1
|
Awazu T, Sakamoto K, Minagi Y, Ohnishi M, Bito T, Matsunaga Y, Iwasaki T, Kawano T. The small GTPase RAB-18 is involved in regulating development/diapause by recruiting the intestinal cholesterol transporter NCR-1 onto the apical side in Caenorhabditis elegans. Biochem Biophys Res Commun 2024; 734:150609. [PMID: 39232459 DOI: 10.1016/j.bbrc.2024.150609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/06/2024]
Abstract
RAB family proteins, which are small GTPases, are integral to the process of eukaryotic membrane trafficking. In the nematode, Caenorhabditis elegans, 31 RAB proteins have been identified through genome sequencing. Using an RNAi screen specifically targeting C. elegans rab genes, we identified multiple genes that are involved in the regulation of larval development, in particular, the rab-18 gene. Our molecular genetic studies resulted in several findings. First, RAB-18 predominantly functions in the intestine to regulate larval development by modulating steroid hormone signaling. Second, the C. elegans cholesterol transporter NCR-1 is a target of RAB-18 in the intestine. Third, the membrane trafficking of NCR-1 to the apical side in intestinal cells is particularly influenced by RAB-18. Finally, RAB-18 and NCR-1 possibly co-localize on membrane vesicles. Our study is the first to demonstrate the relationship between a RAB protein and a cholesterol transporter, in which the RAB protein probably drives the transporter to the apical membrane in the intestine to regulate cholesterol uptake. This study provides insight into the molecular mechanisms underlying human disease stemming from a transport defect of cholesterol and its derivative.
Collapse
Affiliation(s)
- Toshikuni Awazu
- Department of Bioscience, Biotechnology, and Agrochemistry, Faculty of Agriculture, Tottori University, Tottori, Japan
| | - Kanato Sakamoto
- Department of Agricultural Science, Graduate School of Sustainability Science, Tottori University, Tottori, Japan
| | - Yuka Minagi
- Department of Agricultural Science, Graduate School of Sustainability Science, Tottori University, Tottori, Japan
| | - Masumi Ohnishi
- Department of Agricultural Science, Graduate School of Sustainability Science, Tottori University, Tottori, Japan
| | - Tomohiro Bito
- Department of Bioscience, Biotechnology, and Agrochemistry, Faculty of Agriculture, Tottori University, Tottori, Japan; Department of Agricultural Science, Graduate School of Sustainability Science, Tottori University, Tottori, Japan
| | | | - Takashi Iwasaki
- Department of Bioscience, Biotechnology, and Agrochemistry, Faculty of Agriculture, Tottori University, Tottori, Japan; Department of Agricultural Science, Graduate School of Sustainability Science, Tottori University, Tottori, Japan
| | - Tsuyoshi Kawano
- Department of Bioscience, Biotechnology, and Agrochemistry, Faculty of Agriculture, Tottori University, Tottori, Japan; Department of Agricultural Science, Graduate School of Sustainability Science, Tottori University, Tottori, Japan.
| |
Collapse
|
2
|
Li R, Li H, Wang X, Peng Y. Protective Effects of Velvet Antler Methanol Extracts on Hypoxia-Induced Damage in Caenorhabditis elegans through HIF-1 and ECH-8 Mediated Lipid Accumulation. Nutrients 2024; 16:2257. [PMID: 39064700 PMCID: PMC11280314 DOI: 10.3390/nu16142257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/06/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Velvet antler, a traditional tonic widely used in East Asia for its health benefits, is explored in this study for its protective effects against hypoxia-induced damage using Caenorhabditis elegans (C. elegans) as a model. Hypoxia, characterized by low oxygen availability, induces significant physiological stress and potential tissue damage. Our research demonstrates that methanol extracts from velvet antler (MEs) enhance the survival of C. elegans under hypoxic conditions. This enhancement is achieved through the stabilization of hypoxia-inducible factor-1 (HIF-1) and the promotion of lipid accumulation, both of which are crucial for mitigating cellular damage. Specifically, MEs improve mitochondrial function, increase ATP production, and aid in the recovery of physical activity in C. elegans post-hypoxia or following hypoxia-reoxygenation (HR). The pivotal role of HIF-1 is underscored by the loss of these protective effects when HIF-1 function is inhibited. Additionally, our findings reveal that the gene related to lipid metabolism, ech-8, significantly contributes to the lipid accumulation that enhances resilience to hypoxia in C. elegans treated with MEs. These results not only highlight the therapeutic potential of velvet antler in modern medical applications, particularly for conditions involving hypoxic stress, but also provide insights into the molecular mechanisms by which MEs confer protection against hypoxic damage.
Collapse
Affiliation(s)
- Ru Li
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China;
| | - Hongyuan Li
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China;
| | - Xiaohui Wang
- Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China;
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Yinghua Peng
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun 130112, China;
| |
Collapse
|
3
|
Torzone SK, Breen PC, Cohen NR, Simmons KN, Dowen RH. The TWK-26 potassium channel governs nutrient absorption in the C. elegans intestine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.06.592787. [PMID: 38766028 PMCID: PMC11100751 DOI: 10.1101/2024.05.06.592787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Ion channels are necessary for proper water and nutrient absorption in the intestine, which supports cellular metabolism and organismal growth. While a role for Na + co-transporters and pumps in intestinal nutrient absorption is well defined, how individual K + uniporters function to maintain ion homeostasis is poorly understood. Using Caenorhabditis elegans , we show that a gain-of-function mutation in twk-26 , which encodes a two-pore domain K + ion channel orthologous to human KCNK3, facilitates nutrient absorption and suppresses the metabolic and developmental defects displayed by impaired intestinal MAP Kinase (MAPK) signaling. Mutations in drl-1 and flr-4, which encode two components of this MAPK pathway, cause severe growth defects, reduced lipid storage, and a dramatic increase in autophagic lysosomes, which mirror dietary restriction phenotypes. Additionally, these MAPK mutants display structural defects of the intestine and an impaired defecation motor program. We find that activation of TWK-26 reverses the dietary restriction-like state of the MAPK mutants by restoring intestinal nutrient absorption without correcting the intestinal bloating or defecation defects. This study provides unique insight into the mechanisms by which intestinal K + ion channels support intestinal metabolic homeostasis.
Collapse
|
4
|
Lactobacillus pentosus MJM60383 Inhibits Lipid Accumulation in Caenorhabditis elegans Induced by Enterobacter cloacae and Glucose. Int J Mol Sci 2022; 24:ijms24010280. [PMID: 36613723 PMCID: PMC9820548 DOI: 10.3390/ijms24010280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/14/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Gut microbiota are known to play an important role in obesity. Enterobacter cloacae, a Gram-negative bacterium, has been considered a pathogenic bacterium related to obesity in the gut. In this study, we established an obesity model of C. elegans by feeding E. cloacae combined with a high glucose diet (HGD), which significantly induced lipid accumulation. An anti-lipid mechanism study revealed that the fatty acid composition and the expression level of fat metabolism-related genes were altered by feeding E. cloacae to C. elegans under HGD conditions. Lactic acid bacteria that showed antagonistic activity against E. cloacae were used to screen anti-obesity candidates in this model. Among them, L. pentosus MJM60383 (MJM60383) showed good antagonistic activity. C. eleans fed with MJM60383 significantly reduced lipid accumulation and triglyceride content. The ratio of C18:1Δ9/C18:0 was also changed in C. elegans by feeding MJM60383. In addition, the expression level of genes related to fatty acid synthesis was significantly decreased and the genes related to fatty acid β-oxidation were up-regulated by feeding MJM60383. Moreover, MJM60383 also exhibited a high adhesive ability to Caco-2 cells and colonized the gut of C. elegans. Thus, L. pentosus MJM60383 can be a promising candidate for anti-obesity probiotics. To the best of our knowledge, this is the first report that uses E. cloacae combined with a high-glucose diet to study the interactions between individual pathogens and probiotics in C. elegans.
Collapse
|
5
|
Schifano E, Conta G, Preziosi A, Ferrante C, Batignani G, Mancini P, Tomassini A, Sciubba F, Scopigno T, Uccelletti D, Miccheli A. 2-hydroxyisobutyric acid (2-HIBA) modulates ageing and fat deposition in Caenorhabditis elegans. Front Mol Biosci 2022; 9:986022. [PMID: 36533081 PMCID: PMC9749906 DOI: 10.3389/fmolb.2022.986022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 11/07/2022] [Indexed: 06/30/2024] Open
Abstract
High levels of 2-hydroxyisobutyric acid (2-HIBA) were found in urines of patients with obesity and hepatic steatosis, suggesting a potential involvement of this metabolite in clinical conditions. The gut microbial origin of 2-HIBA was hypothesized, however its actual origin and role in biological processes are still not clear. We investigated how treatment with 2-HIBA affected the physiology of the model organism Caenorhabditis elegans, in both standard and high-glucose diet (HGD) growth conditions, by targeted transcriptomic and metabolomic analyses, Coherent Anti-Stokes Raman Scattering (CARS) and two-photon fluorescence microscopy. In standard conditions, 2-HIBA resulted particularly effective to extend the lifespan, delay ageing processes and stimulate the oxidative stress resistance in wild type nematodes through the activation of insulin/IGF-1 signaling (IIS) and p38 MAPK pathways and, consequently, through a reduction of ROS levels. Moreover, variations of lipid accumulation observed in treated worms correlated with transcriptional levels of fatty acid synthesis genes and with the involvement of peptide transporter PEP-2. In HGD conditions, the effect of 2-HIBA on C. elegans resulted in a reduction of the lipid droplets deposition, accordingly with an increase of acs-2 gene transcription, involved in β-oxidation processes. In addition, the pro-longevity effect appeared to be correlated to higher levels of tryptophan, which may play a role in restoring the decreased viability observed in the HGD untreated nematodes.
Collapse
Affiliation(s)
- Emily Schifano
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Giorgia Conta
- Department of Environmental Biology, Sapienza University of Rome, Rome, Italy
- NMR-based Metabolomics Laboratory of Sapienza (NMLab), Sapienza University of Rome, Rome, Italy
| | - Adele Preziosi
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Carino Ferrante
- Department of Physics, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- and Neuro-science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Giovanni Batignani
- Department of Physics, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- and Neuro-science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Patrizia Mancini
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Alberta Tomassini
- Department of Environmental Biology, Sapienza University of Rome, Rome, Italy
- NMR-based Metabolomics Laboratory of Sapienza (NMLab), Sapienza University of Rome, Rome, Italy
| | - Fabio Sciubba
- Department of Environmental Biology, Sapienza University of Rome, Rome, Italy
- NMR-based Metabolomics Laboratory of Sapienza (NMLab), Sapienza University of Rome, Rome, Italy
| | - Tullio Scopigno
- Department of Physics, Sapienza University of Rome, Rome, Italy
- Center for Life Nano- and Neuro-science, Istituto Italiano di Tecnologia, Rome, Italy
| | - Daniela Uccelletti
- Department of Biology and Biotechnology “C. Darwin”, Sapienza University of Rome, Rome, Italy
| | - Alfredo Miccheli
- Department of Environmental Biology, Sapienza University of Rome, Rome, Italy
- NMR-based Metabolomics Laboratory of Sapienza (NMLab), Sapienza University of Rome, Rome, Italy
| |
Collapse
|
6
|
Chauve L, Hodge F, Murdoch S, Masoudzadeh F, Mann HJ, Lopez-Clavijo AF, Okkenhaug H, West G, Sousa BC, Segonds-Pichon A, Li C, Wingett SW, Kienberger H, Kleigrewe K, de Bono M, Wakelam MJO, Casanueva O. Neuronal HSF-1 coordinates the propagation of fat desaturation across tissues to enable adaptation to high temperatures in C. elegans. PLoS Biol 2021; 19:e3001431. [PMID: 34723964 PMCID: PMC8585009 DOI: 10.1371/journal.pbio.3001431] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 11/11/2021] [Accepted: 09/29/2021] [Indexed: 11/18/2022] Open
Abstract
To survive elevated temperatures, ectotherms adjust the fluidity of membranes by fine-tuning lipid desaturation levels in a process previously described to be cell autonomous. We have discovered that, in Caenorhabditis elegans, neuronal heat shock factor 1 (HSF-1), the conserved master regulator of the heat shock response (HSR), causes extensive fat remodeling in peripheral tissues. These changes include a decrease in fat desaturase and acid lipase expression in the intestine and a global shift in the saturation levels of plasma membrane's phospholipids. The observed remodeling of plasma membrane is in line with ectothermic adaptive responses and gives worms a cumulative advantage to warm temperatures. We have determined that at least 6 TAX-2/TAX-4 cyclic guanosine monophosphate (cGMP) gated channel expressing sensory neurons, and transforming growth factor ß (TGF-β)/bone morphogenetic protein (BMP) are required for signaling across tissues to modulate fat desaturation. We also find neuronal hsf-1 is not only sufficient but also partially necessary to control the fat remodeling response and for survival at warm temperatures. This is the first study to show that a thermostat-based mechanism can cell nonautonomously coordinate membrane saturation and composition across tissues in a multicellular animal.
