51
|
Shepherd C, Wangchuk P, Loukas A. Of dogs and hookworms: man's best friend and his parasites as a model for translational biomedical research. Parasit Vectors 2018; 11:59. [PMID: 29370855 PMCID: PMC5785905 DOI: 10.1186/s13071-018-2621-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 01/03/2018] [Indexed: 02/06/2023] Open
Abstract
We present evidence that the dog hookworm (Ancylostoma caninum) is underutilised in the study of host-parasite interactions, particularly as a proxy for the human-hookworm relationship. The inability to passage hookworms through all life stages in vitro means that adult stage hookworms have to be harvested from the gut of their definitive hosts for ex vivo research. This makes study of the human-hookworm interface difficult for technical and ethical reasons. The historical association of humans, dogs and hookworms presents a unique triad of positive evolutionary pressure to drive the A. caninum-canine interaction to reflect that of the human-hookworm relationship. Here we discuss A. caninum as a proxy for human hookworm infection and situate this hookworm model within the current research agenda, including the various 'omics' applications and the search for next generation biologics to treat a plethora of human diseases. Historically, the dog hookworm has been well described on a physiological and biochemical level, with an increasing understanding of its role as a human zoonosis. With its similarity to human hookworm, the recent publications of hookworm genomes and other omics databases, as well as the ready availability of these parasites for ex vivo culture, the dog hookworm presents itself as a valuable tool for discovery and translational research.
Collapse
Affiliation(s)
- Catherine Shepherd
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia.
| | - Phurpa Wangchuk
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, Australia.
| |
Collapse
|
52
|
Offenburger SL, Ho XY, Tachie-Menson T, Coakley S, Hilliard MA, Gartner A. 6-OHDA-induced dopaminergic neurodegeneration in Caenorhabditis elegans is promoted by the engulfment pathway and inhibited by the transthyretin-related protein TTR-33. PLoS Genet 2018; 14:e1007125. [PMID: 29346382 PMCID: PMC5773127 DOI: 10.1371/journal.pgen.1007125] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 11/26/2017] [Indexed: 12/03/2022] Open
Abstract
Oxidative stress is linked to many pathological conditions including the loss of dopaminergic neurons in Parkinson’s disease. The vast majority of disease cases appear to be caused by a combination of genetic mutations and environmental factors. We screened for genes protecting Caenorhabditis elegans dopaminergic neurons from oxidative stress induced by the neurotoxin 6-hydroxydopamine (6-OHDA) and identified the transthyretin-related gene ttr-33. The only described C. elegans transthyretin-related protein to date, TTR-52, has been shown to mediate corpse engulfment as well as axon repair. We demonstrate that TTR-52 and TTR-33 have distinct roles. TTR-33 is likely produced in the posterior arcade cells in the head of C. elegans larvae and is predicted to be a secreted protein. TTR-33 protects C. elegans from oxidative stress induced by paraquat or H2O2 at an organismal level. The increased oxidative stress sensitivity of ttr-33 mutants is alleviated by mutations affecting the KGB-1 MAPK kinase pathway, whereas it is enhanced by mutation of the JNK-1 MAPK kinase. Finally, we provide genetic evidence that the C. elegans cell corpse engulfment pathway is required for the degeneration of dopaminergic neurons after exposure to 6-OHDA. In summary, we describe a new neuroprotective mechanism and demonstrate that TTR-33 normally functions to protect dopaminergic neurons from oxidative stress-induced degeneration, potentially by acting as a secreted sensor or scavenger of oxidative stress. Animals employ multiple mechanisms to prevent their cells from damage by reactive oxygen species, chemically reactive molecules containing oxygen. Oxidative stress, caused by the overabundance of reactive oxygen species or a decreased cellular defence against these chemicals, is linked to a variety of neurodegenerative conditions, including the loss of dopaminergic neurons in Parkinson’s disease. In this study, we discovered a novel protective molecule that functions to prevent dopaminergic neurodegeneration caused by oxidative stress induced by the neurotoxin 6-hydroxydopamine (6-OHDA). We used the nematode C. elegans, a well-characterised model in which mechanisms can be studied on an organismal level. When C. elegans is exposed to 6-OHDA, its dopaminergic neurons gradually die. Our major findings include (i) mutations of the transthyretin-related gene ttr-33 causes highly increased dopaminergic neurodegeneration after 6-OHDA exposure; (ii) TTR-33 is likely produced and secreted by several cells in the head of the animal; (iii) TTR-33 protects against oxidative stress induced by other compounds; (iv) mutations in the KGB-1 MAP kinase stress pathway alleviate dopaminergic neuron loss in the ttr-33 mutant; and (v) the cell corpse engulfment pathway is required for dopaminergic neurodegeneration. We hypothesise that TTR-33 protects dopaminergic neurons against 6-OHDA-induced oxidative stress by acting as an oxygen sensor or scavenger.
Collapse
Affiliation(s)
| | - Xue Yan Ho
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | | | - Sean Coakley
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Massimo A. Hilliard
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Anton Gartner
- School of Life Sciences, University of Dundee, Dundee, United Kingdom
- * E-mail:
| |
Collapse
|
53
|
Doitsidou M, Minevich G, Kroll JR, Soete G, Gowtham S, Korswagen HC, Sebastiaan van Zon J, Hobert O. A Caenorhabditis elegans Zinc Finger Transcription Factor, ztf-6, Required for the Specification of a Dopamine Neuron-Producing Lineage. G3 (BETHESDA, MD.) 2018; 8:17-26. [PMID: 29301976 PMCID: PMC5765345 DOI: 10.1534/g3.117.300132] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/31/2017] [Indexed: 12/12/2022]
Abstract
Invertebrate and vertebrate nervous systems generate different types of dopaminergic neurons in distinct parts of the brain. We have taken a genetic approach to understand how the four functionally related, but lineally unrelated, classes of dopaminergic neurons of the nematode Caenorhabditis elegans, located in distinct parts of its nervous system, are specified. We have identified several genes involved in the generation of a specific dopaminergic neuron type that is generated from the so-called postdeirid lineage, called PDE. Apart from classic proneural genes and components of the mediator complex, we identified a novel, previously uncharacterized zinc finger transcription factor, ztf-6 Loss of ztf-6 has distinct effects in different dopamine neuron-producing neuronal lineages. In the postdeirid lineage, ztf-6 is required for proper cell division patterns and the proper distribution of a critical cell fate determinant, the POP-1/TCF-like transcription factor.
Collapse
Affiliation(s)
- Maria Doitsidou
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center New York, NY 10032
- Centre for Discovery Brain Sciences, University of Edinburgh, EH8 9XD, UK
| | - Gregory Minevich
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center New York, NY 10032
| | - Jason R Kroll
- AMOLF, Amsterdam, 1098 XG, The Netherlands, 3584 CT Utrecht, The Netherlands
| | - Gwen Soete
- Hubrecht Institute, 3584 CT Utrecht, The Netherlands
| | - Sriharsh Gowtham
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center New York, NY 10032
| | | | | | - Oliver Hobert
- Department of Biochemistry and Molecular Biophysics, Columbia University Medical Center New York, NY 10032
- Department of Biological Sciences, Howard Hughes Medical Institute, Columbia University, New York, NY 10027
| |
Collapse
|
54
|
Hsp90-downregulation influences the heat-shock response, innate immune response and onset of oocyte development in nematodes. PLoS One 2017; 12:e0186386. [PMID: 29078207 PMCID: PMC5659845 DOI: 10.1371/journal.pone.0186386] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 09/30/2017] [Indexed: 01/21/2023] Open
Abstract
Hsp90 is a molecular chaperone involved in the regulation and maturation of kinases and transcription factors. In Caenorhabditis elegans, it contributes to the development of fertility, maintenance of muscle structure, the regulation of heat-shock response and dauer state. To understand the consequences of Hsp90-depletion, we studied Hsp90 RNAi-treated nematodes by DNA microarrays and mass spectrometry. We find that upon development of phenotypes the levels of chaperones and Hsp90 cofactors are increased, while specific proteins related to the innate immune response are depleted. In microarrays, we further find many differentially expressed genes related to gonad and larval development. These genes form an expression cluster that is regulated independently from the immune response implying separate pathways of Hsp90-involvement. Using fluorescent reporter strains for the differentially expressed immune response genes skr-5, dod-24 and clec-60 we observe that their activity in intestinal tissues is influenced by Hsp90-depletion. Instead, effects on the development are evident in both gonad arms. After Hsp90-depletion, changes can be observed in early embryos and adults containing fluorescence-tagged versions of SEPA-1, CAV-1 or PUD-1, all of which are downregulated after Hsp90-depletion. Our observations identify molecular events for Hsp90-RNAi induced phenotypes during development and immune responses, which may help to separately investigate independent Hsp90-influenced processes that are relevant during the nematode’s life and development.
Collapse
|
55
|
Hibshman JD, Doan AE, Moore BT, Kaplan RE, Hung A, Webster AK, Bhatt DP, Chitrakar R, Hirschey MD, Baugh LR. daf-16/FoxO promotes gluconeogenesis and trehalose synthesis during starvation to support survival. eLife 2017; 6. [PMID: 29063832 PMCID: PMC5655125 DOI: 10.7554/elife.30057] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2017] [Accepted: 09/25/2017] [Indexed: 12/21/2022] Open
Abstract
daf-16/FoxO is required to survive starvation in Caenorhabditis elegans, but how daf-16IFoxO promotes starvation resistance is unclear. We show that daf-16/FoxO restructures carbohydrate metabolism by driving carbon flux through the glyoxylate shunt and gluconeogenesis and into synthesis of trehalose, a disaccharide of glucose. Trehalose is a well-known stress protectant, capable of preserving membrane organization and protein structure during abiotic stress. Metabolomic, genetic, and pharmacological analyses confirm increased trehalose synthesis and further show that trehalose not only supports survival as a stress protectant but also serves as a glycolytic input. Furthermore, we provide evidence that metabolic cycling between trehalose and glucose is necessary for this dual function of trehalose. This work demonstrates that daf-16/FoxO promotes starvation resistance by shifting carbon metabolism to drive trehalose synthesis, which in turn supports survival by providing an energy source and acting as a stress protectant. Most animals rarely have access to a constant supply of food, and so have evolved ways to cope with times of plenty and times of shortage. Insulin is a hormone that travels throughout the body to signal when an animal is well fed. Insulin signaling inhibits the activity of a protein called FoxO, which otherwise switches on and off hundreds of genes to control the starvation response. The roundworm, Caenorhabditis elegans, has been well studied in the laboratory, and often has to cope with starvation in the wild. These worms can pause their development if no food is available, or divert to a different developmental path if they anticipate that food will be short in future. As with more complex animals, the worm responds to starvation by reducing insulin-like signaling, which in turn activates a FoxO protein called daf-16. When the worms stop feeding, daf-16 is switched on, which is crucial for survival. It was known how daf-16 stops the roundworm’s development, but it was not known how it helps the worms to survive starvation. Now, Hibshman et al. have compared normal roundworm larvae to larvae that are missing the gene for daf-16 to determine how this protein influences the roundworm’s ability to survive starvation. The worms were examined with and without food, to look for which genes were switched on and off by daf-16 during starvation. This revealed that daf-16 controls metabolism, activating a metabolic shortcut that makes the worms produce glucose and begin turning it into another type of sugar, called trehalose. This sugar usually promotes survival in conditions where water is limiting, like dehydration and high salt, but it can also be broken down to release energy. The levels of trehalose in the worms rose within hours of the onset of starvation. To confirm the importance of trehalose in surviving starvation, roundworms with mutations in genes involved in glucose or trehalose production were examined, as was the effect of giving starving worms glucose or trehalose. Disrupting the production of sugars caused the worms to die sooner of starvation, while supplementing with sugar had the opposite effect meaning the worms survived for longer. Taken together, these findings reveal that daf-16 protects against starvation by shifting metabolism towards the production of trehalose. This helps worms to survive by both protecting them from stress and providing them with a source of energy. These findings not only extend the current understanding of how animals respond to starvation, but could also lead to improved understanding of diseases where this response goes wrong, including diabetes and obesity.
