1
|
Phonsiri K, Mavichak R, Panserat S, Boonanuntanasarn S. Differential responses of hepatopancreas transcriptome between fast and slow growth in giant freshwater prawns (Macrobrachium rosenbergii) fed a plant-based diet. Sci Rep 2024; 14:4957. [PMID: 38418833 PMCID: PMC10902295 DOI: 10.1038/s41598-024-54349-6] [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: 02/01/2023] [Accepted: 02/12/2024] [Indexed: 03/02/2024] Open
Abstract
Efficient utilisation of plant-based diets in the giant freshwater prawn, Marcrobrachium rosenbergii, varies according to individual, suggesting that it might be associated with differences in physiological and metabolic responses. Therefore, we aimed to investigate the individual differences in the growth response of shrimp fed to a soybean-based diet (SBM). Two hundred shrimp were fed SBM for 90 days, and specific growth rate (SGR) was determined individually. Fast- and slow-growing shrimp (F-shrimp vs. S-shrimp), with the highest and lowest 5% SGRs, respectively, were sampled to determine haemolymph chemistry and carcass composition. The hepatopancreas of these shrimps were used for transcriptome analysis through RNA sequencing (RNA-Seq). The results showed no significant differences in haemolymph chemistry parameters. In terms of carcass proximate composition, F-shrimp exhibited higher protein composition than did S-shrimp, suggesting that F-shrimp have higher protein anabolism. Using RNA-seq and real-time reverse transcription polymerase chain reaction (qRT-PCR), the expression levels of several genes encoding physiologic and metabolic enzymes were found to be upregulated in F-shrimp compared to in S-shrimp, suggesting that these enzymes/proteins mediated the efficient use of SBM-based diets for growth promotion in shrimp. Various DEGs associated with the immune system were observed, indicating a difference in immune processes between F- and S-shrimp. The expression of several housekeeping genes was found to be upregulated in S-shrimp. Collectively, the upregulated expression of several enzymes associated with physiological and/or metabolic processes and increased protein anabolism may be attributed to the efficient use of SBM for maximal growth in shrimp.
Collapse
Affiliation(s)
- Khanakorn Phonsiri
- School of Animal Technology and Innovation, Institute of Agricultural Technology, Suranaree University of Technology, 111 University Avenue, Muang, Nakhon Ratchasima, 30000, Thailand
| | - Rapeepat Mavichak
- Aquatic Animal Health Research Center, Charoen Pokphand Co. Ltd., Rama 2 Rd., Km 41.5, Bangtorat, Muang Samutsakorn, Samutsakorn, 74000, Thailand
| | - Stephane Panserat
- INRAE, Université de Pau et des Pays de l'Adour, E2S UPPA, NuMéA, Saint-Pée-Sur-Nivelle, France
| | - Surintorn Boonanuntanasarn
- School of Animal Technology and Innovation, Institute of Agricultural Technology, Suranaree University of Technology, 111 University Avenue, Muang, Nakhon Ratchasima, 30000, Thailand.
| |
Collapse
|
2
|
Neuman SD, Levine TP, Bashirullah A. A novel superfamily of bridge-like lipid transfer proteins. Trends Cell Biol 2022; 32:962-974. [PMID: 35491307 PMCID: PMC9588498 DOI: 10.1016/j.tcb.2022.03.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/28/2022] [Accepted: 03/30/2022] [Indexed: 01/21/2023]
Abstract
Lipid transfer proteins mediate nonvesicular transport of lipids at membrane contact sites to regulate the lipid composition of organelle membranes. Recently, a new type of bridge-like lipid transfer protein has emerged; these proteins contain a long hydrophobic groove and can mediate bulk transport of lipids between organelles. Here, we review recent insights into the structure of these proteins and identify a repeating modular unit that we propose to name the repeating β-groove (RBG) domain. This new structural understanding conceptually unifies all the RBG domain-containing lipid transfer proteins as members of an RBG protein superfamily. We also examine the biological functions of these lipid transporters in normal physiology and disease and speculate on the evolutionary origins of RBG proteins in bacteria.
