1
|
Opdensteinen P, Charudattan R, Hong JC, Rosskopf EN, Steinmetz NF. Biochemical and nanotechnological approaches to combat phytoparasitic nematodes. PLANT BIOTECHNOLOGY JOURNAL 2024; 22:2444-2460. [PMID: 38831638 PMCID: PMC11332226 DOI: 10.1111/pbi.14359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 03/09/2024] [Accepted: 04/05/2024] [Indexed: 06/05/2024]
Abstract
The foundation of most food production systems underpinning global food security is the careful management of soil resources. Embedded in the concept of soil health is the impact of diverse soil-borne pests and pathogens, and phytoparasitic nematodes represent a particular challenge. Root-knot nematodes and cyst nematodes are severe threats to agriculture, accounting for annual yield losses of US$157 billion. The control of soil-borne phytoparasitic nematodes conventionally relies on the use of chemical nematicides, which can have adverse effects on the environment and human health due to their persistence in soil, plants, and water. Nematode-resistant plants offer a promising alternative, but genetic resistance is species-dependent, limited to a few crops, and breeding and deploying resistant cultivars often takes years. Novel approaches for the control of phytoparasitic nematodes are therefore required, those that specifically target these parasites in the ground whilst minimizing the impact on the environment, agricultural ecosystems, and human health. In addition to the development of next-generation, environmentally safer nematicides, promising biochemical strategies include the combination of RNA interference (RNAi) with nanomaterials that ensure the targeted delivery and controlled release of double-stranded RNA. Genome sequencing has identified more than 75 genes in root knot and cyst nematodes that have been targeted with RNAi so far. But despite encouraging results, the delivery of dsRNA to nematodes in the soil remains inefficient. In this review article, we describe the state-of-the-art RNAi approaches targeting phytoparasitic nematodes and consider the potential benefits of nanotechnology to improve dsRNA delivery.
Collapse
Affiliation(s)
- Patrick Opdensteinen
- Department of NanoEngineeringUniversity of California, San DiegoLa JollaCaliforniaUSA
- Center for Nano‐ImmunoEngineeringUniversity of California, San DiegoLa JollaCaliforniaUSA
- Shu and K.C. Chien and Peter Farrell CollaboratoryUniversity of California, San DiegoLa JollaCaliforniaUSA
| | | | - Jason C. Hong
- USDA‐ARS‐U.S. Horticultural Research LaboratoryFort PierceFloridaUSA
| | - Erin N. Rosskopf
- USDA‐ARS‐U.S. Horticultural Research LaboratoryFort PierceFloridaUSA
| | - Nicole F. Steinmetz
- Department of NanoEngineeringUniversity of California, San DiegoLa JollaCaliforniaUSA
- Center for Nano‐ImmunoEngineeringUniversity of California, San DiegoLa JollaCaliforniaUSA
- Shu and K.C. Chien and Peter Farrell CollaboratoryUniversity of California, San DiegoLa JollaCaliforniaUSA
- Department of BioengineeringUniversity of California, San DiegoLa JollaCaliforniaUSA
- Department of RadiologyUniversity of California, San DiegoLa JollaCaliforniaUSA
- Institute for Materials Discovery and Design, University of California, San DiegoLa JollaCaliforniaUSA
- Moores Cancer CenterUniversity of California, San DiegoLa JollaCaliforniaUSA
- Center for Engineering in Cancer, Institute of Engineering in MedicineUniversity of California, San DiegoLa JollaCaliforniaUSA
| |
Collapse
|
2
|
Watzinger G, Bennett HL. Ceramide Synthase HYL-2 is Required for Neural Preconditioning to Anoxia in Caenorhabditis elegans . MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001024. [PMID: 38872843 PMCID: PMC11170290 DOI: 10.17912/micropub.biology.001024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 02/13/2024] [Accepted: 05/28/2024] [Indexed: 06/15/2024]
Abstract
Oxygen is vital for neuron development and function, and low oxygen (hypoxia) or 0% oxygen available (anoxia) conditions lead to neuronal dysfunction and death. Nonlethal forms of stress, prior to hypoxic or anoxic (preconditioning) environments protects neurons and increases survival to oxygen deprivation. Hyperpolarization of C. elegans neurons prior to anoxia (neural preconditioning) increases survival, but the cellular and molecular pathways that confer survival are unclear. Here we report that loss in ceramide synthase gene, hyl-2 suppresses increased survival to anoxia in neural preconditioned animals, suggesting that HYL-2 functions upstream of the circuit that regulates neural preconditioning.
Collapse
Affiliation(s)
- Ginger Watzinger
- Department of Biology, Trinity College, Hartford, Connecticut, United States
| | - Heather L Bennett
- Department of Biology, Trinity College, Hartford, Connecticut, United States
| |
Collapse
|
3
|
Shi L, Zhang A, Liu H, Wang H. Deletion of the foxO4 Gene Increases Hypoxia Tolerance in Zebrafish. Int J Mol Sci 2023; 24:ijms24108942. [PMID: 37240290 DOI: 10.3390/ijms24108942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/23/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Oxygen homeostasis is an important organizing principle for understanding development, physiology, disease, and evolution. Under various physiological and pathological states, organisms experience oxygen deficiency or hypoxia. FoxO4 has been recognized as an important transcriptional regulator involved in a variety of cellular functions, including proliferation, apoptosis, differentiation, and stress resistance, but its role in hypoxia adaptation mechanisms in animals is not so clear. To explore the role of foxO4 in the hypoxia response, we detected the expression of foxO4 and the regulatory relationship between Hif1α and foxO4 under hypoxic conditions. It was found that the expression of foxO4 was up-regulated in ZF4 cells and zebrafish tissues after hypoxia treatment, and Hif1α could directly target the HRE of the foxO4 promoter to regulate foxO4 transcription, indicating that foxO4 was involved in the hypoxia response by the Hif1α-mediated pathway. Furthermore, we obtained foxO4 knockout zebrafish and found that the disruption of foxO4 increased the tolerance to hypoxia. Further research found that the oxygen consumption and locomotor activity of foxO4-/- zebrafish were lower than those of WT zebrafish, as was true for NADH content, NADH/NAD+ rate, and expression of mitochondrial respiratory chain complex-related genes. This suggests that disruption of foxO4 reduced the oxygen demand threshold of the organism, which explained why the foxO4-/- zebrafish were more tolerant to hypoxia than WT zebrafish. These results will provide a theoretical basis for further study of the role of foxO4 in the hypoxia response.
Collapse
Affiliation(s)
- Linlin Shi
- Key Lab of Freshwater Animal Breeding/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, Wuhan 430070, China
| | - Axin Zhang
- Key Lab of Freshwater Animal Breeding/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, Wuhan 430070, China
| | - Hong Liu
- Key Lab of Freshwater Animal Breeding/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, Wuhan 430070, China
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, China
| | - Huanling Wang
- Key Lab of Freshwater Animal Breeding/Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Fishery, Huazhong Agricultural University, Wuhan 430070, China
- Engineering Research Center of Green Development for Conventional Aquatic Biological Industry in the Yangtze River Economic Belt, Ministry of Education, Wuhan 430070, China
| |
Collapse
|
4
|
Ding K, Barretto EC, Johnston M, Lee B, Gallo M, Grewal SS. Transcriptome analysis of FOXO-dependent hypoxia gene expression identifies Hipk as a regulator of low oxygen tolerance in Drosophila. G3 (BETHESDA, MD.) 2022; 12:6749561. [PMID: 36200850 PMCID: PMC9713431 DOI: 10.1093/g3journal/jkac263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/16/2022] [Indexed: 12/05/2022]
Abstract
When exposed to low oxygen or hypoxia, animals must alter their metabolism and physiology to ensure proper cell-, tissue-, and whole-body level adaptations to their hypoxic environment. These alterations often involve changes in gene expression. While extensive work has emphasized the importance of the HIF-1 alpha transcription factor on controlling hypoxia gene expression, less is known about other transcriptional mechanisms. We previously identified the transcription factor FOXO as a regulator of hypoxia tolerance in Drosophila larvae and adults. Here, we use an RNA-sequencing approach to identify FOXO-dependent changes in gene expression that are associated with these tolerance effects. We found that hypoxia altered the expression of over 2,000 genes and that ∼40% of these gene expression changes required FOXO. We discovered that hypoxia exposure led to a FOXO-dependent increase in genes involved in cell signaling, such as kinases, GTPase regulators, and regulators of the Hippo/Yorkie pathway. Among these, we identified homeodomain-interacting protein kinase as being required for hypoxia survival. We also found that hypoxia suppresses the expression of genes involved in ribosome synthesis and egg production, and we showed that hypoxia suppresses tRNA synthesis and mRNA translation and reduces female fecundity. Among the downregulated genes, we discovered that FOXO was required for the suppression of many ribosomal protein genes and genes involved in oxidative phosphorylation, pointing to a role for FOXO in limiting energetically costly processes such as protein synthesis and mitochondrial activity upon hypoxic stress. This work uncovers a widespread role for FOXO in mediating hypoxia changes in gene expression.
Collapse
Affiliation(s)
- Kate Ding
- Clark H. Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada.,Department of Biochemistry and Molecular Biology Calgary, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Elizabeth C Barretto
- Clark H. Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada.,Department of Biochemistry and Molecular Biology Calgary, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Michael Johnston
- Clark H. Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada.,Department of Biochemistry and Molecular Biology Calgary, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Byoungchun Lee
- Clark H. Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada.,Department of Biochemistry and Molecular Biology Calgary, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Marco Gallo
- Clark H. Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada.,Department of Biochemistry and Molecular Biology Calgary, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Savraj S Grewal
- Clark H. Smith Brain Tumour Centre, Arnie Charbonneau Cancer Institute, Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada.,Department of Biochemistry and Molecular Biology Calgary, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
5
|
Deciphering the mechanism of anhydrobiosis in the entomopathogenic nematode Heterorhabditis indica through comparative transcriptomics. PLoS One 2022; 17:e0275342. [PMID: 36301967 PMCID: PMC9612587 DOI: 10.1371/journal.pone.0275342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 09/14/2022] [Indexed: 11/07/2022] Open
Abstract
The entomopathogenic nematode, Heterorhabditis indica, is a popular biocontrol agent of high commercial significance. It possesses tremendous genetic architecture to survive desiccation stress by undergoing anhydrobiosis to increase its lifespan-an attribute exploited in the formulation technology. The comparative transcriptome of unstressed and anhydrobiotic H. indica revealed several previously concealed metabolic events crucial for adapting towards the moisture stress. During the induction of anhydrobiosis in the infective juveniles (IJ), 1584 transcripts were upregulated and 340 downregulated. As a strategy towards anhydrobiotic survival, the IJ showed activation of several genes critical to antioxidant defense, detoxification pathways, signal transduction, unfolded protein response and molecular chaperones and ubiquitin-proteasome system. Differential expression of several genes involved in gluconeogenesis - β-oxidation of fatty acids, glyoxylate pathway; glyceroneogenesis; fatty acid biosynthesis; amino-acid metabolism - shikimate pathway, sachharopine pathway, kyneurine pathway, lysine biosynthesis; one-carbon metabolism-polyamine pathway, transsulfuration pathway, folate cycle, methionine cycle, nucleotide biosynthesis; mevalonate pathway; and glyceraldehyde-3-phosphate dehydrogenase were also observed. We report the role of shikimate pathway, sachharopine pathway and glyceroneogenesis in anhydrobiotes, and seven classes of repeat proteins, specifically in H. indica for the first time. These results provide insights into anhydrobiotic survival strategies which can be utilized to strengthen the development of novel formulations with enhanced and sustained shelf-life.