Collapse
Affiliation(s)
- Laetitia Chauve
- Epigenetics Department, Babraham Institute, Cambridge, United Kingdom
| | - Francesca Hodge
- Epigenetics Department, Babraham Institute, Cambridge, United Kingdom
| | - Sharlene Murdoch
- Epigenetics Department, Babraham Institute, Cambridge, United Kingdom
| | | | | | | | | | - Greg West
- Babraham Institute, Cambridge, United Kingdom
| | | | | | - Cheryl Li
- Epigenetics Department, Babraham Institute, Cambridge, United Kingdom
| | | | | | - Karin Kleigrewe
- Bavarian Centre for Biomolecular Mass Spectrometry, Freising, Germany
| | - Mario de Bono
- Institute of Science and Technology, Klosterneuburg, Austria
| | | | - Olivia Casanueva
- Epigenetics Department, Babraham Institute, Cambridge, United Kingdom
| |
Collapse
|
7
|
Zhang J, Hu Y, Wang Y, Fu L, Xu X, Li C, Xu J, Li C, Zhang L, Yang R, Jiang X, Wu Y, Liu P, Zou X, Liang B. mmBCFA C17iso ensures endoplasmic reticulum integrity for lipid droplet growth. J Cell Biol 2021; 220:212690. [PMID: 34623380 PMCID: PMC8563294 DOI: 10.1083/jcb.202102122] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 08/22/2021] [Accepted: 09/21/2021] [Indexed: 12/29/2022] Open
Abstract
In eukaryote cells, lipid droplets (LDs) are key intracellular organelles that dynamically regulate cellular energy homeostasis. LDs originate from the ER and continuously contact the ER during their growth. How the ER affects LD growth is largely unknown. Here, we show that RNAi knockdown of acs-1, encoding an acyl-CoA synthetase required for the biosynthesis of monomethyl branched-chain fatty acids C15iso and C17iso, remarkably prevented LD growth in Caenorhabditis elegans. Dietary C17iso, or complex lipids with C17iso including phosphatidylcholine, phosphatidylethanolamine, and triacylglycerol, could fully restore the LD growth in the acs-1RNAi worms. Mechanistically, C17iso may incorporate into phospholipids to ensure the membrane integrity of the ER so as to maintain the function of ER-resident enzymes such as SCD/stearoyl-CoA desaturase and DGAT2/diacylglycerol acyltransferase for appropriate lipid synthesis and LD growth. Collectively, our work uncovers a unique fatty acid, C17iso, as the side chain of phospholipids for determining the ER homeostasis for LD growth in an intact organism, C. elegans.
Collapse
Affiliation(s)
- Jingjing Zhang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Ying Hu
- School of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Yanli Wang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Lin Fu
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Xiumei Xu
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China.,Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, China
| | - Chunxia Li
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China.,Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jie Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Chengbin Li
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Linqiang Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Rendan Yang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Xue Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yingjie Wu
- Shandong Provincial Hospital, Shandong Laboratory Animal Center, Shandong First Medical University and Shandong Academy of Medical Sciences. Jinan, Shandong, China
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xiaoju Zou
- School of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Bin Liang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| |
Collapse
|
8
|
Cooper JF, Guasp RJ, Arnold ML, Grant BD, Driscoll M. Stress increases in exopher-mediated neuronal extrusion require lipid biosynthesis, FGF, and EGF RAS/MAPK signaling. Proc Natl Acad Sci U S A 2021; 118:e2101410118. [PMID: 34475208 PMCID: PMC8433523 DOI: 10.1073/pnas.2101410118] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 06/23/2021] [Indexed: 01/08/2023] Open
Abstract
In human neurodegenerative diseases, neurons can transfer toxic protein aggregates to surrounding cells, promoting pathology via poorly understood mechanisms. In Caenorhabditis elegans, proteostressed neurons can expel neurotoxic proteins in large, membrane-bound vesicles called exophers. We investigated how specific stresses impact neuronal trash expulsion to show that neuronal exopher production can be markedly elevated by oxidative and osmotic stress. Unexpectedly, we also found that fasting dramatically increases exophergenesis. Mechanistic dissection focused on identifying nonautonomous factors that sense and activate the fasting-induced exopher response revealed that DAF16/FOXO-dependent and -independent processes are engaged. Fasting-induced exopher elevation requires the intestinal peptide transporter PEPT-1, lipid synthesis transcription factors Mediator complex MDT-15 and SBP-1/SREPB1, and fatty acid synthase FASN-1, implicating remotely initiated lipid signaling in neuronal trash elimination. A conserved fibroblast growth factor (FGF)/RAS/MAPK signaling pathway that acts downstream of, or in parallel to, lipid signaling also promotes fasting-induced neuronal exopher elevation. A germline-based epidermal growth factor (EGF) signal that acts through neurons is also required for exopher production. Our data define a nonautonomous network that links food availability changes to remote, and extreme, neuronal homeostasis responses relevant to aggregate transfer biology.
Collapse
Affiliation(s)
- Jason F Cooper
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854
| | - Ryan J Guasp
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854
| | - Meghan Lee Arnold
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854
| | - Barth D Grant
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854
- Rutgers Center for Lipid Research, Rutgers University, New Brunswick, NJ 08901
| | - Monica Driscoll
- Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854;
| |
Collapse
|
9
|
Dysarz J, Fuellen G, Möller S, Luyten W, Schmitz-Linneweber C, Saul N. Genes implicated in Caenorhabditis elegans and human health regulate stress resistance and physical abilities in aged Caenorhabditis elegans. Biol Lett 2021; 17:20200916. [PMID: 34102068 PMCID: PMC8187008 DOI: 10.1098/rsbl.2020.0916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 05/17/2021] [Indexed: 12/26/2022] Open
Abstract
Recently, nine Caenorhabditis elegans genes, grouped into two pathways/clusters, were found to be implicated in healthspan in C. elegans and their homologues in humans, based on literature curation, WormBase data mining and bioinformatics analyses. Here, we further validated these genes experimentally in C. elegans. We downregulated the nine genes via RNA interference (RNAi), and their effects on physical function (locomotion in a swim assay) and on physiological function (survival after heat stress) were analysed in aged nematodes. Swim performance was negatively affected by the downregulation of acox-1.1, pept-1, pak-2, gsk-3 and C25G6.3 in worms with advanced age (twelfth day of adulthood) and heat stress resistance was decreased by RNAi targeting of acox-1.1, daf-22, cat-4, pig-1, pak-2, gsk-3 and C25G6.3 in moderately (seventh day of adulthood) or advanced aged nematodes. Only one gene, sad-1, could not be linked to a health-related function in C. elegans with the bioassays we selected. Thus, most of the healthspan genes could be re-confirmed by health measurements in old worms.
Collapse
Affiliation(s)
- Jessica Dysarz
- Molecular Genetics Group, Institute of Biology, Humboldt University of Berlin, Berlin 10115, Germany
| | - Georg Fuellen
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock 18057, Germany
| | - Steffen Möller
- Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock University Medical Center, Rostock 18057, Germany
| | - Walter Luyten
- Animal Physiology and Neurobiology, Department of Biology, KU Leuven, Leuven 3000, Belgium
| | | | - Nadine Saul
- Molecular Genetics Group, Institute of Biology, Humboldt University of Berlin, Berlin 10115, Germany
| |
Collapse
|
10
|
Zhou JJ, Chun L, Liu JF. A Comprehensive Understanding of Dietary Effects on C. elegans Physiology. Curr Med Sci 2019; 39:679-684. [PMID: 31612382 DOI: 10.1007/s11596-019-2091-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/14/2019] [Indexed: 02/07/2023]
Abstract
Diet has been shown to play an important role in human physiology. It is a predominant exogenous factor regulating the composition of gut microbiota, and dietary intervention holds promise for treatment of diseases such as obesity, type 2 diabetes, and malnutrition. Furthermore, it was reported that diet has significant effects on physiological processes of C. elegans, including reproduction, fat storage, and aging. To reveal novel signaling pathways responsive to different diets, C. elegans and its bacterial diet were used as an interspecies model system to mimic the interaction between host and gut microbiota. Most signaling pathways identified in C. elegans are highly conserved across different species, including humans. A better understanding of these pathways can, therefore, help to develop interventions for human diseases. In this article, we summarize recent achievements on molecular mechanisms underlying the response of C. elegans to different diets and discuss their relevance to human health.
Collapse
Affiliation(s)
- Jie-Jun Zhou
- Collaborative Innovation Center for Brain Science, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Lei Chun
- Collaborative Innovation Center for Brain Science, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Jian-Feng Liu
- Collaborative Innovation Center for Brain Science, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| |
Collapse
|
11
|
Liu S, Wang C, Chen Y, Peng S, Chen X, Tan Z. Association of SLC15A1 polymorphisms with susceptibility to dyslipidaemia in a Chinese Han population. J Clin Pharm Ther 2019; 44:868-874. [PMID: 31454435 DOI: 10.1111/jcpt.13016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 04/03/2019] [Accepted: 07/14/2019] [Indexed: 12/26/2022]
Abstract
WHAT IS KNOWN AND OBJECTIVE Dyslipidaemia is an increasingly serious clinical and public health issue. In this study, we aim to explore the association of genetic polymorphisms in solute carrier transporter (SLC) 15A1 with the risk of dyslipidaemia in a Chinese Han population. METHODS Three single nucleotide polymorphisms (SNPs) in SLC15A1 (rs2297322, rs4646234 and rs1289389) were selected using bioinformatics in a Chinese Han population with 530 participants. Genotyping was conducted with Sequenom MassARRAY. A logistic regression model was used for the analysis of the association between genotypes and dyslipidaemia. SHEsis software was applied to the haplotype analysis. RESULTS AND DISCUSSION The SLC15A1 rs2297322 TT genotype was associated with a lower risk of hypertriglyceridaemia compared with the CC genotype (OR = 0.44, 95% CI = 0.21-0.93, P = .032). The carriers of the SLC15A1 rs1289389 T allele were found to be significantly associated with a lower risk of hypertriglyceridaemia compared with the C allele (OR = 0.54, 95% CI = 0.33-0.88, P = .013). In the recessive model, the carriers of the SLC15A1 rs4646234 CC genotype showed a significantly reduced risk of hypercholesterolaemia (OR = 2.29, 95% CI = 1.23-4.28, P = .009). Haplotype analysis showed that the CTC haplotype composed of SLC15A1 rs2297322, rs4646234 and rs1289389 was associated with a lower risk of hypertriglyceridaemia (OR = 1.58, 95% CI = 1.12-2.24, P = .009), whereas the TTC haplotype was associated with a significantly reduced risk of hypertriglyceridaemia (OR = 0.63, 95% CI = 0.40-0.99, P = .045). WHAT IS NEW AND CONCLUSION SLC15A1 rs2297322 and rs1289389 polymorphisms were associated with alterations in the risk of dyslipidaemia in a Chinese Han population.