Collapse
Affiliation(s)
- Jonathan D Hibshman
- Department of Biology, Duke University, Durham, United States.,University Program in Genetics and Genomics, Duke University, Durham, United States
| | | | - Brad T Moore
- Department of Biology, Duke University, Durham, United States
| | - Rebecca Ew Kaplan
- Department of Biology, Duke University, Durham, United States.,University Program in Genetics and Genomics, Duke University, Durham, United States
| | - Anthony Hung
- Department of Biology, Duke University, Durham, United States
| | - Amy K Webster
- Department of Biology, Duke University, Durham, United States.,University Program in Genetics and Genomics, Duke University, Durham, United States
| | - Dhaval P Bhatt
- Duke Molecular Physiology Institute, Duke University, Durham, United States
| | - Rojin Chitrakar
- Department of Biology, Duke University, Durham, United States
| | - Matthew D Hirschey
- Duke Molecular Physiology Institute, Duke University, Durham, United States.,Department of Medicine, Duke University, Durham, United States.,Department of Pharmacology & Cancer Biology, Duke University, Durham, United States
| | - L Ryan Baugh
- Department of Biology, Duke University, Durham, United States.,University Program in Genetics and Genomics, Duke University, Durham, United States
| |
Collapse
|
56
|
Rapti G, Li C, Shan A, Lu Y, Shaham S. Glia initiate brain assembly through noncanonical Chimaerin-Furin axon guidance in C. elegans. Nat Neurosci 2017; 20:1350-1360. [PMID: 28846083 PMCID: PMC5614858 DOI: 10.1038/nn.4630] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 07/19/2017] [Indexed: 01/08/2023]
Abstract
Brain assembly is hypothesized to begin when pioneer axons extend over non-neuronal cells, forming tracts guiding follower axons. Yet pioneer-neuron identities, their guidance substrates, and their interactions are not well understood. Here, using time-lapse embryonic imaging, genetics, protein-interaction, and functional studies, we uncover the early events of C. elegans brain assembly. We demonstrate that C. elegans glia are key for assembly initiation, guiding pioneer and follower axons using distinct signals. Pioneer sublateral neurons, with unique growth properties, anatomy, and innervation, cooperate with glia to mediate follower-axon guidance. We further identify a Chimaerin (CHIN-1)- Furin (KPC-1) double-mutant that severely disrupts assembly. CHIN-1 and KPC-1 function noncanonically, in glia and pioneer neurons, for guidance-cue trafficking. We exploit this bottleneck to define roles for glial Netrin and Semaphorin in pioneer- and follower-axon guidance, respectively, and for glial and pioneer-neuron Flamingo (CELSR) in follower-axon navigation. Taken together, our studies reveal previously undescribed glial roles in pioneer-axon guidance, suggesting conserved principles of brain assembly.
Collapse
Affiliation(s)
- Georgia Rapti
- Laboratory of Developmental Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Chang Li
- Laboratory of Developmental Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
- These authors contributed equally to this work
| | - Alan Shan
- Laboratory of Developmental Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
- These authors contributed equally to this work
| | - Yun Lu
- Laboratory of Developmental Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| | - Shai Shaham
- Laboratory of Developmental Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065 USA
| |
Collapse
|
57
|
Dawson JA, Methven-Kelley C, Davis GM. atz-1 Influences meiosis to maintain germline chromosomal stability in Caenorhabditis elegans. Cell Biol Int 2017; 41:1160-1168. [PMID: 28696027 DOI: 10.1002/cbin.10821] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/07/2017] [Indexed: 11/07/2022]
Abstract
Exchange of genetic information during meiosis occurs in all sexually reproducing species to produce haploid gametes from diploid cells. This process involves tight coordination of a meiotic specific cohesin complex, the synaptonemal complex, and DNA damage repair mechanisms. In this study, we describe a putative myosin heavy chain protein orthologous to human myosin 1, F28D1.2, which we named Abnormal Transition Zone (atz-1). Deletion of atz-1 results in embryonic lethality and a depleted transition zone, accompanied by reduced expression of the meiotic cohesin protein, REC-8. atz-1 mutants display disorganized and aggregated chromosomal bodies in diakinetic oocytes. In addition to this, atz-1 mutants are hypersensitive to mild inhibition of DNA damage repair, suggesting that DNA replication in atz-1 mutants is impaired. Moreover, the atz-1 mutant phenotype is germline specific and resupplying somatically expressed atz-1 does not rescue the reproductive defects associated with atz-1 mutants. Overall, our data suggest that atz-1 contributes to meiosis and maintains germline chromosomal stability.
Collapse
Affiliation(s)
- Joseph A Dawson
- School of Applied and Biomedical Sciences, Federation University, Churchill, Australia
| | | | - Gregory M Davis
- School of Applied and Biomedical Sciences, Federation University, Churchill, Australia
| |
Collapse
|
58
|
Live-cell confocal microscopy and quantitative 4D image analysis of anchor-cell invasion through the basement membrane in Caenorhabditis elegans. Nat Protoc 2017; 12:2081-2096. [PMID: 28880279 DOI: 10.1038/nprot.2017.093] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Cell invasion through basement membrane (BM) barriers is crucial in development, leukocyte trafficking and the spread of cancer. The mechanisms that direct invasion, despite their importance in normal and disease states, are poorly understood, largely because of the inability to visualize dynamic cell-BM interactions in vivo. This protocol describes multichannel time-lapse confocal imaging of anchor-cell invasion in live Caenorhabditis elegans. Methods presented include outline-slide preparation and worm growth synchronization (15 min), mounting (20 min), image acquisition (20-180 min), image processing (20 min) and quantitative analysis (variable timing). The acquired images enable direct measurement of invasive dynamics including formation of invadopodia and cell-membrane protrusions, and removal of BM. This protocol can be combined with genetic analysis, molecular-activity probes and optogenetic approaches to uncover the molecular mechanisms underlying cell invasion. These methods can also be readily adapted by any worm laboratory for real-time analysis of cell migration, BM turnover and cell-membrane dynamics.
Collapse
|
59
|
Flavin monooxygenases regulate Caenorhabditis elegans axon guidance and growth cone protrusion with UNC-6/Netrin signaling and Rac GTPases. PLoS Genet 2017; 13:e1006998. [PMID: 28859089 PMCID: PMC5597259 DOI: 10.1371/journal.pgen.1006998] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 09/13/2017] [Accepted: 08/25/2017] [Indexed: 01/05/2023] Open
Abstract
The guidance cue UNC-6/Netrin regulates both attractive and repulsive axon guidance. Our previous work showed that in C. elegans, the attractive UNC-6/Netrin receptor UNC-40/DCC stimulates growth cone protrusion, and that the repulsive receptor, an UNC-5:UNC-40 heterodimer, inhibits growth cone protrusion. We have also shown that inhibition of growth cone protrusion downstream of the UNC-5:UNC-40 repulsive receptor involves Rac GTPases, the Rac GTP exchange factor UNC-73/Trio, and the cytoskeletal regulator UNC-33/CRMP, which mediates Semaphorin-induced growth cone collapse in other systems. The multidomain flavoprotein monooxygenase (FMO) MICAL (Molecule Interacting with CasL) also mediates growth cone collapse in response to Semaphorin by directly oxidizing F-actin, resulting in depolymerization. The C. elegans genome does not encode a multidomain MICAL-like molecule, but does encode five flavin monooxygenases (FMO-1, -2, -3, -4, and 5) and another molecule, EHBP-1, similar to the non-FMO portion of MICAL. Here we show that FMO-1, FMO-4, FMO-5, and EHBP-1 may play a role in UNC-6/Netrin directed repulsive guidance mediated through UNC-40 and UNC-5 receptors. Mutations in fmo-1, fmo-4, fmo-5, and ehbp-1 showed VD/DD axon guidance and branching defects, and variably enhanced unc-40 and unc-5 VD/DD axon guidance defects. Developing growth cones in vivo of fmo-1, fmo-4, fmo-5, and ehbp-1 mutants displayed excessive filopodial protrusion, and transgenic expression of FMO-5 inhibited growth cone protrusion. Mutations suppressed growth cone inhibition caused by activated UNC-40 and UNC-5 signaling, and activated Rac GTPase CED-10 and MIG-2, suggesting that these molecules are required downstream of UNC-6/Netrin receptors and Rac GTPases. From these studies we conclude that FMO-1, FMO-4, FMO-5, and EHBP-1 represent new players downstream of UNC-6/Netrin receptors and Rac GTPases that inhibit growth cone filopodial protrusion in repulsive axon guidance. Mechanisms that guide axons to their targets in the developing nervous system have been elucidated, but how these pathways affect behavior of the growth cone of the axon during outgrowth remains poorly understood. We previously showed that the guidance cue UNC-6/Netrin and its receptors UNC-40/DCC and UNC-5 inhibit lamellipodial and filopodial growth cone protrusion to mediate repulsion from UNC-6/Netrin in C. elegans. Here we report a new mechanism downstream of UNC-6/Netrin involving flavin monooxygenase redox enzymes (FMOs). We show that FMOs are normally required for axon guidance and to inhibit growth cone protrusion. Furthermore, we show that they are required for the anti-protrusive effects of activated UNC-40 and UNC-5 receptors, and that they can partially compensate for loss of molecules in the pathway, indicating that they act downstream of UNC-6/Netrin signaling. Based on the function of the FMO-containing MICAL molecules in Drosophila and vertebrates, we speculate that the FMOs might directly oxidize actin, leading to filament disassembly and collapse, and/or lead to the phosphorylation of UNC-33/CRMP, which we show also genetically interacts with the FMOs downstream of UNC-6/Netrin. In conclusion, this is the first evidence that FMOs might act downstream of UNC-6/Netrin signaling in growth cone protrusion and axon repulsion.
Collapse
|
60
|
IGDB-2, an Ig/FNIII protein, binds the ion channel LGC-34 and controls sensory compartment morphogenesis in C. elegans. Dev Biol 2017; 430:105-112. [PMID: 28803967 DOI: 10.1016/j.ydbio.2017.08.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/04/2017] [Accepted: 08/07/2017] [Indexed: 11/21/2022]
Abstract
Sensory organ glia surround neuronal receptive endings (NREs), forming a specialized compartment important for neuronal activity, and reminiscent of glia-ensheathed synapses in the central nervous system. We previously showed that DAF-6, a Patched-related protein, is required in glia of the C. elegans amphid sensory organ to restrict sensory compartment size. LIT-1, a Nemo-like kinase, and SNX-1, a retromer component, antagonize DAF-6 and promote compartment expansion. To further explore the machinery underlying compartment size control, we sought genes whose inactivation restores normal compartment size to daf-6 mutants. We found that mutations in igdb-2, encoding a single-pass transmembrane protein containing Ig-like and fibronectin type III domains, suppress daf-6 mutant defects. IGDB-2 acts in glia, where it localizes to glial membranes surrounding NREs, and, together with LIT-1 and SNX-1, regulates compartment morphogenesis. Immunoprecipitation followed by mass spectrometry demonstrates that IGDB-2 binds to LGC-34, a predicted ligand-gated ion channel, and lgc-34 mutations inhibit igdb-2 suppression of daf-6. Our findings reveal a novel membrane protein complex and suggest possible mechanisms for how sensory compartment size is controlled.
Collapse
|
61
|
Hoang HD, Miller MA. Chemosensory and hyperoxia circuits in C. elegans males influence sperm navigational capacity. PLoS Biol 2017; 15:e2002047. [PMID: 28662030 PMCID: PMC5490939 DOI: 10.1371/journal.pbio.2002047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 05/25/2017] [Indexed: 11/23/2022] Open
Abstract
The sperm’s crucial function is to locate and fuse with a mature oocyte. Under laboratory conditions, Caenorhabditis elegans sperm are very efficient at navigating the hermaphrodite reproductive tract and locating oocytes. Here, we identify chemosensory and oxygen-sensing circuits that affect the sperm’s navigational capacity. Multiple Serpentine Receptor B (SRB) chemosensory receptors regulate Gα pathways in gustatory sensory neurons that extend cilia through the male nose. SRB signaling is necessary and sufficient in these sensory neurons to influence sperm motility parameters. The neuropeptide Y pathway acts together with SRB-13 to antagonize negative effects of the GCY-35 hyperoxia sensor on spermatogenesis. SRB chemoreceptors are not essential for sperm navigation under low oxygen conditions that C. elegans prefers. In ambient oxygen environments, SRB-13 signaling impacts gene expression during spermatogenesis and the sperm’s mitochondria, thereby increasing migration velocity and inhibiting reversals within the hermaphrodite uterus. The SRB-13 transcriptome is highly enriched in genes implicated in pathogen defense, many of which are expressed in diverse tissues. We show that the critical time period for SRB-13 signaling is prior to spermatocyte differentiation. Our results support the model that young C. elegans males sense external environment and oxygen tension, triggering long-lasting downstream signaling events with effects on the sperm’s mitochondria and navigational capacity. Environmental exposures early in male life may alter sperm function and fertility. Habitat loss, disease, climate change, and pollution are thought to negatively affect animal fertility. Sperm are a potential target, but the molecular mechanisms are not understood. The nematode C. elegans is a powerful genetic model to investigate the relationship between environment and male fertility. The hermaphrodite’s transparent epidermis permits the direct visualization of migrating male sperm and fertilization. In this study, we identified multiple serpentine receptor B (SRB) chemosensory receptors that are expressed in amphid sensory neurons, which extend cilia through the male nose. These SRB chemoreceptors are necessary to produce sperm that are efficient at navigating the hermaphrodite reproductive tract to the fertilization site. We show that SRB-13 signaling counteracts the negative effect of GCY-35 O2 sensor activity, thereby maintaining sperm mitochondrial function and navigational capacity in hyperoxic conditions. Of particular interest, SRB-13 acts in early larval stage males prior to testis maturation. We propose that young males respond to specific stressful environments by altering SRB neural circuits, which in turn impact sperm mitochondrial function and motility. This chemosensory mechanism may be part of a systemic response in C. elegans males to external environment and oxygen levels.
