1
|
Jee C, Batsaikhan E. JNK Signaling Positively Regulates Acute Ethanol Tolerance in C. elegans. Int J Mol Sci 2024; 25:6398. [PMID: 38928105 PMCID: PMC11203441 DOI: 10.3390/ijms25126398] [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: 05/12/2024] [Revised: 06/05/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Alcohol use disorder (AUD) is a chronic neurobehavioral condition characterized by a cycle of tolerance development, increased consumption, and reinstated craving and seeking behaviors during withdrawal. Understanding the intricate mechanisms of AUD necessitates reliable animal models reflecting its key features. Caenorhabditis elegans (C. elegans), with its conserved nervous system and genetic tractability, has emerged as a valuable model organism to study AUD. Here, we employ an ethanol vapor exposure model in Caenorhabditis elegans, recapitulating AUD features while maintaining high-throughput scalability. We demonstrate that ethanol vapor exposure induces intoxication-like behaviors, acute tolerance, and ethanol preference, akin to mammalian AUD traits. Leveraging this model, we elucidate the conserved role of c-jun N-terminal kinase (JNK) signaling in mediating acute ethanol tolerance. Mutants lacking JNK signaling components exhibit impaired tolerance development, highlighting JNK's positive regulation. Furthermore, we detect ethanol-induced JNK activation in C. elegans. Our findings underscore the utility of C. elegans with ethanol vapor exposure for studying AUD and offer novel insights into the molecular mechanisms underlying acute ethanol tolerance through JNK signaling.
Collapse
Affiliation(s)
- Changhoon Jee
- Department of Pharmacology, Addiction Science and Toxicology, College of Medicine, University of Tennesse Health Science Center, Memphis, TN 38163, USA;
| | | |
Collapse
|
2
|
Hermeling JCW, Herholz M, Baumann L, Cores EC, Zečić A, Hoppe T, Riemer J, Trifunovic A. Mitochondria-originated redox signalling regulates KLF-1 to promote longevity in Caenorhabditis elegans. Redox Biol 2022; 58:102533. [PMID: 36442394 PMCID: PMC9709155 DOI: 10.1016/j.redox.2022.102533] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 11/07/2022] [Indexed: 11/21/2022] Open
Abstract
Alternations of redox metabolism have been associated with the extension of lifespan in roundworm Caenorhabditis elegans, caused by moderate mitochondrial dysfunction, although the underlying signalling cascades are largely unknown. Previously, we identified transcriptional factor Krüppel-like factor-1 (KLF-1) as the main regulator of cytoprotective longevity-assurance pathways in the C. elegans long-lived mitochondrial mutants. Here, we show that KLF-1 translocation to the nucleus and the activation of the signalling cascade is dependent on the mitochondria-derived hydrogen peroxide (H2O2) produced during late developmental phases where aerobic respiration and somatic mitochondrial biogenesis peak. We further show that mitochondrial-inducible superoxide dismutase-3 (SOD-3), together with voltage-dependent anion channel-1 (VDAC-1), is required for the life-promoting H2O2 signalling that is further regulated by peroxiredoxin-3 (PRDX-3). Increased H2O2 release in the cytoplasm activates the p38 MAPK signalling cascade that induces KLF-1 translocation to the nucleus and the activation of transcription of C. elegans longevity-promoting genes, including cytoprotective cytochrome P450 oxidases. Taken together, our results underline the importance of redox-regulated signalling as the key regulator of longevity-inducing pathways in C. elegans, and position precisely timed mitochondria-derived H2O2 in the middle of it.
Collapse
Affiliation(s)
- Johannes CW Hermeling
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Germany,Institute for Mitochondrial Diseases and Ageing, Medical Faculty, University of Cologne, Cologne, D-50931, Germany
| | - Marija Herholz
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Germany,Institute for Mitochondrial Diseases and Ageing, Medical Faculty, University of Cologne, Cologne, D-50931, Germany
| | - Linda Baumann
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Germany,Institute for Mitochondrial Diseases and Ageing, Medical Faculty, University of Cologne, Cologne, D-50931, Germany
| | - Estela Cepeda Cores
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Germany,Institute for Mitochondrial Diseases and Ageing, Medical Faculty, University of Cologne, Cologne, D-50931, Germany
| | - Aleksandra Zečić
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Germany,Institute for Mitochondrial Diseases and Ageing, Medical Faculty, University of Cologne, Cologne, D-50931, Germany
| | - Thorsten Hoppe
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Germany,Center for Molecular Medicine Cologne (CMMC), Cologne, D-50931, Germany,Institute for Genetics, University of Cologne, Cologne, D-50674, Germany
| | - Jan Riemer
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Germany,Institute for Biochemistry, University of Cologne, Cologne, D-50931, Germany
| | - Aleksandra Trifunovic
- Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), Germany,Institute for Mitochondrial Diseases and Ageing, Medical Faculty, University of Cologne, Cologne, D-50931, Germany,Center for Molecular Medicine Cologne (CMMC), Cologne, D-50931, Germany,Corresponding author. CECAD Research CenterUniversity of Cologne, Joseph-Stelzmann-Str. 26, Cologne, D-50931, Germany.
| |
Collapse
|
3
|
Hwang M, Shrestha C, Kang S, Kim J. MEKK-3 Acts Cooperatively with NSY-1 in SKN-1-Dependent Manner against Oxidative Stress and Aging in Caenorhabditis elegans. BIOLOGY 2022; 11:biology11101526. [PMID: 36290429 PMCID: PMC9598901 DOI: 10.3390/biology11101526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 11/15/2022]
Abstract
Oxidative stress resulting from reactive oxygen species and other toxic metabolites is involved in human diseases, and it plays an important role in aging. In Caenorhabditis elegans, SKN-1 is required for protection against oxidative stress and aging. As p38 mitogen-activated protein kinase signaling is activated in response to oxidative stress, SKN-1 accumulates in intestinal nuclei and induces phase II detoxification genes. However, NSY-1, a well-known mitogen-activated protein kinase kinase kinase (MAPKKK) of C. elegans, acts as a partial regulator of the SKN-1-induced oxidative stress signaling pathway, suggesting that the regulator for optimal activation of SKN-1 remains unknown. Here, we report a MAPKKK, MEKK-3, as a new regulator required for full activation of SKN-1-mediated resistance against oxidative stress and aging. In RNA-interference-based screening, we found that the simultaneous knockdown of mekk-3 and nsy-1 significantly decreased the oxidative stress resistance and survival of SKN-1 transgenic worms. MEKK-3 was induced in response to oxidative stress. Mechanistic analysis revealed that double knockdown of mekk-3 and nsy-1 completely suppressed the nuclear localization of SKN-1. These results were reproduced in mutant worms in which SKN-1 is constitutively localized to intestinal nuclei. In addition, mekk-3 and nsy-1 were required for optimal induction of SKN-1 target genes such as gcs-1 and trx-1. These data indicate that MEKK-3 plays an essential role in the SKN-1-dependent signaling pathway involved in oxidative stress resistance and longevity by cooperating with NSY-1.
Collapse
Affiliation(s)
- Min Hwang
- Department of Pharmacology, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Chandani Shrestha
- Department of Pharmacology, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Shinwon Kang
- Department of Physiology, University of Toronto, Toronto, ON M5S, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON M5G, Canada
| | - Jiyoon Kim
- Department of Pharmacology, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
- Correspondence:
| |
Collapse
|
4
|
Malinow RA, Zhu M, Jin Y, Kim KW. Forward genetic screening identifies novel roles for N-terminal acetyltransferase C and histone deacetylase in C. elegans development. Sci Rep 2022; 12:16438. [PMID: 36180459 PMCID: PMC9525577 DOI: 10.1038/s41598-022-20361-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 09/12/2022] [Indexed: 02/02/2023] Open
Abstract
Coordinating the balance between development and stress responses is critical for organismal survival. However, the cellular signaling controlling this mechanism is not well understood. In Caenorhabditis elegans, it has been hypothesized that a genetic network regulated by NIPI-3/Tibbles may control the balance between animal development and immune response. Using a nipi-3(0) lethality suppressor screen in C. elegans, we reveal a novel role for N-terminal acetyltransferase C complex natc-1/2/3 and histone deacetylase hda-4, in the control of animal development. These signaling proteins act, at least in part, through a PMK-1 p38 MAP kinase pathway (TIR-1-NSY-1-SEK-1-PMK-1), which plays a critical role in the innate immunity against infection. Additionally, using a transcriptional reporter of SEK-1, a signaling molecule within this p38 MAP kinase system that acts directly downstream of C/EBP bZip transcription factor CEBP-1, we find unexpected positive control of sek-1 transcription by SEK-1 along with several other p38 MAP kinase pathway components. Together, these data demonstrate a role for NIPI-3 regulators in animal development, operating, at least in part through a PMK-1 p38 MAPK pathway. Because the C. elegans p38 MAP kinase pathway is well known for its role in cellular stress responses, the novel biological components and mechanisms pertaining to development identified here may also contribute to the balance between stress response and development.
Collapse
Affiliation(s)
- Rose Aria Malinow
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Ming Zhu
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Yishi Jin
- Department of Neurobiology, School of Biological Sciences, University of California, San Diego, La Jolla, CA, 92093, USA.
| | - Kyung Won Kim
- Department of Life Science, Hallym University, Chuncheon, 24252, South Korea.
- Multidisciplinary Genome Institute, Hallym University, Chuncheon, 24252, South Korea.
| |
Collapse
|
5
|
Kim SM, Zou G, Kim H, Kang M, Ahn S, Heo HY, Kim JS, Lim KM, Ausubel FM, Mylonakis E, Gao H, Kim W. Antimicrobial activity of the membrane-active compound nTZDpa is enhanced at low pH. Biomed Pharmacother 2022; 150:112977. [PMID: 35447554 DOI: 10.1016/j.biopha.2022.112977] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/03/2022] [Accepted: 04/12/2022] [Indexed: 11/30/2022] Open
Abstract
The opportunistic human pathogen Staphylococcus aureus can evade antibiotics by acquiring antibiotic resistance genes or by entering into a non-growing dormant state. Moreover, the particular circumstances of a specific infection site, such as acidity or anaerobicity, often weaken antibiotic potency. Decreased bacterial susceptibility combined with diminished antibiotic potency is responsible for high failure rates when treating S. aureus infections. Here, we report that the membrane-active antimicrobial agent nTZDpa does not only exhibit enhanced antibiotic activity against multidrug-resistant Gram-positive pathogens in acidic pH, but also retains antimicrobial potency under anaerobic conditions. This agent completely eradicated highly antibiotic-tolerant cells and biofilms formed by methicillin-resistant S. aureus at pH 5.5 at concentrations at which it was not potent at pH 7.4. Furthermore, nTZDpa was more potent at synergistically potentiating gentamicin killing against antibiotic-tolerant MRSA cells at low pH than at high pH. All-atom molecular dynamics simulations combined with membrane-permeabilization assays revealed that the neutral form of nTZDpa, which contains carboxylic acid, is more effective than the deprotonated form at penetrating the bacterial membrane and plays an essential role in membrane activity. An acidic pH increases the proportion of the neutrally charged nTZDpa, which results in antimicrobial enhancement. Our results provide key insights into rational design of pH-sensitive membrane-active antimicrobials and antibiotic adjuvants that are effective in an infection environment. These findings demonstrate that nTZDpa is a promising lead compound for developing new therapeutics against hard-to-cure infections caused by drug-resistant and -tolerant S. aureus.
Collapse
Affiliation(s)
- Soo Min Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Guijin Zou
- Institute of High Performance Computing, A⁎STAR, Singapore 138632, Singapore
| | - Hyerim Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Minjeong Kang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Soyeon Ahn
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Hee Young Heo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jae-Seok Kim
- Department of Laboratory Medicine, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul 05355, Republic of Korea
| | - Kyung-Min Lim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Frederick M Ausubel
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School of Brown University, Providence, RI 02903, USA
| | - Huajian Gao
- Institute of High Performance Computing, A⁎STAR, Singapore 138632, Singapore; School of Mechanical and Aerospace Engineering, College of Engineering, Nanyang Technological University, Singapore 639789, Singapore
| | - Wooseong Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea.
| |
Collapse
|
6
|
Metformin-ROS-Nrf2 connection in the host defense mechanism against oxidative stress, apoptosis, cancers, and ageing. Biochim Biophys Acta Gen Subj 2022; 1866:130171. [DOI: 10.1016/j.bbagen.2022.130171] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/12/2022] [Accepted: 05/12/2022] [Indexed: 11/18/2022]
|
7
|
Hoerndli FJ, Brockie PJ, Wang R, Mellem JE, Kallarackal A, Doser RL, Pierce DM, Madsen DM, Maricq AV. MAPK signaling and a mobile scaffold complex regulate AMPA receptor transport to modulate synaptic strength. Cell Rep 2022; 38:110577. [PMID: 35354038 PMCID: PMC9965202 DOI: 10.1016/j.celrep.2022.110577] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 01/31/2022] [Accepted: 03/04/2022] [Indexed: 12/27/2022] Open
Abstract
Synaptic plasticity depends on rapid experience-dependent changes in the number of neurotransmitter receptors. Previously, we demonstrated that motor-mediated transport of AMPA receptors (AMPARs) to and from synapses is a critical determinant of synaptic strength. Here, we describe two convergent signaling pathways that coordinate the loading of synaptic AMPARs onto scaffolds, and scaffolds onto motors, thus providing a mechanism for experience-dependent changes in synaptic strength. We find that an evolutionarily conserved JIP-protein scaffold complex and two classes of mitogen-activated protein kinase (MAPK) proteins mediate AMPAR transport by kinesin-1 motors. Genetic analysis combined with in vivo, real-time imaging in Caenorhabditis elegans revealed that CaMKII is required for loading AMPARs onto the scaffold, and MAPK signaling is required for loading the scaffold complex onto motors. Our data support a model where CaMKII signaling and a MAPK-signaling pathway cooperate to facilitate the rapid exchange of AMPARs required for early stages of synaptic plasticity.