Collapse
|
6
|
Lautens MJ, Tan JH, Serrat X, Del Borrello S, Schertzberg MR, Fraser AG. Identification of enzymes that have helminth-specific active sites and are required for Rhodoquinone-dependent metabolism as targets for new anthelmintics. PLoS Negl Trop Dis 2021; 15:e0009991. [PMID: 34843467 PMCID: PMC8659336 DOI: 10.1371/journal.pntd.0009991] [Citation(s) in RCA: 2] [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: 06/11/2021] [Revised: 12/09/2021] [Accepted: 11/11/2021] [Indexed: 11/18/2022] Open
Abstract
Soil transmitted helminths (STHs) are major human pathogens that infect over a billion people. Resistance to current anthelmintics is rising and new drugs are needed. Here we combine multiple approaches to find druggable targets in the anaerobic metabolic pathways STHs need to survive in their mammalian host. These require rhodoquinone (RQ), an electron carrier used by STHs and not their hosts. We identified 25 genes predicted to act in RQ-dependent metabolism including sensing hypoxia and RQ synthesis and found 9 are required. Since all 9 have mammalian orthologues, we used comparative genomics and structural modeling to identify those with active sites that differ between host and parasite. Together, we found 4 genes that are required for RQ-dependent metabolism and have different active sites. Finding these high confidence targets can open up in silico screens to identify species selective inhibitors of these enzymes as new anthelmintics.
Collapse
Affiliation(s)
- Margot J. Lautens
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - June H. Tan
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Xènia Serrat
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | | | | | - Andrew G. Fraser
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
7
|
Hypoxia Tolerant Species: The Wisdom of Nature Translated into Targets for Stroke Therapy. Int J Mol Sci 2021; 22:ijms222011131. [PMID: 34681788 PMCID: PMC8537001 DOI: 10.3390/ijms222011131] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/05/2021] [Accepted: 10/12/2021] [Indexed: 12/13/2022] Open
Abstract
Human neurons rapidly die after ischemia and current therapies for stroke management are limited to restoration of blood flow to prevent further brain damage. Thrombolytics and mechanical thrombectomy are the available reperfusion treatments, but most of the patients remain untreated. Neuroprotective therapies focused on treating the pathogenic cascade of the disease have widely failed. However, many animal species demonstrate that neurons can survive the lack of oxygen for extended periods of time. Here, we reviewed the physiological and molecular pathways inherent to tolerant species that have been described to contribute to hypoxia tolerance. Among them, Foxo3 and Eif5A were reported to mediate anoxic survival in Drosophila and Caenorhabditis elegans, respectively, and those results were confirmed in experimental models of stroke. In humans however, the multiple mechanisms involved in brain cell death after a stroke causes translation difficulties to arise making necessary a timely and coordinated control of the pathological changes. We propose here that, if we were able to plagiarize such natural hypoxia tolerance through drugs combined in a pharmacological cocktail it would open new therapeutic opportunities for stroke and likely, for other hypoxic conditions.
Collapse
|
8
|
Yan J, Sun CL, Shin S, Van Gilst M, Crowder CM. Effect of the mitochondrial unfolded protein response on hypoxic death and mitochondrial protein aggregation. Cell Death Dis 2021; 12:711. [PMID: 34267182 PMCID: PMC8282665 DOI: 10.1038/s41419-021-03979-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/23/2022]
Abstract
Mitochondria are the main oxygen consumers in cells and as such are the primary organelle affected by hypoxia. All hypoxia pathology presumably derives from the initial mitochondrial dysfunction. An early event in hypoxic pathology in C. elegans is disruption of mitochondrial proteostasis with induction of the mitochondrial unfolded protein response (UPRmt) and mitochondrial protein aggregation. Here in C. elegans, we screen through RNAis and mutants that confer either strong resistance to hypoxic cell death or strong induction of the UPRmt to determine the relationship between hypoxic cell death, UPRmt activation, and hypoxia-induced mitochondrial protein aggregation (HIMPA). We find that resistance to hypoxic cell death invariantly mitigated HIMPA. We also find that UPRmt activation invariantly mitigated HIMPA. However, UPRmt activation was neither necessary nor sufficient for resistance to hypoxic death and vice versa. We conclude that UPRmt is not necessarily hypoxia protective against cell death but does protect from mitochondrial protein aggregation, one of the early hypoxic pathologies in C. elegans.
Collapse
Affiliation(s)
- Junyi Yan
- Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, WA, 98109, USA.,Mitochondrial and Metabolism Center, University of Washington School of Medicine, Seattle, WA, 98109, USA.,Department of Anesthesiology, Central Hospital of Changdian, 118214, Dandong, Liaoning, China
| | - Chun-Ling Sun
- Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, WA, 98109, USA.,Mitochondrial and Metabolism Center, University of Washington School of Medicine, Seattle, WA, 98109, USA
| | - Seokyung Shin
- Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, WA, 98109, USA.,Mitochondrial and Metabolism Center, University of Washington School of Medicine, Seattle, WA, 98109, USA.,Department of Anesthesiology and Pain Medicine, Anesthesia and Pain Research Institute Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Marc Van Gilst
- Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, WA, 98109, USA.,Mitochondrial and Metabolism Center, University of Washington School of Medicine, Seattle, WA, 98109, USA
| | - C Michael Crowder
- Department of Anesthesiology and Pain Medicine, University of Washington School of Medicine, Seattle, WA, 98109, USA. .,Mitochondrial and Metabolism Center, University of Washington School of Medicine, Seattle, WA, 98109, USA. .,Department of Genome Science, University of Washington School of Medicine, Seattle, WA, 98109, USA.
| |
Collapse
|
9
|
Bennett HL, McClanahan PD, Fang-Yen C, Kalb RG. Preconditioning of Caenorhabditis elegans to anoxic insult by inactivation of cholinergic, GABAergic and muscle activity. GENES, BRAIN, AND BEHAVIOR 2021; 20:e12713. [PMID: 33155386 DOI: 10.1111/gbb.12713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 10/19/2020] [Accepted: 11/04/2020] [Indexed: 11/26/2022]
Abstract
For most metazoans, oxygen deprivation leads to cell dysfunction and if severe, death. Sublethal stress prior to a hypoxic or anoxic insult ("preconditioning") can protect cells from subsequent oxygen deprivation. The molecular mechanisms by which sublethal stress can buffer against a subsequent toxic insult and the role of the nervous system in the response are not well understood. We studied the role of neuronal activity preconditioning to oxygen deprivation in Caenorhabditis elegans. Animals expressing the histamine gated chloride channels (HisCl1) in select cell populations were used to temporally and spatially inactivate the nervous system or tissue prior to an anoxic insult. We find that inactivation of the nervous system for 3 h prior to the insult confers resistance to a 48-h anoxic insult in 4th-stage larval animals. Experiments show that this resistance can be attributed to loss of activity in cholinergic and GABAergic neurons as well as in body wall muscles. These observations indicate that the nervous system activity can mediate the organism's response to anoxia.
Collapse
Affiliation(s)
- Heather L Bennett
- Department of Pediatrics, Division of Neurology, Research Institute, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biology, Reem-Kayden Center for Science and Computation, Bard College, New York, New York, USA
| | - Patrick D McClanahan
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Christopher Fang-Yen
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Neuroscience, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Robert G Kalb
- Department of Pediatrics, Division of Neurology, Research Institute, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
10
|
Abbass M, Chen Y, Arlt VM, Stürzenbaum SR. Benzo[a]pyrene and Caenorhabditis elegans: defining the genotoxic potential in an organism lacking the classical CYP1A1 pathway. Arch Toxicol 2021; 95:1055-1069. [PMID: 33420596 PMCID: PMC7904753 DOI: 10.1007/s00204-020-02968-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 12/22/2020] [Indexed: 11/29/2022]
Abstract
Benzo[a]pyrene (BaP) is bioactivated in most organisms by the cytochrome P450 (CYP) enzymes, mainly CYP1A1, ultimately resulting in the reactive metabolite BaP-7,8-dihydrodiol-9,10-epoxide (BPDE) capable of covalently binding to DNA and forming adducts. This step has been defined as the key process in cancer initiation in humans. However, limited knowledge is available about the consequences of BaP exposure in organisms lacking this classical CYP1A1 pathway, one example is the model nematode Caenorhabditis elegans. The aim of this study was to define the genotoxic potential of BaP in C. elegans and to advance our understanding of xenobiotic processing in the absence of the CYP1A1 pathway. Exposure to high concentrations of BaP (0-40 µM) significantly affected life cycle endpoints of C. elegans, which were manifested by a reduced reproductive output and shortened life span. An optimised comet assay revealed that DNA damage increased in a dose-dependent manner; however, no bulky DNA adducts (dG-N2-BPDE) were observed by 32P-postlabelling. Global transcriptomic analysis by RNA-Seq identified responsive transcript families, most prominently members of the cyp-35 and UDP-glucuronosyltransferases (UGTs) enzyme families, both of which are linked to xenobiotic metabolism. Strains harbouring mutations in the cyp-35A2 and cyp-35A3 genes were notably less prone to BaP-mediated toxicity, and BaP led to longevity in cyp-35A5 mutants. In summary, BaP induces transcriptional, genotoxic and phenotypic responses in C. elegans, despite the absence of the classical CYP1A1 bioactivation pathway. This provides first evidence that parallel pathways are implicated in BaP metabolism in C. elegans and this seems to be mediated via the cyp-35 pathway.
Collapse
Affiliation(s)
- Mustafa Abbass
- Department of Analytical, Environmental and Forensic Sciences, School of Population Health and Environmental Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Yuzhi Chen
- Department of Analytical, Environmental and Forensic Sciences, School of Population Health and Environmental Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Volker M Arlt
- Department of Analytical, Environmental and Forensic Sciences, School of Population Health and Environmental Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK.,Toxicology Department, GAB Consulting GmbH, 69126, Heidelberg, Germany
| | - Stephen R Stürzenbaum
- Department of Analytical, Environmental and Forensic Sciences, School of Population Health and Environmental Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK.
| |
Collapse
|
11
|
Itani OA, Zhong X, Tang X, Scott BA, Yan JY, Flibotte S, Lim Y, Hsieh AC, Bruce JE, Van Gilst M, Crowder CM. Coordinate Regulation of Ribosome and tRNA Biogenesis Controls Hypoxic Injury and Translation. Curr Biol 2020; 31:128-137.e5. [PMID: 33157031 DOI: 10.1016/j.cub.2020.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 08/21/2020] [Accepted: 10/01/2020] [Indexed: 01/01/2023]
Abstract
The translation machinery is composed of a myriad of proteins and RNAs whose levels must be coordinated to efficiently produce proteins without wasting energy or substrate. However, protein synthesis is clearly not always perfectly tuned to its environment, as disruption of translation machinery components can lengthen lifespan and stress survival. While much has been learned from bacteria and yeast about translational regulation, much less is known in metazoans. In a screen for mutations protecting C. elegans from hypoxic stress, we isolated multiple genes impacting protein synthesis: a ribosomal RNA helicase gene, tRNA biosynthesis genes, and a gene controlling amino acid availability. To define better the mechanisms by which these genes impact protein synthesis, we performed a second screen for suppressors of the conditional developmental arrest phenotype of the RNA helicase mutant and identified genes involved in ribosome biogenesis. Surprisingly, these suppressor mutations restored normal hypoxic sensitivity and protein synthesis to the tRNA biogenesis mutants, but not to the mutant reducing amino acid uptake. Proteomic analysis demonstrated that reduced tRNA biosynthetic activity produces a selective homeostatic reduction in ribosomal subunits, thereby offering a mechanism for the suppression results. Our study uncovers an unrecognized higher-order-translation regulatory mechanism in a metazoan whereby ribosome biogenesis genes communicate with genes controlling tRNA abundance matching the global rate of protein synthesis with available resources.