Collapse
Affiliation(s)
- Shu Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China
| | - Chunyang Wang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China
| | - Yao Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China
| | - Shifang Peng
- Health Management Center, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoping Chen
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China
| | - Zhirong Tan
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, China
| |
Collapse
|
12
|
The Foodborne Strain Lactobacillus fermentum MBC2 Triggers pept-1-Dependent Pro-Longevity Effects in Caenorhabditis elegans. Microorganisms 2019; 7:microorganisms7020045. [PMID: 30736484 PMCID: PMC6406943 DOI: 10.3390/microorganisms7020045] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 01/21/2019] [Accepted: 02/07/2019] [Indexed: 12/16/2022] Open
Abstract
Lactic acid bacteria (LAB) are involved in several food fermentations and many of them provide strain-specific health benefits. Herein, the probiotic potential of the foodborne strain Lactobacillus fermentum MBC2 was investigated through in vitro and in vivo approaches. Caenorhabditis elegans was used as an in vivo model to analyze pro-longevity and anti-aging effects. L. fermentum MBC2 showed a high gut colonization capability compared to E. coli OP50 (OP50) or L.rhamnosus GG (LGG). Moreover, analysis of pumping rate, lipofuscin accumulation, and body bending showed anti-aging effects in L. fermentum MBC2-fed worms. Studies on PEPT-1 mutants demonstrated that pept-1 gene was involved in the anti-aging processes mediated by this bacterial strain through DAF-16, whereas the oxidative stress protection was PEPT-1 independent. Moreover, analysis of acid tolerance, bile tolerance, and antibiotic susceptibility were evaluated. L. fermentum MBC2 exerted beneficial effects on nematode lifespan, influencing energy metabolism and oxidative stress resistance, resulted in being tolerant to acidic pH and able to adhere to Caco-2 cells. Overall, these findings provide new insight for application of this strain in the food industry as a newly isolated functional starter. Furthermore, these results will also shed light on C. elegans molecular players involved in host-microbe interactions.
Collapse
|
13
|
Spanier B, Wallwitz J, Zapoglou D, Idrissou BMG, Fischer C, Troll M, Petzold K, Daniel H. The Reproduction Rate of Peptide Transporter PEPT-1 Deficient C. elegans Is Dependent on Dietary Glutamate Supply. Front Mol Biosci 2018; 5:109. [PMID: 30560135 PMCID: PMC6284198 DOI: 10.3389/fmolb.2018.00109] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/15/2018] [Indexed: 01/19/2023] Open
Abstract
Intestinal absorption of dietary amino acids is mediated via two routes. Free amino acids released by hydrolysis of dietary proteins are taken up by a multitude of amino acid transporters while di- and tripeptides released are taken up by the peptide transporter PEPT-1. Loss of PEPT-1 impairs growth, post-embryonic development and reproduction in Caenorhabditis elegans, and supplementation with a mixture of all L-amino acids only partially rescues fertility. In the present study, we demonstrate that dietary L-glutamate is the responsible amino acid that can increase fertility in hermaphrodite pept-1 worms. This effect was associated with a significantly higher uptake of glutamate/aspartate in pept-1 than in wildtype C. elegans. Furthermore, we found that the intestinal transporter proteins SNF-5 of the solute carrier SLC6 family of nutrient amino acid transporters (NAT) and AAT-6 of the SLC7 family as the light subunit of a heteromeric amino acid transporter (HAT) play a key role in glutamate homeostasis in pept-1 C. elegans. Genes encoding these transporters are highly expressed and upon silencing a 95% reduced fertility (snf-5) and sterility (aat-6) was observed. A subsequent L-glutamate supplementation failed to rescue these phenotypes. Dietary glutamate supplementation did neither influence the feeding frequency, nor did it improve mating efficiency of pept-1 males. Most strikingly, pept-1 were more prone to habituation to repeated gentle touch stimuli than wildtype C. elegans, and dietary glutamate supply was sufficient to alter this behavioral output by restoring the mechanosensory response to wildtype levels. Taken together, our data demonstrate a key role of L-glutamate in amino acid homeostasis in C. elegans lacking the peptide transporter in the intestine and demonstrate its distinct role in reproduction and for neural circuits mediating touch sensitivity.
Collapse
Affiliation(s)
- Britta Spanier
- Nutritional Physiology, Technische Universität München, Munich, Germany
| | | | - Despoina Zapoglou
- Nutritional Physiology, Technische Universität München, Munich, Germany
| | | | - Christine Fischer
- Nutritional Physiology, Technische Universität München, Munich, Germany
| | - Martina Troll
- Nutritional Physiology, Technische Universität München, Munich, Germany
| | - Katrin Petzold
- Nutritional Physiology, Technische Universität München, Munich, Germany
| | - Hannelore Daniel
- Nutritional Physiology, Technische Universität München, Munich, Germany
| |
Collapse
|
14
|
Affiliation(s)
- Weilin Shen
- Division of Bioscience and Biotechnology Faculty of Agriculture Graduate School of Kyushu University 744 Motooka Nishi‐ku Fukuoka 819‐0395 Japan
| | - Toshiro Matsui
- Division of Bioscience and Biotechnology Faculty of Agriculture Graduate School of Kyushu University 744 Motooka Nishi‐ku Fukuoka 819‐0395 Japan
| |
Collapse
|
15
|
Spanier B, Rohm F. Proton Coupled Oligopeptide Transporter 1 (PepT1) Function, Regulation, and Influence on the Intestinal Homeostasis. Compr Physiol 2018; 8:843-869. [DOI: 10.1002/cphy.c170038] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
16
|
Viennois E, Pujada A, Zen J, Merlin D. Function, Regulation, and Pathophysiological Relevance of the POT Superfamily, Specifically PepT1 in Inflammatory Bowel Disease. Compr Physiol 2018; 8:731-760. [PMID: 29687900 DOI: 10.1002/cphy.c170032] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Mammalian members of the proton-coupled oligopeptide transporter family are integral membrane proteins that mediate the cellular uptake of di/tripeptides and peptide-like drugs and couple substrate translocation to the movement of H+ , with the transmembrane electrochemical proton gradient providing the driving force. Peptide transporters are responsible for the (re)absorption of dietary and/or bacterial di- and tripeptides in the intestine and kidney and maintaining homeostasis of neuropeptides in the brain. These proteins additionally contribute to absorption of a number of pharmacologically important compounds. In this overview article, we have provided updated information on the structure, function, expression, localization, and activities of PepT1 (SLC15A1), PepT2 (SLC15A2), PhT1 (SLC15A4), and PhT2 (SLC15A3). Peptide transporters, in particular, PepT1 are discussed as drug-delivery systems in addition to their implications in health and disease. Particular emphasis has been placed on the involvement of PepT1 in the physiopathology of the gastrointestinal tract, specifically, its role in inflammatory bowel diseases. © 2018 American Physiological Society. Compr Physiol 8:731-760, 2018.
Collapse
Affiliation(s)
- Emilie Viennois
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Adani Pujada
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Jane Zen
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Didier Merlin
- Institute for Biomedical Sciences, Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA.,Veterans Affairs Medical Center, Decatur, Georgia, USA
| |
Collapse
|
17
|
Wang B, Wang H, Xiong J, Zhou Q, Wu H, Xia L, Li L, Yu Z. A Proteomic Analysis Provides Novel Insights into the Stress Responses of Caenorhabditis elegans towards Nematicidal Cry6A Toxin from Bacillus thuringiensis. Sci Rep 2017; 7:14170. [PMID: 29074967 PMCID: PMC5658354 DOI: 10.1038/s41598-017-14428-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 10/10/2017] [Indexed: 01/16/2023] Open
Abstract
Cry6A represents a novel family of nematicidal crystal proteins from Bacillus thuringiensis. It has distinctive architecture as well as mechanism of action from Cry5B, a highly focused family of nematicidal crystal proteins, and even from other insecticidal crystal proteins containing the conserved three-domain. However, how nematode defends against Cry6A toxin remains obscure. In this study, the global defense pattern of Caenorhabditis elegans against Cry6Aa2 toxin was investigated by proteomic analysis. In response to Cry6Aa2, 12 proteins with significantly altered abundances were observed from worms, participating in innate immune defense, insulin-like receptor (ILR) signaling pathway, energy metabolism, and muscle assembly. The differentially expressed proteins (DEPs) functioning in diverse biological processes suggest that a variety of defense responses participate in the stress responses of C. elegans to Cry6Aa2. The functional verifications of DEPs suggest that ILR signaling pathway, DIM-1, galectin LEC-6 all are the factors of defense responses to Cry6Aa2. Moreover, Cry6Aa2 also involves in accelerating the metabolic energy production which fulfills the energy demand for the immune responses. In brief, our findings illustrate the global pattern of defense responses of nematode against Cry6A for the first time, and provide a novel insight into the mechanism through which worms respond to Cry6A.
Collapse
Affiliation(s)
- Bing Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, P.R. China
| | - Haiwen Wang
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, P.R. China
| | - Jing Xiong
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, P.R. China
| | - Qiaoni Zhou
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, P.R. China
| | - Huan Wu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, P.R. China
| | - Liqiu Xia
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, P.R. China
| | - Lin Li
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, P.R. China
| | - Ziquan Yu
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Science, Hunan Normal University, Changsha, 410081, P.R. China.
| |
Collapse
|
18
|
Watts JL, Ristow M. Lipid and Carbohydrate Metabolism in Caenorhabditis elegans. Genetics 2017; 207:413-446. [PMID: 28978773 PMCID: PMC5629314 DOI: 10.1534/genetics.117.300106] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 08/02/2017] [Indexed: 12/14/2022] Open
Abstract
Lipid and carbohydrate metabolism are highly conserved processes that affect nearly all aspects of organismal biology. Caenorhabditis elegans eat bacteria, which consist of lipids, carbohydrates, and proteins that are broken down during digestion into fatty acids, simple sugars, and amino acid precursors. With these nutrients, C. elegans synthesizes a wide range of metabolites that are required for development and behavior. In this review, we outline lipid and carbohydrate structures as well as biosynthesis and breakdown pathways that have been characterized in C. elegans We bring attention to functional studies using mutant strains that reveal physiological roles for specific lipids and carbohydrates during development, aging, and adaptation to changing environmental conditions.
Collapse
Affiliation(s)
- Jennifer L Watts
- School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, Washington 99164
| | - Michael Ristow
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology Zurich, 8603 Schwerzenbach-Zurich, Switzerland
| |
Collapse
|
19
|
Di- and tripeptide transport in vertebrates: the contribution of teleost fish models. J Comp Physiol B 2016; 187:395-462. [PMID: 27803975 DOI: 10.1007/s00360-016-1044-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 10/12/2016] [Accepted: 10/20/2016] [Indexed: 02/06/2023]
Abstract
Solute Carrier 15 (SLC15) family, alias H+-coupled oligopeptide cotransporter family, is a group of membrane transporters known for their role in the cellular uptake of di- and tripeptides (di/tripeptides) and peptide-like molecules. Of its members, SLC15A1 (PEPT1) chiefly mediates intestinal absorption of luminal di/tripeptides from dietary protein digestion, while SLC15A2 (PEPT2) mainly allows renal tubular reabsorption of di/tripeptides from ultrafiltration, SLC15A3 (PHT2) and SLC15A4 (PHT1) possibly interact with di/tripeptides and histidine in certain immune cells, and SLC15A5 has unknown function. Our understanding of this family in vertebrates has steadily increased, also due to the surge of genomic-to-functional information from 'non-conventional' animal models, livestock, poultry, and aquaculture fish species. Here, we review the literature on the SLC15 transporters in teleost fish with emphasis on SLC15A1 (PEPT1), one of the solute carriers better studied amongst teleost fish because of its relevance in animal nutrition. We report on the operativity of the transporter, the molecular diversity, and multiplicity of structural-functional solutions of the teleost fish orthologs with respect to higher vertebrates, its relevance at the intersection of the alimentary and osmoregulative functions of the gut, its response under various physiological states and dietary solicitations, and its possible involvement in examples of total body plasticity, such as growth and compensatory growth. By a comparative approach, we also review the few studies in teleost fish on SLC15A2 (PEPT2), SLC15A4 (PHT1), and SLC15A3 (PHT2). By representing the contribution of teleost fish to the knowledge of the physiology of di/tripeptide transport and transporters, we aim to fill the gap between higher and lower vertebrates.