Collapse
Affiliation(s)
- Frédéric J Hoerndli
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA.
| | - Penelope J Brockie
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84112-9458, USA
| | - Rui Wang
- Pathology Department, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Jerry E Mellem
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84112-9458, USA
| | - Angy Kallarackal
- Department of Psychology, Mount Saint Mary's University, Emmitsburg, MD 21727, USA
| | - Rachel L Doser
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - Dayton M Pierce
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | - David M Madsen
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84112-9458, USA
| | - Andres V Maricq
- Department of Neurobiology, University of Utah, Salt Lake City, UT 84112-9458, USA.
| |
Collapse
|
8
|
Wimberly K, Choe KP. An extracellular matrix damage sensor signals through membrane-associated kinase DRL-1 to mediate cytoprotective responses in Caenorhabditis elegans. Genetics 2022; 220:iyab217. [PMID: 34849856 PMCID: PMC9208646 DOI: 10.1093/genetics/iyab217] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/12/2021] [Indexed: 11/13/2022] Open
Abstract
We and others previously identified circumferential bands of collagen named annular furrows as key components of a damage sensor in the cuticle of Caenorhabditis elegans that regulates cytoprotective genes. Mutation or loss of noncollagen secreted proteins OSM-7, OSM-8, and OSM-11 activate the same cytoprotective responses without obvious changes to the cuticle indicating that other extracellular proteins are involved. Here, we used RNAi screening to identify protein kinase DRL-1 as a key modulator of cytoprotective gene expression and stress resistance in furrow and extracellular OSM protein mutants. DRL-1 functions downstream from furrow disruption and is expressed in cells that induce cytoprotective genes. DRL-1 is not required for the expression of cytoprotective genes under basal or oxidative stress conditions consistent with specificity to extracellular signals. DRL-1 was previously shown to regulate longevity via a "Dietary Restriction-Like" state, but it functions downstream from furrow disruption by a distinct mechanism. The kinase domain of DRL-1 is related to mammalian MEKK3, and MEKK3 is recruited to a plasma membrane osmosensor complex by a scaffold protein. In C. elegans, DRL-1 contains an atypical hydrophobic C-terminus with predicted transmembrane domains and is constitutively expressed at or near the plasma membrane where it could function to receive extracellular damage signals for cells that mount cytoprotective responses.
Collapse
Affiliation(s)
- Keon Wimberly
- Department of Biology and Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| | - Keith P Choe
- Department of Biology and Genetics Institute, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
9
|
Host defense mechanisms induce genome instability leading to rapid evolution in an opportunistic fungal pathogen. Infect Immun 2021; 90:e0032821. [PMID: 34898249 DOI: 10.1128/iai.00328-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The ability to generate genetic variation facilitates rapid adaptation in stressful environments. The opportunistic fungal pathogen Candida albicans frequently undergoes large-scale genomic changes, including aneuploidy and loss-of heterozygosity (LOH), following exposure to host environments. However, the specific host factors inducing C. albicans genome instability remain largely unknown. Here, we leveraged the genetic tractability of nematode hosts to investigate whether innate immune components, including antimicrobial peptides (AMPs) and reactive oxygen species (ROS), induced host-associated C. albicans genome instability. C. albicans associated with immunocompetent hosts carried multiple large-scale genomic changes including LOH, whole chromosome, and segmental aneuploidies. In contrast, C. albicans associated with immunocompromised hosts deficient in AMPs or ROS production had reduced LOH frequencies and fewer, if any, additional genomic changes. To evaluate if extensive host-induced genomic changes had long-term consequences for C. albicans adaptation, we experimentally evolved C. albicans in either immunocompetent or immunocompromised hosts and selected for increased virulence. C. albicans evolved in immunocompetent hosts rapidly increased virulence, but not in immunocompromised hosts. Taken together, this work suggests that host-produced ROS and AMPs induces genotypic plasticity in C. albicans which facilitates rapid evolution.
Collapse
|
10
|
Hsieh YW, Xiong R, Chuang CF. Synergistic roles of homeodomain proteins UNC-62 homothorax and MLS-2 HMX/NKX in the specification of olfactory neurons in Caenorhabditis elegans. Genetics 2021; 219:6350488. [PMID: 34849889 DOI: 10.1093/genetics/iyab133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 08/09/2021] [Indexed: 11/13/2022] Open
Abstract
General identity of the Caenorhabditis elegans AWC olfactory neuron pair is specified by the OTX/OTD transcription factor CEH-36 and the HMG-box transcription factor SOX-2, followed by asymmetrical differentiation of the pair into two distinct subtypes, default AWCOFF and induced AWCON, through a stochastic signaling event. The HMX/NKX transcription factor MLS-2 regulates the expression of ceh-36 to specify general AWC identity. However, general AWC identity is lost in only one of the two AWC cells in the majority of mls-2 null mutants displaying defective general AWC identity, suggesting that additional transcription factors have a partially overlapping role with MLS-2 in the specification of general AWC identity. Here, we identify a role of unc-62, encoding a homothorax/Meis/TALE homeodomain protein, in the specification of general AWC identity. As in mls-2 null mutants, unc-62 null mutants showed an incomplete penetrance in loss of general AWC identity. However, unc-62; mls-2 double mutants display a nearly complete penetrance of identity loss in both AWC cells. Thus, unc-62 and mls-2 have a partially overlapping function in the specification of general AWC identity. Furthermore, our genetic results suggest that mls-2 and unc-62 act cell autonomously in promoting the AWCON subtype. Together, our findings reveal the sequential roles of the unc-62 and mls-2 pair in AWC development, specification of general AWC identity in early embryogenesis, and asymmetric differentiation of AWC subtypes in late embryogenesis.
Collapse
Affiliation(s)
- Yi-Wen Hsieh
- Department of Biological Sciences, University of Illinois at Chicago, IL 60607, USA
| | - Rui Xiong
- Department of Biological Sciences, University of Illinois at Chicago, IL 60607, USA
| | - Chiou-Fen Chuang
- Department of Biological Sciences, University of Illinois at Chicago, IL 60607, USA.,Graduate Program in Neuroscience, University of Illinois at Chicago, IL 60607, USA
| |
Collapse
|
11
|
Luo A, Jing H, Yuan L, Wang Y, Xiao H, Zheng Q. Loss of Function of Scavenger Receptor SCAV-5 Protects C. elegans Against Pathogenic Bacteria. Front Cell Infect Microbiol 2021; 11:593745. [PMID: 34414127 PMCID: PMC8370389 DOI: 10.3389/fcimb.2021.593745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 07/08/2021] [Indexed: 11/13/2022] Open
Abstract
Scavenger receptors play a critical role in innate immunity by acting as the pattern-recognition receptors. There are six class B scavenger receptors homologs in C. elegans. However, it remains unclear whether they are required for host defense against bacterial pathogens. Here, we show that, of the six SCAV proteins, only loss of function scav-5 protect C. elegans against pathogenic bacteria S. typhimurium SL1344 and P. aeruginosa PA14 by different mechanism. scav-5 mutants are resistant to S. typhimurium SL1344 due to dietary restriction. While scav-5 acts upstream of or in parallel to tir-1 in conserved PMK-1 p38 MAPK pathway to upregulate the innate immune response to defend worms against P. aeruginosa PA14. This is the first demonstration of a role for SCAV-5 in host defense against pathogenic bacteria. Our results provide an important basis for further elucidating the underlying molecular mechanism by which scav-5 regulates innate immune responses.
Collapse
Affiliation(s)
- Aixiao Luo
- Key Laboratory of the Ministry of Education for Medicinal Plant Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in the Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Huiru Jing
- Key Laboratory of the Ministry of Education for Medicinal Plant Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in the Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Lei Yuan
- Key Laboratory of the Ministry of Education for Medicinal Plant Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in the Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yanzhe Wang
- Key Laboratory of the Ministry of Education for Medicinal Plant Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in the Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Hui Xiao
- Key Laboratory of the Ministry of Education for Medicinal Plant Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in the Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Qian Zheng
- Key Laboratory of the Ministry of Education for Medicinal Plant Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in the Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
12
|
Zhang J, Zhao X, Cappiello JR, Yang Y, Cheng Y, Liu G, Fang W, Luo Y, Zhang Y, Dong J, Zhang L, Sharpless KB. Identification of simple arylfluorosulfates as potent agents against resistant bacteria. Proc Natl Acad Sci U S A 2021; 118:e2103513118. [PMID: 34244433 PMCID: PMC8285976 DOI: 10.1073/pnas.2103513118] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Sulfur fluoride exchange (SuFEx), a next generation of click chemistry, opens an avenue for drug discovery. We report here the discovery and structure-activity relationship studies of a series of arylfluorosulfates, synthesized via SuFEx, as antibacterial agents. Arylfluorosulfates 3, 81, and 101 showed potency to overcome multidrug resistance and were not susceptible to the generation of resistance. They exhibited rapid bactericidal potency and selectively killed gram-positive bacterial strains. These compounds also exhibited the ability to disrupt established bacterial biofilm and kill persisters derived from biofilm. Furthermore, arylfluorosulfate 3 had a synergistic effect with streptomycin and gentamicin. In addition, their anti-MRSA potency was evaluated and determined by the Caenorhabditis elegans model.