Collapse
Affiliation(s)
- Omar A Itani
- Department of Anesthesiology and Pain Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195-6540, USA; Mitochondria and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98105, USA
| | - Xuefei Zhong
- Department of Genome Sciences, University of Washington, 3720 15th Ave NE, Seattle, WA 98105, USA
| | - Xiaoting Tang
- Department of Genome Sciences, University of Washington, 3720 15th Ave NE, Seattle, WA 98105, USA
| | - Barbara A Scott
- Department of Anesthesiology and Pain Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195-6540, USA; Mitochondria and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98105, USA
| | - Jun Yi Yan
- Department of Anesthesiology and Pain Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195-6540, USA; Mitochondria and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98105, USA; Department of Anesthesiology, Central Hospital of Changdian, Dandong, Liaoning 118214, China
| | - Stephane Flibotte
- Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall Vancouver, BC V6T 1Z3, Canada
| | - Yiting Lim
- Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N, Seattle, WA 98109, USA
| | - Andrew C Hsieh
- Department of Genome Sciences, University of Washington, 3720 15th Ave NE, Seattle, WA 98105, USA; Division of Human Biology, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave. N, Seattle, WA 98109, USA; Department of Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195-6420, USA
| | - James E Bruce
- Department of Genome Sciences, University of Washington, 3720 15th Ave NE, Seattle, WA 98105, USA
| | - Marc Van Gilst
- Department of Anesthesiology and Pain Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195-6540, USA; Mitochondria and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98105, USA
| | - C Michael Crowder
- Department of Anesthesiology and Pain Medicine, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195-6540, USA; Mitochondria and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98105, USA; Department of Genome Sciences, University of Washington, 3720 15th Ave NE, Seattle, WA 98105, USA.
| |
Collapse
|
12
|
Heimbucher T, Hog J, Gupta P, Murphy CT. PQM-1 controls hypoxic survival via regulation of lipid metabolism. Nat Commun 2020; 11:4627. [PMID: 33009389 PMCID: PMC7532158 DOI: 10.1038/s41467-020-18369-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 07/31/2020] [Indexed: 12/25/2022] Open
Abstract
Animals have evolved responses to low oxygen conditions to ensure their survival. Here, we have identified the C. elegans zinc finger transcription factor PQM-1 as a regulator of the hypoxic stress response. PQM-1 is required for the longevity of insulin signaling mutants, but surprisingly, loss of PQM-1 increases survival under hypoxic conditions. PQM-1 functions as a metabolic regulator by controlling oxygen consumption rates, suppressing hypoxic glycogen levels, and inhibiting the expression of the sorbitol dehydrogenase-1 SODH-1, a crucial sugar metabolism enzyme. PQM-1 promotes hypoxic fat metabolism by maintaining the expression of the stearoyl-CoA desaturase FAT-7, an oxygen consuming, rate-limiting enzyme in fatty acid biosynthesis. PQM-1 activity positively regulates fat transport to developing oocytes through vitellogenins under hypoxic conditions, thereby increasing survival rates of arrested progeny during hypoxia. Thus, while pqm-1 mutants increase survival of mothers, ultimately this loss is detrimental to progeny survival. Our data support a model in which PQM-1 controls a trade-off between lipid metabolic activity in the mother and her progeny to promote the survival of the species under hypoxic conditions. Animals respond to hypoxic stress by adjusting metabolic processes to balance survival and reproduction. Here the authors identify the transcription factor PQM-1 as a metabolic regulator that balances hypoxic lipid and carbohydrate metabolism in C. elegans to limit somatic integrity and promote progeny survival.
Collapse
Affiliation(s)
- Thomas Heimbucher
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, 08544, USA. .,Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA. .,Bioinformatics and Molecular Genetics, Faculty of Biology, University of Freiburg, Freiburg, 79104, Baden-Wuerttemberg, Germany.
| | - Julian Hog
- Bioinformatics and Molecular Genetics, Faculty of Biology, University of Freiburg, Freiburg, 79104, Baden-Wuerttemberg, Germany
| | - Piyush Gupta
- Bioinformatics and Molecular Genetics, Faculty of Biology, University of Freiburg, Freiburg, 79104, Baden-Wuerttemberg, Germany
| | - Coleen T Murphy
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, 08544, USA. .,Department of Molecular Biology, Princeton University, Princeton, NJ, 08544, USA.
| |
Collapse
|
13
|
Tolerance to Hypoxia Is Promoted by FOXO Regulation of the Innate Immunity Transcription Factor NF-κB/Relish in Drosophila. Genetics 2020; 215:1013-1025. [PMID: 32513813 DOI: 10.1534/genetics.120.303219] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 05/21/2020] [Indexed: 12/14/2022] Open
Abstract
Exposure of tissues and organs to low oxygen (hypoxia) occurs in both physiological and pathological conditions in animals. Under these conditions, organisms have to adapt their physiology to ensure proper functioning and survival. Here, we define a role for the transcription factor Forkhead Box-O (FOXO) as a mediator of hypoxia tolerance in Drosophila We find that upon hypoxia exposure, FOXO transcriptional activity is rapidly induced in both larvae and adults. Moreover, we see that foxo mutant animals show misregulated glucose metabolism in low oxygen and subsequently exhibit reduced hypoxia survival. We identify the innate immune transcription factor, NF-κB/Relish, as a key FOXO target in the control of hypoxia tolerance. We find that expression of Relish and its target genes is increased in a FOXO-dependent manner in hypoxia, and that relish mutant animals show reduced survival in hypoxia. Together, these data indicate that FOXO is a hypoxia-inducible factor that mediates tolerance to low oxygen by inducing immune-like responses.
Collapse
|
14
|
Machiela E, Liontis T, Dues DJ, Rudich PD, Traa A, Wyman L, Kaufman C, Cooper JF, Lew L, Nadarajan S, Senchuk MM, Van Raamsdonk JM. Disruption of mitochondrial dynamics increases stress resistance through activation of multiple stress response pathways. FASEB J 2020; 34:8475-8492. [PMID: 32385951 PMCID: PMC7313680 DOI: 10.1096/fj.201903235r] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 04/05/2020] [Accepted: 04/15/2020] [Indexed: 12/17/2022]
Abstract
Mitochondria are dynamic organelles that can change shape and size depending on the needs of the cell through the processes of mitochondrial fission and fusion. In this work, we investigated the role of mitochondrial dynamics in organismal stress response. By using C. elegans as a genetic model, we could visualize mitochondrial morphology in a live organism with well‐established stress assays and well‐characterized stress response pathways. We found that disrupting mitochondrial fission (DRP1/drp‐1) or fusion (OPA1/eat‐3, MFN/fzo‐1) genes caused alterations in mitochondrial morphology that impacted both mitochondrial function and physiologic rates. While both mitochondrial fission and mitochondrial fusion mutants showed increased sensitivity to osmotic stress and anoxia, surprisingly we found that the mitochondrial fusion mutants eat‐3 and fzo‐1 are more resistant to both heat stress and oxidative stress. In exploring the mechanism of increased stress resistance, we found that disruption of mitochondrial fusion genes resulted in the upregulation of multiple stress response pathways. Overall, this work demonstrates that disrupting mitochondrial dynamics can have opposite effects on resistance to different types of stress. Our results suggest that disruption of mitochondrial fusion activates multiple stress response pathways that enhance resistance to specific stresses.
Collapse
Affiliation(s)
- Emily Machiela
- Laboratory of Aging and Neurodegenerative Disease, Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Thomas Liontis
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.,Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Dylan J Dues
- Laboratory of Aging and Neurodegenerative Disease, Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Paige D Rudich
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.,Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Annika Traa
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.,Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Leslie Wyman
- Laboratory of Aging and Neurodegenerative Disease, Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Corah Kaufman
- Laboratory of Aging and Neurodegenerative Disease, Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Jason F Cooper
- Laboratory of Aging and Neurodegenerative Disease, Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Leira Lew
- Laboratory of Aging and Neurodegenerative Disease, Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | | | - Megan M Senchuk
- Laboratory of Aging and Neurodegenerative Disease, Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Jeremy M Van Raamsdonk
- Laboratory of Aging and Neurodegenerative Disease, Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA.,Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.,Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Genetics, Harvard Medical School, Boston, MA, USA.,Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
15
|
Shao H, Han Z, Krasteva N, Wang D. Identification of signaling cascade in the insulin signaling pathway in response to nanopolystyrene particles. Nanotoxicology 2019; 13:174-188. [DOI: 10.1080/17435390.2018.1530395] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Huimin Shao
- Medical School, Southeast University, Nanjing, China
| | - Zhongyu Han
- Medical School, Southeast University, Nanjing, China
| | - Natalia Krasteva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Science, Sofia, Bulgaria
| | - Dayong Wang
- Medical School, Southeast University, Nanjing, China
| |
Collapse
|
16
|
Doshi S, Price E, Landis J, Barot U, Sabatella M, Lans H, Kalb RG. Neuropeptide signaling regulates the susceptibility of developing C. elegans to anoxia. Free Radic Biol Med 2019; 131:197-208. [PMID: 30529384 DOI: 10.1016/j.freeradbiomed.2018.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/05/2018] [Accepted: 12/05/2018] [Indexed: 02/04/2023]
Abstract
Inadequate delivery of oxygen to organisms during development can lead to cell dysfunction/death and life-long disabilities. Although the susceptibility of developing cells to low oxygen conditions changes with maturation, the cellular and molecular pathways that govern responses to low oxygen are incompletely understood. Here we show that developing Caenorhabditis elegans are substantially more sensitive to anoxia than adult animals and that this sensitivity is controlled by nervous system generated hormones (e.g., neuropeptides). A screen of neuropeptide genes identified and validated nlp-40 and its receptor aex-2 as a key regulator of anoxic survival in developing worms. The survival-promoting action of impaired neuropeptide signaling does not rely on five known stress resistance pathways and is specific to anoxic insult. Together, these data highlight a novel cell non-autonomous pathway that regulates the susceptibility of developing organisms to anoxia.
Collapse
Affiliation(s)
- Shachee Doshi
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Emma Price
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Justin Landis
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Urva Barot
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Mariangela Sabatella
- Department of Molecular Genetics, Erasmus Medical Centre, Oncode Institute, Cancer Genomics Netherlands, Rotterdam 3015 CN, the Netherlands
| | - Hannes Lans
- Department of Molecular Genetics, Erasmus Medical Centre, Oncode Institute, Cancer Genomics Netherlands, Rotterdam 3015 CN, the Netherlands
| | - Robert G Kalb
- Division of Neurology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Neuroscience Graduate Group, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
17
|
Kadekar P, Chaouni R, Clark E, Kazanets A, Roy R. Genome-wide surveys reveal polarity and cytoskeletal regulators mediate LKB1-associated germline stem cell quiescence. BMC Genomics 2018; 19:462. [PMID: 29907081 PMCID: PMC6003023 DOI: 10.1186/s12864-018-4847-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 05/31/2018] [Indexed: 12/22/2022] Open
Abstract
Background Caenorhabditis elegans can endure long periods of environmental stress by altering their development to execute a quiescent state called “dauer”. Previous work has implicated LKB1 - the causative gene in the autosomal dominant, cancer pre-disposing disease called Peutz-Jeghers Syndrome (PJS), and its downstream target AMPK, in the establishment of germline stem cell (GSC) quiescence during the dauer stage. Loss of function mutations in both LKB1/par-4 and AMPK/aak(0) result in untimely GSC proliferation during the onset of the dauer stage, although the molecular mechanism through which these factors regulate quiescence remains unclear. Curiously, the hyperplasia observed in par-4 mutants is more severe than AMPK-compromised dauer larvae, suggesting that par-4 has alternative downstream targets in addition to AMPK to regulate germline quiescence. Results We conducted three genome-wide RNAi screens to identify potential downstream targets of the protein kinases PAR-4 and AMPK that mediate dauer-dependent GSC quiescence. First, we screened to identify genes that phenocopy the par-4-dependent hyperplasia when compromised by RNAi. Two additional RNAi screens were performed to identify genes that suppressed the germline hyperplasia in par-4 and aak(0) dauer larvae, respectively. Interestingly, a subset of the candidates we identified are involved in the regulation of cell polarity and cytoskeletal function downstream of par-4, in an AMPK-independent manner. Moreover, we show that par-4 temporally regulates actin cytoskeletal organization within the dauer germ line at the rachis-adjacent membrane, in an AMPK-independent manner. Conclusion Our data suggest that the regulation of the cytoskeleton and cell polarity may contribute significantly to the tumour suppressor function of LKB1/par-4. Electronic supplementary material The online version of this article (10.1186/s12864-018-4847-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Pratik Kadekar
- Department of Biology, McGill University, 1205 avenue Docteur Penfield, Montreal, Quebec, H3A 1B1, Canada
| | - Rita Chaouni
- Department of Biology, McGill University, 1205 avenue Docteur Penfield, Montreal, Quebec, H3A 1B1, Canada
| | - Emily Clark
- Department of Biology, McGill University, 1205 avenue Docteur Penfield, Montreal, Quebec, H3A 1B1, Canada
| | - Anna Kazanets
- Department of Biology, McGill University, 1205 avenue Docteur Penfield, Montreal, Quebec, H3A 1B1, Canada
| | - Richard Roy
- Department of Biology, McGill University, 1205 avenue Docteur Penfield, Montreal, Quebec, H3A 1B1, Canada.