Collapse
Affiliation(s)
- Hieu D. Hoang
- Department of Cell, Developmental and Integrative Biology, University of Alabama School of Medicine, Birmingham, Alabama, United States of America
| | - Michael A. Miller
- Department of Cell, Developmental and Integrative Biology, University of Alabama School of Medicine, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
62
|
Abstract
Many stem cell niches contain support cells that increase contact with stem cells by enwrapping them in cellular processes. One example is the germ stem cell niche in C. elegans, which is composed of a single niche cell termed the distal tip cell (DTC) that extends cellular processes, constructing an elaborate plexus that enwraps germ stem cells. To identify genes required for plexus formation and to explore the function of this specialized enwrapping behavior, a series of targeted and tissue-specific RNAi screens were performed. Here we identify genes that promote stem cell enwrapment by the DTC plexus, including a set that specifically functions within the DTC, such as the chromatin modifier lin-40/MTA1, and others that act within the germline, such as the 14-3-3 signaling protein par-5. Analysis of genes that function within the germline to mediate plexus development reveal that they are required for expansion of the germ progenitor zone, supporting the emerging idea that germ stem cells signal to the niche to stimulate enwrapping behavior. Examination of wild-type animals with asymmetric plexus formation and animals with reduced DTC plexus elaboration via loss of two candidates including lin-40 indicate that cellular enwrapment promotes GLP-1/Notch signaling and germ stem cell fate. Together, our work identifies novel regulators of cellular enwrapment and suggests that reciprocal signaling between the DTC niche and the germ stem cells promotes enwrapment behavior and stem cell fate.
Collapse
|
63
|
Igual Gil C, Jarius M, von Kries JP, Rohlfing AK. Neuronal Chemosensation and Osmotic Stress Response Converge in the Regulation of aqp-8 in C. elegans. Front Physiol 2017. [PMID: 28649202 PMCID: PMC5465262 DOI: 10.3389/fphys.2017.00380] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Aquaporins occupy an essential role in sustaining the salt/water balance in various cells types and tissues. Here, we present new insights into aqp-8 expression and regulation in Caenorhabditis elegans. We show, that upon exposure to osmotic stress, aqp-8 exhibits a distinct expression pattern within the excretory cell compared to other C. elegans aquaporins expressed. This expression is correlated to the osmolarity of the surrounding medium and can be activated physiologically by osmotic stress or genetically in mutants with constitutively active osmotic stress response. In addition, we found aqp-8 expression to be constitutively active in the TRPV channel mutant osm-9(ok1677). In a genome-wide RNAi screen we identified additional regulators of aqp-8. Many of these regulators are connected to chemosensation by the amphid neurons, e.g., odr-10 and gpa-6, and act as suppressors of aqp-8 expression. We postulate from our results, that aqp-8 plays an important role in sustaining the salt/water balance during a secondary response to hyper-osmotic stress. Upon its activation aqp-8 promotes vesicle docking to the lumen of the excretory cell and thereby enhances the ability to secrete water and transport osmotic active substances or waste products caused by protein damage. In summary, aqp-8 expression and function is tightly regulated by a network consisting of the osmotic stress response, neuronal chemosensation as well as the response to protein damage. These new insights in maintaining the salt/water balance in C. elegans will help to reveal the complex homeostasis network preserved throughout species.
Collapse
Affiliation(s)
- Carla Igual Gil
- Zoophysiology, Institute for Biochemistry and Biology, University PotsdamPotsdam, Germany
| | - Mirko Jarius
- Zoophysiology, Institute for Biochemistry and Biology, University PotsdamPotsdam, Germany
| | - Jens P von Kries
- Leibniz-Institut für Molekulare Pharmakologie (FMP)Berlin, Germany
| | - Anne-Katrin Rohlfing
- Zoophysiology, Institute for Biochemistry and Biology, University PotsdamPotsdam, Germany
| |
Collapse
|
64
|
Evolutionarily conserved TRH neuropeptide pathway regulates growth in Caenorhabditis elegans. Proc Natl Acad Sci U S A 2017; 114:E4065-E4074. [PMID: 28461507 DOI: 10.1073/pnas.1617392114] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
In vertebrates thyrotropin-releasing hormone (TRH) is a highly conserved neuropeptide that exerts the hormonal control of thyroid-stimulating hormone (TSH) levels as well as neuromodulatory functions. However, a functional equivalent in protostomian animals remains unknown, although TRH receptors are conserved in proto- and deuterostomians. Here we identify a TRH-like neuropeptide precursor in Caenorhabditis elegans that belongs to a bilaterian family of TRH precursors. Using CRISPR/Cas9 and RNAi reverse genetics, we show that TRH-like neuropeptides, through the activation of their receptor TRHR-1, promote growth in Celegans TRH-like peptides from pharyngeal motor neurons are required for normal body size, and knockdown of their receptor in pharyngeal muscle cells reduces growth. Mutants deficient for TRH signaling have no defects in pharyngeal pumping or isthmus peristalsis rates, but their growth defect depends on the bacterial diet. In addition to the decrease in growth, trh-1 mutants have a reduced number of offspring. Our study suggests that TRH is an evolutionarily ancient neuropeptide, having its origin before the divergence of protostomes and deuterostomes, and may ancestrally have been involved in the control of postembryonic growth and reproduction.
Collapse
|
65
|
Laranjeiro R, Harinath G, Burke D, Braeckman BP, Driscoll M. Single swim sessions in C. elegans induce key features of mammalian exercise. BMC Biol 2017; 15:30. [PMID: 28395669 PMCID: PMC5385602 DOI: 10.1186/s12915-017-0368-4] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 03/15/2017] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Exercise exerts remarkably powerful effects on metabolism and health, with anti-disease and anti-aging outcomes. Pharmacological manipulation of exercise benefit circuits might improve the health of the sedentary and the aging populations. Still, how exercised muscle signals to induce system-wide health improvement remains poorly understood. With a long-term interest in interventions that promote animal-wide health improvement, we sought to define exercise options for Caenorhabditis elegans. RESULTS Here, we report on the impact of single swim sessions on C. elegans physiology. We used microcalorimetry to show that C. elegans swimming has a greater energy cost than crawling. Animals that swam continuously for 90 min specifically consumed muscle fat supplies and exhibited post-swim locomotory fatigue, with both muscle fat depletion and fatigue indicators recovering within 1 hour of exercise cessation. Quantitative polymerase chain reaction (qPCR) transcript analyses also suggested an increase in fat metabolism during the swim, followed by the downregulation of specific carbohydrate metabolism transcripts in the hours post-exercise. During a 90 min swim, muscle mitochondria matrix environments became more oxidized, as visualized by a localized mitochondrial reduction-oxidation-sensitive green fluorescent protein reporter. qPCR data supported specific transcriptional changes in oxidative stress defense genes during and immediately after a swim. Consistent with potential antioxidant defense induction, we found that a single swim session sufficed to confer protection against juglone-induced oxidative stress inflicted 4 hours post-exercise. CONCLUSIONS In addition to showing that even a single swim exercise bout confers physiological changes that increase robustness, our data reveal that acute swimming-induced changes share common features with some acute exercise responses reported in humans. Overall, our data validate an easily implemented swim experience as C. elegans exercise, setting the foundation for exploiting the experimental advantages of this model to genetically or pharmacologically identify the exercise-associated molecules and signaling pathways that confer system-wide health benefits.
Collapse
Affiliation(s)
- Ricardo Laranjeiro
- Department of Molecular Biology and Biochemistry, Nelson Biological Laboratories, Rutgers, The State University of New Jersey, Piscataway, NJ USA
| | - Girish Harinath
- Department of Molecular Biology and Biochemistry, Nelson Biological Laboratories, Rutgers, The State University of New Jersey, Piscataway, NJ USA
| | - Daniel Burke
- Department of Molecular Biology and Biochemistry, Nelson Biological Laboratories, Rutgers, The State University of New Jersey, Piscataway, NJ USA
| | | | - Monica Driscoll
- Department of Molecular Biology and Biochemistry, Nelson Biological Laboratories, Rutgers, The State University of New Jersey, Piscataway, NJ USA
| |
Collapse
|
66
|
Locomotion Behavior Is Affected by the Gα S Pathway and the Two-Pore-Domain K + Channel TWK-7 Interacting in GABAergic Motor Neurons in Caenorhabditis elegans. Genetics 2017; 206:283-297. [PMID: 28341653 DOI: 10.1534/genetics.116.195669] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 03/19/2017] [Indexed: 01/03/2023] Open
Abstract
Adjusting the efficiency of movement in response to environmental cues is an essential integrative characteristic of adaptive locomotion behavior across species. However, the modulatory molecules and the pathways involved are largely unknown. Recently, we demonstrated that in Caenorhabditis elegans, a loss-of-function of the two-pore-domain potassium (K2P) channel TWK-7 causes a fast, coordinated, and persistent forward crawling behavior in which five central aspects of stimulated locomotion-velocity, direction, wave parameters, duration, and straightness-are affected. Here, we isolated the reduction-of-function allele cau1 of the C. elegans gene kin-2 in a forward genetic screen and showed that it phenocopies the locomotor activity and locomotion behavior of twk-7(null) animals. Kin-2 encodes the negative regulatory subunit of protein kinase A (KIN-1/PKA). Consistently, we found that other gain-of-function mutants of the GαS-KIN-1/PKA pathway resemble kin-2(cau1) and twk-7(null) in locomotion phenotype. Using the powerful genetics of the C. elegans system in combination with cell type-specific approaches and detailed locomotion analyses, we identified TWK-7 as a putative downstream target of the GαS-KIN-1/PKA pathway at the level of the γ-aminobutyric acid (GABA)ergic D-type motor neurons. Due to this epistatic interaction, we suggest that KIN-1/PKA and TWK-7 may share a common pathway that is probably involved in the modulation of both locomotor activity and locomotion behavior during forward crawling.
Collapse
|
67
|
Lok JB, Shao H, Massey HC, Li X. Transgenesis in Strongyloides and related parasitic nematodes: historical perspectives, current functional genomic applications and progress towards gene disruption and editing. Parasitology 2017; 144:327-342. [PMID: 27000743 PMCID: PMC5364836 DOI: 10.1017/s0031182016000391] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 02/10/2016] [Accepted: 02/10/2016] [Indexed: 12/20/2022]
Abstract
Transgenesis for Strongyloides and Parastrongyloides was accomplished in 2006 and is based on techniques derived for Caenorhabditis elegans over two decades earlier. Adaptation of these techniques has been possible because Strongyloides and related parasite genera carry out at least one generation of free-living development, with adult males and females residing in soil contaminated by feces from an infected host. Transgenesis in this group of parasites is accomplished by microinjecting DNA constructs into the syncytia of the distal gonads of free-living females. In Strongyloides stercoralis, plasmid-encoded transgenes are expressed in promoter-regulated fashion in the F1 generation following gene transfer but are silenced subsequently. Stable inheritance and expression of transgenes in S. stercoralis requires their integration into the genome, and stable lines have been derived from integrants created using the piggyBac transposon system. More direct investigations of gene function involving expression of mutant transgene constructs designed to alter intracellular trafficking and developmental regulation have shed light on the function of the insulin-regulated transcription factor Ss-DAF-16. Transgenesis in Strongyloides and Parastrongyloides opens the possibility of powerful new methods for genome editing and transcriptional manipulation in this group of parasites. Proof of principle for one of these, CRISPR/Cas9, is presented in this review.
Collapse
Affiliation(s)
- J B Lok
- Department of Pathobiology,School of Veterinary Medicine,University of Pennsylvania,3800 Spruce Street,Philadelphia,PA 19104,USA
| | - H Shao
- Department of Pathobiology,School of Veterinary Medicine,University of Pennsylvania,3800 Spruce Street,Philadelphia,PA 19104,USA
| | - H C Massey
- Department of Pathobiology,School of Veterinary Medicine,University of Pennsylvania,3800 Spruce Street,Philadelphia,PA 19104,USA
| | - X Li
- Department of Pathobiology,School of Veterinary Medicine,University of Pennsylvania,3800 Spruce Street,Philadelphia,PA 19104,USA
| |
Collapse
|
68
|
Miranda-Vizuete A, Veal EA. Caenorhabditis elegans as a model for understanding ROS function in physiology and disease. Redox Biol 2016; 11:708-714. [PMID: 28193593 PMCID: PMC5304259 DOI: 10.1016/j.redox.2016.12.020] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 12/19/2016] [Indexed: 01/05/2023] Open
Abstract
ROS (reactive oxygen species) are potentially damaging by-products of aerobic metabolism which, unchecked, can have detrimental effects on cell function. However, it is now widely accepted that, at physiological levels, certain ROS play important roles in cell signaling, acting as second messengers to regulate cell choices that contribute to the development, adaptation and survival of plants and animals. Despite important recent advances in the biochemical tools available to study redox-signaling, the molecular mechanisms underlying most of these responses remain poorly understood, particularly in multicellular organisms. As we will review here, C. elegans has emerged as a powerful animal model to elucidate these and other aspects of redox biology.