Collapse
Affiliation(s)
- Jiong Zhang
- Key Laboratory of Organofluorine Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of the Chinese Academy of Sciences, Chinese Academy of Sciences, 200032 Shanghai, China
| | - Xiangxiang Zhao
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 200237 Shanghai, China
| | - John R Cappiello
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | - Yi Yang
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | - Yunfei Cheng
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037
| | - Guang Liu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 200237 Shanghai, China
| | - Wenjing Fang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 200237 Shanghai, China
| | - Yinzhu Luo
- Guangdong Provincial Key Laboratory of Laboratory Animals, Guangdong Laboratory Animals Monitoring Institute, 510663 Guangzhou, China
| | - Yu Zhang
- Guangdong Provincial Key Laboratory of Laboratory Animals, Guangdong Laboratory Animals Monitoring Institute, 510663 Guangzhou, China
| | - Jiajia Dong
- Key Laboratory of Organofluorine Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of the Chinese Academy of Sciences, Chinese Academy of Sciences, 200032 Shanghai, China;
| | - Lixin Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 200237 Shanghai, China;
| | - K Barry Sharpless
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037;
| |
Collapse
|
13
|
Ma J, Wang R, Chen T, Jiang S, Xu A. Protective effects of baicalin in a Caenorhabditis elegans model of Parkinson's disease. Toxicol Res (Camb) 2021; 10:409-417. [PMID: 34141154 DOI: 10.1093/toxres/tfaa107] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/01/2020] [Accepted: 12/07/2020] [Indexed: 12/29/2022] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disorder of the central nervous system. However, the pathogenetic mechanisms of PD are far from understood. The aim of this study was to determine the protective effect of baicalin in a Caenorhabditis elegans model of PD. C. elegans worms were stimulated for 24 h with 6-hydroxydopamine (6-OHDA, 50 mM) and treated with or without baicalin (1, 10, or 100 μM). At all tested concentrations, baicalin improved the reversal and omega turn behavioral phenotypes, as well as the survival, of 6-OHDA-stimulated worms. It also inhibited 6-OHDA-induced oxidative stress by decreasing malondialdehyde levels, increasing superoxide dismutase, glutathione reductase, catalase, and glutathione levels and up-regulating mRNA expression of the antioxidant-related genes sod-1, sod-2, sod-3, daf-2, and daf-16. Additionally, it significantly decreased the expression of the apoptosis-related gene ced-3 and increased that of the anti-apoptosis-related gene ced-9. The expression levels of cleaved caspase-3 and B-cell lymphoma 2 in 6-OHDA-treated worms were reversed by baicalin. Apoptosis was suppressed by 6-OHDA in loss-of-function strains via the p38 mitogen-activated protein kinase (MAPK) signaling pathway. Furthermore, the apoptotic effects of 6-OHDA were blocked in sek-1 and pmk-1 mutants. Finally, the mRNA expression of sek-1 and pmk-1 and the protein expression of p38 MAPK and stress-activated protein kinase/extracellular signal-regulated kinase 1 were up-regulated by 6-OHDA and reversed by baicalin. Baicalin may protect against 6-OHDA injury by inhibiting apoptosis and decreasing oxidative stress through the p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Jing Ma
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, P. R. China
| | - Ranran Wang
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, P. R. China
| | - Ting Chen
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, P. R. China
| | - Shaowei Jiang
- Emergency Department, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, P. R. China
| | - Ajing Xu
- Department of Pharmacy, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, P. R. China
| |
Collapse
|
14
|
Lipopolysaccharide exposure induces oxidative damage in Caenorhabditis elegans: protective effects of carnosine. BMC Pharmacol Toxicol 2020; 21:85. [PMID: 33272314 PMCID: PMC7713333 DOI: 10.1186/s40360-020-00455-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 10/27/2020] [Indexed: 01/17/2023] Open
Abstract
Background The present study was designed to investigate the protective effects and mechanisms of carnosine on lipopolysaccharide (LPS)-induced injury in Caenorhabditis elegans. Methods C. elegans individuals were stimulated for 24 h with LPS (100 μg/mL), with or without carnosine (0.1, 1, 10 mM). The survival rates and behaviors were determined. The activities of superoxide dismutase (SOD), glutathione reductase (GR), and catalase (CAT) and levels of malondialdehyde (MDA) and glutathione (GSH) were determined using the respective kits. Reverse transcription polymerase chain reaction (RT-PCR) was performed to validate the differential expression of sod-1, sod-2, sod-3, daf-16, ced-3, ced-9, sek-1, and pmk-1. Western blotting was used to determine the levels of SEK1, p38 mitogen-activated protein kinase (MAPK), cleaved caspase3, and Bcl-2. C. elegans sek-1 (km2) mutants and pmk-1 (km25) mutants were used to elucidate the role of the p38 MAPK signaling pathway. Results Carnosine improved the survival of LPS-treated C. elegans and rescued behavioral phenotypes. It also restrained oxidative stress by decreasing MDA levels and increasing SOD, GR, CAT, and GSH levels. RT-PCR results showed that carnosine treatment of wild-type C. elegans up-regulated the mRNA expression of the antioxidant-related genes sod-1, sod-2, sod-3, and daf-16. The expression of the anti-apoptosis-related gene ced-9 and apoptosis-related gene ced-3 was reversed by carnosine. In addition, carnosine treatment significantly decreased cleaved caspase3 levels and increased Bcl-2 levels in LPS-treated C. elegans. Apoptosis in the loss-of-function strains of the p38 MAPK signaling pathway was suppressed under LPS stress; however, the apoptotic effects of LPS were blocked in the sek-1 and pmk-1 mutants. The expression levels of sek-1 and pmk-1 mRNAs were up-regulated by LPS and reversed by carnosine. Finally, the expression of p-p38MAPK and SEK1 was significantly increased by LPS, which was reversed by carnosine. Conclusion Carnosine treatment protected against LPS injury by decreasing oxidative stress and inhibiting apoptosis through the p38 MAPK pathway.
Collapse
|
15
|
Yang ZL, Chen JN, Lu YY, Lu M, Wan QL, Wu GS, Luo HR. Inositol polyphosphate multikinase IPMK-1 regulates development through IP3/calcium signaling in Caenorhabditis elegans. Cell Calcium 2020; 93:102327. [PMID: 33316585 DOI: 10.1016/j.ceca.2020.102327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/25/2020] [Accepted: 11/27/2020] [Indexed: 01/17/2023]
Abstract
Inositol polyphosphate multikinase (IPMK) is a conserved protein that initiates the production of inositol phosphate intracellular messengers and is critical for regulating a variety of cellular processes. Here, we report that the C. elegans IPMK-1, which is homologous to the mammalian inositol polyphosphate multikinase, plays a crucial role in regulating rhythmic behavior and development. The deletion mutant ipmk-1(tm2687) displays a long defecation cycle period and retarded postembryonic growth. The expression of functional ipmk-1::GFP was detected in the pharyngeal muscles, amphid sheath cells, the intestine, excretory (canal) cells, proximal gonad, and spermatheca. The expression of IPMK-1 in the intestine was sufficient for the wild-type phenotype. The IP3-kinase activity of IPMK-1 is required for defecation rhythms and postembryonic development. The defective phenotypes of ipmk-1(tm2687) could be rescued by a loss-of-function mutation in type I inositol 5-phosphatase homolog (IPP-5) and improved by a supplemental Ca2+ in the medium. Our work demonstrates that IPMK-1 and the signaling molecule inositol triphosphate (IP3) pathway modulate rhythmic behaviors and development by dynamically regulating the concentration of intracellular Ca2+ in C. elegans. Advances in understanding the molecular regulation of Ca2+ homeostasis and regulation of organism development may lead to therapeutic strategies that modulate Ca2+ signaling to enhance function and counteract disease processes. Unraveling the physiological role of IPMK and the underlying functional mechanism in C. elegans would contribute to understanding the role of IPMK in other species, especially in mammals, and benefit further research on the involvement of IPMK in disease.
Collapse
Affiliation(s)
- Zhong-Lin Yang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Science, Kunming, Yunnan 650201, China; Graduate University of the Chinese Academy of Science, Beijing, 100049, China
| | - Jian-Ning Chen
- Key Laboratory for Aging and Regenerative Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yu-Yang Lu
- Key Laboratory for Aging and Regenerative Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Min Lu
- Key Laboratory for Aging and Regenerative Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Qin-Li Wan
- The Center for Precision Medicine of First Affiliated Hospital, Biomedical Translational Research Institute, Jinan University, Guangzhou, Guangdong 510632, China
| | - Gui-Sheng Wu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Science, Kunming, Yunnan 650201, China; Key Laboratory for Aging and Regenerative Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Huai-Rong Luo
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Science, Kunming, Yunnan 650201, China; Graduate University of the Chinese Academy of Science, Beijing, 100049, China; Key Laboratory for Aging and Regenerative Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
16
|
Yanase S, Yasuda K, Ishii N. Interaction between the ins/IGF-1 and p38 MAPK signaling pathways in molecular compensation of sod genes and modulation related to intracellular ROS levels in C. elegans. Biochem Biophys Rep 2020; 23:100796. [PMID: 32875124 PMCID: PMC7451853 DOI: 10.1016/j.bbrep.2020.100796] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 06/04/2020] [Accepted: 08/02/2020] [Indexed: 01/28/2023] Open
Abstract
Superoxide dismutases, which catalytically remove intracellular superoxide radicals by the disproportionation of molecular oxygen and hydrogen peroxide, are encoded by the sod-1 to -5 genes in the nematode C. elegans. Expression of the sod genes is mutually compensatory for the modulation of intracellular oxidative stress during aging. Interestingly, several-fold higher expression of the sod-1 to -4 was induced in a sod-5 deletion mutant, despite the low expression levels of sod-5 in wild-type animals. Consequently, this molecular compensation facilitated recovery of lifespan in the sod-5 mutant. In previous reports, two transcription factors DAF-16 and SKN-1 are associated with the compensatory expression of sod genes, which are downstream targets of the ins/IGF-1 and p38 MAPK signaling pathways activated under oxidative and heavy metal stresses, respectively. Here, we show that p38 MAPK signaling regulates induction of not only the direct expression of sod-1, -2 and -4 but also the indirect modulation of DAF-16 targets, such as sod-3 and -5 genes. Moreover, a SKN-1 target, the insulin peptide gene ins-5, partially mediates the expression of DAF-16 targets via p38 MAPK signaling. These findings suggest that the interaction of ins/IGF-1 and p38 MAPK signaling pathways plays an important role in the fine-tuning of molecular compensation among sod genes to protect against mitochondrial oxidative damage during aging. Mitochondrial ROS is removed by SODs during aging. Expression of sod genes in C. elegans related to lifespan maintenance. Interaction of the ins/IGF-1 and p38 MAPK signalings regulates the fine-tuning of sod genes expression. ins-5 of SKN-1 target encodes an agonist of ins/IGF-1 signaling.
Collapse
Affiliation(s)
- Sumino Yanase
- Department of Health Science, Daito Bunka University School of Sports & Health Science, 560 Iwadono, Higashi-matsuyama, Saitama, 355-8501, Japan
| | - Kayo Yasuda
- Department of Health Management, Tokai University Undergraduate School of Health Studies, 4-1-1 Kitakaname, Hiratsuka, Kanagawa, 259-1292, Japan
| | - Naoaki Ishii
- Department of Health Management, Tokai University Undergraduate School of Health Studies, 4-1-1 Kitakaname, Hiratsuka, Kanagawa, 259-1292, Japan
| |
Collapse
|
17
|
The Neutrally Charged Diarylurea Compound PQ401 Kills Antibiotic-Resistant and Antibiotic-Tolerant Staphylococcus aureus. mBio 2020; 11:mBio.01140-20. [PMID: 32605985 PMCID: PMC7327171 DOI: 10.1128/mbio.01140-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Membrane-damaging antimicrobial agents have great potential to treat multidrug-resistant or multidrug-tolerant bacteria against which conventional antibiotics are not effective. However, their therapeutic applications are often hampered due to their low selectivity to bacterial over mammalian membranes or their potential for cross-resistance to a broad spectrum of cationic membrane-active antimicrobial agents. We discovered that the diarylurea derivative compound PQ401 has antimicrobial potency against multidrug-resistant and multidrug-tolerant Staphylococcus aureus. PQ401 selectively disrupts bacterial membrane lipid bilayers in comparison to mammalian membranes. Unlike cationic membrane-active antimicrobials, the neutral form of PQ401 rather than its cationic form exhibits maximum membrane activity. Overall, our results demonstrate that PQ401 could be a promising lead compound that overcomes the current limitations of membrane selectivity and cross-resistance. Also, this work provides deeper insight into the design and development of new noncharged membrane-targeting therapeutics to combat hard-to-cure bacterial infections. Resistance or tolerance to traditional antibiotics is a challenging issue in antimicrobial chemotherapy. Moreover, traditional bactericidal antibiotics kill only actively growing bacterial cells, whereas nongrowing metabolically inactive cells are tolerant to and therefore “persist” in the presence of legacy antibiotics. Here, we report that the diarylurea derivative PQ401, previously characterized as an inhibitor of the insulin-like growth factor I receptor, kills both antibiotic-resistant and nongrowing antibiotic-tolerant methicillin-resistant Staphylococcus aureus (MRSA) by lipid bilayer disruption. PQ401 showed several beneficial properties as an antimicrobial lead compound, including rapid killing kinetics, low probability for resistance development, high selectivity to bacterial membranes compared to mammalian membranes, and synergism with gentamicin. In contrast to well-studied membrane-disrupting cationic antimicrobial low-molecular-weight compounds and peptides, molecular dynamic simulations supported by efficacy data demonstrate that the neutral form of PQ401 penetrates and subsequently embeds into bacterial lipid bilayers more effectively than the cationic form. Lastly, PQ401 showed efficacy in both the Caenorhabditis elegans and Galleria mellonella models of MRSA infection. These data suggest that PQ401 may be a lead candidate for repurposing as a membrane-active antimicrobial and has potential for further development as a human antibacterial therapeutic for difficult-to-treat infections caused by both drug-resistant and -tolerant S. aureus.
Collapse
|
18
|
Sphingosine kinase and p38 MAP kinase signaling promote resistance to arsenite-induced lethality in Caenorhabditis elegan. Mol Cell Toxicol 2019. [DOI: 10.1007/s13273-019-0045-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
19
|
Tharmalingam N, Khader R, Fuchs BB, Mylonakis E. The Anti-virulence Efficacy of 4-(1,3-Dimethyl-2,3-Dihydro-1H-Benzimidazol-2-yl)Phenol Against Methicillin-Resistant Staphylococcus aureus. Front Microbiol 2019; 10:1557. [PMID: 31379761 PMCID: PMC6653400 DOI: 10.3389/fmicb.2019.01557] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 06/21/2019] [Indexed: 12/24/2022] Open
Abstract
Antimicrobial drug discovery against drug-resistant bacteria is an urgent need. Beyond agents with direct antibacterial activity, anti-virulent molecules may also be viable compounds to defend against bacterial pathogenesis. Using a high throughput screen (HTS) that utilized Caenorhabditis elegans infected with methicillin-resistant Staphylococcus aureus (MRSA) strain of MW2, we identified 4-(1,3-dimethyl-2,3-dihydro-1H-benzimidazol-2-yl)phenol (BIP). Interestingly, BIP had no in vitro inhibition activity against MW2, at least up to 64 μg/ml. The lack of direct antimicrobial activity suggests that BIP could inhibit bacterial virulence factors. To explore the possible anti-virulence effect of the identified molecule, we first performed real-time PCR to examine changes in virulence expression. BIP was highly active against MRSA virulence factors at sub-lethal concentrations and down-regulated virulence regulator genes, such as agrA and codY. However, the benzimidazole derivatives omeprazole and pantoprazole did not down-regulate virulence genes significantly, compared to BIP. Moreover, the BIP-pretreated MW2 cells were more vulnerable to macrophage-mediated killing, as confirmed by intracellular killing and live/dead staining assays, and less efficient in establishing a lethal infection in the invertebrate host Galleria mellonella (p = 0.0131). We tested the cytotoxicity of BIP against human red blood cells (RBCs), and it did not cause hemolysis at the highest concentration tested (64 μg/ml). Taken together, our findings outline the potential anti-virulence activity of BIP that was identified through a C. elegans-based, whole animal based, screen.