| |
Collapse
|
18
|
King SD, Gray CF, Song L, Nechushtai R, Gumienny TL, Mittler R, Padilla PA. The cisd gene family regulates physiological germline apoptosis through ced-13 and the canonical cell death pathway in Caenorhabditis elegans. Cell Death Differ 2018; 26:162-178. [PMID: 29666474 PMCID: PMC6294797 DOI: 10.1038/s41418-018-0108-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 02/13/2018] [Accepted: 03/05/2018] [Indexed: 11/09/2022] Open
Abstract
Programmed cell death, which occurs through a conserved core molecular pathway, is important for fundamental developmental and homeostatic processes. The human iron-sulfur binding protein NAF-1/CISD2 binds to Bcl-2 and its disruption in cells leads to an increase in apoptosis. Other members of the CDGSH iron sulfur domain (CISD) family include mitoNEET/CISD1 and Miner2/CISD3. In humans, mutations in CISD2 result in Wolfram syndrome 2, a disease in which the patients display juvenile diabetes, neuropsychiatric disorders and defective platelet aggregation. The C. elegans genome contains three previously uncharacterized cisd genes that code for CISD-1, which has homology to mitoNEET/CISD1 and NAF-1/CISD2, and CISD-3.1 and CISD-3.2, both of which have homology to Miner2/CISD3. Disrupting the function of the cisd genes resulted in various germline abnormalities including distal tip cell migration defects and a significant increase in the number of cell corpses within the adult germline. This increased germ cell death is blocked by a gain-of-function mutation of the Bcl-2 homolog CED-9 and requires functional caspase CED-3 and the APAF-1 homolog CED-4. Furthermore, the increased germ cell death is facilitated by the pro-apoptotic, CED-9-binding protein CED-13, but not the related EGL-1 protein. This work is significant because it places the CISD family members as regulators of physiological germline programmed cell death acting through CED-13 and the core apoptotic machinery.
Collapse
Affiliation(s)
- Skylar D King
- Department of Biological Sciences, University of North Texas, Denton, TX, 76203, USA
| | - Chipo F Gray
- Department of Biological Sciences, University of North Texas, Denton, TX, 76203, USA
| | - Luhua Song
- Department of Biological Sciences, University of North Texas, Denton, TX, 76203, USA
| | - Rachel Nechushtai
- Alexander Silberman Institute of Life Sciences, Hebrew University of Jerusalem, Edmond J. Safra Campus at Givat Ram, Jerusalem, 91904, Israel
| | - Tina L Gumienny
- Department of Biology, Texas Woman's University, Denton, TX, 76204, USA
| | - Ron Mittler
- Department of Biological Sciences, University of North Texas, Denton, TX, 76203, USA
| | - Pamela A Padilla
- Department of Biological Sciences, University of North Texas, Denton, TX, 76203, USA.
| |
Collapse
|
19
|
Glucose or Altered Ceramide Biosynthesis Mediate Oxygen Deprivation Sensitivity Through Novel Pathways Revealed by Transcriptome Analysis in Caenorhabditis elegans. G3-GENES GENOMES GENETICS 2016; 6:3149-3160. [PMID: 27507791 PMCID: PMC5068937 DOI: 10.1534/g3.116.031583] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Individuals with type 2 diabetes display metabolic abnormalities, such as hyperglycemia, increased free fatty acids, insulin resistance, and altered ceramide levels, that contribute to vascular dysfunctions and compromised oxygen delivery. Caenorhabditis elegans fed a glucose-supplemented diet or with altered ceramide metabolism, due to a hyl-2 mutation, are sensitive to oxygen deprivation (anoxia). Our experiments showed that the combination of these factors further decreased the anoxia survival. RNA-sequencing analysis was performed to assess how a glucose-supplemented diet and/or a hyl-2 mutation altered the transcriptome. Comparison analysis of transcripts associated with anoxia-sensitive animals [hyl-2(tm2031) mutation or a glucose diet] revealed 199 common transcripts encoded by genes with known or predicted functions involving innate immunity, cuticle function (collagens), or xenobiotic and endobiotic phase I and II detoxification system. Use of RNA interference (RNAi) to target gene products of the xenobiotic and endobiotic phase I and II detoxification system (UDP-glycosyltransferase and Cytochrome p450 genes; ugt-15, ugt-18, ugt-19, ugt-41, ugt-63, cyp-13A12, cyp-25A1, and cyp-33C8) increased anoxia survival in wild-type animals fed a standard diet. Anoxia sensitivity of the hyl-2(tm2031) animals was suppressed by RNAi of cyp-25A1 or cyp-33C8 genes. A glucose diet fed to the P0 hermaphrodite decreased the anoxia survival of its F1 embryos; however, the RNAi of ugt-63 and cyp-33C8 suppressed anoxia sensitivity. These studies provide evidence that the detoxification system impacts oxygen deprivation responses and that C. elegans can be used to model the conserved detoxification system.
Collapse
|
20
|
Dey S, Proulx SR, Teotónio H. Adaptation to Temporally Fluctuating Environments by the Evolution of Maternal Effects. PLoS Biol 2016; 14:e1002388. [PMID: 26910440 PMCID: PMC4766184 DOI: 10.1371/journal.pbio.1002388] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 01/21/2016] [Indexed: 01/27/2023] Open
Abstract
All organisms live in temporally fluctuating environments. Theory predicts that the evolution of deterministic maternal effects (i.e., anticipatory maternal effects or transgenerational phenotypic plasticity) underlies adaptation to environments that fluctuate in a predictably alternating fashion over maternal-offspring generations. In contrast, randomizing maternal effects (i.e., diversifying and conservative bet-hedging), are expected to evolve in response to unpredictably fluctuating environments. Although maternal effects are common, evidence for their adaptive significance is equivocal since they can easily evolve as a correlated response to maternal selection and may or may not increase the future fitness of offspring. Using the hermaphroditic nematode Caenorhabditis elegans, we here show that the experimental evolution of maternal glycogen provisioning underlies adaptation to a fluctuating normoxia-anoxia hatching environment by increasing embryo survival under anoxia. In strictly alternating environments, we found that hermaphrodites evolved the ability to increase embryo glycogen provisioning when they experienced normoxia and to decrease embryo glycogen provisioning when they experienced anoxia. At odds with existing theory, however, populations facing irregularly fluctuating normoxia-anoxia hatching environments failed to evolve randomizing maternal effects. Instead, adaptation in these populations may have occurred through the evolution of fitness effects that percolate over multiple generations, as they maintained considerably high expected growth rates during experimental evolution despite evolving reduced fecundity and reduced embryo survival under one or two generations of anoxia. We develop theoretical models that explain why adaptation to a wide range of patterns of environmental fluctuations hinges on the existence of deterministic maternal effects, and that such deterministic maternal effects are more likely to contribute to adaptation than randomizing maternal effects.
Collapse
Affiliation(s)
- Snigdhadip Dey
- Institut de Biologie de l´École Normale Supérieure, INSERM U1024, CNRS UMR 8197, Paris, France
| | - Stephen R. Proulx
- Department of Ecology, Evolution, and Marine Biology, University of California, Santa Barbara, Santa Barbara, California, United States of America
| | - Henrique Teotónio
- Institut de Biologie de l´École Normale Supérieure, INSERM U1024, CNRS UMR 8197, Paris, France
| |
Collapse
|
21
|
Glycogen Fuels Survival During Hyposmotic-Anoxic Stress in Caenorhabditis elegans. Genetics 2015; 201:65-74. [PMID: 26116152 PMCID: PMC4566277 DOI: 10.1534/genetics.115.179416] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 06/18/2015] [Indexed: 11/18/2022] Open
Abstract
Oxygen is an absolute requirement for multicellular life. Animals that are deprived of oxygen for sufficient periods of time eventually become injured and die. This is largely due to the fact that, without oxygen, animals are unable to generate sufficient quantities of energy. In human diseases triggered by oxygen deprivation, such as heart attack and stroke, hyposmotic stress and cell swelling (edema) arise in affected tissues as a direct result of energetic failure. Edema independently enhances tissue injury in these diseases by incompletely understood mechanisms, resulting in poor clinical outcomes. Here, we present investigations into the effects of osmotic stress during complete oxygen deprivation (anoxia) in the genetically tractable nematode Caenorhabditis elegans. Our findings demonstrate that nematode survival of a hyposmotic environment during anoxia (hyposmotic anoxia) depends on the nematode’s ability to engage in glycogen metabolism. We also present results of a genome-wide screen for genes affecting glycogen content and localization in the nematode, showing that nematode survival of hyposmotic anoxia depends on a large number of these genes. Finally, we show that an inability to engage in glycogen synthesis results in suppression of the enhanced survival phenotype observed in daf-2 insulin-like pathway mutants, suggesting that alterations in glycogen metabolism may serve as a basis for these mutants’ resistance to hyposmotic anoxia.
Collapse
|
22
|
Glucose induces sensitivity to oxygen deprivation and modulates insulin/IGF-1 signaling and lipid biosynthesis in Caenorhabditis elegans. Genetics 2015; 200:167-84. [PMID: 25762526 DOI: 10.1534/genetics.115.174631] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 03/02/2015] [Indexed: 12/15/2022] Open
Abstract
Diet is a central environmental factor that contributes to the phenotype and physiology of individuals. At the root of many human health issues is the excess of calorie intake relative to calorie expenditure. For example, the increasing amount of dietary sugars in the human diet is contributing to the rise of obesity and type 2 diabetes. Individuals with obesity and type 2 diabetes have compromised oxygen delivery, and thus it is of interest to investigate the impact a high-sugar diet has on oxygen deprivation responses. By utilizing the Caenorhabditis elegans genetic model system, which is anoxia tolerant, we determined that a glucose-supplemented diet negatively impacts responses to anoxia and that the insulin-like signaling pathway, through fatty acid and ceramide synthesis, modulates anoxia survival. Additionally, a glucose-supplemented diet alters lipid localization and initiates a positive chemotaxis response. Use of RNA-sequencing analysis to compare gene expression responses in animals fed either a standard or glucose-supplemented diet revealed that glucose impacts the expression of genes involved with multiple cellular processes including lipid and carbohydrate metabolism, stress responses, cell division, and extracellular functions. Several of the genes we identified show homology to human genes that are differentially regulated in response to obesity or type 2 diabetes, suggesting that there may be conserved gene expression responses between C. elegans fed a glucose-supplemented diet and a diabetic and/or obesity state observed in humans. These findings support the utility of the C. elegans model for understanding the molecular mechanisms regulating dietary-induced metabolic diseases.