Collapse
Affiliation(s)
- Sarah D Neuman
- Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI 53705-2222, USA
| | - Tim P Levine
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
| | - Arash Bashirullah
- Division of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, WI 53705-2222, USA.
| |
Collapse
|
3
|
Nishioka K, Imai Y, Yoshino H, Li Y, Funayama M, Hattori N. Clinical Manifestations and Molecular Backgrounds of Parkinson's Disease Regarding Genes Identified From Familial and Population Studies. Front Neurol 2022; 13:764917. [PMID: 35720097 PMCID: PMC9201061 DOI: 10.3389/fneur.2022.764917] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Over the past 20 years, numerous robust analyses have identified over 20 genes related to familial Parkinson's disease (PD), thereby uncovering its molecular underpinnings and giving rise to more sophisticated approaches to investigate its pathogenesis. α-Synuclein is a major component of Lewy bodies (LBs) and behaves in a prion-like manner. The discovery of α-Synuclein enables an in-depth understanding of the pathology behind the generation of LBs and dopaminergic neuronal loss. Understanding the pathophysiological roles of genes identified from PD families is uncovering the molecular mechanisms, such as defects in dopamine biosynthesis and metabolism, excessive oxidative stress, dysfunction of mitochondrial maintenance, and abnormalities in the autophagy–lysosome pathway, involved in PD pathogenesis. This review summarizes the current knowledge on familial PD genes detected by both single-gene analyses obeying the Mendelian inheritance and meta-analyses of genome-wide association studies (GWAS) from genome libraries of PD. Studying the functional role of these genes might potentially elucidate the pathological mechanisms underlying familial PD and sporadic PD and stimulate future investigations to decipher the common pathways between the diseases.
Collapse
Affiliation(s)
- Kenya Nishioka
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
- *Correspondence: Kenya Nishioka
| | - Yuzuru Imai
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
- Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Yuzuru Imai
| | - Hiroyo Yoshino
- Research Institute for Diseases of Old Age, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Yuanzhe Li
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Manabu Funayama
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
- Research Institute for Diseases of Old Age, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
- Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Research Institute for Diseases of Old Age, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| |
Collapse
|
4
|
Insertional mutagenesis in Chlamydomonas reinhardtii: An effective strategy for the identification of new genes involved in the DNA damage response. Eur J Protistol 2021; 82:125855. [PMID: 34954500 DOI: 10.1016/j.ejop.2021.125855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 11/26/2021] [Accepted: 12/02/2021] [Indexed: 11/23/2022]
Abstract
The formation of double-strand breaks in DNA represents a serious stress for all types of organisms and requires a precisely regulated and organized DNA damage response (DDR) to maintain genetic information and genome integrity. Chlamydomonas reinhardtii possesses the characteristics of both plants and animals and is therefore suitable for the identification of novel genes connected to a wide spectrum of metabolic pathways, including DDR. One very effective tool for the detection and subsequent characterization of new mutants in C. reinhardtii is insertional mutagenesis. We isolated several insertion mutants sensitive to DNA-damaging agents that had disrupted or completely deleted genes with putative functions in the DDR. In most of the analysed mutants, we identified various changes at both ends and even inside the inserted cassette. Using recent information from databases, we were also able to supplement the characteristics of the previously described mutant with a pleiotropic phenotype. In addition, we confirmed the effectiveness of hairpin-PCR as a strategy for the identification of insertion flanking sites and as a tool for the detection of changes at the site of insertion, thus enabling a better understanding of insertion events.
Collapse
|
5
|
Leonzino M, Reinisch KM, De Camilli P. Insights into VPS13 properties and function reveal a new mechanism of eukaryotic lipid transport. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:159003. [PMID: 34216812 PMCID: PMC8325632 DOI: 10.1016/j.bbalip.2021.159003] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/07/2021] [Accepted: 06/09/2021] [Indexed: 02/06/2023]
Abstract
The occurrence of protein mediated lipid transfer between intracellular membranes has been known since the late 1960's. Since these early discoveries, numerous proteins responsible for such transport, which often act at membrane contact sites, have been identified. Typically, they comprise a lipid harboring module thought to shuttle back and forth between the two adjacent bilayers. Recently, however, studies of the chorein domain protein family, which includes VPS13 and ATG2, has led to the identification of a novel mechanism of lipid transport between organelles in eukaryotic cells mediated by a rod-like protein bridge with a hydrophobic groove through which lipids can slide. This mechanism is ideally suited for bulk transport of bilayer lipids to promote membrane growth. Here we describe how studies of VPS13 led to the discovery of this new mechanism, summarize properties and known roles of VPS13 proteins, and discuss how their dysfunction may lead to disease.