Collapse
Affiliation(s)
| | | | | | - Eleftherios Mylonakis
- Department of Medicine, Division of Infectious Diseases, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI, United States
| |
Collapse
|
20
|
Foley SJ, Wu Z, Politz SM. A C. elegans MAP kinase pathway is required for wild-type display of an L1-specific surface antigen ( srf-6 is nsy-1 III). MICROPUBLICATION BIOLOGY 2019; 2019:10.17912/micropub.biology.000129. [PMID: 32550420 PMCID: PMC7252279 DOI: 10.17912/micropub.biology.000129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Stephen J. Foley
- Department of Chemistry and Biochemistry, Worcester Polytechnic Institute, Worcester, MA
| | - Zheyang Wu
- Department of Mathematical Sciences, Worcester Polytechnic Institute, Worcester, MA
| | - Samuel M. Politz
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA,
Correspondence to: Samuel M. Politz ()
| |
Collapse
|
21
|
Shin H, Kaplan REW, Duong T, Fakieh R, Reiner DJ. Ral Signals through a MAP4 Kinase-p38 MAP Kinase Cascade in C. elegans Cell Fate Patterning. Cell Rep 2018; 24:2669-2681.e5. [PMID: 30184501 PMCID: PMC6484852 DOI: 10.1016/j.celrep.2018.08.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/18/2018] [Accepted: 08/06/2018] [Indexed: 12/24/2022] Open
Abstract
C. elegans vulval precursor cell (VPC) fates are patterned by an epidermal growth factor (EGF) gradient. High-dose EGF induces 1° VPC fate, and lower dose EGF contributes to 2° fate in support of LIN-12/Notch. We previously showed that the EGF 2°-promoting signal is mediated by LET-60/Ras switching effectors, from the canonical Raf-MEK-ERK mitogen-activated protein (MAP) kinase cascade that promotes 1° fate to the non-canonical RalGEF-Ral that promotes 2° fate. Of oncogenic Ras effectors, RalGEF-Ral is by far the least well understood. We use genetic analysis to identify an effector cascade downstream of C. elegans RAL-1/Ral, starting with an established Ral binding partner, Exo84 of the exocyst complex. Additionally, RAL-1 signals through GCK-2, a citron-N-terminal-homology-domain-containing MAP4 kinase, and PMK-1/p38 MAP kinase cascade to promote 2° fate. Our study delineates a Ral-dependent developmental signaling cascade in vivo, thus providing the mechanism by which lower EGF dose is transduced.
Collapse
Affiliation(s)
- Hanna Shin
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA
| | - Rebecca E W Kaplan
- Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tam Duong
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA
| | - Razan Fakieh
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA
| | - David J Reiner
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA; Department of Medical Physiology, College of Medicine, Texas A&M University, College Station, TX 77843, USA; Department of Pharmacology and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
22
|
Sphingosine Kinase Regulates Neuropeptide Secretion During the Oxidative Stress-Response Through Intertissue Signaling. J Neurosci 2018; 38:8160-8176. [PMID: 30082417 DOI: 10.1523/jneurosci.0536-18.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 07/19/2018] [Accepted: 07/20/2018] [Indexed: 12/12/2022] Open
Abstract
The Nrf2 antioxidant transcription factor promotes redox homeostasis in part through reciprocal signaling between neurons and neighboring cells, but the signals involved in intertissue signaling in response to Nrf2 activation are not well defined. In Caenorhabditis elegans, activation of SKN-1/Nrf2 in the intestine negatively regulates neuropeptide secretion from motor neurons. Here, we show that sphingosine kinase (SPHK-1) functions downstream of SKN-1/Nrf2 in the intestine to regulate neuropeptide secretion from motor neurons during the oxidative stress response in C. elegans hermaphrodites. SPHK-1 localizes to mitochondria in the intestine and SPHK-1 mitochondrial localization and kinase activity are essential for its function in regulating motor neuron function. SPHK-1 is recruited to mitochondria from cytosolic pools and its mitochondrial abundance is negatively regulated by acute or chronic SKN-1 activation. Finally, the regulation of motor function by SKN-1 requires the activation of the p38 MAPK cascade in the intestine and occurs through controlling the biogenesis or maturation of dense core vesicles in motor neurons. These findings show that the inhibition of SPHK-1 in the intestine by SKN-1 negatively regulates neuropeptide secretion from motor neurons, revealing a new mechanism by which SPHK-1 signaling mediates its effects on neuronal function in response to oxidative stress.SIGNIFICANCE STATEMENT Neurons are highly susceptible to damage by oxidative stress, yet have limited capacity to activate the SKN-1/Nrf2 oxidative stress response, relying instead on astrocytes to provide redox homeostasis. In Caenorhabditis elegans, intertissue signaling from the intestine plays a key role in regulating neuronal function during the oxidative stress response. Here, through a combination of genetic, behavioral, and fluorescent imaging approaches, we found that sphingosine kinase functions in the SKN-1/Nrf2 pathway in the intestine to regulate neuropeptide biogenesis and secretion in motor neurons. These results implicate sphingolipid signaling as a new component of the oxidative stress response and suggest that C. elegans may be a genetically tractable model to study non-cell-autonomous oxidative stress signaling to neurons.
Collapse
|
23
|
Kimura Y, Tsutsumi K, Konno A, Ikegami K, Hameed S, Kaneko T, Kaplan OI, Teramoto T, Fujiwara M, Ishihara T, Blacque OE, Setou M. Environmental responsiveness of tubulin glutamylation in sensory cilia is regulated by the p38 MAPK pathway. Sci Rep 2018; 8:8392. [PMID: 29849065 PMCID: PMC5976657 DOI: 10.1038/s41598-018-26694-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 05/16/2018] [Indexed: 01/09/2023] Open
Abstract
Glutamylation is a post-translational modification found on tubulin that can alter the interaction between microtubules (MTs) and associated proteins. The molecular mechanisms regulating tubulin glutamylation in response to the environment are not well understood. Here, we show that in the sensory cilia of Caenorhabditis elegans, tubulin glutamylation is upregulated in response to various signals such as temperature, osmolality, and dietary conditions. Similarly, tubulin glutamylation is modified in mammalian photoreceptor cells following light adaptation. A tubulin glutamate ligase gene ttll-4, which is essential for tubulin glutamylation of axonemal MTs in sensory cilia, is activated by p38 MAPK. Amino acid substitution of TTLL-4 has revealed that a Thr residue (a putative MAPK-phosphorylation site) is required for enhancement of tubulin glutamylation. Intraflagellar transport (IFT), a bidirectional trafficking system specifically observed along axonemal MTs, is required for the formation, maintenance, and function of sensory cilia. Measurement of the velocity of IFT particles revealed that starvation accelerates IFT, which was also dependent on the Thr residue of TTLL-4. Similarly, starvation-induced attenuation of avoidance behaviour from high osmolality conditions was also dependent on ttll-4. Our data suggest that a novel evolutionarily conserved regulatory system exists for tubulin glutamylation in sensory cilia in response to the environment.
Collapse
Affiliation(s)
- Yoshishige Kimura
- Department of Cellular & Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
- Department of Liberal Arts and Sciences, Kanagawa University of Human Services, 1-10-1 Heisei-cho, Yokosuka, Kanagawa, 238-8522, Japan
| | - Koji Tsutsumi
- Department of Cellular & Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
- Division of Cell Biology, Department of Biosciences, School of Science, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0373, Japan
| | - Alu Konno
- Department of Cellular & Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Koji Ikegami
- Department of Cellular & Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
- Department of Anatomy and Developmental Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Hiroshima, 734-8553, Japan
| | - Saira Hameed
- Department of Cellular & Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan
| | - Tomomi Kaneko
- Department of Liberal Arts and Sciences, Kanagawa University of Human Services, 1-10-1 Heisei-cho, Yokosuka, Kanagawa, 238-8522, Japan
| | - Oktay Ismail Kaplan
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin, 4, Ireland
- Abdullah Gul Universitesi, Doga Bilimleri Fakultesi, Sumer Kampusu, 38090, Kocasinan, Kayseri, Turkey
| | - Takayuki Teramoto
- Department of Biology, Faculty of Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Manabi Fujiwara
- Department of Biology, Faculty of Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Takeshi Ishihara
- Department of Biology, Faculty of Sciences, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka, 819-0395, Japan
| | - Oliver E Blacque
- School of Biomolecular and Biomedical Science, University College Dublin, Belfield, Dublin, 4, Ireland
| | - Mitsutoshi Setou
- Department of Cellular & Molecular Anatomy, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan.
- Department of Systems Molecular Anatomy, Institute for Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Hamamatsu, Shizuoka, 431-3192, Japan.
- The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan.
- Department of Anatomy, The University of Hong Kong, Hong Kong, China.
- Division of Neural Systematics, National Institute for Physiological Sciences, 38 Nishigonaka Myodaiji, Okazaki, Aichi, 444-8585, Japan.
| |
Collapse
|
24
|
Tharmalingam N, Rajmuthiah R, Kim W, Fuchs BB, Jeyamani E, Kelso MJ, Mylonakis E. Antibacterial Properties of Four Novel Hit Compounds from a Methicillin-Resistant Staphylococcus aureus-Caenorhabditis elegans High-Throughput Screen. Microb Drug Resist 2018; 24:666-674. [PMID: 29461939 DOI: 10.1089/mdr.2017.0250] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
There is an urgent need for the discovery of effective new antimicrobial agents to combat the rise of bacterial drug resistance. High-throughput screening (HTS) in whole-animal infection models is a powerful tool for identifying compounds that show antibacterial activity and low host toxicity. In this report, we characterize the activities of four novel antistaphylococcal compounds identified from an HTS campaign conducted using Caenorhabditis elegans nematodes infected with methicillin-resistant Staphylococcus aureus (MRSA). The hit compounds included an N-hydroxy indole-1, a substituted melamine derivative-2, N-substituted indolic alkyl isothiocyanate-3, and p-difluoromethylsulfide analog-4 of the well-known protonophore carbonyl cyanide m-chlorophenyl hydrazone. Minimal inhibitory concentrations (MICs) of the four compounds ranged from 2 to 8 μg/ml against MRSA-MW2 and Enterococcus faecium and all were bacteriostatic. The compounds were mostly inactive against Gram-negative pathogens, with only 1 and 4 showing slight activity (MIC = 32 μg/ml) against Acinetobacter baumanii. Compounds 2 and 3 (but not 1 or 4) were found to perturb MRSA membranes. In phagocytosis assays, compounds 1, 2, and 4 inhibited the growth of internalized MRSA in macrophages, whereas compound 3 showed a remarkable ability to clear intracellular MRSA at its MIC (p < 0.001). None of the compounds showed hemolytic activity at concentrations below 64 μg/ml (p = 0.0021). Compounds 1, 2, and 4 (but not 3) showed synergistic activity against MRSA with ciprofloxacin, while compound 3 synergized with erythromycin, gentamicin, streptomycin, and vancomycin. In conclusion, we describe four new antistaphylococcal compounds that warrant further study as novel antibacterial agents against Gram-positive organisms.
