turing Centre for Living Systems, Aix-Marseille University, Marseille, France
| | - Jonathan J Ewbank
- INSERM, CNRS, CIML, Turing Centre for Living Systems, Aix-Marseille University, Marseille, France.
| | | |
Collapse
|