Collapse
|
20
|
Nutrient Transporter Expression in the Jejunum in Relation to Body Mass Index in Patients Undergoing Bariatric Surgery. Nutrients 2016; 8:nu8110683. [PMID: 27801863 PMCID: PMC5133071 DOI: 10.3390/nu8110683] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/11/2016] [Accepted: 10/26/2016] [Indexed: 12/20/2022] Open
Abstract
Nutrient tranters (NT) facilitate nutrient absorption and contribute to the regulation of circulating nutrients. In this cross-sectional study, we determined the associations between the level of obesity; mRNA abundance for NTs; and serum concentrations of amino acids, short-chain fatty acids, and glucose in patients with morbid obesity undergoing a Roux-en-Y gastric bypass. Proximal jejunal samples were obtained at the time of surgery from 42 patients (90% female, age = 42.6 ± 11.9 years, pre-operative body mass index (BMI) = 55.5 ± 11.3 kg/m²) undergoing a Roux-en-Y gastric bypass. RNA was extracted from the jejunal mucosa and quantitative real-time-PCR was performed for the NTs studied. BMI negatively correlated with jejunal mRNA abundance of the amino acid NTs TauT (r = -0.625, p < 0.0001), ASCT2 (r = -0.320, p = 0.039), LAT1 (r = -0.304, p = 0.05). BMI positively correlated with jejunal mRNA abundance of the lactate/short-chain fatty acid NT SMCT1 (r = 0.543, p = 0.0002). Serum concentrations of the short-chain fatty acids, butyric, valeric, and isocaproic acid correlated positively with BMI (n = 30) (r = 0.45, r = 0.44, r = 0.36, p ≤ 0.05; respectively). Lower jejunal mRNA abundance for the amino acid NTs TauT, ASCT2, and LAT1 could protect against further obesity-related elevations in circulating amino acids. The positive correlation between BMI and the jejunal mRNA abundance of the high-affinity short-chain fatty acid/monocarboxylate transporter SMCT1 is intriguing and requires further investigation.
Collapse
|
21
|
Iron Overload Coordinately Promotes Ferritin Expression and Fat Accumulation in Caenorhabditis elegans. Genetics 2016; 203:241-53. [PMID: 27017620 DOI: 10.1534/genetics.116.186742] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/20/2016] [Indexed: 01/22/2023] Open
Abstract
The trace element iron is crucial for living organisms, since it plays essential roles in numerous cellular functions. Systemic iron overload and the elevated level of ferritin, a ubiquitous intracellular protein that stores and releases iron to maintain the iron homeostasis in cells, has long been epidemiologically associated with obesity and obesity-related diseases. However, the underlying mechanisms of this association remain unclear. Here, using Caenorhabditis elegans, we show that iron overload induces the expression of sgk-1, encoding the serum and glucocorticoid-inducible kinase, to promote the level of ferritin and fat accumulation. Mutation of cyp-23A1, encoding a homolog of human cytochrome P450 CYP7B1 that is related to neonatal hemochromatosis, further enhances the elevated expression of ftn-1, sgk-1, and fat accumulation. sgk-1 positively regulates the expression of acs-20 and vit-2, genes encoding homologs of the mammalian FATP1/4 fatty acid transport proteins and yolk lipoproteins, respectively, to facilitate lipid uptake and translocation for storage under iron overload. This study reveals a completely novel pathway in which sgk-1 plays a central role to synergistically regulate iron and lipid homeostasis, offering not only experimental evidence supporting a previously unverified link between iron and obesity, but also novel insights into the pathogenesis of iron and obesity-related human metabolic diseases.
Collapse
|
22
|
Kern T, Kutzner E, Eisenreich W, Fuchs TM. Pathogen-nematode interaction: Nitrogen supply of Listeria monocytogenes during growth in Caenorhabditis elegans. ENVIRONMENTAL MICROBIOLOGY REPORTS 2016; 8:20-29. [PMID: 26478569 DOI: 10.1111/1758-2229.12344] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 10/08/2015] [Indexed: 06/05/2023]
Abstract
Listeria monocytogenes is a Gram-positive facultatively intracellular human pathogen. Due to its saprophytic lifestyle, L. monocytogenes is assumed to infect and proliferate within soil organisms such as Caenorhabditis elegans. However, little is known about the nutrient usages and metabolite fluxes in this bacterium-nematode interaction. Here, we established a nematode colonization model for L. monocytogenes and a method for the efficient separation of the pathogen from the nematodal gut. Following (15)N labelling of C. elegans and gas chromatography-mass spectrometry-based (15)N isotopologue analysis, we detected a high basal metabolic rate of the nematode, and observed a significant metabolic flux from nitrogenous compounds of the nematode to listerial proteins during proliferation of the pathogen in the worm's intestine. For comparison, we also measured the N fluxes from the gut content into listerial proteins using completely (15)N-labelled Escherichia coli OP50 as food for C. elegans. In both settings, L. monocytogenes prefers the direct incorporation of histidine, arginine and lysine over their de novo biosynthesis. Our data suggest that colonization of nematodes is a strategy of L. monocytogenes to increase its access to N-rich nutrients.
Collapse
Affiliation(s)
- Tanja Kern
- Lehrstuhl für Mikrobielle Ökologie, Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München, Weihenstephaner Berg 3, 85354, Freising, Germany
| | - Erika Kutzner
- Lehrstuhl für Biochemie, Technische Universität München, D-85747, Garching, Germany
| | - Wolfgang Eisenreich
- Lehrstuhl für Biochemie, Technische Universität München, D-85747, Garching, Germany
| | - Thilo M Fuchs
- Lehrstuhl für Mikrobielle Ökologie, Zentralinstitut für Ernährungs- und Lebensmittelforschung (ZIEL), Technische Universität München, Weihenstephaner Berg 3, 85354, Freising, Germany
| |
Collapse
|
23
|
Mohandas N, Young ND, Jabbar A, Korhonen PK, Koehler AV, Hall RS, Hu M, Hofmann A, Gasser RB. The complement of family M1 aminopeptidases of Haemonchus contortus--Biotechnological implications. Biotechnol Adv 2015; 34:65-76. [PMID: 26597954 DOI: 10.1016/j.biotechadv.2015.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 10/04/2015] [Accepted: 10/10/2015] [Indexed: 01/03/2023]
Abstract
Although substantial research has been focused on the 'hidden antigen' H11 of Haemonchus contortus as a vaccine against haemonchosis in small ruminants, little is know about this and related aminopeptidases. In the present article, we reviewed genomic and transcriptomic data sets to define, for the first time, the complement of aminopeptidases (designated Hc-AP-1 to Hc-AP-13) of the family M1 with homologues in Caenorhabditis elegans, characterised by zinc-binding (HEXXH) and exo-peptidase (GAMEN) motifs. The three previously published H11 isoforms (accession nos. X94187, FJ481146 and AJ249941) had most sequence similarity to Hc-AP-2 and Hc-AP-8, whereas unpublished isoforms (accession nos. AJ249942 and AJ311316) were both most similar to Hc-AP-3. The aminopeptidases characterised here had homologues in C. elegans. Hc-AP-1 to Hc-AP-8 were most similar in amino acid sequence (28-41%) to C. elegans T07F10.1; Hc-AP-9 and Hc-AP-10 to C. elegans PAM-1 (isoform b) (53-54% similar); Hc-AP-11 and Hc-AP-12 to C. elegans AC3.5 and Y67D8C.9 (26% and 50% similar, respectively); and Hc-AP-13 to C. elegans C42C1.11 and ZC416.6 (50-58% similar). Comparative analysis suggested that Hc-AP-1 to Hc-AP-8 play roles in digestion, metabolite excretion, neuropeptide processing and/or osmotic regulation, with Hc-AP-4 and Hc-AP-7 having male-specific functional roles. The analysis also indicated that Hc-AP-9 and Hc-AP-10 might be involved in the degradation of cyclin (B3) and required to complete meiosis. Hc-AP-11 represents a leucyl/cystinyl aminopeptidase, predicted to have metallopeptidase and zinc ion binding activity, whereas Hc-AP-12 likely encodes an aminopeptidase Q homologue also with these activities and a possible role in gonad function. Finally, Hc-AP-13 is predicted to encode an aminopeptidase AP-1 homologue of C. elegans with hydrolase activity, suggested to operate, possibly synergistically with a PEPT-1 ortholog, as an oligopeptide transporter in the gut for protein uptake and normal development and/or reproduction of the worm. An appraisal of structure-based amino acid sequence alignments revealed that all conceptually translated Hc-AP proteins, with the exception of Hc-AP-12, adopt a topology similar to those observed for the two subgroups of mammalian M1 aminopeptidases, which possess either three (I, II and IV) or four (I-IV) domains. In contrast, Hc-AP-12 lacks the N-terminal domain (I), but possesses a substantially expanded domain III. Although further work needs to be done to assess amino acid sequence conservation of the different aminopeptidases among individual worms within and among H. contortus populations, we hope that these insights will support future localisation, structural and functional studies of these molecules in H. contortus as well as facilitate future assessments of a recombinant subunit or cocktail vaccine against haemonchosis.
Collapse
Affiliation(s)
- Namitha Mohandas
- The University of Melbourne, Faculty of Veterinary and Agricultural Sciences, Parkville, Victoria 3010, Australia
| | - Neil D Young
- The University of Melbourne, Faculty of Veterinary and Agricultural Sciences, Parkville, Victoria 3010, Australia
| | - Abdul Jabbar
- The University of Melbourne, Faculty of Veterinary and Agricultural Sciences, Parkville, Victoria 3010, Australia
| | - Pasi K Korhonen
- The University of Melbourne, Faculty of Veterinary and Agricultural Sciences, Parkville, Victoria 3010, Australia
| | - Anson V Koehler
- The University of Melbourne, Faculty of Veterinary and Agricultural Sciences, Parkville, Victoria 3010, Australia
| | - Ross S Hall
- The University of Melbourne, Faculty of Veterinary and Agricultural Sciences, Parkville, Victoria 3010, Australia
| | - Min Hu
- State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei, China
| | - Andreas Hofmann
- The University of Melbourne, Faculty of Veterinary and Agricultural Sciences, Parkville, Victoria 3010, Australia; Structural Chemistry Program, Eskitis Institute for Drug Discovery, Griffith University, Brisbane, Queensland 4111, Australia.
| | - Robin B Gasser
- The University of Melbourne, Faculty of Veterinary and Agricultural Sciences, Parkville, Victoria 3010, Australia; State Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei, China.
| |
Collapse
|
24
|
Dialogue between E. coli free radical pathways and the mitochondria of C. elegans. Proc Natl Acad Sci U S A 2015; 112:12456-61. [PMID: 26392561 DOI: 10.1073/pnas.1517448112] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The microbial world presents a complex palette of opportunities and dangers to animals, which have developed surveillance and response strategies to hints of microbial intent. We show here that the mitochondrial homeostatic response pathway of the nematode Caenorhabditis elegans responds to Escherichia coli mutations that activate free radical detoxification pathways. Activation of C. elegans mitochondrial responses could be suppressed by additional mutations in E. coli, suggesting that C. elegans responds to products of E. coli to anticipate challenges to its mitochondrion. Out of 50 C. elegans gene inactivations known to mediate mitochondrial defense, we found that 7 genes were required for C. elegans response to a free radical producing E. coli mutant, including the bZip transcription factor atfs-1 (activating transcription factor associated with stress). An atfs-1 loss-of-function mutant was partially resistant to the effects of free radical-producing E. coli mutant, but a constitutively active atfs-1 mutant growing on wild-type E. coli inappropriately activated the pattern of mitochondrial responses normally induced by an E. coli free radical pathway mutant. Carbonylated proteins from free radical-producing E. coli mutant may directly activate the ATFS-1/bZIP transcription factor to induce mitochondrial stress response: feeding C. elegans with H2O2-treated E. coli induces the mitochondrial unfolded protein response, and inhibition of a gut peptide transporter partially suppressed C. elegans response to free radical damaged E. coli.