Collapse
Affiliation(s)
- Marianna Leonzino
- Department of Neuroscience, Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration and Repair, Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, USA; Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA; CNR Institute of Neuroscience, Milan, Italy and Humanitas Clinical and Research Center, Rozzano, MI, Italy.
| | - Karin M Reinisch
- Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| | - Pietro De Camilli
- Department of Neuroscience, Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration and Repair, Kavli Institute for Neuroscience, Yale School of Medicine, New Haven, USA; Department of Cell Biology, Yale School of Medicine, New Haven, CT, USA; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
6
|
Ugur B, Hancock-Cerutti W, Leonzino M, De Camilli P. Role of VPS13, a protein with similarity to ATG2, in physiology and disease. Curr Opin Genet Dev 2020; 65:61-68. [PMID: 32563856 PMCID: PMC7746581 DOI: 10.1016/j.gde.2020.05.027] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 05/06/2020] [Accepted: 05/21/2020] [Indexed: 12/12/2022]
Abstract
The evolutionarily conserved VPS13 family proteins have been implicated in several cellular processes. Mutations in each of the four human VPS13s cause neurodevelopmental or neurodegenerative disorders. Until recently, the molecular function of VPS13 remained elusive. Genetic, functional and structural studies have now revealed that VPS13 acts at contact sites between intracellular organelles to transport lipids by a novel mechanism: direct transfer between bilayers via a hydrophobic channel that spans its entire rod-like N-terminal half. Predicted similarities to the autophagy protein ATG2 suggested a similar role for ATG2 that has now been confirmed by structural and functional studies. Here, after a brief review of this evidence, we discuss what is known of human VPS13 proteins in physiology and disease.
Collapse
Affiliation(s)
- Berrak Ugur
- Departments of Neuroscience and Cell Biology, Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration and Repair, Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - William Hancock-Cerutti
- Departments of Neuroscience and Cell Biology, Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration and Repair, Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Marianna Leonzino
- Departments of Neuroscience and Cell Biology, Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration and Repair, Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Pietro De Camilli
- Departments of Neuroscience and Cell Biology, Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration and Repair, Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
7
|
Muñoz-Braceras S, Tornero-Écija AR, Vincent O, Escalante R. VPS13A is closely associated with mitochondria and is required for efficient lysosomal degradation. Dis Model Mech 2019; 12:dmm036681. [PMID: 30709847 PMCID: PMC6398486 DOI: 10.1242/dmm.036681] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 01/22/2019] [Indexed: 12/11/2022] Open
Abstract
Members of the VPS13 family are associated with various human diseases. In particular, the loss of function of VPS13A leads to chorea-acanthocytosis (ChAc), a rare neurodegenerative disease without available curative treatments. Autophagy has been considered a promising therapeutic target because the absence of VPS13A causes a defective autophagy flux. However, the mechanistic details of this deficiency are unknown. Here, we identified Rab7A as an interactor of one of the VPS13 family members in Dictyostelium discoideum and showed that this interaction is conserved between the human homologs VPS13A and RAB7A in HeLa cells. As RAB7A is a key player in endosome trafficking, we addressed the possible function of VPS13A in endosome dynamics and lysosome degradation. Our results suggest that the decrease in autophagy observed in the absence of VPS13A may be the result of a more general defect in endocytic trafficking and lysosomal degradation. Unexpectedly, we found that VPS13A is closely localized to mitochondria, suggesting that the role of VPS13A in the endolysosomal pathway might be related to inter-organelle communication. We show that VPS13A localizes at the interface between mitochondria-endosomes and mitochondria-endoplasmic reticulum and that the presence of membrane contact sites is altered in the absence of VPS13A. Based on these findings, we propose that therapeutic strategies aimed at modulating the endolysosomal pathway could be beneficial in the treatment of ChAc.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Sandra Muñoz-Braceras
- Instituto de Investigaciones Biomédicas Alberto Sols, Department of Experimental Models of Human Diseases, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma Madrid (UAM), 28029-Madrid, Spain