Collapse
Affiliation(s)
- Antonio Miranda-Vizuete
- Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Sevilla, Spain.
| | - Elizabeth A Veal
- Institute for Cell and Molecular Biosciences, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK; Institute for Ageing, Newcastle University, Framlington Place, Newcastle upon Tyne NE2 4HH, UK.
| |
Collapse
|
69
|
Whole-Organism Developmental Expression Profiling Identifies RAB-28 as a Novel Ciliary GTPase Associated with the BBSome and Intraflagellar Transport. PLoS Genet 2016; 12:e1006469. [PMID: 27930654 PMCID: PMC5145144 DOI: 10.1371/journal.pgen.1006469] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 11/08/2016] [Indexed: 01/18/2023] Open
Abstract
Primary cilia are specialised sensory and developmental signalling devices extending from the surface of most eukaryotic cells. Defects in these organelles cause inherited human disorders (ciliopathies) such as retinitis pigmentosa and Bardet-Biedl syndrome (BBS), frequently affecting many physiological and developmental processes across multiple organs. Cilium formation, maintenance and function depend on intracellular transport systems such as intraflagellar transport (IFT), which is driven by kinesin-2 and IFT-dynein motors and regulated by the Bardet-Biedl syndrome (BBS) cargo-adaptor protein complex, or BBSome. To identify new cilium-associated genes, we employed the nematode C. elegans, where ciliogenesis occurs within a short timespan during late embryogenesis when most sensory neurons differentiate. Using whole-organism RNA-Seq libraries, we discovered a signature expression profile highly enriched for transcripts of known ciliary proteins, including FAM-161 (FAM161A orthologue), CCDC-104 (CCDC104), and RPI-1 (RP1/RP1L1), which we confirm are cilium-localised in worms. From a list of 185 candidate ciliary genes, we uncover orthologues of human MAP9, YAP, CCDC149, and RAB28 as conserved cilium-associated components. Further analyses of C. elegans RAB-28, recently associated with autosomal-recessive cone-rod dystrophy, reveal that this small GTPase is exclusively expressed in ciliated neurons where it dynamically associates with IFT trains. Whereas inactive GDP-bound RAB-28 displays no IFT movement and diffuse localisation, GTP-bound (activated) RAB-28 concentrates at the periciliary membrane in a BBSome-dependent manner and undergoes bidirectional IFT. Functional analyses reveal that whilst cilium structure, sensory function and IFT are seemingly normal in a rab-28 null allele, overexpression of predicted GDP or GTP locked variants of RAB-28 perturbs cilium and sensory pore morphogenesis and function. Collectively, our findings present a new approach for identifying ciliary proteins, and unveil RAB28, a GTPase most closely related to the BBS protein RABL4/IFT27, as an IFT-associated cargo with BBSome-dependent cell autonomous and non-autonomous functions at the ciliary base. Ciliopathies are genetic disorders that arise from loss or mutation of genes that encode proteins which play roles in the biology of cilia, organelles found on most of the cells in the human body. Ciliopathy-associated ailments include–but are not limited to–kidney dysfunction, blindness, skeletal abnormalities, as well as brain disorders. Although a great number of cilium-targeted proteins are known, it is thought that a large proportion remain unidentified. Here, we use a developmental gene expression series to discover novel cilia genes in the nematode Caenorhabditis elegans. We present several cilium-localised proteins resulting from our analysis, including RAB-28, a GTPase previously implicated in the degenerative eye disease known as cone-rod dystrophy. Through live videomicroscopy, we show that RAB-28 undergoes bidirectional transport within the cilium. A RAB-28 inactivating mutation results in loss of transport, while an activating mutation results in stronger localisation at the ciliary base and robust transport, although overexpression results in a variety of cilia-related defects. Both the wild type and activating mutant proteins require the Bardet-Biedl Syndrome-related complex of proteins for their transport, linking RAB-28 to an established ciliary transport machinery.
Collapse
|
70
|
Noormohammadi A, Khodakarami A, Gutierrez-Garcia R, Lee HJ, Koyuncu S, König T, Schindler C, Saez I, Fatima A, Dieterich C, Vilchez D. Somatic increase of CCT8 mimics proteostasis of human pluripotent stem cells and extends C. elegans lifespan. Nat Commun 2016; 7:13649. [PMID: 27892468 PMCID: PMC5133698 DOI: 10.1038/ncomms13649] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 10/19/2016] [Indexed: 12/14/2022] Open
Abstract
Human embryonic stem cells can replicate indefinitely while maintaining their undifferentiated state and, therefore, are immortal in culture. This capacity may demand avoidance of any imbalance in protein homeostasis (proteostasis) that would otherwise compromise stem cell identity. Here we show that human pluripotent stem cells exhibit enhanced assembly of the TRiC/CCT complex, a chaperonin that facilitates the folding of 10% of the proteome. We find that ectopic expression of a single subunit (CCT8) is sufficient to increase TRiC/CCT assembly. Moreover, increased TRiC/CCT complex is required to avoid aggregation of mutant Huntingtin protein. We further show that increased expression of CCT8 in somatic tissues extends Caenorhabditis elegans lifespan in a TRiC/CCT-dependent manner. Ectopic expression of CCT8 also ameliorates the age-associated demise of proteostasis and corrects proteostatic deficiencies in worm models of Huntington's disease. Our results suggest proteostasis is a common principle that links organismal longevity with hESC immortality.
Collapse
Affiliation(s)
- Alireza Noormohammadi
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, Cologne 50931, Germany
| | - Amirabbas Khodakarami
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, Cologne 50931, Germany
| | - Ricardo Gutierrez-Garcia
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, Cologne 50931, Germany
| | - Hyun Ju Lee
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, Cologne 50931, Germany
| | - Seda Koyuncu
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, Cologne 50931, Germany
| | - Tim König
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, Cologne 50931, Germany
| | - Christina Schindler
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, Cologne 50931, Germany
| | - Isabel Saez
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, Cologne 50931, Germany
| | - Azra Fatima
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, Cologne 50931, Germany
| | - Christoph Dieterich
- Section of Bioinformatics and Systems Cardiology, Department of Internal Medicine III and Klaus Tschira Institute for Computational Cardiology, Neuenheimer Feld 669, University Hospital, Heidelberg 69120, Germany
| | - David Vilchez
- Cologne Excellence Cluster for Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Joseph Stelzmann Strasse 26, Cologne 50931, Germany
| |
Collapse
|
71
|
A novel approach using C. elegans DNA damage-induced apoptosis to characterize the dynamics of uptake transporters for therapeutic drug discoveries. Sci Rep 2016; 6:36026. [PMID: 27786254 PMCID: PMC5081529 DOI: 10.1038/srep36026] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 10/10/2016] [Indexed: 11/08/2022] Open
Abstract
Organic cation transporter (OCT) function is critical for cellular homeostasis. C. elegans lacking OCT-1 displays a shortened lifespan and increased susceptibility to oxidative stress. We show that these phenotypes can be rescued by downregulating the OCT-1 paralogue, OCT-2. Herein, we delineate a biochemical pathway in C. elegans where uptake of genotoxic chemotherapeutics such as doxorubicin and cisplatin, and subsequent DNA damage-induced apoptosis of germ cells, are dependent exclusively upon OCT-2. We characterized OCT-2 as the main uptake transporter for doxorubicin, as well as a number of other therapeutic agents and chemical compounds, some identified through ligand-protein docking analyses. We provide insights into the conserved features of the structure and function and gene regulation of oct-1 and oct-2 in distinct tissues of C. elegans. Importantly, our innovative approach of exploiting C. elegans uptake transporters in combination with defective DNA repair pathways will have broad applications in medicinal chemistry.
Collapse
|
72
|
Caenorhabditis elegans AGXT-1 is a mitochondrial and temperature-adapted ortholog of peroxisomal human AGT1: New insights into between-species divergence in glyoxylate metabolism. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1864:1195-1205. [PMID: 27179589 DOI: 10.1016/j.bbapap.2016.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 04/27/2016] [Accepted: 05/10/2016] [Indexed: 11/23/2022]
|
73
|
Tomioka M, Naito Y, Kuroyanagi H, Iino Y. Splicing factors control C. elegans behavioural learning in a single neuron by producing DAF-2c receptor. Nat Commun 2016; 7:11645. [PMID: 27198602 PMCID: PMC4876481 DOI: 10.1038/ncomms11645] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 04/17/2016] [Indexed: 01/18/2023] Open
Abstract
Alternative splicing generates protein diversity essential for neuronal properties. However, the precise mechanisms underlying this process and its relevance to physiological and behavioural functions are poorly understood. To address these issues, we focused on a cassette exon of the Caenorhabditis elegans insulin receptor gene daf-2, whose proper variant expression in the taste receptor neuron ASER is critical for taste-avoidance learning. We show that inclusion of daf-2 exon 11.5 is restricted to specific neuron types, including ASER, and is controlled by a combinatorial action of evolutionarily conserved alternative splicing factors, RBFOX, CELF and PTB families of proteins. Mutations of these factors cause a learning defect, and this defect is relieved by DAF-2c (exon 11.5+) isoform expression only in a single neuron ASER. Our results provide evidence that alternative splicing regulation of a single critical gene in a single critical neuron is essential for learning ability in an organism. Little is known about the molecular mechanisms regulating neuron-specific alternative splicing. Here, the authors identify a combination of RNA-binding proteins regulating neuron-specific expression of the C. elegans insulin receptor isoform DAF-2c and find disrupting these factors leads to learning deficits.
Collapse
Affiliation(s)
- Masahiro Tomioka
- Molecular Genetics Research Laboratory, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yasuki Naito
- Molecular Genetics Research Laboratory, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan.,Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Hidehito Kuroyanagi
- Laboratory of Gene Expression, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Yuichi Iino
- Molecular Genetics Research Laboratory, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan.,Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
74
|
Hirani N, Westenberg M, Seed PT, Petalcorin MIR, Dolphin CT. C. elegans flavin-containing monooxygenase-4 is essential for osmoregulation in hypotonic stress. Biol Open 2016; 5:537-49. [PMID: 27010030 PMCID: PMC4874355 DOI: 10.1242/bio.017400] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Studies in Caenorhabditis elegans have revealed osmoregulatory systems engaged when worms experience hypertonic conditions, but less is known about measures employed when faced with hypotonic stress. Inactivation of fmo-4, which encodes flavin-containing monooxygenase-4, results in dramatic hypoosmotic hypersensitivity; worms are unable to prevent overwhelming water influx and swell rapidly, finally rupturing due to high internal hydrostatic pressure. fmo-4 is expressed prominently in hypodermis, duct and pore cells but is excluded from the excretory cell. Thus, FMO-4 plays a crucial osmoregulatory role by promoting clearance of excess water that enters during hypotonicity, perhaps by synthesizing an osmolyte that acts to establish an osmotic gradient from excretory cell to duct and pore cells. C. elegans FMO-4 contains a C-terminal extension conserved in all nematode FMO-4s. The coincidently numbered human FMO4 also contains an extended C-terminus with features similar to those of FMO-4. Although these shared sequence characteristics suggest potential orthology, human FMO4 was unable to rescue the fmo-4 osmoregulatory defect. Intriguingly, however, mammalian FMO4 is expressed predominantly in the kidney - an appropriate site if it too is, or once was, involved in osmoregulation.
Collapse
Affiliation(s)
- Nisha Hirani
- Institute of Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Marcel Westenberg
- Institute of Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Paul T Seed
- Division of Women's Health, King's College London, St Thomas' Hospital, London SE1 7EH, UK
| | - Mark I R Petalcorin
- Institute of Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Colin T Dolphin
- Institute of Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| |
Collapse
|
75
|
Thompson KW, Joshi P, Dymond JS, Gorrepati L, Smith HE, Krause MW, Eisenmann DM. The Paired-box protein PAX-3 regulates the choice between lateral and ventral epidermal cell fates in C. elegans. Dev Biol 2016; 412:191-207. [PMID: 26953187 PMCID: PMC4846358 DOI: 10.1016/j.ydbio.2016.03.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 03/01/2016] [Accepted: 03/01/2016] [Indexed: 01/12/2023]
Abstract
The development of the single cell layer skin or hypodermis of Caenorhabditis elegans is an excellent model for understanding cell fate specification and differentiation. Early in C. elegans embryogenesis, six rows of hypodermal cells adopt dorsal, lateral or ventral fates that go on to display distinct behaviors during larval life. Several transcription factors are known that function in specifying these major hypodermal cell fates, but our knowledge of the specification of these cell types is sparse, particularly in the case of the ventral hypodermal cells, which become Vulval Precursor Cells and form the vulval opening in response to extracellular signals. Previously, the gene pvl-4 was identified in a screen for mutants with defects in vulval development. We found by whole genome sequencing that pvl-4 is the Paired-box gene pax-3, which encodes the sole PAX-3 transcription factor homolog in C. elegans. pax-3 mutants show embryonic and larval lethality, and body morphology abnormalities indicative of hypodermal cell defects. We report that pax-3 is expressed in ventral P cells and their descendants during embryogenesis and early larval stages, and that in pax-3 reduction-of-function animals the ventral P cells undergo a cell fate transformation and express several markers of the lateral seam cell fate. Furthermore, forced expression of pax-3 in the lateral hypodermal cells causes them to lose expression of seam cell markers. We propose that pax-3 functions in the ventral hypodermal cells to prevent these cells from adopting the lateral seam cell fate. pax-3 represents the first gene required for specification solely of the ventral hypodermal fate in C. elegans providing insights into cell type diversification.