Collapse
|
25
|
Daniel H, Zietek T. Taste and move: glucose and peptide transporters in the gastrointestinal tract. Exp Physiol 2015; 100:1441-50. [DOI: 10.1113/ep085029] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 06/26/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Hannelore Daniel
- Nutritional Physiology; Technische Universität München; 85350 Freising Germany
- ZIEL Institute for Food & Health; 85350 Freising Germany
| | - Tamara Zietek
- Nutritional Physiology; Technische Universität München; 85350 Freising Germany
- ZIEL Institute for Food & Health; 85350 Freising Germany
| |
Collapse
|
26
|
Impact of a Complex Food Microbiota on Energy Metabolism in the Model Organism Caenorhabditis elegans. BIOMED RESEARCH INTERNATIONAL 2015; 2015:621709. [PMID: 25961031 PMCID: PMC4417589 DOI: 10.1155/2015/621709] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 11/11/2014] [Indexed: 02/03/2023]
Abstract
The nematode Caenorhabditis elegans is widely used as a model system for research on aging, development, and host-pathogen interactions. Little is currently known about the mechanisms underlying the effects exerted by foodborne microbes. We took advantage of C. elegans to evaluate the impact of foodborne microbiota on well characterized physiological features of the worms. Foodborne lactic acid bacteria (LAB) consortium was used to feed nematodes and its composition was evaluated by 16S rDNA analysis and strain typing before and after colonization of the nematode gut. Lactobacillus delbrueckii, L. fermentum, and Leuconostoc lactis were identified as the main species and shown to display different worm gut colonization capacities. LAB supplementation appeared to decrease nematode lifespan compared to the animals fed with the conventional Escherichia coli nutrient source or a probiotic bacterial strain. Reduced brood size was also observed in microbiota-fed nematodes. Moreover, massive accumulation of lipid droplets was revealed by BODIPY staining. Altered expression of nhr-49, pept-1, and tub-1 genes, associated with obesity phenotypes, was demonstrated by RT-qPCR. Since several pathways are evolutionarily conserved in C. elegans, our results highlight the nematode as a valuable model system to investigate the effects of a complex microbial consortium on host energy metabolism.
Collapse
|
27
|
Sheng M, Hosseinzadeh A, Muralidharan SV, Gaur R, Selstam E, Tuck S. Aberrant fat metabolism in Caenorhabditis elegans mutants with defects in the defecation motor program. PLoS One 2015; 10:e0124515. [PMID: 25849533 PMCID: PMC4388766 DOI: 10.1371/journal.pone.0124515] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 03/16/2015] [Indexed: 01/08/2023] Open
Abstract
The molecular mechanisms by which dietary fatty acids are absorbed by the intestine, and the way in which the process is regulated are poorly understood. In a genetic screen for mutations affecting fat accumulation in the intestine of Caenorhabditis elegans, nematode worms, we have isolated mutations in the aex-5 gene, which encodes a Kex2/subtilisin-family, Ca2+-sensitive proprotein convertase known to be required for maturation of certain neuropeptides, and for a discrete step in an ultradian rhythmic phenomenon called the defecation motor program. We demonstrate that aex-5 mutants have markedly lower steady-state levels of fat in the intestine, and that this defect is associated with a significant reduction in the rate at which labeled fatty acid derivatives are taken up from the intestinal lumen. Other mutations affecting the defecation motor program also affect steady-state levels of triglycerides, suggesting that the program is required per se for the proper accumulation of neutral lipids. Our results suggest that an important function of the defecation motor program in C. elegans is to promote the uptake of an important class of dietary nutrients. They also imply that modulation of the program might be one way in which worms adjust nutrient uptake in response to altered metabolic status.
Collapse
Affiliation(s)
- Ming Sheng
- Umeå Center for Molecular Medicine, Umeå University, SE-901 87 Umeå, Sweden
| | - Ava Hosseinzadeh
- Umeå Center for Molecular Medicine, Umeå University, SE-901 87 Umeå, Sweden
| | | | - Rahul Gaur
- Umeå Center for Molecular Medicine, Umeå University, SE-901 87 Umeå, Sweden
| | - Eva Selstam
- Department of Plant Physiology, Umeå University, SE-901 87 Umeå, Sweden
| | - Simon Tuck
- Umeå Center for Molecular Medicine, Umeå University, SE-901 87 Umeå, Sweden
| |
Collapse
|
28
|
|
29
|
Geillinger KE, Kuhlmann K, Eisenacher M, Giesbertz P, Meyer HE, Daniel H, Spanier B. Intestinal amino acid availability via PEPT-1 affects TORC1/2 signaling and the unfolded protein response. J Proteome Res 2014; 13:3685-92. [PMID: 24999909 DOI: 10.1021/pr5002669] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The intestinal peptide transporter PEPT-1 plays an important role in development, growth, reproduction, and stress tolerance in Caenorhabditis elegans, as revealed by the severe phenotype of the pept-1-deficient strain. The reduced number of offspring and increased stress resistance were shown to result from changes in the insulin/IGF-signaling cascade. To further elucidate the regulatory network behind the phenotypic alterations in PEPT1-deficient animals, a quantitative proteome analysis combined with transcriptome profiling was applied. Various target genes of XBP-1, the major mediator of the unfolded protein response, were found to be downregulated at the mRNA and protein levels, accompanied by a reduction of spliced xbp-1 mRNA. Proteome analysis also revealed a markedly reduced content of numerous ribosomal proteins. This was associated with a reduction in the protein synthesis rate in pept-1 C. elegans, a process that is strictly regulated by the TOR (target of rapamycine) complex, the cellular sensor for free amino acids. These data argue for a central role of PEPT-1 in cellular amino acid homeostasis. In PEPT-1 deficiency, amino acid levels dropped systematically, leading to alterations in protein synthesis and in the IRE-1/XBP-1 pathway.
Collapse
Affiliation(s)
- Kerstin E Geillinger
- ZIEL Research Center of Nutrition and Food Sciences, Molecular Nutrition and Biochemistry Unit, Technische Universität München , Gregor-Mendel-Str. 2, 85350 Freising, Germany
| | | | | | | | | | | | | |
Collapse
|
30
|
Romano A, Barca A, Storelli C, Verri T. Teleost fish models in membrane transport research: the PEPT1(SLC15A1) H+-oligopeptide transporter as a case study. J Physiol 2013; 592:881-97. [PMID: 23981715 DOI: 10.1113/jphysiol.2013.259622] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Human genes for passive, ion-coupled transporters and exchangers are included in the so-called solute carrier (SLC) gene series, to date consisting of 52 families and 398 genes. Teleost fish genes for SLC proteins have also been described in the last two decades, and catalogued in preliminary SLC-like form in 50 families and at least 338 genes after systematic GenBank database mining (December 2010-March 2011). When the kinetic properties of the expressed proteins are studied in detail, teleost fish SLC transporters always reveal extraordinary 'molecular diversity' with respect to the mammalian counterparts, which reflects peculiar adaptation of the protein to the physiology of the species and/or to the environment where the species lives. In the case of the H+ -oligopeptide transporter PEPT1(SLC15A1), comparative analysis of diverse teleost fish orthologs has shown that the protein may exhibit very eccentric properties in terms of pH dependence (e.g., the adaptation of zebrafish PEPT1 to alkaline pH), temperature dependence (e.g., the adaptation of icefish PEPT1 to sub-zero temperatures) and/or substrate specificity (e.g., the species-specificity of PEPT1 for the uptake of l-lysine-containing peptides). The revelation of such peculiarities is providing new contributions to the discussion on PEPT1 in both basic (e.g., molecular structure-function analyses) and applied research (e.g., optimizing diets to enhance growth of commercially valuable fish).
Collapse
Affiliation(s)
- Alessandro Romano
- Laboratory of General Physiology, Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Provinciale Lecce-Monteroni, I-73100 Lecce, Italy.
| | | | | | | |
Collapse
|
31
|
Spanier B. Transcriptional and functional regulation of the intestinal peptide transporter PEPT1. J Physiol 2013; 592:871-9. [PMID: 23959672 DOI: 10.1113/jphysiol.2013.258889] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Dietary proteins are cleaved within the intestinal lumen to oligopeptides which are further processed to small peptides (di- and tripeptides) and free amino acids. Although the transport of amino acids is mediated by several specific amino acid transporters, the proton-coupled uptake of the more than 8000 different di- and tripeptides is performed by the high-capacity/low-affinity peptide transporter isoform PEPT1 (SLC15A1). Its wide substrate tolerance also allows the transport of a repertoire of structurally closely related compounds and drugs, which explains their high oral bioavailability and brings PEPT1 into focus for medical and pharmaceutical approaches. Although the first evidence for the interplay of nutrient supply and PEPT1 expression and function was described over 20 years ago, many aspects of the molecular processes controlling its transcription and translation and modifying its transporter properties are still awaiting discovery. The present review summarizes the recent knowledge on the factors modulating PEPT1 expression and function in Caenorhabditis elegans, Danio rerio, Mus musculus and Homo sapiens, with focus on dietary ingredients, transcription factors and functional modulators, such as the sodium-proton exchanger NHE3 and selected scaffold proteins.
Collapse
Affiliation(s)
- Britta Spanier
- Biochemistry, Technische Universität München, ZIEL Research Center of Nutrition and Food Sciences, Gregor-Mendel-Straße 2, D-85350 Freising, Germany.
| |
Collapse
|
32
|
Ding YH, Du YG, Luo S, Li YX, Li TM, Yoshina S, Wang X, Klage K, Mitani S, Ye K, Dong MQ. Characterization of PUD-1 and PUD-2, two proteins up-regulated in a long-lived daf-2 mutant. PLoS One 2013; 8:e67158. [PMID: 23799143 PMCID: PMC3683130 DOI: 10.1371/journal.pone.0067158] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Accepted: 05/14/2013] [Indexed: 01/30/2023] Open
Abstract
C. elegans PUD-1 and PUD-2, two proteins up-regulated in daf-2(loss-of-function) (PUD), are homologous 17-kD proteins with a large abundance increase in long-lived daf-2 mutant animals of reduced insulin signaling. In this study, we show that both PUD-1 and PUD-2 are abundantly expressed in the intestine and hypodermis, and form a heterodimer. We have solved their crystal structure to 1.9-Å resolution and found that both proteins adopt similar β-sandwich folds in the V-shaped dimer. In contrast, their homologs PUD-3, PUD-4, PUDL-1 and PUDL-2 are all monomeric proteins with distinct expression patterns in C. elegans. Thus, the PUD-1/PUD-2 heterodimer probably has a function distinct from their family members. Neither overexpression nor deletion of pud-1 and pud-2 affected the lifespan of WT or daf-2 mutant animals, suggesting that their induction in daf-2 worms does not contribute to longevity. Curiously, deletion of pud-1 and pud-2 was associated with a protective effect against paralysis induced by the amyloid β-peptide (1-42), which further enhanced the protection conferred by daf-2(RNAi) against Aβ.