Collapse
Affiliation(s)
- Nagendran Tharmalingam
- 1 Infectious Diseases Division, Department of Medicine, Warren Alpert Medical School of Brown University , Rhode Island Hospital, Providence, Rhode Island
| | - Rajmohan Rajmuthiah
- 1 Infectious Diseases Division, Department of Medicine, Warren Alpert Medical School of Brown University , Rhode Island Hospital, Providence, Rhode Island
| | - Wooseong Kim
- 1 Infectious Diseases Division, Department of Medicine, Warren Alpert Medical School of Brown University , Rhode Island Hospital, Providence, Rhode Island
| | - Beth Burgwyn Fuchs
- 1 Infectious Diseases Division, Department of Medicine, Warren Alpert Medical School of Brown University , Rhode Island Hospital, Providence, Rhode Island
| | - Elamparithi Jeyamani
- 2 Massachusetts General Hospital , Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | - Michael J Kelso
- 3 Illawarra Health and Medical Research Institute and School of Chemistry, University of Wollongong , Wollongong, New South Wales
| | - Eleftherios Mylonakis
- 1 Infectious Diseases Division, Department of Medicine, Warren Alpert Medical School of Brown University , Rhode Island Hospital, Providence, Rhode Island
| |
Collapse
|
25
|
Crawley O, Giles AC, Desbois M, Kashyap S, Birnbaum R, Grill B. A MIG-15/JNK-1 MAP kinase cascade opposes RPM-1 signaling in synapse formation and learning. PLoS Genet 2017; 13:e1007095. [PMID: 29228003 PMCID: PMC5754208 DOI: 10.1371/journal.pgen.1007095] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 01/04/2018] [Accepted: 11/01/2017] [Indexed: 12/18/2022] Open
Abstract
The Pam/Highwire/RPM-1 (PHR) proteins are conserved intracellular signaling hubs that regulate synapse formation and axon termination. The C. elegans PHR protein, called RPM-1, acts as a ubiquitin ligase to inhibit the DLK-1 and MLK-1 MAP kinase pathways. We have identified several kinases that are likely to form a new MAP kinase pathway that suppresses synapse formation defects, but not axon termination defects, in the mechanosensory neurons of rpm-1 mutants. This pathway includes: MIG-15 (MAP4K), NSY-1 (MAP3K), JKK-1 (MAP2K) and JNK-1 (MAPK). Transgenic overexpression of kinases in the MIG-15/JNK-1 pathway is sufficient to impair synapse formation in wild-type animals. The MIG-15/JNK-1 pathway functions cell autonomously in the mechanosensory neurons, and these kinases localize to presynaptic terminals providing further evidence of a role in synapse development. Loss of MIG-15/JNK-1 signaling also suppresses defects in habituation to repeated mechanical stimuli in rpm-1 mutants, a behavioral deficit that is likely to arise from impaired glutamatergic synapse formation. Interestingly, habituation results are consistent with the MIG-15/JNK-1 pathway functioning as a parallel opposing pathway to RPM-1. These findings indicate the MIG-15/JNK-1 pathway can restrict both glutamatergic synapse formation and short-term learning.
Collapse
Affiliation(s)
- Oliver Crawley
- Department of Neuroscience, The Scripps Research Institute, Scripps Florida, Jupiter, Florida, United States of America
| | - Andrew C. Giles
- Department of Neuroscience, The Scripps Research Institute, Scripps Florida, Jupiter, Florida, United States of America
| | - Muriel Desbois
- Department of Neuroscience, The Scripps Research Institute, Scripps Florida, Jupiter, Florida, United States of America
| | - Sudhanva Kashyap
- Department of Neuroscience, The Scripps Research Institute, Scripps Florida, Jupiter, Florida, United States of America
| | - Rayna Birnbaum
- Harriet L. Wilkes Honors College, Florida Atlantic University, Jupiter, FL, United States of America
| | - Brock Grill
- Department of Neuroscience, The Scripps Research Institute, Scripps Florida, Jupiter, Florida, United States of America
| |
Collapse
|
26
|
Alqadah A, Hsieh YW, Xiong R, Chuang CF. Stochastic left-right neuronal asymmetry in Caenorhabditis elegans. Philos Trans R Soc Lond B Biol Sci 2017; 371:rstb.2015.0407. [PMID: 27821536 DOI: 10.1098/rstb.2015.0407] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2016] [Indexed: 12/28/2022] Open
Abstract
Left-right asymmetry in the nervous system is observed across species. Defects in left-right cerebral asymmetry are linked to several neurological diseases, but the molecular mechanisms underlying brain asymmetry in vertebrates are still not very well understood. The Caenorhabditis elegans left and right amphid wing 'C' (AWC) olfactory neurons communicate through intercellular calcium signalling in a transient embryonic gap junction neural network to specify two asymmetric subtypes, AWCOFF (default) and AWCON (induced), in a stochastic manner. Here, we highlight the molecular mechanisms that establish and maintain stochastic AWC asymmetry. As the components of the AWC asymmetry pathway are highly conserved, insights from the model organism C. elegans may provide a window onto how brain asymmetry develops in humans.This article is part of the themed issue 'Provocative questions in left-right asymmetry'.
Collapse
Affiliation(s)
- Amel Alqadah
- Department of Biological Sciences, University of Illinois at Chicago, IL 60607, USA
| | - Yi-Wen Hsieh
- Department of Biological Sciences, University of Illinois at Chicago, IL 60607, USA
| | - Rui Xiong
- Department of Biological Sciences, University of Illinois at Chicago, IL 60607, USA
| | - Chiou-Fen Chuang
- Department of Biological Sciences, University of Illinois at Chicago, IL 60607, USA
| |
Collapse
|
27
|
Alqadah A, Hsieh YW, Morrissey ZD, Chuang CF. Asymmetric development of the nervous system. Dev Dyn 2017; 247:124-137. [PMID: 28940676 DOI: 10.1002/dvdy.24595] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 09/09/2017] [Accepted: 09/18/2017] [Indexed: 12/13/2022] Open
Abstract
The human nervous system consists of seemingly symmetric left and right halves. However, closer observation of the brain reveals anatomical and functional lateralization. Defects in brain asymmetry correlate with several neurological disorders, yet our understanding of the mechanisms used to establish lateralization in the human central nervous system is extremely limited. Here, we review left-right asymmetries within the nervous system of humans and several model organisms, including rodents, Zebrafish, chickens, Xenopus, Drosophila, and the nematode Caenorhabditis elegans. Comparing and contrasting mechanisms used to develop left-right asymmetry in the nervous system can provide insight into how the human brain is lateralized. Developmental Dynamics 247:124-137, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Amel Alqadah
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Yi-Wen Hsieh
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois
| | - Zachery D Morrissey
- Graduate Program in Neuroscience, University of Illinois at Chicago, Chicago, Illinois
| | - Chiou-Fen Chuang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois.,Graduate Program in Neuroscience, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
28
|
The SEK-1 p38 MAP Kinase Pathway Modulates Gq Signaling in Caenorhabditis elegans. G3-GENES GENOMES GENETICS 2017; 7:2979-2989. [PMID: 28696924 PMCID: PMC5592925 DOI: 10.1534/g3.117.043273] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Gq is a heterotrimeric G protein that is widely expressed in neurons and regulates neuronal activity. To identify pathways regulating neuronal Gq signaling, we performed a forward genetic screen in Caenorhabditis elegans for suppressors of activated Gq. One of the suppressors is an allele of sek-1, which encodes a mitogen-activated protein kinase kinase (MAPKK) in the p38 MAPK pathway. Here, we show that sek-1 mutants have a slow locomotion rate and that sek-1 acts in acetylcholine neurons to modulate both locomotion rate and Gq signaling. Furthermore, we find that sek-1 acts in mature neurons to modulate locomotion. Using genetic and behavioral approaches, we demonstrate that other components of the p38 MAPK pathway also play a positive role in modulating locomotion and Gq signaling. Finally, we find that mutants in the SEK-1 p38 MAPK pathway partially suppress an activated mutant of the sodium leak channel, NCA-1/NALCN, a downstream target of Gq signaling. Our results suggest that the SEK-1 p38 pathway may modulate the output of Gq signaling through NCA-1(unc-77).
Collapse
|
29
|
Activity of a novel protonophore against methicillin-resistant Staphylococcus aureus. Future Med Chem 2017; 9:1401-1411. [PMID: 28771026 DOI: 10.4155/fmc-2017-0047] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
AIM Compound 1-(4-chlorophenyl)-4,4,4-trifluoro-3-hydroxy-2-buten-1-one (compound 1) was identified as a hit against methicillin-resistant Staphylococcus aureus (MRSA) strain MW2. METHODS & RESULTS The MIC of compound 1 against MRSA was 4 μg/ml. The compound showed enhanced activity at acidic pH by lowering bacterial intracellular pH and exhibited no lysis of human red blood cells at up to 64 μg/ml and its IC50 against HepG2 cells was 32 μg/ml. The compound reduced 1-log10 colony forming units of intracellular MRSA in macrophages and prolonged the survival of MRSA-infected Caenorhabditis elegans (p = 0.0015) and Galleria mellonella (p = 0.0002). CONCLUSION Compound 1 is a protonophore with potent in vitro and in vivo activity against MRSA and no toxicity in mammalian cells up to 8 μg/ml that warrants further investigation as a novel antibacterial.
Collapse
|
30
|
Wang J, Du H, Nie Y, Wang Y, Dai H, Wang M, Wang D, Xu A. Mitochondria and MAPK cascades modulate endosulfan-induced germline apoptosis in Caenorhabditis elegans. Toxicol Res (Camb) 2017; 6:412-419. [PMID: 30090509 PMCID: PMC6062295 DOI: 10.1039/c7tx00046d] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 04/11/2017] [Indexed: 12/27/2022] Open
Abstract
Endosulfan as a new member of persistent organic pollutants has been shown to induce apoptosis in various animal models. However, the mechanism underlying endosulfan-induced apoptosis has not been well elucidated thus far. Caenorhabditis elegans N2 wild type and mutant strains were used in the present study to clarify the roles of the mitochondria, the insulin/insulin-like growth factor-1 (IGF-1) signaling pathway, and mitogen-activated protein kinase (MAPK) cascades in α-endosulfan-induced apoptosis. Our results demonstrated a dose- and time-dependent increase of apoptosis in the meiotic zone of the gonad of C. elegans exposed to graded concentrations of endosulfan. The expression levels of sod-3, localized in the mitochondrial matrix, increased greatly after endosulfan exposure. A significant increase in germ cell apoptosis was observed in abnormal methyl viologen sensitivity-1 (mev-1(kn-1)) mutants (with abnormal mitochondrial respiratory chain complex II and higher ROS levels) compared to that in N2 at equal endosulfan concentrations. We found that the insulin/IGF-1 signaling pathway and its downstream Ras/ERK/MAPK did not participate in the endosulfan-induced apoptosis. However, the apoptosis in the loss-of-function strains of JNK and p38 MAPK signaling pathways was completely or mildly suppressed under endosulfan stress. The apoptotic effects of endosulfan were blocked in the mutants of jnk-1/JNK-MAPK, sek-1/MAP2K, and pmk-1/p38-MAPK, suggesting that these downstream genes play an essential role in endosulfan-induced germ cell apoptosis. In contrast, the mkk-4/MAP2K and nsy-1/MAP3K were only partially involved in the apoptosis induction. Our data provide evidence that endosulfan increases germ cell apoptosis, which is regulated by mitochondrial function, JNK and p38 MAPK cascades. These findings contribute to the understanding of the signal transduction pathways involved in endosulfan-induced apoptosis.
Collapse
Affiliation(s)
- Jingjing Wang
- School of Environmental Science and Optoelectronic Technology , University of Science and Technology of China , Hefei , Anhui 230026 , P. R. China
- Key Laboratory of Ion Beam Bioengineering , Hefei Institutes of Physical Science , Chinese Academy of Sciences and Anhui Province , Hefei , Anhui 230031 , P. R. China .
| | - Hua Du
- Key Laboratory of Ion Beam Bioengineering , Hefei Institutes of Physical Science , Chinese Academy of Sciences and Anhui Province , Hefei , Anhui 230031 , P. R. China .
| | - Yaguang Nie
- Key Laboratory of Ion Beam Bioengineering , Hefei Institutes of Physical Science , Chinese Academy of Sciences and Anhui Province , Hefei , Anhui 230031 , P. R. China .
| | - Yun Wang
- School of Life Sciences , University of Science and Technology of China , Hefei , Anhui 230027 , P. R. China
| | - Hui Dai
- Key Laboratory of Ion Beam Bioengineering , Hefei Institutes of Physical Science , Chinese Academy of Sciences and Anhui Province , Hefei , Anhui 230031 , P. R. China .
| | - Mudi Wang
- Key Laboratory of Ion Beam Bioengineering , Hefei Institutes of Physical Science , Chinese Academy of Sciences and Anhui Province , Hefei , Anhui 230031 , P. R. China .
| | - Dayan Wang
- School of Environmental Science and Optoelectronic Technology , University of Science and Technology of China , Hefei , Anhui 230026 , P. R. China
- Key Laboratory of Ion Beam Bioengineering , Hefei Institutes of Physical Science , Chinese Academy of Sciences and Anhui Province , Hefei , Anhui 230031 , P. R. China .
| | - An Xu
- Key Laboratory of Ion Beam Bioengineering , Hefei Institutes of Physical Science , Chinese Academy of Sciences and Anhui Province , Hefei , Anhui 230031 , P. R. China .
| |
Collapse
|
31
|
Lee TY, Yoon KH, Lee JI. NGT-3D: a simple nematode cultivation system to study Caenorhabditis elegans biology in 3D. Biol Open 2016; 5:529-34. [PMID: 26962047 PMCID: PMC4890662 DOI: 10.1242/bio.015743] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The nematode Caenorhabditiselegans is one of the premier experimental model organisms today. In the laboratory, they display characteristic development, fertility, and behaviors in a two dimensional habitat. In nature, however, C. elegans is found in three dimensional environments such as rotting fruit. To investigate the biology of C. elegans in a 3D controlled environment we designed a nematode cultivation habitat which we term the nematode growth tube or NGT-3D. NGT-3D allows for the growth of both nematodes and the bacteria they consume. Worms show comparable rates of growth, reproduction and lifespan when bacterial colonies in the 3D matrix are abundant. However, when bacteria are sparse, growth and brood size fail to reach levels observed in standard 2D plates. Using NGT-3D we observe drastic deficits in fertility in a sensory mutant in 3D compared to 2D, and this defect was likely due to an inability to locate bacteria. Overall, NGT-3D will sharpen our understanding of nematode biology and allow scientists to investigate questions of nematode ecology and evolutionary fitness in the laboratory. Summary: We have designed a three dimensional nematode habitat for use in the lab, called NGT-3D, where growth, fertility and lifespan are comparable with the traditional two dimensional lab environment.