Collapse
|
23
|
Shi Z, Yu H, Sun Y, Yang C, Lian H, Cai P. The Energy Metabolism in Caenorhabditis elegans under The Extremely Low-Frequency Electromagnetic Field Exposure. Sci Rep 2015; 5:8471. [PMID: 25683579 PMCID: PMC4329544 DOI: 10.1038/srep08471] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 01/21/2015] [Indexed: 02/08/2023] Open
Abstract
A literal mountain of documentation generated in the past five decades showing unmistakable health hazards associated with extremely low-frequency electromagnetic fields (ELF-EMFs) exposure. However, the relation between energy mechanism and ELF-EMF exposure is poorly understood. In this study, Caenorhabditis elegans was exposed to 50 Hz ELF-EMF at intensities of 0.5, 1, 2, and 3 mT, respectively. Their metabolite variations were analyzed by GC-TOF/MS-based metabolomics. Although minimal metabolic variations and no regular pattern were observed, the contents of energy metabolism-related metabolites such as pyruvic acid, fumaric acid, and L-malic acid were elevated in all the treatments. The expressions of nineteen related genes that encode glycolytic enzymes were analyzed by using quantitative real-time PCR. Only genes encoding GAPDH were significantly upregulated (P < 0.01), and this result was further confirmed by western blot analysis. The enzyme activity of GAPDH was increased (P < 0.01), whereas the total intracellular ATP level was decreased. While no significant difference in lifespan, hatching rate and reproduction, worms exposed to ELF-EMF exhibited less food consumption compared with that of the control (P < 0.01). In conclusion, C. elegans exposed to ELF-EMF have enhanced energy metabolism and restricted dietary, which might contribute to the resistance against exogenous ELF-EMF stress.
Collapse
Affiliation(s)
- Zhenhua Shi
- 1] Physical Environment Group, Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, P. R. China [2] University of the Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, P. R. China
| | - Hui Yu
- Physical Environment Group, Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, P. R. China
| | - Yongyan Sun
- 1] Physical Environment Group, Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, P. R. China [2] University of the Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, P. R. China
| | - Chuanjun Yang
- Physical Environment Group, Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, P. R. China
| | - Huiyong Lian
- Physical Environment Group, Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, P. R. China
| | - Peng Cai
- Physical Environment Group, Key Laboratory of Urban Environment and Health, Institute of Urban Environment, Chinese Academy of Sciences, 1799 Jimei Road, Xiamen 361021, P. R. China
| |
Collapse
|
24
|
Flibotte JJ, Jablonski AM, Kalb RG. Oxygen sensing neurons and neuropeptides regulate survival after anoxia in developing C. elegans. PLoS One 2014; 9:e101102. [PMID: 24967811 PMCID: PMC4072718 DOI: 10.1371/journal.pone.0101102] [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: 11/13/2013] [Accepted: 06/02/2014] [Indexed: 11/18/2022] Open
Abstract
Hypoxic brain injury remains a major source of neurodevelopmental impairment for both term and preterm infants. The perinatal period is a time of rapid transition in oxygen environments and developmental resetting of oxygen sensing. The relationship between neural oxygen sensing ability and hypoxic injury has not been studied. The oxygen sensing circuitry in the model organism C. elegans is well understood. We leveraged this information to investigate the effects of impairments in oxygen sensing on survival after anoxia. There was a significant survival advantage in developing worms specifically unable to sense oxygen shifts below their preferred physiologic range via genetic ablation of BAG neurons, which appear important for conferring sensitivity to anoxia. Oxygen sensing that is mediated through guanylate cyclases (gcy-31, 33, 35) is unlikely to be involved in conferring this sensitivity. Additionally, animals unable to process or elaborate neuropeptides displayed a survival advantage after anoxia. Based on these data, we hypothesized that elaboration of neuropeptides by BAG neurons sensitized animals to anoxia, but further experiments indicate that this is unlikely to be true. Instead, it seems that neuropeptides and signaling from oxygen sensing neurons operate through independent mechanisms, each conferring sensitivity to anoxia in wild type animals.
Collapse
Affiliation(s)
- John J. Flibotte
- Department of Pediatrics, Division of Neonatology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Angela M. Jablonski
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Robert G. Kalb
- Department of Pediatrics, Division of Neurology, Research Institute, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
25
|
Johnson DW, Llop JR, Farrell SF, Yuan J, Stolzenburg LR, Samuelson AV. The Caenorhabditis elegans Myc-Mondo/Mad complexes integrate diverse longevity signals. PLoS Genet 2014; 10:e1004278. [PMID: 24699255 PMCID: PMC3974684 DOI: 10.1371/journal.pgen.1004278] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 02/18/2014] [Indexed: 11/29/2022] Open
Abstract
The Myc family of transcription factors regulates a variety of biological processes, including the cell cycle, growth, proliferation, metabolism, and apoptosis. In Caenorhabditis elegans, the "Myc interaction network" consists of two opposing heterodimeric complexes with antagonistic functions in transcriptional control: the Myc-Mondo:Mlx transcriptional activation complex and the Mad:Max transcriptional repression complex. In C. elegans, Mondo, Mlx, Mad, and Max are encoded by mml-1, mxl-2, mdl-1, and mxl-1, respectively. Here we show a similar antagonistic role for the C. elegans Myc-Mondo and Mad complexes in longevity control. Loss of mml-1 or mxl-2 shortens C. elegans lifespan. In contrast, loss of mdl-1 or mxl-1 increases longevity, dependent upon MML-1:MXL-2. The MML-1:MXL-2 and MDL-1:MXL-1 complexes function in both the insulin signaling and dietary restriction pathways. Furthermore, decreased insulin-like/IGF-1 signaling (ILS) or conditions of dietary restriction increase the accumulation of MML-1, consistent with the notion that the Myc family members function as sensors of metabolic status. Additionally, we find that Myc family members are regulated by distinct mechanisms, which would allow for integrated control of gene expression from diverse signals of metabolic status. We compared putative target genes based on ChIP-sequencing data in the modENCODE project and found significant overlap in genomic DNA binding between the major effectors of ILS (DAF-16/FoxO), DR (PHA-4/FoxA), and Myc family (MDL-1/Mad/Mxd) at common target genes, which suggests that diverse signals of metabolic status converge on overlapping transcriptional programs that influence aging. Consistent with this, there is over-enrichment at these common targets for genes that function in lifespan, stress response, and carbohydrate metabolism. Additionally, we find that Myc family members are also involved in stress response and the maintenance of protein homeostasis. Collectively, these findings indicate that Myc family members integrate diverse signals of metabolic status, to coordinate overlapping metabolic and cytoprotective transcriptional programs that determine the progression of aging.
Collapse
Affiliation(s)
- David W. Johnson
- University of Rochester, Department of Biomedical Genetics, Rochester, New York, United States of America
| | - Jesse R. Llop
- University of Rochester, Department of Biomedical Genetics, Rochester, New York, United States of America
| | - Sara F. Farrell
- University of Rochester, Department of Biomedical Genetics, Rochester, New York, United States of America
| | - Jie Yuan
- Rochester Institute of Technology, Computer Science Department, Rochester, New York, United States of America
| | - Lindsay R. Stolzenburg
- Northwestern University, Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Andrew V. Samuelson
- University of Rochester, Department of Biomedical Genetics, Rochester, New York, United States of America
| |
Collapse
|
26
|
Ghose P, Park EC, Tabakin A, Salazar-Vasquez N, Rongo C. Anoxia-reoxygenation regulates mitochondrial dynamics through the hypoxia response pathway, SKN-1/Nrf, and stomatin-like protein STL-1/SLP-2. PLoS Genet 2013; 9:e1004063. [PMID: 24385935 PMCID: PMC3873275 DOI: 10.1371/journal.pgen.1004063] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 11/12/2013] [Indexed: 12/04/2022] Open
Abstract
Many aerobic organisms encounter oxygen-deprived environments and thus must have adaptive mechanisms to survive such stress. It is important to understand how mitochondria respond to oxygen deprivation given the critical role they play in using oxygen to generate cellular energy. Here we examine mitochondrial stress response in C. elegans, which adapt to extreme oxygen deprivation (anoxia, less than 0.1% oxygen) by entering into a reversible suspended animation state of locomotory arrest. We show that neuronal mitochondria undergo DRP-1-dependent fission in response to anoxia and undergo refusion upon reoxygenation. The hypoxia response pathway, including EGL-9 and HIF-1, is not required for anoxia-induced fission, but does regulate mitochondrial reconstitution during reoxygenation. Mutants for egl-9 exhibit a rapid refusion of mitochondria and a rapid behavioral recovery from suspended animation during reoxygenation; both phenotypes require HIF-1. Mitochondria are significantly larger in egl-9 mutants after reoxygenation, a phenotype similar to stress-induced mitochondria hyperfusion (SIMH). Anoxia results in mitochondrial oxidative stress, and the oxidative response factor SKN-1/Nrf is required for both rapid mitochondrial refusion and rapid behavioral recovery during reoxygenation. In response to anoxia, SKN-1 promotes the expression of the mitochondrial resident protein Stomatin-like 1 (STL-1), which helps facilitate mitochondrial dynamics following anoxia. Our results suggest the existence of a conserved anoxic stress response involving changes in mitochondrial fission and fusion. Oxygen deprivation plays a role in multiple human diseases ranging from heart attack, ischemic stroke, and traumatic injury. Aerobic organisms use oxygen to generate cellular energy in mitochondria; thus, oxygen deprivation results in energy depletion. Low oxygen can be catastrophic in tissues like the nervous system, which has high-energy demands and few glycolytic reserves. By contrast, other cells, including stem cells and cancerous cells within tumors, adapt and thrive in low oxygen. We are just beginning to understand how different organisms and even different cell types within the same organism respond to low oxygen conditions. The response of mitochondria to oxygen deprivation is particularly critical given their role in aerobic energy production. In addition, mitochondria actively injure cells during oxygen deprivation through the generation of reactive oxygen species, the disruption of calcium homeostasis, and the activation of cell death pathways. Here we use a genetic approach to show that mitochondria undergo fission during oxygen deprivation and refusion upon oxygen restoration. The hypoxia response pathway and the oxidative stress response pathway together modulate this response. We identify a new factor, stomatin-like protein, as a promoter of mitochondrial fusion in response to oxygen deprivation stress. Our findings uncover a new mechanism – regulated mitochondrial dynamics – by which cells adapt to oxygen deprivation stress.