Collapse
Affiliation(s)
- Kenneth W Thompson
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| | - Pradeep Joshi
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| | - Jessica S Dymond
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| | - Lakshmi Gorrepati
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| | - Harold E Smith
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 5 Center Drive, Bethesda, MD 20892, USA.
| | - Michael W Krause
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, 5 Center Drive, Bethesda, MD 20892, USA.
| | - David M Eisenmann
- Department of Biological Sciences, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD 21250, USA.
| |
Collapse
|
76
|
Diapause is associated with a change in the polarity of secretion of insulin-like peptides. Nat Commun 2016; 7:10573. [PMID: 26838180 PMCID: PMC4742890 DOI: 10.1038/ncomms10573] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 12/30/2015] [Indexed: 12/19/2022] Open
Abstract
The insulin/IGF-1 signalling (IIS) pathway plays an important role in the regulation of larval diapause, the long-lived growth arrest state called dauer arrest, in Caenorhabditis elegans. In this nematode, 40 insulin-like peptides (ILPs) have been identified as putative ligands of the IIS pathway; however, it remains unknown how ILPs modulate larval diapause. Here we show that the secretory polarity of INS-35 and INS-7, which suppress larval diapause, is changed in the intestinal epithelial cells at larval diapause. These ILPs are secreted from the intestine into the body cavity during larval stages. In contrast, they are secreted into the intestinal lumen and degraded during dauer arrest, only to be secreted into the body cavity again when the worms return to developmental growth. The process that determines the secretory polarity of INS-35 and INS-7, thus, has an important role in the modulation of larval diapause. Insulin-like peptides INS-7 and INS-35 suppress larval diapause in Caenorhabditis elegans via unknown mechanism. Here, Matsunaga et al. show that the secretory polarity of both peptides changes in diapause, when these peptides are secreted into the intestinal lumen instead of the body cavity like in other larval stages.
Collapse
|
77
|
Leung A, Hua K, Ramachandran P, Hingwing K, Wu M, Koh PL, Hawkins N. C. elegans HAM-1 functions in the nucleus to regulate asymmetric neuroblast division. Dev Biol 2015; 410:56-69. [PMID: 26703426 DOI: 10.1016/j.ydbio.2015.12.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 12/08/2015] [Accepted: 12/10/2015] [Indexed: 01/19/2023]
Abstract
All 302 neurons in the C. elegans hermaphrodite arise through asymmetric division of neuroblasts. During embryogenesis, the C. elegans ham-1 gene is required for several asymmetric neuroblast divisions in lineages that generate both neural and apoptotic cells. By antibody staining, endogenous HAM-1 is found exclusively at the cell cortex in many cells during embryogenesis and is asymmetrically localized in dividing cells. Here we show that in transgenic embryos expressing a functional GFP::HAM-1 fusion protein, GFP expression is also detected in the nucleus, in addition to the cell cortex. Consistent with the nuclear localization is the presence of a putative DNA binding winged-helix domain within the N-terminus of HAM-1. Through a deletion analysis we determined that the C-terminus of the protein is required for nuclear localization and we identified two nuclear localization sequences (NLSs). A subcellular fractionation experiment from wild type embryos, followed by Western blotting, revealed that endogenous HAM-1 is primarily found in the nucleus. Our analysis also showed that the N-terminus is necessary for cortical localization. While ham-1 function is essential for asymmetric division in the lineage that generates the PLM mechanosensory neuron, we showed that cortical localization may not required. Thus, our results suggest that there is a nuclear function for HAM-1 in regulating asymmetric neuroblast division and that the requirement for cortical localization may be lineage dependent.
Collapse
Affiliation(s)
- Amy Leung
- Department of Molecular Biology and Biochemistry Simon Fraser University, Burnaby, BC, Canada
| | - Khang Hua
- Department of Molecular Biology and Biochemistry Simon Fraser University, Burnaby, BC, Canada
| | | | - Kyla Hingwing
- Department of Molecular Biology and Biochemistry Simon Fraser University, Burnaby, BC, Canada
| | - Maria Wu
- Department of Molecular Biology and Biochemistry Simon Fraser University, Burnaby, BC, Canada
| | - Pei Luan Koh
- Department of Molecular Biology and Biochemistry Simon Fraser University, Burnaby, BC, Canada
| | - Nancy Hawkins
- Department of Molecular Biology and Biochemistry Simon Fraser University, Burnaby, BC, Canada.
| |
Collapse
|
78
|
RAB-10 Regulates Dendritic Branching by Balancing Dendritic Transport. PLoS Genet 2015; 11:e1005695. [PMID: 26633194 PMCID: PMC4669152 DOI: 10.1371/journal.pgen.1005695] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 10/31/2015] [Indexed: 11/19/2022] Open
Abstract
The construction of a large dendritic arbor requires robust growth and the precise delivery of membrane and protein cargoes to specific subcellular regions of the developing dendrite. How the microtubule-based vesicular trafficking and sorting systems are regulated to distribute these dendritic development factors throughout the dendrite is not well understood. Here we identify the small GTPase RAB-10 and the exocyst complex as critical regulators of dendrite morphogenesis and patterning in the C. elegans sensory neuron PVD. In rab-10 mutants, PVD dendritic branches are reduced in the posterior region of the cell but are excessive in the distal anterior region of the cell. We also demonstrate that the dendritic branch distribution within PVD depends on the balance between the molecular motors kinesin-1/UNC-116 and dynein, and we propose that RAB-10 regulates dendrite morphology by balancing the activity of these motors to appropriately distribute branching factors, including the transmembrane receptor DMA-1. Building a complex dendritic arbor requires tremendous cellular growth, and how membrane and protein components are transported to support a rapidly growing, polarized dendrite remains unclear. We have identified the small GTPase RAB-10 as a key regulator of this process, providing insight into both dendritic development and the control of trafficking by small GTPases.
Collapse
|
79
|
Abstract
TRIM-NHL proteins are key regulators of developmental transitions, for example promoting differentiation, while inhibiting cell growth and proliferation, in stem and progenitor cells. Abnormalities in these proteins have been also associated with human diseases, particularly affecting muscular and neuronal functions, making them potential targets for therapeutic intervention. The purpose of this review is to provide a systematic and comprehensive summary on the most studied TRIM-NHL proteins, highlighting examples where connections were established between structural features, molecular functions and biological outcomes.
Collapse
Affiliation(s)
- Cristina Tocchini
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland
| | - Rafal Ciosk
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland.
| |
Collapse
|
80
|
Luck AN, Anderson KG, McClung CM, VerBerkmoes NC, Foster JM, Michalski ML, Slatko BE. Tissue-specific transcriptomics and proteomics of a filarial nematode and its Wolbachia endosymbiont. BMC Genomics 2015; 16:920. [PMID: 26559510 PMCID: PMC4642636 DOI: 10.1186/s12864-015-2083-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 10/15/2015] [Indexed: 11/12/2022] Open
Abstract
Background Filarial nematodes cause debilitating human diseases. While treatable, recent evidence suggests drug resistance is developing, necessitating the development of novel targets and new treatment options. Although transcriptomic and proteomic studies around the nematode life cycle have greatly enhanced our knowledge, whole organism approaches have not provided spatial resolution of gene expression, which can be gained by examining individual tissues. Generally, due to their small size, tissue dissection of human-infecting filarial nematodes remains extremely challenging. However, canine heartworm disease is caused by a closely related and much larger filarial nematode, Dirofilaria immitis. As with many other filarial nematodes, D. immitis contains Wolbachia, an obligate bacterial endosymbiont present in the hypodermis and developing oocytes within the uterus. Here, we describe the first concurrent tissue-specific transcriptomic and proteomic profiling of a filarial nematode (D. immitis) and its Wolbachia (wDi) in order to better understand tissue functions and identify tissue-specific antigens that may be used for the development of new diagnostic and therapeutic tools. Methods Adult D. immitis worms were dissected into female body wall (FBW), female uterus (FU), female intestine (FI), female head (FH), male body wall (MBW), male testis (MT), male intestine (MI), male head (MH) and 10.1186/s12864-015-2083-2 male spicule (MS) and used to prepare transcriptomic and proteomic libraries. Results Transcriptomic and proteomic analysis of several D. immitis tissues identified many biological functions enriched within certain tissues. Hierarchical clustering of the D. immitis tissue transcriptomes, along with the recently published whole-worm adult male and female D. immitis transcriptomes, revealed that the whole-worm transcriptome is typically dominated by transcripts originating from reproductive tissue. The uterus appeared to have the most variable transcriptome, possibly due to age. Although many functions are shared between the reproductive tissues, the most significant differences in gene expression were observed between the uterus and testis. Interestingly, wDi gene expression in the male and female body wall is fairly similar, yet slightly different to that of Wolbachia gene expression in the uterus. Proteomic methods verified 32 % of the predicted D. immitis proteome, including over 700 hypothetical proteins of D. immitis. Of note, hypothetical proteins were among some of the most abundant Wolbachia proteins identified, which may fulfill some important yet still uncharacterized biological function. Conclusions The spatial resolution gained from this parallel transcriptomic and proteomic analysis adds to our understanding of filarial biology and serves as a resource with which to develop future therapeutic strategies against filarial nematodes and their Wolbachia endosymbionts. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2083-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ashley N Luck
- Genome Biology Division, New England Biolabs, Inc., 240 County Road, Ipswich, MA, 01938, USA
| | - Kathryn G Anderson
- Department of Biology and Microbiology, University of Wisconsin Oshkosh, Oshkosh, WI, 54901, USA
| | - Colleen M McClung
- Chemical Biology Division, New England Biolabs, Inc., 240 County Road, Ipswich, MA, 01938, USA
| | - Nathan C VerBerkmoes
- Chemical Biology Division, New England Biolabs, Inc., 240 County Road, Ipswich, MA, 01938, USA
| | - Jeremy M Foster
- Genome Biology Division, New England Biolabs, Inc., 240 County Road, Ipswich, MA, 01938, USA
| | - Michelle L Michalski
- Department of Biology and Microbiology, University of Wisconsin Oshkosh, Oshkosh, WI, 54901, USA
| | - Barton E Slatko
- Genome Biology Division, New England Biolabs, Inc., 240 County Road, Ipswich, MA, 01938, USA.
| |
Collapse
|
81
|
Rompay LV, Borghgraef C, Beets I, Caers J, Temmerman L. New genetic regulators question relevance of abundant yolk protein production in C. elegans. Sci Rep 2015; 5:16381. [PMID: 26553710 PMCID: PMC4639837 DOI: 10.1038/srep16381] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 09/24/2015] [Indexed: 11/25/2022] Open
Abstract
Vitellogenesis or maternal yolk formation is considered critical to the reproduction of egg-laying animals. In invertebrates, however, most of its regulatory genes are still unknown. Via a combined mapping and whole-genome sequencing strategy, we performed a forward genetic screen to isolate novel regulators of yolk production in the nematode model system Caenorhabditis elegans. In addition to isolating new alleles of rab-35, rab-10 and M04F3.2, we identified five mutant alleles corresponding to three novel regulatory genes potently suppressing the expression of a GFP-based yolk reporter. We confirmed that mutations in vrp-1, ceh-60 and lrp-2 disrupt endogenous yolk protein synthesis at the transcriptional and translational level. In contrast to current beliefs, our discovered set of mutants with strongly reduced yolk proteins did not show serious reproduction defects. This raises questions as to whether yolk proteins per se are needed for ultimate reproductive success.
Collapse
Affiliation(s)
- Liesbeth Van Rompay
- Functional Genomics and Proteomics, Department of Biology, KU Leuven, Naamsestraat 59 bus 2465, 3000 Leuven, Belgium
| | - Charline Borghgraef
- Functional Genomics and Proteomics, Department of Biology, KU Leuven, Naamsestraat 59 bus 2465, 3000 Leuven, Belgium
| | - Isabel Beets
- Functional Genomics and Proteomics, Department of Biology, KU Leuven, Naamsestraat 59 bus 2465, 3000 Leuven, Belgium
| | - Jelle Caers
- Functional Genomics and Proteomics, Department of Biology, KU Leuven, Naamsestraat 59 bus 2465, 3000 Leuven, Belgium
| | - Liesbet Temmerman
- Functional Genomics and Proteomics, Department of Biology, KU Leuven, Naamsestraat 59 bus 2465, 3000 Leuven, Belgium
| |
Collapse
|
82
|
Sammut M, Cook SJ, Nguyen KCQ, Felton T, Hall DH, Emmons SW, Poole RJ, Barrios A. Glia-derived neurons are required for sex-specific learning in C. elegans. Nature 2015; 526:385-390. [PMID: 26469050 PMCID: PMC4650210 DOI: 10.1038/nature15700] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 09/10/2015] [Indexed: 12/14/2022]
Abstract
Sex differences in behaviour extend to cognitive-like processes such as learning, but the underlying dimorphisms in neural circuit development and organization that generate these behavioural differences are largely unknown. Here we define at the single-cell level-from development, through neural circuit connectivity, to function-the neural basis of a sex-specific learning in the nematode Caenorhabditis elegans. We show that sexual conditioning, a form of associative learning, requires a pair of male-specific interneurons whose progenitors are fully differentiated glia. These neurons are generated during sexual maturation and incorporated into pre-exisiting sex-shared circuits to couple chemotactic responses to reproductive priorities. Our findings reveal a general role for glia as neural progenitors across metazoan taxa and demonstrate that the addition of sex-specific neuron types to brain circuits during sexual maturation is an important mechanism for the generation of sexually dimorphic plasticity in learning.