| | - Alba R Tornero-Écija
- Instituto de Investigaciones Biomédicas Alberto Sols, Department of Experimental Models of Human Diseases, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma Madrid (UAM), 28029-Madrid, Spain
| | - Olivier Vincent
- Instituto de Investigaciones Biomédicas Alberto Sols, Department of Experimental Models of Human Diseases, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma Madrid (UAM), 28029-Madrid, Spain
| | - Ricardo Escalante
- Instituto de Investigaciones Biomédicas Alberto Sols, Department of Experimental Models of Human Diseases, Consejo Superior de Investigaciones Científicas (CSIC)/Universidad Autónoma Madrid (UAM), 28029-Madrid, Spain
| |
Collapse
|
8
|
Yeshaw WM, van der Zwaag M, Pinto F, Lahaye LL, Faber AI, Gómez-Sánchez R, Dolga AM, Poland C, Monaco AP, van IJzendoorn SC, Grzeschik NA, Velayos-Baeza A, Sibon OC. Human VPS13A is associated with multiple organelles and influences mitochondrial morphology and lipid droplet motility. eLife 2019; 8:43561. [PMID: 30741634 PMCID: PMC6389287 DOI: 10.7554/elife.43561] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/10/2019] [Indexed: 02/03/2023] Open
Abstract
The VPS13A gene is associated with the neurodegenerative disorder Chorea Acanthocytosis. It is unknown what the consequences are of impaired function of VPS13A at the subcellular level. We demonstrate that VPS13A is a peripheral membrane protein, associated with mitochondria, the endoplasmic reticulum and lipid droplets. VPS13A is localized at sites where the endoplasmic reticulum and mitochondria are in close contact. VPS13A interacts with the ER residing protein VAP-A via its FFAT domain. Interaction with mitochondria is mediated via its C-terminal domain. In VPS13A-depleted cells, ER-mitochondria contact sites are decreased, mitochondria are fragmented and mitophagy is decreased. VPS13A also localizes to lipid droplets and affects lipid droplet motility. In VPS13A-depleted mammalian cells lipid droplet numbers are increased. Our data, together with recently published data from others, indicate that VPS13A is required for establishing membrane contact sites between various organelles to enable lipid transfer required for mitochondria and lipid droplet related processes.
Collapse
Affiliation(s)
- Wondwossen M Yeshaw
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marianne van der Zwaag
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Francesco Pinto
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Liza L Lahaye
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anita Ie Faber
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Rubén Gómez-Sánchez
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Amalia M Dolga
- Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), Faculty of Science and Engineering, University of Groningen, Groningen, The Netherlands
| | - Conor Poland
- Wellcome Trust Centre for Human Genetics, Oxford, United Kingdom
| | - Anthony P Monaco
- Wellcome Trust Centre for Human Genetics, Oxford, United Kingdom.,Office of the President, Tufts University, Medford, United States
| | - Sven Cd van IJzendoorn
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Nicola A Grzeschik
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Ody Cm Sibon
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
9
|
Kumar N, Leonzino M, Hancock-Cerutti W, Horenkamp FA, Li P, Lees JA, Wheeler H, Reinisch KM, De Camilli P. VPS13A and VPS13C are lipid transport proteins differentially localized at ER contact sites. J Cell Biol 2018; 217:3625-3639. [PMID: 30093493 PMCID: PMC6168267 DOI: 10.1083/jcb.201807019] [Citation(s) in RCA: 389] [Impact Index Per Article: 55.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 07/13/2018] [Accepted: 07/17/2018] [Indexed: 01/07/2023] Open
Abstract
Mutations in the human VPS13 genes are responsible for neurodevelopmental and neurodegenerative disorders including chorea acanthocytosis (VPS13A) and Parkinson's disease (VPS13C). The mechanisms of these diseases are unknown. Genetic studies in yeast hinted that Vps13 may have a role in lipid exchange between organelles. In this study, we show that the N-terminal portion of VPS13 is tubular, with a hydrophobic cavity that can solubilize and transport glycerolipids between membranes. We also show that human VPS13A and VPS13C bind to the ER, tethering it to mitochondria (VPS13A), to late endosome/lysosomes (VPS13C), and to lipid droplets (both VPS13A and VPS13C). These findings identify VPS13 as a lipid transporter between the ER and other organelles, implicating defects in membrane lipid homeostasis in neurological disorders resulting from their mutations. Sequence and secondary structure similarity between the N-terminal portions of Vps13 and other proteins such as the autophagy protein ATG2 suggest lipid transport roles for these proteins as well.