Collapse
Affiliation(s)
- Piya Ghose
- The Waksman Institute, Department of Genetics, Rutgers The State University of New Jersey, Piscataway, New Jersey, United States of America
- The Graduate Program in Neuroscience, Rutgers The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Eun Chan Park
- The Waksman Institute, Department of Genetics, Rutgers The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Alexandra Tabakin
- The Waksman Institute, Department of Genetics, Rutgers The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Nathaly Salazar-Vasquez
- The Waksman Institute, Department of Genetics, Rutgers The State University of New Jersey, Piscataway, New Jersey, United States of America
- The Graduate Program in Genetics and Microbiology, Rutgers The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Christopher Rongo
- The Waksman Institute, Department of Genetics, Rutgers The State University of New Jersey, Piscataway, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
27
|
A cytoprotective perspective on longevity regulation. Trends Cell Biol 2013; 23:409-20. [PMID: 23726168 DOI: 10.1016/j.tcb.2013.04.007] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 04/17/2013] [Accepted: 04/18/2013] [Indexed: 02/07/2023]
Abstract
There are many mechanisms of lifespan extension, including the disruption of insulin/insulin-like growth factor 1 (IGF-1) signaling, metabolism, translation, and feeding. Despite the disparate functions of these pathways, inhibition of each induces responses that buffer stress and damage. Here, emphasizing data from genetic analyses in Caenorhabditis elegans, we explore the effectors and upstream regulatory components of numerous cytoprotective mechanisms activated as major elements of longevity programs, including detoxification, innate immunity, proteostasis, and oxidative stress response. We show that their induction underpins longevity extension across functionally diverse triggers and across species. Intertwined with the evolution of longevity, cytoprotective pathways are coupled to the surveillance of core cellular components, with important implications in normal and aberrant responses to drugs, chemicals, and pathogens.
Collapse
|
28
|
Iranon NN, Miller DL. Interactions between oxygen homeostasis, food availability, and hydrogen sulfide signaling. Front Genet 2012; 3:257. [PMID: 23233860 PMCID: PMC3516179 DOI: 10.3389/fgene.2012.00257] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 11/04/2012] [Indexed: 12/19/2022] Open
Abstract
The ability to sense and respond to stressful conditions is essential to maintain organismal homeostasis. It has long been recognized that stress response factors that improve survival in changing conditions can also influence longevity. In this review, we discuss different strategies used by animals in response to decreased O(2) (hypoxia) to maintain O(2) homeostasis, and consider interactions between hypoxia responses, nutritional status, and H(2)S signaling. O(2) is an essential environmental nutrient for almost all metazoans as it plays a fundamental role in development and cellular metabolism. However, the physiological response(s) to hypoxia depend greatly on the amount of O(2) available. Animals must sense declining O(2) availability to coordinate fundamental metabolic and signaling pathways. It is not surprising that factors involved in the response to hypoxia are also involved in responding to other key environmental signals, particularly food availability. Recent studies in mammals have also shown that the small gaseous signaling molecule hydrogen sulfide (H(2)S) protects against cellular damage and death in hypoxia. These results suggest that H(2)S signaling also integrates with hypoxia response(s). Many of the signaling pathways that mediate the effects of hypoxia, food deprivation, and H(2)S signaling have also been implicated in the control of lifespan. Understanding how these pathways are coordinated therefore has the potential to reveal new cellular and organismal homeostatic mechanisms that contribute to longevity assurance in animals.
Collapse
Affiliation(s)
- Nicole N Iranon
- Department of Biochemistry, University of Washington School of Medicine Seattle, WA, USA ; Molecular and Cellular Biology Graduate Program, University of Washington School of Medicine Seattle, WA, USA
| | | |
Collapse
|
29
|
Abstract
The nematode Caenorhabditis elegans is a model organism that has seen extensive use over the last four decades in multiple areas of investigation. In this study we explore the response of the nematode Caenorhabditis elegans to acute anoxia using gas-chromatography mass-spectrometry (GC-MS). We focus on the readily-accessible worm exometabolome to show that C. elegans are mixed acid fermenters that utilize several metabolic pathways in unconventional ways to remove reducing equivalents - including partial reversal of branched-chain amino acid catabolism and a potentially novel use of the glyoxylate pathway. In doing so, we provide detailed methods for the collection and analysis of excreted metabolites that, with minimal adjustment, should be applicable to many other species. We also describe a procedure for collecting highly volatile compounds from C. elegans. We are distributing our mass spectral library in an effort to facilitate wider use of metabolomics.
Collapse
|
30
|
Yuan Y, Kadiyala CS, Ching TT, Hakimi P, Saha S, Xu H, Yuan C, Mullangi V, Wang L, Fivenson E, Hanson RW, Ewing R, Hsu AL, Miyagi M, Feng Z. Enhanced energy metabolism contributes to the extended life span of calorie-restricted Caenorhabditis elegans. J Biol Chem 2012; 287:31414-26. [PMID: 22810224 DOI: 10.1074/jbc.m112.377275] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Caloric restriction (CR) markedly extends life span and improves the health of a broad number of species. Energy metabolism fundamentally contributes to the beneficial effects of CR, but the underlying mechanisms that are responsible for this effect remain enigmatic. A multidisciplinary approach that involves quantitative proteomics, immunochemistry, metabolic quantification, and life span analysis was used to determine how CR, which occurs in the Caenorhabditis elegans eat-2 mutants, modifies energy metabolism of the worm, and whether the observed modifications contribute to the CR-mediated physiological responses. A switch to fatty acid metabolism as an energy source and an enhanced rate of energy metabolism by eat-2 mutant nematodes were detected. Life span analyses validated the important role of these previously unknown alterations of energy metabolism in the CR-mediated longevity of nematodes. As observed in mice, the overexpression of the gene for the nematode analog of the cytosolic form of phosphoenolpyruvate carboxykinase caused a marked extension of the life span in C. elegans, presumably by enhancing energy metabolism via an altered rate of cataplerosis of tricarboxylic acid cycle anions. We conclude that an increase, not a decrease in fuel consumption, via an accelerated oxidation of fuels in the TCA cycle is involved in life span regulation; this mechanism may be conserved across phylogeny.
Collapse
Affiliation(s)
- Yiyuan Yuan
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Ischemic preconditioning: the role of mitochondria and aging. Exp Gerontol 2011; 47:1-7. [PMID: 22100642 DOI: 10.1016/j.exger.2011.11.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2011] [Revised: 10/24/2011] [Accepted: 11/02/2011] [Indexed: 11/22/2022]
Abstract
Aging represents a triple threat for myocardial infarction (MI). Not only does the incidence of MI increase with age, but the heart becomes more susceptible to MI induced damage and protective interventions such as ischemic preconditioning (IPC) become less effective. Therefore, any rational therapeutic strategy must be built around the ability to combat the detrimental effects of ischemia in aged individuals. To accomplish this, we need to develop a better understanding of how ischemic damage, protection, and aging are linked. In this regard, mitochondria have emerged as a common theme. First, mitochondria contribute to cell damage during ischemia-reperfusion (IR) and are central to cell death. Second, the protective signaling pathways activated by IPC converge on mitochondria, and the opening of mitochondrial ion channels alone is sufficient to elicit protection. Finally, mitochondria clearly influence the aging process, and specific defects in mitochondrial activity are associated with age-related functional decline. This review will summarize the effects of aging on myocardial IR injury and discuss relevant and emerging strategies to protect against MI with an emphasis on mitochondrial function.
Collapse
|
32
|
Rahman MM, Stuchlick O, El-Karim EG, Stuart R, Kipreos ET, Wells L. Intracellular protein glycosylation modulates insulin mediated lifespan in C.elegans. Aging (Albany NY) 2011; 2:678-90. [PMID: 20952811 PMCID: PMC2993798 DOI: 10.18632/aging.100208] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
O-linked-β-N-acetylglucosamine (O-GlcNAc) modification is a regulatory, nuclear and cytoplasmic post-translational glycosylation of proteins associated with age-related diseases such as Alzheimer's, Parkinson's, and type II diabetes. Global elevation of O-GlcNAc levels on intracellular proteins can induce insulin resistance, the hallmark of type II diabetes, in mammalian systems. In C. elegans, attenuation of the insulin-like signal transduction pathway increases adult lifespan of the nematode. We demonstrate that the O-GlcNAc cycling enzymes OGT and OGA, which add and remove O-GlcNAc respectively, modulate lifespan in C. elegans. Median adult lifespan is increased in an oga-1 deletion strain while median adult life span is decreased upon ogt-1 deletion. The O-GlcNAc-mediated effect on nematode lifespan is dependent on the FoxO transcription factor DAF-16. DAF-16 is a key factor in the insulin-like signal transduction pathway to regulate reproductive development, lifespan, stress tolerance, and dauer formation in C. elegans. Our data indicates that O-GlcNAc cycling selectively influences only a subset of DAF-16 mediated phenotypes, including lifespan and oxidative stress resistance. We performed an affinity purification of O-GlcNAc-modified proteins and observed that a high percentage of these proteins are regulated by insulin signaling and/or impact insulin pathway functional outcomes, suggesting that the O-GlcNAc modification may control downstream effectors to modulate insulin pathway mediated cellular processes.
Collapse
|
33
|
Hulme SE, Whitesides GM. Die Chemie und der Wurm: Caenorhabditis elegans als Plattform für das Zusammenführen von chemischer und biologischer Forschung. Angew Chem Int Ed Engl 2011. [DOI: 10.1002/ange.201005461] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
34
|
Hulme SE, Whitesides GM. Chemistry and the Worm: Caenorhabditis elegans as a Platform for Integrating Chemical and Biological Research. Angew Chem Int Ed Engl 2011; 50:4774-807. [DOI: 10.1002/anie.201005461] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Indexed: 12/15/2022]
|
35
|
Environmental and genetic preconditioning for long-term anoxia responses requires AMPK in Caenorhabditis elegans. PLoS One 2011; 6:e16790. [PMID: 21304820 PMCID: PMC3033420 DOI: 10.1371/journal.pone.0016790] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2010] [Accepted: 01/14/2011] [Indexed: 01/04/2023] Open
Abstract
Background Preconditioning environments or therapeutics, to suppress the cellular damage associated with severe oxygen deprivation, is of interest to our understanding of diseases associated with oxygen deprivation. Wildtype C. elegans exposed to anoxia enter into a state of suspended animation in which energy-requiring processes reversibly arrest. C. elegans at all developmental stages survive 24-hours of anoxia exposure however, the ability of adult hermaphrodites to survive three days of anoxia significantly decreases. Mutations in the insulin-like signaling receptor (daf-2) and LIN-12/Notch (glp-1) lead to an enhanced long-term anoxia survival phenotype. Methodology/Principal Findings In this study we show that the combined growth environment of 25°C and a diet of HT115 E. coli will precondition adult hermaphrodites to survive long-term anoxia; many of these survivors have normal movement after anoxia treatment. Animals fed the drug metformin, which induces a dietary-restriction like state in animals and activates AMPK in mammalian cell culture, have a higher survival rate when exposed to long-term anoxia. Mutations in genes encoding components of AMPK (aak-2, aakb-1, aakb-2, aakg-2) suppress the environmentally and genetically induced long-term anoxia survival phenotype. We further determine that there is a correlation between the animals that survive long-term anoxia and increased levels of carminic acid staining, which is a fluorescent dye that incorporates in with carbohydrates such as glycogen. Conclusions/Significance We conclude that small changes in growth conditions such as increased temperature and food source can influence the physiology of the animal thus affecting the responses to stress such as anoxia. Furthermore, this supports the idea that metformin should be further investigated as a therapeutic tool for treatment of oxygen-deprived tissues. Finally, the capacity for an animal to survive long bouts of severe oxygen deprivation is likely dependent on specific subunits of the heterotrimeric protein AMPK and energy stores such as carbohydrates.
Collapse
|
36
|
Regulation of anoxic death in Caenorhabditis elegans by mammalian apoptosis signal-regulating kinase (ASK) family proteins. Genetics 2011; 187:785-92. [PMID: 21212236 DOI: 10.1534/genetics.110.124883] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Cells and organisms face anoxia in a wide variety of contexts, including ischemia and hibernation. Cells respond to anoxic conditions through multiple signaling pathways. We report that NSY-1, the Caenorhabditis elegans ortholog of mammalian apoptosis signal-regulating kinase (ASK) family of MAP kinase (MAPK) kinase kinases (MAP3Ks), regulates viability of animals in anoxia. Loss-of-function mutations of nsy-1 increased survival under anoxic conditions, and increased survival was also observed in animals with mutations in tir-1 and the MAPK kinase (MAP2K) sek-1, which are upstream and downstream factors of NSY-1, respectively. Consistent with these findings, anoxia was found to activate the p38 MAPK ortholog PMK-1, and this was suppressed in nsy-1 and tir-1 mutant animals. Furthermore, double-mutant analysis showed that the insulin-signaling pathway, which also regulates viability in anoxia, functioned in parallel to NSY-1. These results suggest that the TIR-1-NSY-1-SEK-1-PMK-1 pathway plays important roles in the reponse to anoxia in C. elegans.