Collapse
Affiliation(s)
- Michele Sammut
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT
| | - Steven J Cook
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Ken C Q Nguyen
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Terry Felton
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT
| | - David H Hall
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Scott W Emmons
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Richard J Poole
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT
| | - Arantza Barrios
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT
| |
Collapse
|
83
|
Karnik R, Beer MA. Identification of Predictive Cis-Regulatory Elements Using a Discriminative Objective Function and a Dynamic Search Space. PLoS One 2015; 10:e0140557. [PMID: 26465884 PMCID: PMC4605740 DOI: 10.1371/journal.pone.0140557] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 09/28/2015] [Indexed: 01/06/2023] Open
Abstract
The generation of genomic binding or accessibility data from massively parallel sequencing technologies such as ChIP-seq and DNase-seq continues to accelerate. Yet state-of-the-art computational approaches for the identification of DNA binding motifs often yield motifs of weak predictive power. Here we present a novel computational algorithm called MotifSpec, designed to find predictive motifs, in contrast to over-represented sequence elements. The key distinguishing feature of this algorithm is that it uses a dynamic search space and a learned threshold to find discriminative motifs in combination with the modeling of motifs using a full PWM (position weight matrix) rather than k-mer words or regular expressions. We demonstrate that our approach finds motifs corresponding to known binding specificities in several mammalian ChIP-seq datasets, and that our PWMs classify the ChIP-seq signals with accuracy comparable to, or marginally better than motifs from the best existing algorithms. In other datasets, our algorithm identifies novel motifs where other methods fail. Finally, we apply this algorithm to detect motifs from expression datasets in C. elegans using a dynamic expression similarity metric rather than fixed expression clusters, and find novel predictive motifs.
Collapse
Affiliation(s)
- Rahul Karnik
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States of America
| | - Michael A. Beer
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, United States of America
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD, United States of America
- * E-mail:
| |
Collapse
|
84
|
Metabolome and proteome changes with aging in Caenorhabditis elegans. Exp Gerontol 2015; 72:67-84. [PMID: 26390854 DOI: 10.1016/j.exger.2015.09.013] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 09/15/2015] [Accepted: 09/16/2015] [Indexed: 01/13/2023]
Abstract
To expand the understanding of aging in the model organism Caenorhabditis elegans, global quantification of metabolite and protein levels in young and aged nematodes was performed using mass spectrometry. With age, there was a decreased abundance of proteins functioning in transcription termination, mRNA degradation, mRNA stability, protein synthesis, and proteasomal function. Furthermore, there was altered S-adenosyl methionine metabolism as well as a decreased abundance of the S-adenosyl methionine synthetase (SAMS-1) protein. Other aging-related changes included alterations in free fatty acid levels and composition, decreased levels of ribosomal proteins, decreased levels of NADP-dependent isocitrate dehydrogenase (IDH1), a shift in the cellular redox state, an increase in sorbitol content, alterations in free amino acid levels, and indications of altered muscle function and sarcoplasmic reticulum Ca(2+) homeostasis. There were also decreases in pyrimidine and purine metabolite levels, most markedly nitrogenous bases. Supplementing the culture medium with cytidine (a pyrimidine nucleoside) or hypoxanthine (a purine base) increased lifespan slightly, suggesting that aging-induced alterations in ribonucleotide metabolism affect lifespan. An age-related increase in body size, lipotoxicity from ectopic yolk lipoprotein accumulation, a decline in NAD(+) levels, and mitochondrial electron transport chain dysfunction may explain many of these changes. In addition, dietary restriction in aged worms resulting from sarcopenia of the pharyngeal pump likely decreases the abundance of SAMS-1, possibly leading to decreased phosphatidylcholine levels, larger lipid droplets, and ER and mitochondrial stress. The complementary use of proteomics and metabolomics yielded unique insights into the molecular processes altered with age in C. elegans.
Collapse
|
85
|
Miller EV, Grandi LN, Giannini JA, Robinson JD, Powell JR. The Conserved G-Protein Coupled Receptor FSHR-1 Regulates Protective Host Responses to Infection and Oxidative Stress. PLoS One 2015; 10:e0137403. [PMID: 26360906 PMCID: PMC4567296 DOI: 10.1371/journal.pone.0137403] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Accepted: 08/17/2015] [Indexed: 01/08/2023] Open
Abstract
The innate immune system’s ability to sense an infection is critical so that it can rapidly respond if pathogenic microorganisms threaten the host, but otherwise maintain a quiescent baseline state to avoid causing damage to the host or to commensal microorganisms. One important mechanism for discriminating between pathogenic and non-pathogenic bacteria is the recognition of cellular damage caused by a pathogen during the course of infection. In Caenorhabditis elegans, the conserved G-protein coupled receptor FSHR-1 is an important constituent of the innate immune response. FSHR-1 activates the expression of antimicrobial infection response genes in infected worms and delays accumulation of the ingested pathogen Pseudomonas aeruginosa. FSHR-1 is central not only to the worm’s survival of infection by multiple pathogens, but also to the worm’s survival of xenobiotic cadmium and oxidative stresses. Infected worms produce reactive oxygen species to fight off the pathogens; FSHR-1 is required at the site of infection for the expression of detoxifying genes that protect the host from collateral damage caused by this defense response. Finally, the FSHR-1 pathway is important for the ability of worms to discriminate pathogenic from benign bacteria and subsequently initiate an aversive learning program that promotes selective pathogen avoidance.
Collapse
Affiliation(s)
- Elizabeth V. Miller
- Department of Biology, Gettysburg College, Gettysburg, Pennsylvania, United States of America
| | - Leah N. Grandi
- Department of Biology, Gettysburg College, Gettysburg, Pennsylvania, United States of America
| | - Jennifer A. Giannini
- Department of Biology, Gettysburg College, Gettysburg, Pennsylvania, United States of America
| | - Joseph D. Robinson
- Department of Biology, Gettysburg College, Gettysburg, Pennsylvania, United States of America
| | - Jennifer R. Powell
- Department of Biology, Gettysburg College, Gettysburg, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
86
|
Mok DZL, Sternberg PW, Inoue T. Morphologically defined sub-stages of C. elegans vulval development in the fourth larval stage. BMC DEVELOPMENTAL BIOLOGY 2015; 15:26. [PMID: 26066484 PMCID: PMC4464634 DOI: 10.1186/s12861-015-0076-7] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/03/2015] [Indexed: 01/06/2023]
Abstract
Background During the fourth larval (L4) stage, vulval cells of C. elegans undergo extensive morphogenesis accompanied by changes in gene expression. This phase of vulval development, occurring after the well-studied induction of vulval cells, is not well understood but is potentially a useful context in which to study how a complex temporal sequence of events is regulated during development. However, a system for precisely describing different phases of vulval development in the L4 stage has been lacking. Results We defined ten sub-stages of L4 based on morphological criteria as observed using Nomarski microscopy (L4.0 ~ L4.9). Precise timing of each sub-stage at 20 °C was determined. We also re-examined the timing of expression for several gene expression markers, and correlated the sub-stages with the timing of other developmental events in the vulva and the uterus. Conclusions This scheme allows the developmental timing of an L4 individual to be determined at approximately one-hour resolution without the need to resort to time course experiments. These well-defined developmental stages will enable more precise description of gene expression and other developmental events.
Collapse
Affiliation(s)
- Darren Z L Mok
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Blk MD7, #02-06, Singapore, 117597, Singapore
| | - Paul W Sternberg
- HHMI and Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Takao Inoue
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Blk MD7, #02-06, Singapore, 117597, Singapore. .,HHMI and Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| |
Collapse
|
87
|
Gorrepati L, Krause MW, Chen W, Brodigan TM, Correa-Mendez M, Eisenmann DM. Identification of Wnt Pathway Target Genes Regulating the Division and Differentiation of Larval Seam Cells and Vulval Precursor Cells in Caenorhabditis elegans. G3 (BETHESDA, MD.) 2015; 5:1551-66. [PMID: 26048561 PMCID: PMC4528312 DOI: 10.1534/g3.115.017715] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/18/2015] [Indexed: 12/29/2022]
Abstract
The evolutionarily conserved Wnt/β-catenin signaling pathway plays a fundamental role during metazoan development, regulating numerous processes including cell fate specification, cell migration, and stem cell renewal. Wnt ligand binding leads to stabilization of the transcriptional effector β-catenin and upregulation of target gene expression to mediate a cellular response. During larval development of the nematode Caenorhabditis elegans, Wnt/β-catenin pathways act in fate specification of two hypodermal cell types, the ventral vulval precursor cells (VPCs) and the lateral seam cells. Because little is known about targets of the Wnt signaling pathways acting during larval VPC and seam cell differentiation, we sought to identify genes regulated by Wnt signaling in these two hypodermal cell types. We conditionally activated Wnt signaling in larval animals and performed cell type-specific "mRNA tagging" to enrich for VPC and seam cell-specific mRNAs, and then used microarray analysis to examine gene expression compared to control animals. Two hundred thirty-nine genes activated in response to Wnt signaling were identified, and we characterized 50 genes further. The majority of these genes are expressed in seam and/or vulval lineages during normal development, and reduction of function for nine genes caused defects in the proper division, fate specification, fate execution, or differentiation of seam cells and vulval cells. Therefore, the combination of these techniques was successful at identifying potential cell type-specific Wnt pathway target genes from a small number of cells and at increasing our knowledge of the specification and behavior of these C. elegans larval hypodermal cells.
Collapse
Affiliation(s)
- Lakshmi Gorrepati
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland 21250
| | | | - Weiping Chen
- Intramural Research Program, NIDDK, Bethesda, Maryland 20814
| | | | - Margarita Correa-Mendez
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland 21250
| | - David M Eisenmann
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland 21250
| |
Collapse
|
88
|
Hench J, Henriksson J, Abou-Zied AM, Lüppert M, Dethlefsen J, Mukherjee K, Tong YG, Tang L, Gangishetti U, Baillie DL, Bürglin TR. The Homeobox Genes of Caenorhabditis elegans and Insights into Their Spatio-Temporal Expression Dynamics during Embryogenesis. PLoS One 2015; 10:e0126947. [PMID: 26024448 PMCID: PMC4448998 DOI: 10.1371/journal.pone.0126947] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 04/09/2015] [Indexed: 11/18/2022] Open
Abstract
Homeobox genes play crucial roles for the development of multicellular eukaryotes. We have generated a revised list of all homeobox genes for Caenorhabditis elegans and provide a nomenclature for the previously unnamed ones. We show that, out of 103 homeobox genes, 70 are co-orthologous to human homeobox genes. 14 are highly divergent, lacking an obvious ortholog even in other Caenorhabditis species. One of these homeobox genes encodes 12 homeodomains, while three other highly divergent homeobox genes encode a novel type of double homeodomain, termed HOCHOB. To understand how transcription factors regulate cell fate during development, precise spatio-temporal expression data need to be obtained. Using a new imaging framework that we developed, Endrov, we have generated spatio-temporal expression profiles during embryogenesis of over 60 homeobox genes, as well as a number of other developmental control genes using GFP reporters. We used dynamic feedback during recording to automatically adjust the camera exposure time in order to increase the dynamic range beyond the limitations of the camera. We have applied the new framework to examine homeobox gene expression patterns and provide an analysis of these patterns. The methods we developed to analyze and quantify expression data are not only suitable for C. elegans, but can be applied to other model systems or even to tissue culture systems.