Collapse
Affiliation(s)
- Nikit Kumar
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Marianna Leonzino
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT
- Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT
| | - William Hancock-Cerutti
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT
- Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT
| | - Florian A Horenkamp
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - PeiQi Li
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Joshua A Lees
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Heather Wheeler
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT
- Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT
| | - Karin M Reinisch
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Pietro De Camilli
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT
- Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
10
|
Rzepnikowska W, Flis K, Muñoz-Braceras S, Menezes R, Escalante R, Zoladek T. Yeast and other lower eukaryotic organisms for studies of Vps13 proteins in health and disease. Traffic 2017; 18:711-719. [PMID: 28846184 DOI: 10.1111/tra.12523] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 08/23/2017] [Accepted: 08/23/2017] [Indexed: 12/25/2022]
Abstract
Human Vps13 proteins are associated with several diseases, including the neurodegenerative disorder Chorea-acanthocytosis (ChAc), yet the biology of these proteins is still poorly understood. Studies in Saccharomyces cerevisiae, Dictyostelium discoideum, Tetrahymena thermophila and Drosophila melanogaster point to the involvement of Vps13 in cytoskeleton organization, vesicular trafficking, autophagy, phagocytosis, endocytosis, proteostasis, sporulation and mitochondrial functioning. Recent findings show that yeast Vps13 binds to phosphatidylinositol lipids via 4 different regions and functions at membrane contact sites, enlarging the list of Vps13 functions. This review describes the great potential of simple eukaryotes to decipher disease mechanisms in higher organisms and highlights novel insights into the pathological role of Vps13 towards ChAc.
Collapse
Affiliation(s)
- Weronika Rzepnikowska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Krzysztof Flis
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | | - Regina Menezes
- iBET, Instituto de Biologia Experimental e Tecnológica, Oeiras, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Ricardo Escalante
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain
| | - Teresa Zoladek
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
11
|
Pappas SS, Bonifacino J, Danek A, Dauer WT, De M, De Franceschi L, DiPaolo G, Fuller R, Haucke V, Hermann A, Kornmann B, Landwehrmeyer B, Levin J, Neiman AM, Rudnicki DD, Sibon O, Velayos-Baeza A, Vonk JJ, Walker RH, Weisman LS, Albin RL. Eighth International Chorea-Acanthocytosis Symposium: Summary of Workshop Discussion and Action Points. Tremor Other Hyperkinet Mov (N Y) 2017; 7:428. [PMID: 28224046 PMCID: PMC5313633 DOI: 10.7916/d8xd127w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 01/10/2017] [Indexed: 02/05/2023] Open
Abstract
Chorea-Acanthocytosis (ChAc) is a rare hereditary neurological disorder characterized by abnormal movements, red blood cell pathology, and progressive neurodegeneration. Little is understood of the pathogenesis of ChAc and related disorders (collectively Neuroacanthocytosis). The Eighth International Chorea-Acanthocytosis Symposium was held in May 2016 in Ann Arbor, MI, USA, and focused on molecular mechanisms driving ChAc pathophysiology. Accompanying the meeting, members of the neuroacanthocytosis research community and other invited scientists met in a workshop to discuss the current understanding and next steps needed to better understand ChAc pathogenesis. These discussions identified several broad and critical needs for advancing ChAc research and patient care, and led to the definition of 18 specific action points related to functional and molecular studies, animal models, and clinical research. These action points, described below, represent tractable research goals to pursue for the next several years.
Collapse
Affiliation(s)
- Samuel S. Pappas
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Juan Bonifacino
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Adrian Danek
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität, Munich, Germany
| | - William T. Dauer
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Neurology Service, VAAAHS, University of Michigan, Ann Arbor, MI, USA
- Udall Centre, University of Michigan, Ann Arbor, MI, USA
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Mithu De
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Lucia De Franceschi
- Department of Medicine, University of Verona and Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | | | - Robert Fuller
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Volker Haucke
- Department of Molecular Pharmacology and Cell Biology, Leibniz Institut für Molekulare Pharmakologie, Berlin, Germany
| | - Andreas Hermann
- Department of Neurology, Technische Universität, Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | | | | | - Johannes Levin
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Aaron M. Neiman
- Department of Biochemistry and Cell Biology, Stony Brook University, New York, NY, USA
| | | | - Ody Sibon
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Gronigen, The Netherlands
| | | | - Jan J. Vonk
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Gronigen, The Netherlands
| | - Ruth H. Walker
- Department of Neurology, James J. Peters VAMC, Bronx, NY, USA
- Department of Neurology, Mount Sinai School of Medicine, New York, NY, USA
| | - Lois S. Weisman
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Roger L. Albin
- Department of Neurology, University of Michigan, Ann Arbor, MI, USA
- Neurology Service, VAAAHS, University of Michigan, Ann Arbor, MI, USA
- Udall Centre, University of Michigan, Ann Arbor, MI, USA
- GRECC, VAAAHS, University of Michigan, Ann Arbor, MI, USA,
- Michigan Alzheimer's Disease Center, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
12
|
Abstract
Yeast Vps13 is a member of a conserved protein family that includes human homologues associated with neurodegenerative and developmental disorders. In this issue, De et al. (2017. J. Cell Biol. https://doi.org/10.1083/jcb.201606078) establish direct roles for Vps13 and its surprising binding partner, the calcium-binding centrin Cdc31, in trans-Golgi network (TGN) to endosome traffic and TGN homotypic fusion.