Collapse
Affiliation(s)
- Yue-He Ding
- National Institute of Biological Sciences, Beijing, Beijing, China
- Graduate Program in Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yun-Guang Du
- National Institute of Biological Sciences, Beijing, Beijing, China
| | - Shukun Luo
- National Institute of Biological Sciences, Beijing, Beijing, China
| | - Yu-Xin Li
- National Institute of Biological Sciences, Beijing, Beijing, China
| | - Tie-Mei Li
- National Institute of Biological Sciences, Beijing, Beijing, China
| | - Sawako Yoshina
- Department of Physiology, Tokyo Women's Medical University, Tokyo, Japan
| | - Xing Wang
- National Institute of Biological Sciences, Beijing, Beijing, China
| | - Karsten Klage
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Shohei Mitani
- Department of Physiology, Tokyo Women's Medical University, Tokyo, Japan
| | - Keqiong Ye
- National Institute of Biological Sciences, Beijing, Beijing, China
- * E-mail: (KY); (M-QD)
| | - Meng-Qiu Dong
- National Institute of Biological Sciences, Beijing, Beijing, China
- Graduate Program in Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- * E-mail: (KY); (M-QD)
| |
Collapse
|
33
|
Hunter SE, Finnegan EF, Zisoulis DG, Lovci MT, Melnik-Martinez KV, Yeo GW, Pasquinelli AE. Functional genomic analysis of the let-7 regulatory network in Caenorhabditis elegans. PLoS Genet 2013; 9:e1003353. [PMID: 23516374 PMCID: PMC3597506 DOI: 10.1371/journal.pgen.1003353] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Accepted: 01/16/2013] [Indexed: 12/21/2022] Open
Abstract
The let-7 microRNA (miRNA) regulates cellular differentiation across many animal species. Loss of let-7 activity causes abnormal development in Caenorhabditis elegans and unchecked cellular proliferation in human cells, which contributes to tumorigenesis. These defects are due to improper expression of protein-coding genes normally under let-7 regulation. While some direct targets of let-7 have been identified, the genome-wide effect of let-7 insufficiency in a developing animal has not been fully investigated. Here we report the results of molecular and genetic assays aimed at determining the global network of genes regulated by let-7 in C. elegans. By screening for mis-regulated genes that also contribute to let-7 mutant phenotypes, we derived a list of physiologically relevant potential targets of let-7 regulation. Twenty new suppressors of the rupturing vulva or extra seam cell division phenotypes characteristic of let-7 mutants emerged. Three of these genes, opt-2, prmt-1, and T27D12.1, were found to associate with Argonaute in a let-7–dependent manner and are likely novel direct targets of this miRNA. Overall, a complex network of genes with various activities is subject to let-7 regulation to coordinate developmental timing across tissues during worm development. In the past decade, microRNAs (miRNAs) have become recognized as key regulators of gene expression in many biological pathways. These small, non-coding RNAs target specific protein-coding genes for repression. The specificity is mediated by partial base-pairing interactions between the 22 nucleotide miRNA and sequences in the target messenger RNA (mRNA). The use of imperfect base-pairing means that a single miRNA can regulate many different mRNAs, but it also means that identifying these targets is not straightforward. One of the first discovered miRNAs, let-7, generally promotes cellular differentiation pathways through a repertoire of targets that is yet to be fully described. Here we utilized molecular and genetic approaches to identify biologically relevant targets of the let-7 miRNA in Caenorhabditis elegans. Our analyses indicate that let-7 regulates a large cast of genes, both directly and indirectly. Loss of let-7 activity in C. elegans results in multiple developmental abnormalities and, ultimately, death. We uncovered new targets of let-7 that contribute to these phenotypes when they fail to be properly regulated. Given the highly conserved nature of let-7 from worms to humans, our studies highlight new genes and pathways potentially under let-7 regulation across species.
Collapse
Affiliation(s)
- Shaun E. Hunter
- Division of Biology, University of California San Diego, La Jolla, California, United States of America
| | - Emily F. Finnegan
- Division of Biology, University of California San Diego, La Jolla, California, United States of America
| | - Dimitrios G. Zisoulis
- Division of Biology, University of California San Diego, La Jolla, California, United States of America
| | - Michael T. Lovci
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
- Stem Cell Program, University of California San Diego, La Jolla, California, United States of America
- Institute for Genomic Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Katya V. Melnik-Martinez
- Division of Biology, University of California San Diego, La Jolla, California, United States of America
| | - Gene W. Yeo
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, California, United States of America
- Stem Cell Program, University of California San Diego, La Jolla, California, United States of America
- Institute for Genomic Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Amy E. Pasquinelli
- Division of Biology, University of California San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
34
|
Webster CM, Deline ML, Watts JL. Stress response pathways protect germ cells from omega-6 polyunsaturated fatty acid-mediated toxicity in Caenorhabditis elegans. Dev Biol 2012; 373:14-25. [PMID: 23064027 DOI: 10.1016/j.ydbio.2012.10.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2012] [Revised: 09/24/2012] [Accepted: 10/01/2012] [Indexed: 12/31/2022]
Abstract
Polyunsaturated fatty acids serve both structural and functional roles as membrane components and precursors for a number of different factors involved in inflammation and signaling. These fatty acids are required in the human diet, although excess dietary intake of omega-6 fatty polyunsaturated fatty acids may have a negative influence on human health. In the model nematode, Caenorhabditis elegans, dietary exposure to dihomo-gamma-linolenic acid (DGLA), an omega-6 fatty acid, causes the destruction of germ cells and leads to sterility. In this study we used genetic and microscopic approaches to further characterize this phenomenon. We found that strains carrying mutations in genes involved in lipid homeostasis enhanced sterility phenotypes, while mutations reducing the activity of the conserved insulin/IGF signaling pathway suppressed sterility phenotypes. Exposure to a mild heat stress prior to omega-6 fatty acid treatment led to an adaptive or hormetic response, resulting in less sterility. Mutations in skn-1 and knockdown of genes encoding phase II detoxification enzymes led to increased sterility in the presence of dietary DGLA. Thus, detoxification systems and genetic changes that increase overall stress responses protect the germ cells from destruction. Microscopic analyses revealed that dietary DGLA leads to deterioration of germ cell membranes in the proliferative and transition zones of the developing germ line. Together, these data demonstrate that specific omega-6 polyunsaturated fatty acids, or molecules derived from them, are transported to the germ line where they disrupt the rapidly expanding germ cell membranes, leading to germ cell death.
Collapse
Affiliation(s)
- Christopher M Webster
- School of Molecular Biosciences and Center for Reproductive Biology, Washington State University, Pullman, WA 99614-6340, USA
| | | | | |
Collapse
|
35
|
Shafaq-Zadah M, Brocard L, Solari F, Michaux G. AP-1 is required for the maintenance of apico-basal polarity in the C. elegans intestine. Development 2012; 139:2061-70. [DOI: 10.1242/dev.076711] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Epithelial tubes perform functions that are essential for the survival of multicellular organisms. Understanding how their polarised features are maintained is therefore crucial. By analysing the function of the clathrin adaptor AP-1 in the C. elegans intestine, we found that AP-1 is required for epithelial polarity maintenance. Depletion of AP-1 subunits does not affect epithelial polarity establishment or the formation of the intestinal lumen. However, the loss of AP-1 affects the polarised distribution of both apical and basolateral transmembrane proteins. Moreover, it triggers de novo formation of ectopic apical lumens between intestinal cells along the lateral membranes later during embryogenesis. We also found that AP-1 is specifically required for the apical localisation of the small GTPase CDC-42 and the polarity determinant PAR-6. Our results demonstrate that AP-1 controls an apical trafficking pathway required for the maintenance of epithelial polarity in vivo in a tubular epithelium.
Collapse
Affiliation(s)
- Massiullah Shafaq-Zadah
- INSERM Avenir group, F-35043 Rennes, France
- CNRS, UMR6061, Institut de Génétique et Développement de Rennes, F-35043 Rennes, France
- Université de Rennes 1, UEB, IFR140, Faculté de Médecine, F-35043 Rennes, France
| | - Lysiane Brocard
- INSERM Avenir group, F-35043 Rennes, France
- CNRS, UMR6061, Institut de Génétique et Développement de Rennes, F-35043 Rennes, France
- Université de Rennes 1, UEB, IFR140, Faculté de Médecine, F-35043 Rennes, France
| | - Florence Solari
- CNRS UMR5534, Centre de Génétique et de Physiologie Moléculaires et Cellulaires, Université Claude Bernard Lyon 1 F-69622 Villeurbanne, France
| | - Grégoire Michaux
- INSERM Avenir group, F-35043 Rennes, France
- CNRS, UMR6061, Institut de Génétique et Développement de Rennes, F-35043 Rennes, France
- Université de Rennes 1, UEB, IFR140, Faculté de Médecine, F-35043 Rennes, France
| |
Collapse
|
36
|
Overexpression of Akt1 enhances adipogenesis and leads to lipoma formation in zebrafish. PLoS One 2012; 7:e36474. [PMID: 22623957 PMCID: PMC3356305 DOI: 10.1371/journal.pone.0036474] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Accepted: 04/04/2012] [Indexed: 12/22/2022] Open
Abstract
Background Obesity is a complex, multifactorial disorder influenced by the interaction of genetic, epigenetic, and environmental factors. Obesity increases the risk of contracting many chronic diseases or metabolic syndrome. Researchers have established several mammalian models of obesity to study its underlying mechanism. However, a lower vertebrate model for conveniently performing drug screening against obesity remains elusive. The specific aim of this study was to create a zebrafish obesity model by over expressing the insulin signaling hub of the Akt1 gene. Methodology/Principal Findings Skin oncogenic transformation screening shows that a stable zebrafish transgenic of Tg(krt4Hsa.myrAkt1)cy18 displays severely obese phenotypes at the adult stage. In Tg(krt4:Hsa.myrAkt1)cy18, the expression of exogenous human constitutively active Akt1 (myrAkt1) can activate endogenous downstream targets of mTOR, GSK-3α/β, and 70S6K. During the embryonic to larval transitory phase, the specific over expression of myrAkt1 in skin can promote hypertrophic and hyperplastic growth. From 21 hour post-fertilization (hpf) onwards, myrAkt1 transgene was ectopically expressed in several mesenchymal derived tissues. This may be the result of the integration position effect. Tg(krt4:Hsa.myrAkt1)cy18 caused a rapid increase of body weight, hyperplastic growth of adipocytes, abnormal accumulation of fat tissues, and blood glucose intolerance at the adult stage. Real-time RT-PCR analysis showed the majority of key genes on regulating adipogenesis, adipocytokine, and inflammation are highly upregulated in Tg(krt4:Hsa.myrAkt1)cy18. In contrast, the myogenesis- and skeletogenesis-related gene transcripts are significantly downregulated in Tg(krt4:Hsa.myrAkt1)cy18, suggesting that excess adipocyte differentiation occurs at the expense of other mesenchymal derived tissues. Conclusion/Significance Collectively, the findings of this study provide direct evidence that Akt1 signaling plays an important role in balancing normal levels of fat tissue in vivo. The obese zebrafish examined in this study could be a new powerful model to screen novel drugs for the treatment of human obesity.
Collapse
|
37
|
Fuchs TM, Eisenreich W, Kern T, Dandekar T. Toward a Systemic Understanding of Listeria monocytogenes Metabolism during Infection. Front Microbiol 2012; 3:23. [PMID: 22347216 PMCID: PMC3271275 DOI: 10.3389/fmicb.2012.00023] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2011] [Accepted: 01/13/2012] [Indexed: 02/03/2023] Open
Abstract
Listeria monocytogenes is a foodborne human pathogen that can cause invasive infection in susceptible animals and humans. For proliferation within hosts, this facultative intracellular pathogen uses a reservoir of specific metabolic pathways, transporter, and enzymatic functions whose expression requires the coordinated activity of a complex regulatory network. The highly adapted metabolism of L. monocytogenes strongly depends on the nutrient composition of various milieus encountered during infection. Transcriptomic and proteomic studies revealed the spatial-temporal dynamic of gene expression of this pathogen during replication within cultured cells or in vivo. Metabolic clues are the utilization of unusual C(2)- and C(3)-bodies, the metabolism of pyruvate, thiamine availability, the uptake of peptides, the acquisition or biosynthesis of certain amino acids, and the degradation of glucose-phosphate via the pentose phosphate pathway. These examples illustrate the interference of in vivo conditions with energy, carbon, and nitrogen metabolism, thus affecting listerial growth. The exploitation, analysis, and modeling of the available data sets served as a first attempt to a systemic understanding of listerial metabolism during infection. L. monocytogenes might serve as a model organism for systems biology of a Gram-positive, facultative intracellular bacterium.