Collapse
Affiliation(s)
- Tong Young Lee
- Division of Biological Science and Technology, College of Science and Technology, Yonsei University, Wonju 220-710, South Korea
| | - Kyoung-Hye Yoon
- Division of Biological Science and Technology, College of Science and Technology, Yonsei University, Wonju 220-710, South Korea
| | - Jin Il Lee
- Division of Biological Science and Technology, College of Science and Technology, Yonsei University, Wonju 220-710, South Korea
| |
Collapse
|
32
|
Andrusiak MG, Jin Y. Context Specificity of Stress-activated Mitogen-activated Protein (MAP) Kinase Signaling: The Story as Told by Caenorhabditis elegans. J Biol Chem 2016; 291:7796-804. [PMID: 26907690 DOI: 10.1074/jbc.r115.711101] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Stress-associated p38 and JNK mitogen-activated protein (MAP) kinase signaling cascades trigger specific cellular responses and are involved in multiple disease states. At the root of MAP kinase signaling complexity is the differential use of common components on a context-specific basis. The roundwormCaenorhabditis eleganswas developed as a system to study genes required for development and nervous system function. The powerful genetics ofC. elegansin combination with molecular and cellular dissections has led to a greater understanding of how p38 and JNK signaling affects many biological processes under normal and stress conditions. This review focuses on the studies revealing context specificity of different stress-activated MAPK components inC. elegans.
Collapse
Affiliation(s)
| | - Yishi Jin
- From the Howard Hughes Medical Institute and the Neurobiology Section, Division of Biological Sciences, University of California, San Diego, La Jolla, California 92093
| |
Collapse
|
33
|
Fukuzono T, Pastuhov SI, Fukushima O, Li C, Hattori A, Iemura SI, Natsume T, Shibuya H, Hanafusa H, Matsumoto K, Hisamoto N. Chaperone complex BAG2-HSC70 regulates localization of Caenorhabditis elegans leucine-rich repeat kinase LRK-1 to the Golgi. Genes Cells 2016; 21:311-24. [PMID: 26853528 DOI: 10.1111/gtc.12338] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 12/15/2015] [Indexed: 01/15/2023]
Abstract
Mutations in LRRK2 are linked to autosomal dominant forms of Parkinson's disease. We identified two human proteins that bind to LRRK2: BAG2 and HSC70, which are known to form a chaperone complex. We characterized the role of their Caenorhabditis elegans homologues, UNC-23 and HSP-1, in the regulation of LRK-1, the sole homologue of human LRRK2. In C. elegans, LRK-1 determines the polarized sorting of synaptic vesicle (SV) proteins to the axons by excluding SV proteins from the dendrite-specific transport machinery in the Golgi. In unc-23 mutants, SV proteins are localized to both presynaptic and dendritic endings in neurons, a phenotype also observed in lrk-1 deletion mutants. Furthermore, we isolated mutations in the hsp-1 gene that can suppress the unc-23, but not the lrk-1 defect. We show that UNC-23 determines LRK-1 localization to the Golgi apparatus in cooperation with HSP-1. These results describe a chaperone-dependent mechanism through which LRK-1 localization is regulated.
Collapse
Affiliation(s)
- Takashi Fukuzono
- Division of Biological Science, Graduate School of Science, Nagoya University, Chikusa-ku, Nagoya, 464-8602, Japan
| | - Strahil Iv Pastuhov
- Division of Biological Science, Graduate School of Science, Nagoya University, Chikusa-ku, Nagoya, 464-8602, Japan
| | - Okinobu Fukushima
- Division of Biological Science, Graduate School of Science, Nagoya University, Chikusa-ku, Nagoya, 464-8602, Japan
| | - Chun Li
- Division of Biological Science, Graduate School of Science, Nagoya University, Chikusa-ku, Nagoya, 464-8602, Japan
| | - Ayuna Hattori
- Division of Biological Science, Graduate School of Science, Nagoya University, Chikusa-ku, Nagoya, 464-8602, Japan
| | - Shun-ichiro Iemura
- National Institutes of Advanced Industrial Science and Technology, Molecular Profiling Research Center for Drug Discovery (Molprof), Kohtoh-ku, Tokyo, 135-0064, Japan
| | - Tohru Natsume
- National Institutes of Advanced Industrial Science and Technology, Molecular Profiling Research Center for Drug Discovery (Molprof), Kohtoh-ku, Tokyo, 135-0064, Japan
| | - Hiroshi Shibuya
- Department of Molecular Cell Biology, Medical Research Institute, Tokyo Medical and Dental University, Chiyoda-ku, Tokyo, 101-0062, Japan
| | - Hiroshi Hanafusa
- Division of Biological Science, Graduate School of Science, Nagoya University, Chikusa-ku, Nagoya, 464-8602, Japan
| | - Kunihiro Matsumoto
- Division of Biological Science, Graduate School of Science, Nagoya University, Chikusa-ku, Nagoya, 464-8602, Japan
| | - Naoki Hisamoto
- Division of Biological Science, Graduate School of Science, Nagoya University, Chikusa-ku, Nagoya, 464-8602, Japan
| |
Collapse
|
34
|
Alqadah A, Hsieh YW, Schumacher JA, Wang X, Merrill SA, Millington G, Bayne B, Jorgensen EM, Chuang CF. SLO BK Potassium Channels Couple Gap Junctions to Inhibition of Calcium Signaling in Olfactory Neuron Diversification. PLoS Genet 2016; 12:e1005654. [PMID: 26771544 PMCID: PMC4714817 DOI: 10.1371/journal.pgen.1005654] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 10/16/2015] [Indexed: 01/09/2023] Open
Abstract
The C. elegans AWC olfactory neuron pair communicates to specify asymmetric subtypes AWCOFF and AWCON in a stochastic manner. Intercellular communication between AWC and other neurons in a transient NSY-5 gap junction network antagonizes voltage-activated calcium channels, UNC-2 (CaV2) and EGL-19 (CaV1), in the AWCON cell, but how calcium signaling is downregulated by NSY-5 is only partly understood. Here, we show that voltage- and calcium-activated SLO BK potassium channels mediate gap junction signaling to inhibit calcium pathways for asymmetric AWC differentiation. Activation of vertebrate SLO-1 channels causes transient membrane hyperpolarization, which makes it an important negative feedback system for calcium entry through voltage-activated calcium channels. Consistent with the physiological roles of SLO-1, our genetic results suggest that slo-1 BK channels act downstream of NSY-5 gap junctions to inhibit calcium channel-mediated signaling in the specification of AWCON. We also show for the first time that slo-2 BK channels are important for AWC asymmetry and act redundantly with slo-1 to inhibit calcium signaling. In addition, nsy-5-dependent asymmetric expression of slo-1 and slo-2 in the AWCON neuron is necessary and sufficient for AWC asymmetry. SLO-1 and SLO-2 localize close to UNC-2 and EGL-19 in AWC, suggesting a role of possible functional coupling between SLO BK channels and voltage-activated calcium channels in AWC asymmetry. Furthermore, slo-1 and slo-2 regulate the localization of synaptic markers, UNC-2 and RAB-3, in AWC neurons to control AWC asymmetry. We also identify the requirement of bkip-1, which encodes a previously identified auxiliary subunit of SLO-1, for slo-1 and slo-2 function in AWC asymmetry. Together, these results provide an unprecedented molecular link between gap junctions and calcium pathways for terminal differentiation of olfactory neurons.
Collapse
Affiliation(s)
- Amel Alqadah
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Yi-Wen Hsieh
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Jennifer A. Schumacher
- Division of Developmental Biology, Cincinnati Children’s Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - Xiaohong Wang
- Division of Developmental Biology, Cincinnati Children’s Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - Sean A. Merrill
- Department of Biology and Howard Hughes Medical Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Grethel Millington
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Brittany Bayne
- Division of Developmental Biology, Cincinnati Children’s Hospital Research Foundation, Cincinnati, Ohio, United States of America
| | - Erik M. Jorgensen
- Department of Biology and Howard Hughes Medical Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Chiou-Fen Chuang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
35
|
Intracellular and Extracellular Expression of Bacillus thuringiensis Crystal Protein Cry5B in Lactococcus lactis for Use as an Anthelminthic. Appl Environ Microbiol 2015; 82:1286-94. [PMID: 26682852 PMCID: PMC4751831 DOI: 10.1128/aem.02365-15] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 11/22/2015] [Indexed: 12/20/2022] Open
Abstract
The Bacillus thuringiensis crystal (Cry) protein Cry5B (140 kDa) and a truncated version of the protein, tCry5B (79 kDa), are lethal to nematodes. Genes encoding the two proteins were separately cloned into a high-copy-number vector with a strong constitutive promoter (pTRK593) in Lactococcus lactis for potential oral delivery against parasitic nematode infections. Western blots using a Cry5B-specific antibody revealed that constitutively expressed Cry5B and tCry5B were present in both cells and supernatants. To increase production, cry5B was cloned into the high-copy-number plasmid pMSP3535H3, carrying a nisin-inducible promoter. Immunoblotting revealed that 3 h after nisin induction, intracellular Cry5B was strongly induced at 200 ng/ml nisin, without adversely affecting cell viability or cell membrane integrity. Both Cry5B genes were also cloned into plasmid pTRK1061, carrying a promoter and encoding a transcriptional activator that invoke low-level expression of prophage holin and lysin genes in Lactococcus lysogens, resulting in a leaky phenotype. Cry5B and tCry5B were actively expressed in the lysogenic strain L. lactis KP1 and released into cell supernatants without affecting culture growth. Lactate dehydrogenase (LDH) assays indicated that Cry5B, but not LDH, leaked from the bacteria. Lastly, using intracellular lysates from L. lactis cultures expressing both Cry5B and tCry5B, in vivo challenges of Caenorhabditis elegans worms demonstrated that the Cry proteins were biologically active. Taken together, these results indicate that active Cry5B proteins can be expressed intracellularly in and released extracellularly from L. lactis, showing potential for future use as an anthelminthic that could be delivered orally in a food-grade microbe.
Collapse
|
36
|
Caenorhabditis elegans as a Model to Study the Molecular and Genetic Mechanisms of Drug Addiction. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 137:229-52. [PMID: 26810004 DOI: 10.1016/bs.pmbts.2015.10.019] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Drug addiction takes a massive toll on society. Novel animal models are needed to test new treatments and understand the basic mechanisms underlying addiction. Rodent models have identified the neurocircuitry involved in addictive behavior and indicate that rodents possess some of the same neurobiologic mechanisms that mediate addiction in humans. Recent studies indicate that addiction is mechanistically and phylogenetically ancient and many mechanisms that underlie human addiction are also present in invertebrates. The nematode Caenorhabditis elegans has conserved neurobiologic systems with powerful molecular and genetic tools and a rapid rate of development that enables cost-effective translational discovery. Emerging evidence suggests that C. elegans is an excellent model to identify molecular mechanisms that mediate drug-induced behavior and potential targets for medications development for various addictive compounds. C. elegans emit many behaviors that can be easily quantitated including some that involve interactions with the environment. Ethanol (EtOH) is the best-studied drug-of-abuse in C. elegans and at least 50 different genes/targets have been identified as mediating EtOH's effects and polymorphisms in some orthologs in humans are associated with alcohol use disorders. C. elegans has also been shown to display dopamine and cholinergic system-dependent attraction to nicotine and demonstrate preference for cues previously associated with nicotine. Cocaine and methamphetamine have been found to produce dopamine-dependent reward-like behaviors in C. elegans. These behavioral tests in combination with genetic/molecular manipulations have led to the identification of dozens of target genes/systems in C. elegans that mediate drug effects. The one target/gene identified as essential for drug-induced behavioral responses across all drugs of abuse was the cat-2 gene coding for tyrosine hydroxylase, which is consistent with the role of dopamine neurotransmission in human addiction. Overall, C. elegans can be used to model aspects of drug addiction and identify systems and molecular mechanisms that mediate drug effects. The findings are surprisingly consistent with analogous findings in higher-level organisms. Further, model refinement is warranted to improve model validity and increase utility for medications development.