Collapse
Affiliation(s)
- Jürgen Hench
- Dept. of Biosciences and Nutrition & Center for Biosciences, Karolinska Institutet, Hälsovägen 7, Novum, SE-141 83, Huddinge, Sweden
- School of Life Sciences, Södertörns Högskola, Huddinge, Sweden
| | - Johan Henriksson
- Dept. of Biosciences and Nutrition & Center for Biosciences, Karolinska Institutet, Hälsovägen 7, Novum, SE-141 83, Huddinge, Sweden
- School of Life Sciences, Södertörns Högskola, Huddinge, Sweden
| | - Akram M. Abou-Zied
- Dept. of Biosciences and Nutrition & Center for Biosciences, Karolinska Institutet, Hälsovägen 7, Novum, SE-141 83, Huddinge, Sweden
- School of Life Sciences, Södertörns Högskola, Huddinge, Sweden
| | - Martin Lüppert
- Dept. of Biosciences and Nutrition & Center for Biosciences, Karolinska Institutet, Hälsovägen 7, Novum, SE-141 83, Huddinge, Sweden
- School of Life Sciences, Södertörns Högskola, Huddinge, Sweden
| | - Johan Dethlefsen
- Dept. of Biosciences and Nutrition & Center for Biosciences, Karolinska Institutet, Hälsovägen 7, Novum, SE-141 83, Huddinge, Sweden
- School of Life Sciences, Södertörns Högskola, Huddinge, Sweden
| | - Krishanu Mukherjee
- Dept. of Biosciences and Nutrition & Center for Biosciences, Karolinska Institutet, Hälsovägen 7, Novum, SE-141 83, Huddinge, Sweden
- School of Life Sciences, Södertörns Högskola, Huddinge, Sweden
| | - Yong Guang Tong
- Dept. of Biosciences and Nutrition & Center for Biosciences, Karolinska Institutet, Hälsovägen 7, Novum, SE-141 83, Huddinge, Sweden
- School of Life Sciences, Södertörns Högskola, Huddinge, Sweden
| | - Lois Tang
- Dept. of Biosciences and Nutrition & Center for Biosciences, Karolinska Institutet, Hälsovägen 7, Novum, SE-141 83, Huddinge, Sweden
- School of Life Sciences, Södertörns Högskola, Huddinge, Sweden
| | - Umesh Gangishetti
- Dept. of Biosciences and Nutrition & Center for Biosciences, Karolinska Institutet, Hälsovägen 7, Novum, SE-141 83, Huddinge, Sweden
| | - David L. Baillie
- Dept. of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, V5A 1S6, Canada
| | - Thomas R. Bürglin
- Dept. of Biosciences and Nutrition & Center for Biosciences, Karolinska Institutet, Hälsovägen 7, Novum, SE-141 83, Huddinge, Sweden
- School of Life Sciences, Södertörns Högskola, Huddinge, Sweden
| |
Collapse
|
89
|
Abstract
Proteomic analysis of rare cells in heterogeneous environments presents difficult challenges. Systematic methods are needed to enrich, identify, and quantify proteins expressed in specific cells in complex biological systems including multicellular plants and animals. Here, we have engineered a Caenorhabditis elegans phenylalanyl-tRNA synthetase capable of tagging proteins with the reactive noncanonical amino acid p-azido-L-phenylalanine. We achieved spatiotemporal selectivity in the labeling of C. elegans proteins by controlling expression of the mutant synthetase using cell-selective (body wall muscles, intestinal epithelial cells, neurons, and pharyngeal muscle) or state-selective (heat-shock) promoters in several transgenic lines. Tagged proteins are distinguished from the rest of the protein pool through bioorthogonal conjugation of the azide side chain to probes that permit visualization and isolation of labeled proteins. By coupling our methodology with stable-isotope labeling of amino acids in cell culture (SILAC), we successfully profiled proteins expressed in pharyngeal muscle cells, and in the process, identified proteins not previously known to be expressed in these cells. Our results show that tagging proteins with spatiotemporal selectivity can be achieved in C. elegans and illustrate a convenient and effective approach for unbiased discovery of proteins expressed in targeted subsets of cells.
Collapse
|
90
|
Grants JM, Goh GYS, Taubert S. The Mediator complex of Caenorhabditis elegans: insights into the developmental and physiological roles of a conserved transcriptional coregulator. Nucleic Acids Res 2015; 43:2442-53. [PMID: 25634893 PMCID: PMC4344494 DOI: 10.1093/nar/gkv037] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The Mediator multiprotein complex (‘Mediator’) is an important transcriptional coregulator that is evolutionarily conserved throughout eukaryotes. Although some Mediator subunits are essential for the transcription of all protein-coding genes, others influence the expression of only subsets of genes and participate selectively in cellular signaling pathways. Here, we review the current knowledge of Mediator subunit function in the nematode Caenorhabditis elegans, a metazoan in which established and emerging genetic technologies facilitate the study of developmental and physiological regulation in vivo. In this nematode, unbiased genetic screens have revealed critical roles for Mediator components in core developmental pathways such as epidermal growth factor (EGF) and Wnt/β-catenin signaling. More recently, important roles for C. elegans Mediator subunits have emerged in the regulation of lipid metabolism and of systemic stress responses, engaging conserved transcription factors such as nuclear hormone receptors (NHRs). We emphasize instances where similar functions for individual Mediator subunits exist in mammals, highlighting parallels between Mediator subunit action in nematode development and in human cancer biology. We also discuss a parallel between the association of the Mediator subunit MED12 with several human disorders and the role of its C. elegans ortholog mdt-12 as a regulatory hub that interacts with numerous signaling pathways.
Collapse
Affiliation(s)
- Jennifer M Grants
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada Centre for Molecular Medicine and Therapeutics, Child & Family Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Grace Y S Goh
- Centre for Molecular Medicine and Therapeutics, Child & Family Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada Graduate Program in Cell and Developmental Biology, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Stefan Taubert
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada Centre for Molecular Medicine and Therapeutics, Child & Family Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada Graduate Program in Cell and Developmental Biology, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| |
Collapse
|
91
|
Ewald CY, Landis JN, Porter Abate J, Murphy CT, Blackwell TK. Dauer-independent insulin/IGF-1-signalling implicates collagen remodelling in longevity. Nature 2014; 519:97-101. [PMID: 25517099 PMCID: PMC4352135 DOI: 10.1038/nature14021] [Citation(s) in RCA: 225] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 10/27/2014] [Indexed: 01/04/2023]
Abstract
Interventions that delay ageing mobilize mechanisms that protect and repair cellular components, but it is unknown how these interventions might slow the functional decline of extracellular matrices, which are also damaged during ageing. Reduced insulin/IGF-1 signalling (rIIS) extends lifespan across the evolutionary spectrum, and in juvenile Caenorhabditis elegans also allows the transcription factor DAF-16/FOXO to induce development into dauer, a diapause that withstands harsh conditions. It has been suggested that rIIS delays C. elegans ageing through activation of dauer-related processes during adulthood, but some rIIS conditions confer robust lifespan extension unaccompanied by any dauer-like traits. Here we show that rIIS can promote C. elegans longevity through a program that is genetically distinct from the dauer pathway, and requires the Nrf (NF-E2-related factor) orthologue SKN-1 acting in parallel to DAF-16. SKN-1 is inhibited by IIS and has been broadly implicated in longevity, but is rendered dispensable for rIIS lifespan extension by even mild activity of dauer-related processes. When IIS is decreased under conditions that do not induce dauer traits, SKN-1 most prominently increases expression of collagens and other extracellular matrix genes. Diverse genetic, nutritional, and pharmacological pro-longevity interventions delay an age-related decline in collagen expression. These collagens mediate adulthood extracellular matrix remodelling, and are needed for ageing to be delayed by interventions that do not involve dauer traits. By genetically delineating a dauer-independent rIIS ageing pathway, our results show that IIS controls a broad set of protective mechanisms during C. elegans adulthood, and may facilitate elucidation of processes of general importance for longevity. The importance of collagen production in diverse anti-ageing interventions implies that extracellular matrix remodelling is a generally essential signature of longevity assurance, and that agents promoting extracellular matrix youthfulness may have systemic benefit.
Collapse
Affiliation(s)
- Collin Y Ewald
- 1] Joslin Diabetes Center, One Joslin Place, Boston, Massachusetts 02215, USA [2] Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, Massachusetts 02138, USA [3] Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, Massachusetts 02215, USA
| | - Jess N Landis
- Department of Molecular Biology, Lewis-Sigler Institute for Integrative Genomics, Princeton University, 148 Carl Icahn Laboratory, Washington Road, Princeton, New Jersey 08544, USA
| | - Jess Porter Abate
- 1] Joslin Diabetes Center, One Joslin Place, Boston, Massachusetts 02215, USA [2] Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, Massachusetts 02138, USA [3] Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, Massachusetts 02215, USA
| | - Coleen T Murphy
- Department of Molecular Biology, Lewis-Sigler Institute for Integrative Genomics, Princeton University, 148 Carl Icahn Laboratory, Washington Road, Princeton, New Jersey 08544, USA
| | - T Keith Blackwell
- 1] Joslin Diabetes Center, One Joslin Place, Boston, Massachusetts 02215, USA [2] Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, Massachusetts 02138, USA [3] Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, Massachusetts 02215, USA
| |
Collapse
|
92
|
Yuan W, Liu Y, Lok JB, Stoltzfus JD, Gasser RB, Lei W, Fang R, Zhao J, Hu M. Exploring features and function of Ss-riok-3, an enigmatic kinase gene from Strongyloides stercoralis. Parasit Vectors 2014; 7:561. [PMID: 25477034 PMCID: PMC4265397 DOI: 10.1186/s13071-014-0561-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 11/23/2014] [Indexed: 02/01/2023] Open
Abstract
Background Right open reading frame protein kinase 3 (RIOK-3) belongs to the atypical kinase family. Unlike the other two members, RIOK-1 and RIOK-2, which are conserved from Archaea to humans, RIOK-3 occurs only in multicellular organisms. Studies on HeLa cells indicate that human RIOK-3 is a component of the 40S small ribosome subunit and supports cancer cell growth and survival. However, almost nothing is known about the function of RIOK-3. We explored the functional role of RIOK-3 encoding gene from Strongyloides stercoralis, a parasitic nematode of humans and dogs. Methods To analyze the gene and promoter structure of Ss-riok-3, RACE-PCR and Genome-walker PCR were performed to isolate the full length cDNA, gDNA and promoter region of Ss-riok-3. RNA-seq was conducted to assess the transcript abundance of Ss-riok-3 in different stages of S. stercoralis. Transgenesis was employed to determine the anatomic expression patterns of Ss-riok-3. Results The RIOK-3 protein-encoding gene (designated Ss-riok-3) of S. stercoralis was characterized. The full-length complementary and genomic DNAs of the RIOK-3 encoding gene (riok-3) were isolated from this nematode. The cDNA of Ss-riok-3 is 1,757 bp in length, including a 23 bp 5’-UTR, a 36 bp 3’-UTR and a 1,698 bp coding region encoding a protein of 565 amino acids (aa) containing a RIO kinase domain. RNA sequencing (RNA-seq) analysis revealed that Ss-riok-3 is transcribed in all developmental stages of S. stercoralis assessed, with transcripts being particularly abundant in parasitic females. Gene structure analysis revealed that Ss-riok-3 contains no intron. The putative promoter contains conserved promoter elements, including four TATA, two GATA, one inverse GATA and one inverse CAAT boxes. The promoter of Ss-riok-3 drives GFP expression in the head neuron, intestine and body wall muscle of transgenic S. stercoralis larvae, and the TATA boxes present in the 3’-UTR of the gene immediately upstream of Ss-riok-3 initiate transcription. Conclusions The characterization of the RIOK-3 encoding gene from S. stercoralis provides a sound foundation for investigating in detail its function in the development and reproduction of this important pathogen. Electronic supplementary material The online version of this article (doi:10.1186/s13071-014-0561-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wang Yuan
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, College of Veterinary Medicine, Huazhong Agricultural University, 1 Shizishan Street, Wuhan, 430070, China.
| | - Yingying Liu
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, College of Veterinary Medicine, Huazhong Agricultural University, 1 Shizishan Street, Wuhan, 430070, China.
| | - James B Lok
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA, 19104, USA.
| | - Jonathan D Stoltzfus
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA, 19104, USA. .,Department of Biology, Hollins University, Roanoke, VI, 24020, USA.
| | - Robin B Gasser
- Faculty of Veterinary and Agricultural Sciences, The University of Melbourne, Corner of Flemington Road and Park Drive, Parkville, VI, 3010, Australia.
| | - Weiqiang Lei
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, College of Veterinary Medicine, Huazhong Agricultural University, 1 Shizishan Street, Wuhan, 430070, China.
| | - Rui Fang
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, College of Veterinary Medicine, Huazhong Agricultural University, 1 Shizishan Street, Wuhan, 430070, China.
| | - Junlong Zhao
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, College of Veterinary Medicine, Huazhong Agricultural University, 1 Shizishan Street, Wuhan, 430070, China.
| | - Min Hu
- State Key Laboratory of Agricultural Microbiology, Key Laboratory of Development of Veterinary Diagnostic Products, Ministry of Agriculture, College of Veterinary Medicine, Huazhong Agricultural University, 1 Shizishan Street, Wuhan, 430070, China.
| |
Collapse
|
93
|
Spencer WC, McWhirter R, Miller T, Strasbourger P, Thompson O, Hillier LW, Waterston RH, Miller DM. Isolation of specific neurons from C. elegans larvae for gene expression profiling. PLoS One 2014; 9:e112102. [PMID: 25372608 PMCID: PMC4221280 DOI: 10.1371/journal.pone.0112102] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 10/13/2014] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The simple and well-described structure of the C. elegans nervous system offers an unprecedented opportunity to identify the genetic programs that define the connectivity and function of individual neurons and their circuits. A correspondingly precise gene expression map of C. elegans neurons would facilitate the application of genetic methods toward this goal. Here we describe a powerful new approach, SeqCeL (RNA-Seq of C. elegans cells) for producing gene expression profiles of specific larval C. elegans neurons. METHODS AND RESULTS We have exploited available GFP reporter lines for FACS isolation of specific larval C. elegans neurons for RNA-Seq analysis. Our analysis showed that diverse classes of neurons are accessible to this approach. To demonstrate the applicability of this strategy to rare neuron types, we generated RNA-Seq profiles of the NSM serotonergic neurons that occur as a single bilateral pair of cells in the C. elegans pharynx. These data detected >1,000 NSM enriched transcripts, including the majority of previously known NSM-expressed genes. SIGNIFICANCE This work offers a simple and robust protocol for expression profiling studies of post-embryonic C. elegans neurons and thus provides an important new method for identifying candidate genes for key roles in neuron-specific development and function.