Collapse
|
37
|
Butler JA, Ventura N, Johnson TE, Rea SL. Long‐lived mitochondrial (Mit) mutants of Caenorhabditis elegansutilize a novel metabolism. FASEB J 2010. [DOI: 10.1096/fj.10.162941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Jeffrey A. Butler
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science Center at San Antonio San Antonio Texas USA
- Department of PhysiologyUniversity of Texas Health Science Center at San Antonio San Antonio Texas USA
- Department of Integrative PhysiologyInstitute for Behavioral Genetics, University of Colorado Boulder Colorado USA
| | - Natascia Ventura
- Department of Experimental Medicine and Biochemical SciencesUniversity of Rome Tor Vergata Rome Italy
| | - Thomas E. Johnson
- Department of Integrative PhysiologyInstitute for Behavioral Genetics, University of Colorado Boulder Colorado USA
| | - Shane L. Rea
- Barshop Institute for Longevity and Aging StudiesUniversity of Texas Health Science Center at San Antonio San Antonio Texas USA
- Department of PhysiologyUniversity of Texas Health Science Center at San Antonio San Antonio Texas USA
- Department of Integrative PhysiologyInstitute for Behavioral Genetics, University of Colorado Boulder Colorado USA
| |
Collapse
|
38
|
Abstract
The simple nematode worm Caenorhabditis elegans has been instrumental in deciphering the molecular mechanisms underlying apoptosis. Beyond apoptosis, several paradigms of non-apoptotic cell death, either genetically or extrinsically triggered, have also been described in C. elegans. Remarkably, non-apoptotic cell death in worms and pathological cell death in humans share numerous key features and mechanistic aspects. Such commonalities suggest that similarly to apoptosis, non-apoptotic cell death mechanisms are also conserved, and render the worm a useful organism, in which to model and dissect human pathologies. Indeed, the genetic malleability and the sophisticated molecular tools available for C. elegans have contributed decisively to advance our understanding of non-apoptotic cell death. Here, we review the literature on the various types of non-apoptotic cell death in C. elegans and discuss the implications, relevant to pathological conditions in humans.
Collapse
Affiliation(s)
- Manolis Vlachos
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete, Greece
| | | |
Collapse
|
39
|
Butler JA, Ventura N, Johnson TE, Rea SL. Long-lived mitochondrial (Mit) mutants of Caenorhabditis elegans utilize a novel metabolism. FASEB J 2010; 24:4977-88. [PMID: 20732954 DOI: 10.1096/fj.10-162941] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The Caenorhabditis elegans mitochondrial (Mit) mutants have disrupted mitochondrial electron transport chain (ETC) functionality, yet, surprisingly, they are long lived. We have previously proposed that Mit mutants supplement their energy needs by exploiting alternate energy production pathways normally used by wild-type animals only when exposed to hypoxic conditions. We have also proposed that longevity in the Mit mutants arises as a property of their new metabolic state. If longevity does arise as a function of metabolic state, we would expect to find a common metabolic signature among these animals. To test these predictions, we established a novel approach monitoring the C. elegans exometabolism as a surrogate marker for internal metabolic events. Using HPLC-ultraviolet-based metabolomics and multivariate analyses, we show that long-lived clk-1(qm30) and isp-1(qm150) Mit mutants have a common metabolic profile that is distinct from that of aerobically cultured wild-type animals and, unexpectedly, wild-type animals cultured under severe oxygen deprivation. Moreover, we show that 2 short-lived mitochondrial ETC mutants, mev-1(kn1) and ucr-2.3(pk732), also share a common metabolic signature that is unique. We show that removal of soluble fumarate reductase unexpectedly increases health span in several genetically defined Mit mutants, identifying at least 1 alternate energy production pathway, malate dismutation, that is operative in these animals. Our study suggests long-lived, genetically specified Mit mutants employ a novel metabolism and that life span may well arise as a function of metabolic state.
Collapse
Affiliation(s)
- Jeffrey A Butler
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | | | | | | |
Collapse
|
40
|
Powell-Coffman JA. Hypoxia signaling and resistance in C. elegans. Trends Endocrinol Metab 2010; 21:435-40. [PMID: 20335046 DOI: 10.1016/j.tem.2010.02.006] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Revised: 02/14/2010] [Accepted: 02/18/2010] [Indexed: 11/20/2022]
Abstract
In normal development and homeostasis and in many disease states, cells and tissues must overcome the challenge of oxygen deprivation (hypoxia). The nematode C. elegans is emerging as an increasingly powerful system in which to understand how animals adapt to moderate hypoxia and survive extreme hypoxic insults. This review provides an overview of C. elegans responses to hypoxia, ranging from adaptation and arrest to death, and highlights some of the recent studies that have provided important insights into hypoxia signaling and resistance. Many of the key genes and pathways are evolutionarily conserved, and C. elegans hypoxia research promises to inform our understanding of oxygen-sensitive signaling and survival in mammalian development and disease.
Collapse
Affiliation(s)
- Jo Anne Powell-Coffman
- Genetics, Development, and Cell Biology Department, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
41
|
Avery L. Caenorhabditis elegans behavioral genetics: where are the knobs? BMC Biol 2010; 8:69. [PMID: 20504291 PMCID: PMC2882361 DOI: 10.1186/1741-7007-8-69] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2010] [Accepted: 05/25/2010] [Indexed: 01/17/2023] Open
Abstract
Thousands of behavioral mutants of Caenorhabditis elegans have been studied. I suggest a set of criteria by which some genes important in the evolution of behavior might be recognized, and identify neuropeptide signaling pathways as candidates.
Collapse
Affiliation(s)
- Leon Avery
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX 75390-9148, USA.
| |
Collapse
|
42
|
Mabon ME, Scott BA, Crowder CM. Divergent mechanisms controlling hypoxic sensitivity and lifespan by the DAF-2/insulin/IGF-receptor pathway. PLoS One 2009; 4:e7937. [PMID: 19936206 PMCID: PMC2775958 DOI: 10.1371/journal.pone.0007937] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Accepted: 10/22/2009] [Indexed: 11/19/2022] Open
Abstract
Organisms and their cells vary greatly in their tolerance of low oxygen environments (hypoxia). A delineation of the determinants of hypoxia tolerance is incomplete, despite intense interest for its implications in diseases such as stroke and myocardial infarction. The insulin/IGF-1 receptor (IGFR) signaling pathway controls survival of Caenorhabditis elegans from a variety of stressors including aging, hyperthermia, and hypoxia. daf-2 encodes a C. elegans IGFR homolog whose primary signaling pathway modulates the activity of the FOXO transcription factor DAF-16. DAF-16 regulates the transcription of a large number of genes, some of which have been shown to control aging. To identify genes that selectively regulate hypoxic sensitivity, we compared the whole-organismal transcriptomes of three daf-2 reduction-of-function alleles, all of which are hypoxia resistant, thermotolerant, and long lived, but differ in their rank of severities for these phenotypes. The transcript levels of 172 genes were increased in the most hypoxia resistant daf-2 allele, e1370, relative to the other alleles whereas transcripts from only 10 genes were decreased in abundance. RNAi knockdown of 6 of the 10 genes produced a significant increase in organismal survival after hypoxic exposure as would be expected if down regulation of these genes by the e1370 mutation was responsible for hypoxia resistance. However, RNAi knockdown of these genes did not prolong lifespan. These genes definitively separate the mechanisms of hypoxic sensitivity and lifespan and identify biological strategies to survive hypoxic injury.
Collapse
Affiliation(s)
- Meghann E. Mabon
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- The Division of Biology & Biomedical Sciences, Program in Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Barbara A. Scott
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - C. Michael Crowder
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- The Division of Biology & Biomedical Sciences, Program in Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
43
|
Zhang Y, Shao Z, Zhai Z, Shen C, Powell-Coffman JA. The HIF-1 hypoxia-inducible factor modulates lifespan in C. elegans. PLoS One 2009; 4:e6348. [PMID: 19633713 PMCID: PMC2711329 DOI: 10.1371/journal.pone.0006348] [Citation(s) in RCA: 142] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2009] [Accepted: 06/17/2009] [Indexed: 12/26/2022] Open
Abstract
During normal development or during disease, animal cells experience hypoxic (low oxygen) conditions, and the hypoxia-inducible factor (HIF) transcription factors implement most of the critical changes in gene expression that enable animals to adapt to this stress. Here, we examine the roles of HIF-1 in post-mitotic aging. We examined the effects of HIF-1 over-expression and of hif-1 loss-of-function mutations on longevity in C. elegans, a powerful genetic system in which adult somatic cells are post-mitotic. We constructed transgenic lines that expressed varying levels of HIF-1 protein and discovered a positive correlation between HIF-1 expression levels and lifespan. The data further showed that HIF-1 acted in parallel to the SKN-1/NRF and DAF-16/FOXO transcription factors to promote longevity. HIF-1 over-expression also conferred increased resistance to heat and oxidative stress. We isolated and characterized additional hif-1 mutations, and we found that each of 3 loss-of-function mutations conferred increased longevity in normal lab culture conditions, but, unlike HIF-1 over-expression, a hif-1 deletion mutation did not extend the lifespan of daf-16 or skn-1 mutants. We conclude that HIF-1 over-expression and hif-1 loss-of-function mutations promote longevity by different pathways. These data establish HIF-1 as one of the key stress-responsive transcription factors that modulate longevity in C. elegans and advance our understanding of the regulatory networks that link oxygen homeostasis and aging.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, Iowa, United States of America
| | - Zhiyong Shao
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, Iowa, United States of America
| | - Zhiwei Zhai
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, Iowa, United States of America
| | - Chuan Shen
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, Iowa, United States of America
| | - Jo Anne Powell-Coffman
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, Iowa, United States of America
- * E-mail:
| |
Collapse
|
44
|
Menuz V, Howell KS, Gentina S, Epstein S, Riezman I, Fornallaz-Mulhauser M, Hengartner MO, Gomez M, Riezman H, Martinou JC. Protection of C. elegans from anoxia by HYL-2 ceramide synthase. Science 2009; 324:381-4. [PMID: 19372430 DOI: 10.1126/science.1168532] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Oxygen deprivation is rapidly deleterious for most organisms. However, Caenorhabditis elegans has developed the ability to survive anoxia for at least 48 hours. Mutations in the DAF-2/DAF-16 insulin-like signaling pathway promote such survival. We describe a pathway involving the HYL-2 ceramide synthase that acts independently of DAF-2. Loss of the ceramide synthase gene hyl-2 results in increased sensitivity of C. elegans to anoxia. C. elegans has two ceramide synthases, hyl-1 and hyl-2, that participate in ceramide biogenesis and affect its ability to survive anoxic conditions. In contrast to hyl-2(lf) mutants, hyl-1(lf) mutants are more resistant to anoxia than normal animals. HYL-1 and HYL-2 have complementary specificities for fatty acyl chains. These data indicate that specific ceramides produced by HYL-2 confer resistance to anoxia.