Collapse
|
37
|
Alqadah A, Hsieh YW, Vidal B, Chang C, Hobert O, Chuang CF. Postmitotic diversification of olfactory neuron types is mediated by differential activities of the HMG-box transcription factor SOX-2. EMBO J 2015; 34:2574-89. [PMID: 26341465 DOI: 10.15252/embj.201592188] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 08/06/2015] [Indexed: 12/23/2022] Open
Abstract
Diversification of neuron classes is essential for functions of the olfactory system, but the underlying mechanisms that generate individual olfactory neuron types are only beginning to be understood. Here we describe a role of the highly conserved HMG-box transcription factor SOX-2 in postmitotic specification and alternative differentiation of the Caenorhabditis elegans AWC and AWB olfactory neurons. We show that SOX-2 partners with different transcription factors to diversify postmitotic olfactory cell types. SOX-2 functions cooperatively with the OTX/OTD transcription factor CEH-36 to specify an AWC "ground state," and functions with the LIM homeodomain factor LIM-4 to suppress this ground state and drive an AWB identity instead. Our findings provide novel insights into combinatorial codes that drive terminal differentiation programs in the nervous system and reveal a biological function of the deeply conserved Sox2 protein that goes beyond its well-known role in stem cell biology.
Collapse
Affiliation(s)
- Amel Alqadah
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA Molecular and Developmental Biology Graduate Program, University of Cincinnati, Cincinnati, OH, USA
| | - Yi-Wen Hsieh
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Berta Vidal
- Department of Biological Sciences, Department of Biochemistry and Molecular Biophysics, Howard Hughes Medical Institute Columbia University, New York, NY, USA
| | - Chieh Chang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Oliver Hobert
- Department of Biological Sciences, Department of Biochemistry and Molecular Biophysics, Howard Hughes Medical Institute Columbia University, New York, NY, USA
| | - Chiou-Fen Chuang
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
38
|
Rajamuthiah R, Jayamani E, Conery AL, Fuchs BB, Kim W, Johnston T, Vilcinskas A, Ausubel FM, Mylonakis E. A Defensin from the Model Beetle Tribolium castaneum Acts Synergistically with Telavancin and Daptomycin against Multidrug Resistant Staphylococcus aureus. PLoS One 2015; 10:e0128576. [PMID: 26062137 PMCID: PMC4465704 DOI: 10.1371/journal.pone.0128576] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 04/28/2015] [Indexed: 01/19/2023] Open
Abstract
The red flour beetle Tribolium castaneum is a common insect pest and has been established as a model beetle to study insect development and immunity. This study demonstrates that defensin 1 from T. castaneum displays in vitro and in vivo antimicrobial activity against drug resistant Staphylococcus aureus strains. The minimum inhibitory concentration (MIC) of defensin 1 against 11 reference and clinical staphylococcal isolates was between 16–64 μg/ml. The putative mode of action of the defensin peptide is disruption of the bacterial cell membrane. The antibacterial activity of defensin 1 was attenuated by salt concentrations of 1.56 mM and 25 mM for NaCl and CaCl2 respectively. Treatment of defensin 1 with the reducing agent dithiothreitol (DTT) at concentrations 1.56 to 3.13 mM abolished the antimicrobial activity of the peptide. In the presence of subinhibitory concentrations of antibiotics that also target the bacterial cell envelope such as telavancin and daptomycin, the MIC of the peptide was as low as 1 μg/ml. Moreover, when tested against an S. aureus strain that was defective in D-alanylation of the cell wall, the MIC of the peptide was 0.5 μg/ml. Defensin 1 exhibited no toxicity against human erythrocytes even at 400 μg/ml. The in vivo activity of the peptide was validated in a Caenorhabditis elegans-MRSA liquid infection assay. These results suggest that defensin 1 behaves similarly to other cationic AMPs in its mode of action against S. aureus and that the activity of the peptide can be enhanced in combination with other antibiotics with similar modes of action or with compounds that have the ability to decrease D-alanylation of the bacterial cell wall.
Collapse
Affiliation(s)
- Rajmohan Rajamuthiah
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, 02903, United States of America
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, United States of America
| | - Elamparithi Jayamani
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, 02903, United States of America
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, United States of America
| | - Annie L. Conery
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, United States of America
| | - Beth Burgwyn Fuchs
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, 02903, United States of America
| | - Wooseong Kim
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, 02903, United States of America
| | - Tatiana Johnston
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, 02903, United States of America
| | - Andreas Vilcinskas
- Institute of Phytopathology and Applied Zoology, Justus-Liebig University, Heinrich-Buff-Ring 26–32, 35392, Giessen, Germany
| | - Frederick M. Ausubel
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, United States of America
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI, 02903, United States of America
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, United States of America
- * E-mail:
| |
Collapse
|
39
|
Rajamuthiah R, Fuchs BB, Conery AL, Kim W, Jayamani E, Kwon B, Ausubel FM, Mylonakis E. Repurposing salicylanilide anthelmintic drugs to combat drug resistant Staphylococcus aureus. PLoS One 2015; 10:e0124595. [PMID: 25897961 PMCID: PMC4405337 DOI: 10.1371/journal.pone.0124595] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2014] [Accepted: 03/16/2015] [Indexed: 01/10/2023] Open
Abstract
Staphylococcus aureus is a Gram-positive bacterium that has become the leading cause of hospital acquired infections in the US. Repurposing Food and Drug Administration (FDA) approved drugs for antimicrobial therapy involves lower risks and costs compared to de novo development of novel antimicrobial agents. In this study, we examined the antimicrobial properties of two commercially available anthelmintic drugs. The FDA approved drug niclosamide and the veterinary drug oxyclozanide displayed strong in vivo and in vitro activity against methicillin resistant S. aureus (minimum inhibitory concentration (MIC): 0.125 and 0.5 μg/ml respectively; minimum effective concentration: ≤ 0.78 μg/ml for both drugs). The two drugs were also effective against another Gram-positive bacteria Enterococcus faecium (MIC 0.25 and 2 μg/ml respectively), but not against the Gram-negative species Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter aerogenes. The in vitro antimicrobial activity of niclosamide and oxyclozanide were determined against methicillin, vancomycin, linezolid or daptomycin resistant S. aureus clinical isolates, with MICs at 0.0625-0.5 and 0.125-2 μg/ml for niclosamide and oxyclozanide respectively. A time-kill study demonstrated that niclosamide is bacteriostatic, whereas oxyclozanide is bactericidal. Interestingly, oxyclozanide permeabilized the bacterial membrane but neither of the anthelmintic drugs exhibited demonstrable toxicity to sheep erythrocytes. Oxyclozanide was non-toxic to HepG2 human liver carcinoma cells within the range of its in vitro MICs but niclosamide displayed toxicity even at low concentrations. These data show that the salicylanilide anthelmintic drugs niclosamide and oxyclozanide are suitable candidates for mechanism of action studies and further clinical evaluation for treatment of staphylococcal infections.
Collapse
Affiliation(s)
- Rajmohan Rajamuthiah
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Beth Burgwyn Fuchs
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Annie L. Conery
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Wooseong Kim
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Elamparithi Jayamani
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Bumsup Kwon
- Division of Neurology, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Frederick M. Ausubel
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
40
|
Pagano DJ, Kingston ER, Kim DH. Tissue expression pattern of PMK-2 p38 MAPK is established by the miR-58 family in C. elegans. PLoS Genet 2015; 11:e1004997. [PMID: 25671546 PMCID: PMC4335502 DOI: 10.1371/journal.pgen.1004997] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 01/12/2015] [Indexed: 11/18/2022] Open
Abstract
Analyses of gene expression profiles in evolutionarily diverse organisms have revealed a role for microRNAs in tuning tissue-specific gene expression. Here, we show that the relatively abundant and constitutively expressed miR-58 family of microRNAs sharply defines the tissue-specific expression of the broadly transcribed gene encoding PMK-2 p38 MAPK in Caenorhabditis elegans. Whereas PMK-2 functions redundantly with PMK-1 in the nervous system to regulate neuronal development and behavioral responses to pathogenic bacteria, the miR-58, miR-80, miR-81, and miR-82 microRNAs function redundantly to destabilize pmk-2 mRNA in non-neuronal cells with switch-like potency. Our data suggest a role for the miR-58 family in the establishment of neuronal-specific gene expression in C. elegans, and support a more general role for microRNAs in the establishment of tissue-specific gene expression.
Collapse
Affiliation(s)
- Daniel J. Pagano
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Elena R. Kingston
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Dennis H. Kim
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
41
|
Desalermos A, Tan X, Rajamuthiah R, Arvanitis M, Wang Y, Li D, Kourkoumpetis TK, Fuchs BB, Mylonakis E. A multi-host approach for the systematic analysis of virulence factors in Cryptococcus neoformans. J Infect Dis 2014; 211:298-305. [PMID: 25114160 DOI: 10.1093/infdis/jiu441] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
A multi-host approach was followed to screen a library of 1201 signature-tagged deletion strains of Cryptococcus neoformans mutants to identify previously unknown virulence factors. The primary screen was performed using a Caenorhabditis elegans-C. neoformans infection assay. The hits among these strains were reconfirmed as less virulent than the wild type in the insect Galleria mellonella-C. neoformans infection assay. After this 2-stage screen, and to prioritize hits, we performed serial evaluations of the selected strains, using the C. elegans model. All hit strains identified through these studies were validated in a murine model of systemic cryptococcosis. Twelve strains were identified through a stepwise screening assay. Among them, 4 (CSN1201, SRE1, RDI1, and YLR243W) were previously discovered, providing proof of principle for this approach, while the role of the remaining 8 genes (CKS101, CNC5600, YOL003C, CND1850, MLH3, HAP502, MSL5, and CNA2580) were not previously described in cryptococcal virulence. The multi-host approach is an efficient method of studying the pathogenesis of C. neoformans. We used diverse model hosts, C. elegans, G. mellonella, and mice, with physiological differences and identified 12 genes associated with mammalian infection. Our approach may be suitable for large pathogenesis screens.
Collapse
Affiliation(s)
- Athanasios Desalermos
- Division of Infectious diseases, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Xiaojiang Tan
- Division of Infectious diseases, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Rajmohan Rajamuthiah
- Division of Infectious diseases, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Marios Arvanitis
- Division of Infectious diseases, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Yan Wang
- Division of Infectious diseases, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Dedong Li
- Division of Infectious diseases, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | | | - Beth Burgwyn Fuchs
- Division of Infectious diseases, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Eleftherios Mylonakis
- Division of Infectious diseases, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
42
|
Hsieh YW, Alqadah A, Chuang CF. Asymmetric neural development in the Caenorhabditis elegans olfactory system. Genesis 2014; 52:544-54. [PMID: 24478264 PMCID: PMC4065219 DOI: 10.1002/dvg.22744] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 01/17/2014] [Accepted: 01/23/2014] [Indexed: 02/02/2023]
Abstract
Asymmetries in the nervous system have been observed throughout the animal kingdom. Deviations of brain asymmetries are associated with a variety of neurodevelopmental disorders; however, there has been limited progress in determining how normal asymmetry is established in vertebrates. In the Caenorhabditis elegans chemosensory system, two pairs of morphologically symmetrical neurons exhibit molecular and functional asymmetries. This review focuses on the development of antisymmetry of the pair of amphid wing "C" (AWC) olfactory neurons, from transcriptional regulation of general cell identity, establishment of asymmetry through neural network formation and calcium signaling, to the maintenance of asymmetry throughout the life of the animal. Many of the factors that are involved in AWC development have homologs in vertebrates, which may potentially function in the development of vertebrate brain asymmetry.
Collapse
Affiliation(s)
- Yi-Wen Hsieh
- Division of Developmental Biology, Cincinnati Children’s Hospital Research Foundation, Cincinnati, OH, USA
| | - Amel Alqadah
- Division of Developmental Biology, Cincinnati Children’s Hospital Research Foundation, Cincinnati, OH, USA
- Molecular and Developmental Biology Graduate Program, University of Cincinnati, OH, USA
| | - Chiou-Fen Chuang
- Division of Developmental Biology, Cincinnati Children’s Hospital Research Foundation, Cincinnati, OH, USA
| |
Collapse
|
43
|
Rajamuthiah R, Fuchs BB, Jayamani E, Kim Y, Larkins-Ford J, Conery A, Ausubel FM, Mylonakis E. Whole animal automated platform for drug discovery against multi-drug resistant Staphylococcus aureus. PLoS One 2014; 9:e89189. [PMID: 24586584 PMCID: PMC3929655 DOI: 10.1371/journal.pone.0089189] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Accepted: 01/15/2014] [Indexed: 11/19/2022] Open
Abstract
Staphylococcus aureus, the leading cause of hospital-acquired infections in the United States, is also pathogenic to the model nematode Caenorhabditis elegans. The C. elegans-S. aureus infection model was previously carried out on solid agar plates where the bacteriovorous C. elegans feeds on a lawn of S. aureus. However, agar-based assays are not amenable to large scale screens for antibacterial compounds. We have developed a high throughput liquid screening assay that uses robotic instrumentation to dispense a precise amount of methicillin resistant S. aureus (MRSA) and worms in 384-well assay plates, followed by automated microscopy and image analysis. In validation of the liquid assay, an MRSA cell wall defective mutant, MW2ΔtarO, which is attenuated for killing in the agar-based assay, was found to be less virulent in the liquid assay. This robust assay with a Z'-factor consistently greater than 0.5 was utilized to screen the Biomol 4 compound library consisting of 640 small molecules with well characterized bioactivities. As proof of principle, 27 of the 30 clinically used antibiotics present in the library conferred increased C. elegans survival and were identified as hits in the screen. Surprisingly, the antihelminthic drug closantel was also identified as a hit in the screen. In further studies, we confirmed the anti-staphylococcal activity of closantel against vancomycin-resistant S. aureus isolates and other Gram-positive bacteria. The liquid C. elegans-S. aureus assay described here allows screening for anti-staphylococcal compounds that are not toxic to the host.