Collapse
Affiliation(s)
- W. Clay Spencer
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Rebecca McWhirter
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Tyne Miller
- Program in Neuroscience, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Pnina Strasbourger
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Owen Thompson
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - LaDeana W. Hillier
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Robert H. Waterston
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - David M. Miller
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
- Program in Neuroscience, Vanderbilt University, Nashville, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
94
|
In situ imaging in C. elegans reveals developmental regulation of microtubule dynamics. Dev Cell 2014; 29:203-16. [PMID: 24780738 DOI: 10.1016/j.devcel.2014.03.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 12/12/2013] [Accepted: 03/13/2014] [Indexed: 11/20/2022]
Abstract
Microtubules (MTs) are cytoskeletal polymers that undergo dynamic instability, the stochastic transition between growth and shrinkage phases. MT dynamics are required for diverse cellular processes and, while intrinsic to tubulin, are highly regulated. However, little is known about how MT dynamics facilitate or are regulated by tissue biogenesis and differentiation. We imaged MT dynamics in a smooth muscle-like lineage in intact developing Caenorhabditis elegans. All aspects of MT dynamics change significantly as stem-like precursors exit mitosis and, secondarily, as they differentiate. We found that suppression, but not enhancement, of dynamics perturbs differentiated muscle function in vivo. Distinct ensembles of MT-associated proteins are specifically required for tissue biogenesis versus tissue function. A CLASP family MT stabilizer and the depolymerizing kinesin MCAK are differentially required for MT dynamics in the precursor or differentiated cells, respectively. All of these multidimensional phenotypic comparisons were facilitated by a data display method called the diamond graph.
Collapse
|
95
|
Wong MC, Kennedy WP, Schwarzbauer JE. Transcriptionally regulated cell adhesion network dictates distal tip cell directionality. Dev Dyn 2014; 243:999-1010. [PMID: 24811939 DOI: 10.1002/dvdy.24146] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 04/30/2014] [Accepted: 05/02/2014] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND The mechanisms that govern directional changes in cell migration are poorly understood. The migratory paths of two distal tip cells (DTC) determine the U-shape of the C. elegans hermaphroditic gonad. The morphogenesis of this organ provides a model system to identify genes necessary for the DTCs to execute two stereotyped turns. RESULTS Using candidate genes for RNAi knockdown in a DTC-specific strain, we identified two transcriptional regulators required for DTC turning: cbp-1, the CBP/p300 transcriptional coactivator homologue, and let-607, a CREBH transcription factor homologue. Further screening of potential target genes uncovered a network of integrin adhesion-related genes that have roles in turning and are dependent on cbp-1 and let-607 for expression. These genes include src-1/Src kinase, tln-1/talin, pat-2/α integrin and nmy-2, a nonmuscle myosin heavy chain. CONCLUSIONS Transcriptional regulation by means of cbp-1 and let-607 is crucial for determining directional changes during DTC migration. These regulators coordinate a gene network that is necessary for integrin-mediated adhesion. Overall, these results suggest that directional changes in cell migration rely on the precise gene regulation of adhesion.
Collapse
Affiliation(s)
- Ming-Ching Wong
- Department of Molecular Biology, Princeton University, Princeton, New Jersey
| | | | | |
Collapse
|
96
|
Depuydt G, Xie F, Petyuk VA, Smolders A, Brewer HM, Camp DG, Smith RD, Braeckman BP. LC-MS proteomics analysis of the insulin/IGF-1-deficient Caenorhabditis elegans daf-2(e1370) mutant reveals extensive restructuring of intermediary metabolism. J Proteome Res 2014; 13:1938-56. [PMID: 24555535 PMCID: PMC3993954 DOI: 10.1021/pr401081b] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Indexed: 12/11/2022]
Abstract
The insulin/IGF-1 receptor is a major known determinant of dauer formation, stress resistance, longevity, and metabolism in Caenorhabditis elegans. In the past, whole-genome transcript profiling was used extensively to study differential gene expression in response to reduced insulin/IGF-1 signaling, including the expression levels of metabolism-associated genes. Taking advantage of the recent developments in quantitative liquid chromatography mass spectrometry (LC-MS)-based proteomics, we profiled the proteomic changes that occur in response to activation of the DAF-16 transcription factor in the germline-less glp-4(bn2);daf-2(e1370) receptor mutant. Strikingly, the daf-2 profile suggests extensive reorganization of intermediary metabolism, characterized by the upregulation of many core intermediary metabolic pathways. These include glycolysis/gluconeogenesis, glycogenesis, pentose phosphate cycle, citric acid cycle, glyoxylate shunt, fatty acid β-oxidation, one-carbon metabolism, propionate and tyrosine catabolism, and complexes I, II, III, and V of the electron transport chain. Interestingly, we found simultaneous activation of reciprocally regulated metabolic pathways, which is indicative of spatiotemporal coordination of energy metabolism and/or extensive post-translational regulation of these enzymes. This restructuring of daf-2 metabolism is reminiscent to that of hypometabolic dauers, allowing the efficient and economical utilization of internal nutrient reserves and possibly also shunting metabolites through alternative energy-generating pathways to sustain longevity.
Collapse
Affiliation(s)
- Geert Depuydt
- Biology
Department, Ghent University, Proeftuinstraat 86 N1, B-9000 Ghent, Belgium
| | - Fang Xie
- Biological
Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Vladislav A. Petyuk
- Biological
Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Arne Smolders
- Biology
Department, Ghent University, Proeftuinstraat 86 N1, B-9000 Ghent, Belgium
| | - Heather M. Brewer
- Biological
Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - David G. Camp
- Biological
Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Richard D. Smith
- Biological
Sciences Division and Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Bart P. Braeckman
- Biology
Department, Ghent University, Proeftuinstraat 86 N1, B-9000 Ghent, Belgium
| |
Collapse
|
97
|
The ubiquitin proteasome system in Caenorhabditis elegans and its regulation. Redox Biol 2014; 2:333-47. [PMID: 24563851 PMCID: PMC3926112 DOI: 10.1016/j.redox.2014.01.007] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/08/2014] [Accepted: 01/10/2014] [Indexed: 11/20/2022] Open
Abstract
Protein degradation constitutes a major cellular function that is responsible for maintenance of the normal cellular physiology either through the degradation of normal proteins or through the elimination of damaged proteins. The Ubiquitin–Proteasome System (UPS)1 is one of the main proteolytic systems that orchestrate protein degradation. Given that up- and down- regulation of the UPS system has been shown to occur in various normal (such as ageing) and pathological (such as neurodegenerative diseases) processes, the exogenous modulation of the UPS function and activity holds promise of (a) developing new therapeutic interventions against various diseases and (b) establishing strategies to maintain cellular homeostasis. Since the proteasome genes are evolutionarily conserved, their role can be dissected in simple model organisms, such as the nematode, Caenorhabditis elegans. In this review, we survey findings on the redox regulation of the UPS in C. elegans showing that the nematode is an instrumental tool in the identification of major players in the UPS pathway. Moreover, we specifically discuss UPS-related genes that have been modulated in the nematode and in human cells and have resulted in similar effects thus further exhibiting the value of this model in the study of the UPS. UPS is one of the main proteolytic systems that orchestrate protein degradation. Proteasome function can be dissected in Caenorhabditis elegans. Nematodes can be used in the identification of major players in the UPS pathway.
Collapse
|
98
|
Salzberg Y, Díaz-Balzac CA, Ramirez-Suarez NJ, Attreed M, Tecle E, Desbois M, Kaprielian Z, Bülow HE. Skin-derived cues control arborization of sensory dendrites in Caenorhabditis elegans. Cell 2013; 155:308-20. [PMID: 24120132 DOI: 10.1016/j.cell.2013.08.058] [Citation(s) in RCA: 125] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 07/18/2013] [Accepted: 08/26/2013] [Indexed: 11/25/2022]
Abstract
Sensory dendrites depend on cues from their environment to pattern their growth and direct them toward their correct target tissues. Yet, little is known about dendrite-substrate interactions during dendrite morphogenesis. Here, we describe MNR-1/menorin, which is part of the conserved Fam151 family of proteins and is expressed in the skin to control the elaboration of "menorah"-like dendrites of mechanosensory neurons in Caenorhabditis elegans. We provide biochemical and genetic evidence that MNR-1 acts as a contact-dependent or short-range cue in concert with the neural cell adhesion molecule SAX-7/L1CAM in the skin and through the neuronal leucine-rich repeat transmembrane receptor DMA-1 on sensory dendrites. Our data describe an unknown pathway that provides spatial information from the skin substrate to pattern sensory dendrite development nonautonomously.
Collapse
Affiliation(s)
- Yehuda Salzberg
- Department of Genetics, Albert Einstein College of Medicine of Yeshiva University, Bronx, NY 10461, USA
| | | | | | | | | | | | | | | |
Collapse
|
99
|
Gissendanner CR, Cardin D, Dubose CJ, El Sayed M, Harmson JS, Praslicka B, Rowan BG. C. elegans nuclear receptor NHR-6 functionally interacts with the jun-1 transcription factor during spermatheca development. Genesis 2013; 52:29-38. [PMID: 24178943 DOI: 10.1002/dvg.22723] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 10/23/2013] [Accepted: 10/24/2013] [Indexed: 12/27/2022]
Abstract
The NR4A nuclear receptor NHR-6 is an essential regulator of spermatheca organogenesis in C. elegans. In this study, we perform a focused, RNAi-based screen to identify modifiers of partial nhr-6 loss of function. Ninety-eight genes that encode signaling proteins expressed in the spermatheca were screened for enhancement of the nhr-6 RNAi phenotype. We identify the C. elegans gene jun-1, which encodes the homolog of the Jun transcription factor, as a strong enhancer of nhr-6 partial loss of function. We show that nhr-6 and jun-1 function together to regulate development of the spermatheca and are necessary for generating an organ with the normal number of cells. jun-1 is expressed in all cells of the developing spermatheca. We also provide evidence that NHR-6 and JUN-1 can physically interact in a yeast two-hybrid assay. Our results provide in vivo evidence that NR4A nuclear receptor and Jun transcription factor interactions are essential in regulating developmental processes in metazoans.
Collapse
Affiliation(s)
- Chris R Gissendanner
- Department of Basic Pharmaceutical Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, Louisiana, 71209
| | | | | | | | | | | | | |
Collapse
|
100
|
Gu S, Goszczynski B, McGhee JD, Fire AZ. Unusual DNA packaging characteristics in endoreduplicated Caenorhabditis elegans oocytes defined by in vivo accessibility to an endogenous nuclease activity. Epigenetics Chromatin 2013; 6:37. [PMID: 24279402 PMCID: PMC3819648 DOI: 10.1186/1756-8935-6-37] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 09/17/2013] [Indexed: 01/29/2023] Open
Abstract
Background Germ cells in animals are highly specialized to preserve the genome. A distinct set of chromatin structures must be properly established in germ cells to maintain cell fate and genome integrity. We describe DNA-surface interactions in activated Caenorhabditis elegans oocytes that are revealed through the activity of an endogenous nuclease ('endocleavage’). Results Our analysis began with an unexpected observation that a majority (>50%) of DNA from ovulated but unfertilized C. elegans oocytes can be recovered in fragments of approximately 500 base pairs or shorter, cleaved at regular intervals (10 to 11 nt) along the DNA helix. In some areas of the genome, DNA cleavage patterns in these endoreduplicated oocytes appear consistent from cell-to-cell, indicating coherent rotational positioning of the DNA in chromatin. Particularly striking in this analysis are arrays of sensitive sites with a periodicity of approximately 10 bp that persist for several hundred base pairs of genomic DNA, longer than a single nucleosome core. Genomic regions with a strong bias toward a 10-nt periodic occurrence of A(n)/T(n) (so-called PATC regions) appear to exhibit a high degree of rotational constraint in endocleavage phasing, with a strong tendency for the periodic A(n)/T(n) sites to remain on the face of the helix protected from nuclease digestion. Conclusion The present analysis provides evidence for an unusual structure in C. elegans oocytes in which genomic DNA and associated protein structures are coherently linked.
Collapse
|