Collapse
Affiliation(s)
- Thilo M. Fuchs
- Abteilung Mikrobiologie, Zentralinstitut für Ernährungs- und Lebensmittelforschung, Technische Universität MünchenFreising, Germany
- Lehrstuhl für Mikrobielle Ökologie, Department Biowissenschaften, Wissenschaftszentrum Weihenstephan, Technische Universität MünchenFreising, Germany
| | | | - Tanja Kern
- Abteilung Mikrobiologie, Zentralinstitut für Ernährungs- und Lebensmittelforschung, Technische Universität MünchenFreising, Germany
| | - Thomas Dandekar
- Abteilung Bioinformatik, Theodor-Boveri-Institut (Biozentrum), Universität WürzburgWürzburg, Germany
| |
Collapse
|
38
|
Nerveless and gutsy: intestinal nutrient sensing from invertebrates to humans. Semin Cell Dev Biol 2012; 23:614-20. [PMID: 22248674 PMCID: PMC3712190 DOI: 10.1016/j.semcdb.2012.01.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 12/27/2011] [Accepted: 01/04/2012] [Indexed: 12/22/2022]
Abstract
The increasingly recognized role of gastrointestinal signals in the regulation of food intake, insulin production and peripheral nutrient storage has prompted a surge of interest in studying how the gastrointestinal tract senses and responds to nutritional information. Identification of metabolically important intestinal nutrient sensors could provide potential new drug targets for the treatment of diabetes, obesity and gastrointestinal disorders. From a more fundamental perspective, the study of intestinal chemosensation is revealing novel, non-neuronal modes of communication involving differentiated epithelial cells. It is also identifying signalling mechanisms downstream of not only canonical receptors but also nutrient transporters, thereby supporting a chemosensory role for “transceptors” in the intestine. This review describes known and proposed mechanisms of intestinal carbohydrate, protein and lipid sensing, best characterized in mammalian systems. It also highlights the potential of invertebrate model systems such as C. elegans and Drosophila melanogaster by summarizing known examples of molecular evolutionary conservation. Recently developed genetic tools in Drosophila, an emerging model system for the study of physiology and metabolism, allow the temporal, spatial and high-throughput manipulation of putative intestinal sensors. Hence, fruit flies may prove particularly suited to the study of the link between intestinal nutrient sensing and metabolic homeostasis.
Collapse
|
39
|
Barros AGDA, Liu J, Lemieux GA, Mullaney BC, Ashrafi K. Analyses of C. elegans fat metabolic pathways. Methods Cell Biol 2012; 107:383-407. [PMID: 22226531 DOI: 10.1016/b978-0-12-394620-1.00013-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In Caenorhabdatis elegans as in other animals, fat regulation reflects the outcome of behavioral, physiological, and metabolic processes. The amenability of C. elegans to experimentation has led to utilization of this organism for elucidating the complex homeostatic mechanisms that underlie energy balance in intact organisms. The optical advantages of C. elegans further offer the possibility of studying cell biological mechanisms of fat uptake, transport, storage, and utilization, perhaps in real time. Here, we discuss the rationale as well as advantages and potential pitfalls of methods used thus far to study metabolism and fat regulation, specifically triglyceride metabolism, in C. elegans. We provide detailed methods for visualization of fat depots in fixed animals using histochemical stains and in live animals by vital dyes. Protocols are provided and discussed for chloroform-based extraction of total lipids from C. elegans homogenates used to assess total triglyceride or phospholipid content by methods such as thin-layer chromatography or used to obtain fatty acid profiles by methods such as gas chromatography/mass spectrometry. Additionally, protocols are provided for the determination of rates of intestinal fatty acid uptake and fatty acid breakdown by β-oxidation. Finally, we discuss methods for determining rates of de novo fat synthesis and Raman scattering approaches that have recently been employed to investigate C. elegans lipids without reliance on invasive techniques. As the C. elegans fat field is relatively new, we anticipate that the indicated methods will likely be improved upon and expanded as additional researchers enter this field.
Collapse
|
40
|
Hindlet P, Barraud C, Boschat L, Farinotti R, Bado A, Buyse M. Rosiglitazone and metformin have opposite effects on intestinal absorption of oligopeptides via the proton-dependent PepT1 transporter. Mol Pharmacol 2011; 81:319-27. [PMID: 22108913 DOI: 10.1124/mol.111.073874] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The intestinal H(+)/peptide cotransporter 1 (PepT1) plays a major role in nitrogen supply to the body by mediating intestinal absorption of di- and tripeptides. Previous studies have reported that in animal models of type 2 diabetes/obesity, PepT1 activity and expression were markedly reduced. This prompted us to investigate the effects of two antidiabetic drugs, rosiglitazone and metformin, on PepT1 activity/expression in a murine diet-induced obesity model. C57BL/6J male mice were fed a high-fat diet (HFD) or a standard chow for 6 weeks and then were treated for 7 days with metformin (250 mg/kg/day) and/or rosiglitazone (8 mg/kg/day). For in vitro studies, Caco-2 enterocyte-like cells were treated for 7 days with metformin (10 mM) and/or rosiglitazone (10 μM). A 7-day rosiglitazone treatment increased PepT1 activity and prevented the 2-fold HFD-induced reduction in PepT1 transport. Metformin alone did not modify PepT1 activity but counteracted rosiglitazone-induced PepT1-mediated transport. As with the in vivo studies, rosiglitazone treatment up-regulated PepT1 transport activity with concomitant induction of S6 ribosomal protein activation in vitro. Furthermore, metformin decreased PepT1 expression (mRNA and protein) and its transport activity. The effect of metformin was linked to a reduction of phosphorylated S6 ribosomal protein (active form) and of phosphorylated 4E-BP1 (inactive form), a translation repressor. These data demonstrate that two antidiabetic drugs exert opposite effects on the PepT1 transport function probably through direct action on enterocytes. In our type 2 diabetes/obesity model, rosiglitazone, a peroxisome proliferator-activated receptor-γ agonist compensated for the HFD-induced PepT1 down-regulation, whereas metformin reversed rosiglitazone activity at the translational level.
Collapse
Affiliation(s)
- Patrick Hindlet
- Clinical Pharmacy Department (EA4123), Paris-Sud 11 University, Châtenay-Malabry, France.
| | | | | | | | | | | |
Collapse
|
41
|
Benner J, Daniel H, Spanier B. A glutathione peroxidase, intracellular peptidases and the TOR complexes regulate peptide transporter PEPT-1 in C. elegans. PLoS One 2011; 6:e25624. [PMID: 21980510 PMCID: PMC3182239 DOI: 10.1371/journal.pone.0025624] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 09/08/2011] [Indexed: 11/18/2022] Open
Abstract
The intestinal peptide transporter PEPT-1 in Caenorhabditis elegans is a rheogenic H+-dependent carrier responsible for the absorption of di- and tripeptides. Transporter-deficient pept-1(lg601) worms are characterized by impairments in growth, development and reproduction and develop a severe obesity like phenotype. The transport function of PEPT-1 as well as the influx of free fatty acids was shown to be dependent on the membrane potential and on the intracellular pH homeostasis, both of which are regulated by the sodium-proton exchanger NHX-2. Since many membrane proteins commonly function as complexes, there could be proteins that possibly modulate PEPT-1 expression and function. A systematic RNAi screening of 162 genes that are exclusively expressed in the intestine combined with a functional transport assay revealed four genes with homologues existing in mammals as predicted PEPT-1 modulators. While silencing of a glutathione peroxidase surprisingly caused an increase in PEPT-1 transport function, silencing of the ER to Golgi cargo transport protein and of two cytosolic peptidases reduced PEPT-1 transport activity and this even corresponded with lower PEPT-1 protein levels. These modifications of PEPT-1 function by gene silencing of homologous genes were also found to be conserved in the human epithelial cell line Caco-2/TC7 cells. Peptidase inhibition, amino acid supplementation and RNAi silencing of targets of rapamycin (TOR) components in C. elegans supports evidence that intracellular peptide hydrolysis and amino acid concentration are a part of a sensing system that controls PEPT-1 expression and function and that involves the TOR complexes TORC1 and TORC2.
Collapse
Affiliation(s)
- Jacqueline Benner
- ZIEL Research Center of Nutrition and Food Sciences, Abteilung Biochemie, Technische Universität München, Freising, Germany
| | - Hannelore Daniel
- ZIEL Research Center of Nutrition and Food Sciences, Abteilung Biochemie, Technische Universität München, Freising, Germany
| | - Britta Spanier
- ZIEL Research Center of Nutrition and Food Sciences, Abteilung Biochemie, Technische Universität München, Freising, Germany
- * E-mail:
| |
Collapse
|
42
|
Knight AJ, Behm CA. Minireview: the role of the vacuolar ATPase in nematodes. Exp Parasitol 2011; 132:47-55. [PMID: 21959022 DOI: 10.1016/j.exppara.2011.09.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 09/08/2011] [Accepted: 09/13/2011] [Indexed: 11/28/2022]
Abstract
The vacuolar ATPase enzyme complex (V-ATPase) pumps protons across membranes, energised by hydrolysis of ATP. It is involved in many physiological processes and has been implicated in many different diseases. While the broader functions of V-ATPases have been reviewed extensively, the role of this complex in nematodes specifically has not. Here, the essential role of the V-ATPase in nematode nutrition, osmoregulation, synthesis of the cuticle, neurobiology and reproduction is discussed. Based on the requirement of V-ATPase activity, or components of the V-ATPase, for these processes, the potential of the V-ATPase as a drug target for nematode parasites, which cause a significant burden to human health and agriculture, is also discussed. The V-ATPase has all the characteristics of a suitable drug target against nematodes, however the challenge will be to develop a high-throughput assay with which to test potential inhibitors.
Collapse
Affiliation(s)
- Alison J Knight
- Research School of Biology, College of Medicine, Biology and Environment, Australian National University, Canberra ACT 0200, Australia
| | | |
Collapse
|
43
|
Verri T, Terova G, Dabrowski K, Saroglia M. Peptide transport and animal growth: the fish paradigm. Biol Lett 2011; 7:597-600. [PMID: 21389019 DOI: 10.1098/rsbl.2010.1164] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Protein digestion products are transported from the intestinal lumen into the enterocyte both in the form of free amino acids (AAs), by a large variety of brush border membrane AA transporters, and in the form of di/tripeptides, by a single brush border membrane transporter known as PEPtide Transporter 1 (PEPT1). Recent data indicate that, at least in teleost fish, PEPT1 plays a significant role in animal growth by operating, at the gastrointestinal level, as part of an integrated response network to food availability that directly supports body weight. Notably, PEPT1 responds to both fasting and refeeding and is involved in a phenomenon known as compensatory growth (a phase of accelerated growth when food levels are restored after a period of growth depression). In particular, PEPT1 expression decreases during fasting and increases during refeeding, which is the opposite of what observed so far in mammals and birds. These findings in teleost fish document, to our knowledge, for the first time in a vertebrate model, a direct correlation between the expression of an intestinal transporter, such as PEPT1, primarily involved in the uptake of dietary protein degradation products and animal growth.
Collapse
Affiliation(s)
- Tiziano Verri
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy.
| | | | | | | |
Collapse
|
44
|
Martin FPJ, Spanier B, Collino S, Montoliu I, Kolmeder C, Giesbertz P, Affolter M, Kussmann M, Daniel H, Kochhar S, Rezzi S. Metabotyping of Caenorhabditis elegans and their culture media revealed unique metabolic phenotypes associated to amino acid deficiency and insulin-like signaling. J Proteome Res 2011; 10:990-1003. [PMID: 21275419 DOI: 10.1021/pr100703a] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Insulin/IGF-like signaling (IIS) and nutrient sensing are among the most potent regulators of health status and aging. Here, a global view of the metabolic changes in C. elegans with impaired function of IIS represented by daf-2 and daf-16 and the intestinal di- and tripeptide transport pept-1 was generated using (1)H nuclear magnetic resonance spectroscopic analysis of worm extracts and spent culture media. We showed that specific metabolic profiles were significantly associated with each type of mutant. On the basis of the metabonomics data, selected underlying processes were further investigated using proteomic and transcriptomic approaches. The observed changes suggest a decreased activity of the one carbon metabolism in pept-1(lg601) mutants. Higher concentration of branched-chain amino acids (BCAA) and altered transcript levels of genes involved in BCAA metabolism were observed in long-living strains daf-2(e1370) and daf-2(e1370);pept-1(lg601) when compared to wild types and daf-16(m26);daf-2(e1370);pept-1(lg601) C. elegans, suggesting a DAF-16-dependent mechanism.