Collapse
Affiliation(s)
- Rajmohan Rajamuthiah
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Beth Burgwyn Fuchs
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Elamparithi Jayamani
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Younghoon Kim
- Department of Animal Science, Chonbuk National University, Jeonju, Republic of Korea
| | - Jonah Larkins-Ford
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Annie Conery
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Frederick M. Ausubel
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
44
|
Sasakura H, Tsukada Y, Takagi S, Mori I. Japanese studies on neural circuits and behavior of Caenorhabditis elegans. Front Neural Circuits 2013; 7:187. [PMID: 24348340 PMCID: PMC3842693 DOI: 10.3389/fncir.2013.00187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Accepted: 11/03/2013] [Indexed: 01/25/2023] Open
Abstract
The nematode Caenorhabditis elegans is an ideal organism for studying neural plasticity and animal behaviors. A total of 302 neurons of a C. elegans hermaphrodite have been classified into 118 neuronal groups. This simple neural circuit provides a solid basis for understanding the mechanisms of the brains of higher animals, including humans. Recent studies that employ modern imaging and manipulation techniques enable researchers to study the dynamic properties of nervous systems with great precision. Behavioral and molecular genetic analyses of this tiny animal have contributed greatly to the advancement of neural circuit research. Here, we will review the recent studies on the neural circuits of C. elegans that have been conducted in Japan. Several laboratories have established unique and clever methods to study the underlying neuronal substrates of behavioral regulation in C. elegans. The technological advances applied to studies of C. elegans have allowed new approaches for the studies of complex neural systems. Through reviewing the studies on the neuronal circuits of C. elegans in Japan, we will analyze and discuss the directions of neural circuit studies.
Collapse
Affiliation(s)
- Hiroyuki Sasakura
- Laboratory of Molecular Neurobiology, Division of Biological Science, Nagoya University Nagoya, Japan
| | - Yuki Tsukada
- Laboratory of Molecular Neurobiology, Division of Biological Science, Nagoya University Nagoya, Japan
| | - Shin Takagi
- Laboratory of Brain Function and Structure, Division of Biological Science, Nagoya University Nagoya, Japan
| | - Ikue Mori
- Laboratory of Molecular Neurobiology, Division of Biological Science, Nagoya University Nagoya, Japan
| |
Collapse
|
45
|
Alqadah A, Hsieh YW, Chuang CF. microRNA function in left-right neuronal asymmetry: perspectives from C. elegans. Front Cell Neurosci 2013; 7:158. [PMID: 24065887 PMCID: PMC3779813 DOI: 10.3389/fncel.2013.00158] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 09/01/2013] [Indexed: 11/13/2022] Open
Abstract
Left-right asymmetry in anatomical structures and functions of the nervous system is present throughout the animal kingdom. For example, language centers are localized in the left side of the human brain, while spatial recognition functions are found in the right hemisphere in the majority of the population. Disruption of asymmetry in the nervous system is correlated with neurological disorders. Although anatomical and functional asymmetries are observed in mammalian nervous systems, it has been a challenge to identify the molecular basis of these asymmetries. C. elegans has emerged as a prime model organism to investigate molecular asymmetries in the nervous system, as it has been shown to display functional asymmetries clearly correlated to asymmetric distribution and regulation of biologically relevant molecules. Small non-coding RNAs have been recently implicated in various aspects of neural development. Here, we review cases in which microRNAs are crucial for establishing left-right asymmetries in the C. elegans nervous system. These studies may provide insight into how molecular and functional asymmetries are established in the human brain.
Collapse
Affiliation(s)
- Amel Alqadah
- Division of Developmental Biology, Cincinnati Children's Hospital Research Foundation Cincinnati, OH, USA ; Molecular and Developmental Biology Graduate Program, University of Cincinnati Cincinnati, OH, USA
| | | | | |
Collapse
|
46
|
Tao L, Xie Q, Ding YH, Li ST, Peng S, Zhang YP, Tan D, Yuan Z, Dong MQ. CAMKII and calcineurin regulate the lifespan of Caenorhabditis elegans through the FOXO transcription factor DAF-16. eLife 2013; 2:e00518. [PMID: 23805378 PMCID: PMC3691573 DOI: 10.7554/elife.00518] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 05/24/2013] [Indexed: 01/03/2023] Open
Abstract
The insulin-like signaling pathway maintains a relatively short wild-type lifespan in Caenorhabditis elegans by phosphorylating and inactivating DAF-16, the ortholog of the FOXO transcription factors of mammalian cells. DAF-16 is phosphorylated by the AKT kinases, preventing its nuclear translocation. Calcineurin (PP2B phosphatase) also limits the lifespan of C. elegans, but the mechanism through which it does so is unknown. Herein, we show that TAX-6•CNB-1 and UNC-43, the C. elegans Calcineurin and Ca(2+)/calmodulin-dependent kinase type II (CAMKII) orthologs, respectively, also regulate lifespan through DAF-16. Moreover, UNC-43 regulates DAF-16 in response to various stress conditions, including starvation, heat or oxidative stress, and cooperatively contributes to lifespan regulation by insulin signaling. However, unlike insulin signaling, UNC-43 phosphorylates and activates DAF-16, thus promoting its nuclear localization. The phosphorylation of DAF-16 at S286 by UNC-43 is removed by TAX-6•CNB-1, leading to DAF-16 inactivation. Mammalian FOXO3 is also regulated by CAMKIIA and Calcineurin. DOI:http://dx.doi.org/10.7554/eLife.00518.001.
Collapse
Affiliation(s)
- Li Tao
- Graduate Program in Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China ; National Institute of Biological Sciences, Beijing , Beijing , China
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Simske JS. Claudins reign: The claudin/EMP/PMP22/γ channel protein family in C. elegans. Tissue Barriers 2013; 1:e25502. [PMID: 24665403 PMCID: PMC3879130 DOI: 10.4161/tisb.25502] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Revised: 06/19/2013] [Accepted: 06/21/2013] [Indexed: 01/06/2023] Open
Abstract
The claudin family of integral membrane proteins was identified as the major protein component of the tight junctions in all vertebrates. Since their identification, claudins, and their associated pfam00822 superfamily of proteins have been implicated in a wide variety of cellular processes. Claudin homologs have been identified in invertebrates as well, including Drosophila and C. elegans. Recent studies demonstrate that the C. elegans claudins, clc-1-clc- 5, and similar proteins in the greater PMP22/EMP/claudin/voltage-gated calcium channel γ subunit family, including nsy-4, and vab-9, while highly divergent at a sequence level from each other and from the vertebrate claudins, in many cases play roles similar to those traditionally assigned to their vertebrate homologs. These include regulating cell adhesion and passage of small molecules through the paracellular space, channel activity, protein aggregation, sensitivity to pore-forming toxins, intercellular signaling, cell fate specification and dynamic changes in cell morphology. Study of claudin superfamily proteins in C. elegans should continue to provide clues as to how claudin family protein function has been adapted to perform diverse functions at specialized cell-cell contacts in metazoans.
Collapse
|
48
|
Chew YL, Fan X, Götz J, Nicholas HR. Aging in the nervous system of Caenorhabditis elegans. Commun Integr Biol 2013; 6:e25288. [PMID: 24255742 PMCID: PMC3829903 DOI: 10.4161/cib.25288] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 06/04/2013] [Accepted: 06/05/2013] [Indexed: 11/28/2022] Open
Abstract
It has recently been described that aging in C. elegans is accompanied by the progressive development of morphological changes in the nervous system. These include novel outgrowths from the cell body or axonal process, as well as blebbing and beading along the length of the axon. The formation of these structures is regulated by numerous molecular players including members of the well-conserved insulin/insulin growth factor-like (IGF)-1 signaling and mitogen-activated protein (MAP) kinase pathways. This review summarizes the recent literature on neuronal aging in C. elegans, including our own findings, which indicate a role for protein with tau-like repeats (PTL-1), the homolog of mammalian tau and MAP2/4, in maintaining neuronal integrity during aging.
Collapse
Affiliation(s)
- Yee Lian Chew
- School of Molecular Bioscience; University of Sydney; Sydney, NSW Australia
| | | | | | | |
Collapse
|
49
|
Mertenskötter A, Keshet A, Gerke P, Paul RJ. The p38 MAPK PMK-1 shows heat-induced nuclear translocation, supports chaperone expression, and affects the heat tolerance of Caenorhabditis elegans. Cell Stress Chaperones 2013; 18:293-306. [PMID: 23117578 PMCID: PMC3631094 DOI: 10.1007/s12192-012-0382-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Revised: 10/17/2012] [Accepted: 10/19/2012] [Indexed: 01/09/2023] Open
Abstract
The p38 mitogen-activated protein kinase PMK-1 of Caenorhabditis elegans has been associated with heavy metal, oxidative and pathogen stress. Pmk-1 is part of an operon comprising three p38 homologues, with pmk-1 expression suggested to be regulated by the operon promoter. There are contradictory reports about the cellular localization of PMK-1. We were interested to study principles of pmk-1 expression and to analyze the role of PMK-1 under heat stress. Using a translational GFP reporter, we found pmk-1 expression to be driven by a promoter in front of pmk-1. PMK-1 was detected in intestinal cells and neurons, with a cytoplasmic localization at moderate temperature. Increasing temperature above 32 °C, however, induced a nuclear translocation of PMK-1 as well as PMK-1 accumulation near to apical membranes. Testing survival rates revealed 34-35 °C as critical temperature range, where short-term survival severely decreased. Mutants of the PMK-1 pathway (pmk-1Δ, sek-1Δ, mek-1Δ) as well as a mutant of JNK pathway (jnk-1Δ) showed significantly lower survival rates than wild-type or mutants of other pathways (kgb-1Δ, daf-2Δ). Rescue and overexpression experiments verified the negative effects of pmk-1Δ on heat tolerance. Studying gene expression by RNA-seq and semi-quantitative reverse transcriptase polymerase chain reaction revealed positive effects of the PMK-1 pathway on the expression of genes for chaperones, protein biosynthesis, protein degradation, and other functional categories. Thus, the PMK-1 pathway is involved in the heat stress responses of C. elegans, possibly by a PMK-1-mediated activation of the transcription factor SKN-1 and/or an indirect or direct PMK-1-dependent activation (hyperphosphorylation) of heat-shock factor 1.
Collapse
Affiliation(s)
- Ansgar Mertenskötter
- Institute of Zoophysiology, University of Münster, Schlossplatz 8, 48143, Münster, Germany.
| | | | | | | |
Collapse
|
50
|
Alterations in gene expression in Caenorhabditis elegans associated with organophosphate pesticide intoxication and recovery. BMC Genomics 2013; 14:291. [PMID: 23631360 PMCID: PMC3760450 DOI: 10.1186/1471-2164-14-291] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 04/20/2013] [Indexed: 01/13/2023] Open
Abstract
Background The principal toxicity of acute organophosphate (OP) pesticide poisoning is the disruption of neurotransmission through inhibition of acetylcholinesterase (AChE). However, other mechanisms leading to persistent effects and neurodegeneration remain controversial and difficult to detect. Because Caenorhabditis elegans is relatively resistant to OP lethality—particularly through the inhibition of AChE—studies in this nematode provide an opportunity to observe alterations in global gene expression following OP exposure that cannot be readily observed in less resistant organisms. Results We exposed cultures of worms in axenic, defined medium to dichlorvos under three exposure protocols. In the first, worms were exposed continuously throughout the experiment. In the second and third, the worms were exposed for either 2 or 8 h, the dichlorvos was washed out of the culture, and the worms were allowed to recover. We then analyzed gene expression using whole genome microarrays from RNA obtained from worms sampled at multiple time points throughout the exposure. The worms showed a time-dependent increase in the expression of genes involved in stress responses. Early in the exposure, the predominant effect was on metabolic processes, while at later times, an immune-like response and cellular repair mechanisms dominated the expression pattern. Following removal of dichlorvos, the gene expression in the worms appeared to relatively rapidly return to steady-state levels. Conclusion The changes in gene expression observed in the worms following exposure to dichlorvos point towards two potential mechanisms of toxicity: inhibition of AChE and mitochondrial disruption.
Collapse
|