Collapse
Affiliation(s)
- Vincent Menuz
- Department of Cell Biology, University of Geneva, CH-1211 Geneva 4, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Mendenhall AR, LeBlanc MG, Mohan DP, Padilla PA. Reduction in ovulation or male sex phenotype increases long-term anoxia survival in a daf-16-independent manner in Caenorhabditis elegans. Physiol Genomics 2008; 36:167-78. [PMID: 19050081 DOI: 10.1152/physiolgenomics.90278.2008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Identifying genotypes and phenotypes that enhance an organism's ability to survive stress is of interest. We used Caenorhabditis elegans mutants, RNA interference (RNAi), and the chemical 5-fluorodeoxyuridine (FUDR) to test the hypothesis that a reduction in progeny would increase oxygen deprivation (anoxia) survival. In the hermaphrodite gonad, germ line processes such as spermatogenesis and oogenesis can be simultaneously as well as independently disrupted by genetic mutations. We analyzed genetic mutants [glp-1(q158), glp-4(bn2ts), plc-1(rx1), ksr-1(ku68), fog-2(q71), fem-3(q20), spe-9(hc52ts), fer-15(hc15ts)] with reduced progeny production due to various reproductive defects. Furthermore, we used RNAi to inhibit the function of gene products in the RTK/Ras/MAPK signaling pathway, which is known to be involved in a variety of developmental processes including gonad function. We determined that reduced progeny production or complete sterility enhanced anoxia survival except in the case of sterile hermaphrodites [spe-9(hc52ts), fer-15(hc15ts)] undergoing oocyte maturation and ovulation as exhibited by the presence of laid unfertilized oocytes. Furthermore, the fog-2(q71) long-term anoxia survival phenotype was suppressed when oocyte maturation and ovulation were induced by mating with males that have functional or nonfunctional sperm. The mutants with a reduced progeny production survive long-term anoxia in a daf-16- and hif-1-independent manner. Finally, we determined that wild-type males were able to survive long-term anoxia in a daf-16-independent manner. Together, these results suggest that the insulin signaling pathway is not the only mechanism to survive oxygen deprivation and that altering gonad function, in particular oocyte maturation and ovulation, leads to a physiological state conducive for oxygen deprivation survival.
Collapse
|
46
|
Abstract
The sensitivity of an organism to hypoxic injury varies widely across species and among cell types. However, a systematic description of the determinants of metazoan hypoxic sensitivity is lacking. Toward this end, we screened a whole-genome RNAi library for genes that promote hypoxic sensitivity in Caenorhabditis elegans. RNAi knockdown of 198 genes conferred an invariant hypoxia-resistant phenotype (Hyp-r). Eighty-six per cent of these hyp genes had strong homologs in other organisms, 73 with human reciprocal orthologs. The hyp genes were distributed among multiple functional categories. Transcription factors, chromatin modifying enzymes, and intracellular signaling proteins were highly represented. RNAi knockdown of about half of the genes produced no apparent deleterious phenotypes. The hyp genes had significant overlap with previously identified life span extending genes. Testing of the RNAi's in a mutant background defective in somatic RNAi machinery showed that most genes function in somatic cells to control hypoxic sensitivity. DNA microarray analysis identified a subset of the hyp genes that may be hypoxia regulated. siRNA knockdown of human orthologs of the hyp genes conferred hypoxia resistance to transformed human cells for 40% of the genes tested, indicating extensive evolutionary conservation of the hypoxic regulatory activities. The results of the screen provide the first systematic picture of the genetic determinants of hypoxic sensitivity. The number and diversity of genes indicates a surprisingly nonredundant genetic network promoting hypoxic sensitivity.
Collapse
|
47
|
Samokhvalov V, Scott BA, Crowder CM. Autophagy protects against hypoxic injury in C. elegans. Autophagy 2008; 4:1034-41. [PMID: 18849662 DOI: 10.4161/auto.6994] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Macroautophagy has been implicated in a variety of pathological processes. Hypoxic/ischemic cellular injury is one such process in which autophagy has emerged as an important regulator. In general, autophagy is induced after a hypoxic/ischemic insult; however, whether the induction of autophagy promotes cell death or recovery is controversial and appears to be context dependent. We have developed C. elegans as a genetically tractable model for the study of hypoxic cell injury. Both necrosis and apoptosis are mechanisms of cell death following hypoxia in C. elegans. However, the role of autophagy in hypoxic injury in C. elegans has not been examined. Here, we found that RNAi knockdown of the C. elegans homologs of beclin 1/Atg6 (bec-1) and LC3/Atg8 (lgg-1, lgg-2), and mutation of Atg1 (unc-51) decreased animal survival after a severe hypoxic insult. Acute inhibition of autophagy by the type III phosphatidylinositol 3-kinase inhibitors, 3-methyladenine and Wortmannin, also sensitized animals to hypoxic death. Hypoxia-induced neuronal and myocyte injury as well as necrotic cellular morphology were increased by RNAi knockdown of BEC-1. Hypoxia increased the expression of a marker of autophagosomes in a bec-1-dependent manner. Finally, we found that the hypoxia hypersensitive phenotype of bec-1(RNAi) animals could be blocked by loss-of-function mutations in either the apoptosis or necrosis pathway. These results argue that inhibition of autophagy sensitizes C. elegans and its cells to hypoxic injury and that this sensitization is blocked or circumvented when either of the two major cell-death mechanisms is inhibited.
Collapse
Affiliation(s)
- Victor Samokhvalov
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110-1010, USA
| | | | | |
Collapse
|
48
|
Hajeri VA, Stewart AM, Moore LL, Padilla PA. Genetic analysis of the spindle checkpoint genes san-1, mdf-2, bub-3 and the CENP-F homologues hcp-1 and hcp-2 in Caenorhabditis elegans. Cell Div 2008; 3:6. [PMID: 18248670 PMCID: PMC2265278 DOI: 10.1186/1747-1028-3-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2007] [Accepted: 02/04/2008] [Indexed: 11/18/2022] Open
Abstract
Background The spindle checkpoint delays the onset of anaphase until all sister chromatids are aligned properly at the metaphase plate. To investigate the role san-1, the MAD3 homologue, has in Caenorhabditis elegans embryos we used RNA interference (RNAi) to identify genes synthetic lethal with the viable san-1(ok1580) deletion mutant. Results The san-1(ok1580) animal has low penetrating phenotypes including an increased incidence of males, larvae arrest, slow growth, protruding vulva, and defects in vulva morphogenesis. We found that the viability of san-1(ok1580) embryos is significantly reduced when HCP-1 (CENP-F homologue), MDF-1 (MAD-1 homologue), MDF-2 (MAD-2 homologue) or BUB-3 (predicted BUB-3 homologue) are reduced by RNAi. Interestingly, the viability of san-1(ok1580) embryos is not significantly reduced when the paralog of HCP-1, HCP-2, is reduced. The phenotype of san-1(ok1580);hcp-1(RNAi) embryos includes embryonic and larval lethality, abnormal organ development, and an increase in abnormal chromosome segregation (aberrant mitotic nuclei, anaphase bridging). Several of the san-1(ok1580);hcp-1(RNAi) animals displayed abnormal kinetochore (detected by MPM-2) and microtubule structure. The survival of mdf-2(RNAi);hcp-1(RNAi) embryos but not bub-3(RNAi);hcp-1(RNAi) embryos was also compromised. Finally, we found that san-1(ok1580) and bub-3(RNAi), but not hcp-1(RNAi) embryos, were sensitive to anoxia, suggesting that like SAN-1, BUB-3 has a functional role as a spindle checkpoint protein. Conclusion Together, these data suggest that in the C. elegans embryo, HCP-1 interacts with a subset of the spindle checkpoint pathway. Furthermore, the fact that san-1(ok1580);hcp-1(RNAi) animals had a severe viability defect whereas in the san-1(ok1580);hcp-2(RNAi) and san-1(ok1580);hcp-2(ok1757) animals the viability defect was not as severe suggesting that hcp-1 and hcp-2 are not completely redundant.
Collapse
Affiliation(s)
- Vinita A Hajeri
- Department of Biological Sciences, University of North Texas, Denton, TX, USA.
| | | | | | | |
Collapse
|
49
|
Hoogewijs D, Houthoofd K, Matthijssens F, Vandesompele J, Vanfleteren JR. Selection and validation of a set of reliable reference genes for quantitative sod gene expression analysis in C. elegans. BMC Mol Biol 2008; 9:9. [PMID: 18211699 PMCID: PMC2254638 DOI: 10.1186/1471-2199-9-9] [Citation(s) in RCA: 292] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2007] [Accepted: 01/22/2008] [Indexed: 11/10/2022] Open
Abstract
Background In the nematode Caenorhabditis elegans the conserved Ins/IGF-1 signaling pathway regulates many biological processes including life span, stress response, dauer diapause and metabolism. Detection of differentially expressed genes may contribute to a better understanding of the mechanism by which the Ins/IGF-1 signaling pathway regulates these processes. Appropriate normalization is an essential prerequisite for obtaining accurate and reproducible quantification of gene expression levels. The aim of this study was to establish a reliable set of reference genes for gene expression analysis in C. elegans. Results Real-time quantitative PCR was used to evaluate the expression stability of 12 candidate reference genes (act-1, ama-1, cdc-42, csq-1, eif-3.C, mdh-1, gpd-2, pmp-3, tba-1, Y45F10D.4, rgs-6 and unc-16) in wild-type, three Ins/IGF-1 pathway mutants, dauers and L3 stage larvae. After geNorm analysis, cdc-42, pmp-3 and Y45F10D.4 showed the most stable expression pattern and were used to normalize 5 sod expression levels. Significant differences in mRNA levels were observed for sod-1 and sod-3 in daf-2 relative to wild-type animals, whereas in dauers sod-1, sod-3, sod-4 and sod-5 are differentially expressed relative to third stage larvae. Conclusion Our findings emphasize the importance of accurate normalization using stably expressed reference genes. The methodology used in this study is generally applicable to reliably quantify gene expression levels in the nematode C. elegans using quantitative PCR.
Collapse
Affiliation(s)
- David Hoogewijs
- Department of Biology and Center for Molecular Phylogeny and Evolution, Ghent University, B-9000 Ghent, Belgium.
| | | | | | | | | |
Collapse
|
50
|
Dasgupta N, Patel AM, Scott BA, Crowder CM. Hypoxic preconditioning requires the apoptosis protein CED-4 in C. elegans. Curr Biol 2007; 17:1954-9. [PMID: 17980592 DOI: 10.1016/j.cub.2007.10.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2007] [Revised: 09/27/2007] [Accepted: 10/01/2007] [Indexed: 10/22/2022]
Abstract
Hypoxic preconditioning (HP) is a rapid and reversible proadaptive response to mild hypoxic exposure with such a response protecting cells from subsequent hypoxic or ischemic insult. HP mechanisms are of great interest because of their therapeutic potential and insight into metabolic adaptation and cell death. HP has been widely demonstrated in the vertebrate subphylum but not in invertebrates. Here, we report that the nematode Caenorhabditis elegans has a potent HP mechanism that protects the organism as well as its neurons and myocytes from hypoxic injury. The time course of C. elegans HP was consistent with vertebrate-delayed HP, appearing 16 hr after preconditioning and lasting at least 36 hr. The apoptosis pathway has been proposed as either a trigger or target of HP. Testing of mutations in the canonical C. elegans apoptosis pathway showed that in general, genes in this pathway are not required for HP. However, loss-of-function mutations in ced-4, which encodes an Apaf-1 homolog, completely blocked HP. RNAi silencing of ced-4 in adult animals immediately preceding preconditioning blocked HP, indicating that CED-4 is required in adults during or after preconditioning. CED-4/Apaf-1 is essential for HP in C. elegans and acts through a mechanism independent of the classical apoptosis pathway.
Collapse
Affiliation(s)
- Nupur Dasgupta
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | | | |
Collapse
|