Collapse
|
45
|
Mullaney BC, Blind RD, Lemieux GA, Perez CL, Elle IC, Faergeman NJ, Van Gilst MR, Ingraham HA, Ashrafi K. Regulation of C. elegans fat uptake and storage by acyl-CoA synthase-3 is dependent on NR5A family nuclear hormone receptor nhr-25. Cell Metab 2010; 12:398-410. [PMID: 20889131 PMCID: PMC2992884 DOI: 10.1016/j.cmet.2010.08.013] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2009] [Revised: 04/28/2010] [Accepted: 06/23/2010] [Indexed: 01/09/2023]
Abstract
Acyl-CoA synthases are important for lipid synthesis and breakdown, generation of signaling molecules, and lipid modification of proteins, highlighting the challenge of understanding metabolic pathways within intact organisms. From a C. elegans mutagenesis screen, we found that loss of ACS-3, a long-chain acyl-CoA synthase, causes enhanced intestinal lipid uptake, de novo fat synthesis, and accumulation of enlarged, neutral lipid-rich intestinal depots. Here, we show that ACS-3 functions in seam cells, epidermal cells anatomically distinct from sites of fat uptake and storage, and that acs-3 mutant phenotypes require the nuclear hormone receptor NHR-25, a key regulator of C. elegans molting. Our findings suggest that ACS-3-derived long-chain fatty acyl-CoAs, perhaps incorporated into complex ligands such as phosphoinositides, modulate NHR-25 function, which in turn regulates an endocrine program of lipid uptake and synthesis. These results reveal a link between acyl-CoA synthase function and an NR5A family nuclear receptor in C. elegans.
Collapse
Affiliation(s)
- Brendan C Mullaney
- Graduate Program in Neuroscience, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA; UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Raymond D Blind
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - George A Lemieux
- Department of Anatomy, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Carissa L Perez
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Molecular and Cellular Biology Program, University of Washington, Seattle, WA 98195, USA
| | - Ida C Elle
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Nils J Faergeman
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Marc R Van Gilst
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Holly A Ingraham
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kaveh Ashrafi
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94158, USA; UCSF Diabetes Center, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
46
|
Spanier B, Rubio-Aliaga I, Hu H, Daniel H. Altered signalling from germline to intestine pushes daf-2;pept-1 Caenorhabditis elegans into extreme longevity. Aging Cell 2010; 9:636-46. [PMID: 20550516 DOI: 10.1111/j.1474-9726.2010.00591.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The insulin-like signalling pathway is a central regulator of development, metabolism, stress resistance and lifespan in eukaryotes. Caenorhabditis elegans daf-2(e1370) animals with a loss-of-function mutation in the insulin-like receptor live twice as long as wild-type animals, and the additional knockout of the intestinal di- and tripeptide transporter pept-1 further increases lifespan by 60%. In assessing the underlying molecular mechanisms for this phenomenon, microarray-based transcriptome data sets of daf-2(e1370) and daf-2(e1370);pept-1(lg601) animals were compared with a focus on genes that showed significantly higher changes in expression levels in daf-2;pept-1 than in daf-2. We identified 187 genes with at least fourfold decreased transcript levels and 170 with more than a fourfold increase. A large fraction of the down-regulated genes encode proteins involved in germline proliferation and reproduction. The DAF-9/DAF-12 signalling cascade was identified as a prime pathway that mediates the longevity of daf-2;pept-1 with a strict dependance on DAF-16. Loss of DAF-9/DAF-12 or KRI-1 reduces the lifespan of daf-2;pept-1 to that of the daf-2 mutant. Amongst the DAF-16 target genes, numerous enzymes involved in the defence of reactive oxygen species were with increased expression level in daf-2;pept-1. On a functional level, it was demonstrated that amongst those, a high de novo synthesis rate of glutathione is most important for the longevity phenotype of this strain. Taken together, a close interdependence of endocrine hormone signalling from germline to intestine was identified as an essential element in the control of the extreme longevity of C. elegans lacking a proper function of the insulin receptor and lacking the intestinal peptide transporter.
Collapse
Affiliation(s)
- Britta Spanier
- ZIEL Research Center of Nutrition and Food Sciences, Technische Universität München, Freising, Germany.
| | | | | | | |
Collapse
|
47
|
Elle IC, Olsen LCB, Pultz D, Rødkaer SV, Faergeman NJ. Something worth dyeing for: molecular tools for the dissection of lipid metabolism in Caenorhabditis elegans. FEBS Lett 2010; 584:2183-93. [PMID: 20371247 DOI: 10.1016/j.febslet.2010.03.046] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Revised: 03/30/2010] [Accepted: 03/30/2010] [Indexed: 10/19/2022]
Abstract
The nematode Caenorhabditis elegans (C. elegans) has during the last decade emerged as an invaluable eukaryotic model organism to understand the metabolic and neuro-endocrine regulation of lipid accumulation. The fundamental pathways of food intake, digestion, metabolism, and signalling are evolutionary conserved between mammals and worms making C. elegans a genetically and metabolically extremely tractable model to decipher new regulatory mechanisms of lipid storage and to understand how nutritional and genetic perturbations can lead to obesity and other metabolic diseases. Besides providing an overview of the most important regulatory mechanisms of lipid accumulation in C. elegans, we also critically assess the current methodologies to monitor lipid storage and content as various methods differ in their applicability, consistency, and simplicity.
Collapse
Affiliation(s)
- Ida Coordt Elle
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense M, Denmark
| | | | | | | | | |
Collapse
|
48
|
Ballard SL, Jarolimova J, Wharton KA. Gbb/BMP signaling is required to maintain energy homeostasis in Drosophila. Dev Biol 2009; 337:375-85. [PMID: 19914231 DOI: 10.1016/j.ydbio.2009.11.011] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Revised: 10/19/2009] [Accepted: 11/06/2009] [Indexed: 12/31/2022]
Abstract
The coordination of animal growth and development requires adequate nutrients. During times of insufficient food, developmental progression is slowed and stored energy is utilized to ensure that cell and tissue survival are maintained. Here, we report our finding that the Gbb/BMP signaling pathway, known to play an important role in many developmental processes in both vertebrates and invertebrates, is critical in the Drosophila larval fat body for regulating energy homeostasis. Animals with mutations in the Drosophila BMP-5,7 orthologue, glass bottom boat (gbb), or in its signaling components, display phenotypes similar to nutrient-deprived and Tor mutant larvae. These phenotypes include a developmental delay with reduced overall growth, a transparent appearance, and altered total lipid, glucose and trehalose levels. We find that Gbb/BMP signaling is required in the larval fat body for maintaining proper metabolism, yet interestingly, following nutrient deprivation larvae in turn show a loss of BMP signaling in fat body cells indicating that Gbb/BMP signaling is a central player in homeostasis. Finally, despite strong phenotypic similarities between nutrient-compromised animals and gbb mutants, distinct differences are observed in the expression of a group of starvation responsive genes. Overall, our results implicate Gbb/BMP signaling as a new pathway critical for positive regulation of nutrient storage and energy homeostasis during development.
Collapse
Affiliation(s)
- Shannon L Ballard
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02912, USA
| | | | | |
Collapse
|
49
|
Brooks KK, Liang B, Watts JL. The influence of bacterial diet on fat storage in C. elegans. PLoS One 2009; 4:e7545. [PMID: 19844570 PMCID: PMC2760100 DOI: 10.1371/journal.pone.0007545] [Citation(s) in RCA: 220] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2009] [Accepted: 09/30/2009] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND The nematode Caenorhabditis elegans has emerged as an important model for studies of the regulation of fat storage. C. elegans feed on bacteria, and various strains of E. coli are commonly used in research settings. However, it is not known whether particular bacterial diets affect fat storage and metabolism. METHODOLOGY/PRINCIPAL FINDINGS Fat staining of fixed nematodes, as well as biochemical analysis of lipid classes, revealed considerable differences in fat stores in C. elegans growing on four different E. coli strains. Fatty acid composition and carbohydrate levels differ in the E. coli strains examined in these studies, however these nutrient differences did not appear to have a causative effect on fat storage levels in worms. Analysis of C. elegans strains carrying mutations disrupting neuroendocrine and other fat-regulatory pathways demonstrated that the intensity of Nile Red staining of live worms does not correlate well with biochemical methods of fat quantification. Several neuroendocrine pathway mutants and eating defective mutants show higher or lower fat storage levels than wild type, however, these mutants still show differences in fat stores when grown on different bacterial strains. Of all the mutants tested, only pept-1 mutants, which lack a functional intestinal peptide transporter, fail to show differential fat stores. Furthermore, fatty acid analysis of triacylglycerol stores reveals an inverse correlation between total fat stores and the levels of 15-methylpalmitic acid, derived from leucine catabolism. CONCLUSIONS These studies demonstrate that nutritional cues perceived in the intestine regulate fat storage levels independently of neuroendocrine cues. The involvement of peptide transport and the accumulation of a fatty acid product derived from an amino acid suggest that specific peptides or amino acids may provide nutritional signals regulating fat metabolism and fat storage levels.
Collapse
Affiliation(s)
- Kyleann K. Brooks
- School of Molecular Biosciences, Washington State University, Pullman, Washington, United States of America
| | - Bin Liang
- School of Molecular Biosciences, Washington State University, Pullman, Washington, United States of America
| | - Jennifer L. Watts
- School of Molecular Biosciences, Washington State University, Pullman, Washington, United States of America
- * E-mail:
| |
Collapse
|
50
|
Allman E, Johnson D, Nehrke K. Loss of the apical V-ATPase a-subunit VHA-6 prevents acidification of the intestinal lumen during a rhythmic behavior in C. elegans. Am J Physiol Cell Physiol 2009; 297:C1071-81. [PMID: 19741196 DOI: 10.1152/ajpcell.00284.2009] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In Caenorhabditis elegans, oscillations of intestinal pH contribute to the rhythmic defecation behavior, but the acid-base transport mechanisms that facilitate proton movement are not well understood. Here, we demonstrate that VHA-6, an intestine-specific a-subunit of the H(+)-K(+)-ATPase complex (V-ATPase), resides in the apical membrane of the intestinal epithelial cells and is required for luminal acidification. Disruption of the vha-6 gene led to early developmental arrest; the arrest phenotype could be complemented by expression of a fluorescently labeled vha-6 transgene. To study the contribution of vha-6 to pH homeostasis in larval worms, we used a partial reduction of function through postembryonic single-generation RNA interference. We demonstrate that the inability to fully acidify the intestinal lumen coincides with a defect in pH recovery of the intestinal epithelial cells, suggesting that VHA-6 is essential for proton pumping following defecation. Moreover, intestinal dipeptide accumulation and fat storage are compromised by the loss of VHA-6, suggesting that luminal acidification promotes nutrient uptake in worms, as well as in mammals. Since acidified intracellular vesicles and autofluorescent storage granules are indistinguishable between the vha-6 mutant and controls, it is likely that the nutrient-restricted phenotype is due to a loss of plasma membrane V-ATPase activity specifically. These data establish a simple genetic model for proton pump-driven acidification. Since defecation occurs at 45-s intervals in worms, this model represents an opportunity to study acute regulation of V-ATPase activity on a short time scale and may be useful in the study of alternative treatments for acid-peptic disorders.
Collapse
Affiliation(s)
- Erik Allman
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York 14642, USA
| | | | | |
Collapse
|