Collapse
Affiliation(s)
- Margaret D Myers
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095
| | - Gregory S Payne
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095
| |
Collapse
|
13
|
De M, Oleskie AN, Ayyash M, Dutta S, Mancour L, Abazeed ME, Brace EJ, Skiniotis G, Fuller RS. The Vps13p-Cdc31p complex is directly required for TGN late endosome transport and TGN homotypic fusion. J Cell Biol 2017; 216:425-439. [PMID: 28122955 PMCID: PMC5294781 DOI: 10.1083/jcb.201606078] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 11/04/2016] [Accepted: 01/11/2017] [Indexed: 01/09/2023] Open
Abstract
VPS13 proteins are widely conserved in eukaryotes and associated with human neurodegenerative and neurodevelopmental diseases. De et al. describe the lipid specificity and structure of yeast Vps13p, providing insight into its role in both TGN late endosome transport and TGN homotypic fusion. Yeast VPS13 is the founding member of a eukaryotic gene family of growing interest in cell biology and medicine. Mutations in three of four human VPS13 genes cause autosomal recessive neurodegenerative or neurodevelopmental disease, making yeast Vps13p an important structural and functional model. Using cell-free reconstitution with purified Vps13p, we show that Vps13p is directly required both for transport from the trans-Golgi network (TGN) to the late endosome/prevacuolar compartment (PVC) and for TGN homotypic fusion. Vps13p must be in complex with the small calcium-binding protein Cdc31p to be active. Single-particle electron microscopic analysis of negatively stained Vps13p indicates that this 358-kD protein is folded into a compact rod-shaped density (20 × 4 nm) with a loop structure at one end with a circular opening ∼6 nm in diameter. Vps13p exhibits ATP-stimulated binding to yeast membranes and specific interactions with phosphatidic acid and phosphorylated forms of phosphatidyl inositol at least in part through the binding affinities of conserved N- and C-terminal domains.
Collapse
Affiliation(s)
- Mithu De
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Austin N Oleskie
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109.,Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Mariam Ayyash
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Somnath Dutta
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109.,Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Liliya Mancour
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109.,Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Mohamed E Abazeed
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109.,Medical Scientist Training Program, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Eddy J Brace
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Georgios Skiniotis
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109.,Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109
| | - Robert S Fuller
- Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI 48109
| |
Collapse
|
14
|
Vonk JJ, Yeshaw WM, Pinto F, Faber AIE, Lahaye LL, Kanon B, van der Zwaag M, Velayos-Baeza A, Freire R, van IJzendoorn SC, Grzeschik NA, Sibon OCM. Drosophila Vps13 Is Required for Protein Homeostasis in the Brain. PLoS One 2017; 12:e0170106. [PMID: 28107480 PMCID: PMC5249141 DOI: 10.1371/journal.pone.0170106] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/10/2016] [Indexed: 11/22/2022] Open
Abstract
Chorea-Acanthocytosis is a rare, neurodegenerative disorder characterized by progressive loss of locomotor and cognitive function. It is caused by loss of function mutations in the Vacuolar Protein Sorting 13A (VPS13A) gene, which is conserved from yeast to human. The consequences of VPS13A dysfunction in the nervous system are still largely unspecified. In order to study the consequences of VPS13A protein dysfunction in the ageing central nervous system we characterized a Drosophila melanogaster Vps13 mutant line. The Drosophila Vps13 gene encoded a protein of similar size as human VPS13A. Our data suggest that Vps13 is a peripheral membrane protein located to endosomal membranes and enriched in the fly head. Vps13 mutant flies showed a shortened life span and age associated neurodegeneration. Vps13 mutant flies were sensitive to proteotoxic stress and accumulated ubiquitylated proteins. Levels of Ref(2)P, the Drosophila orthologue of p62, were increased and protein aggregates accumulated in the central nervous system. Overexpression of the human Vps13A protein in the mutant flies partly rescued apparent phenotypes. This suggests a functional conservation of human VPS13A and Drosophila Vps13. Our results demonstrate that Vps13 is essential to maintain protein homeostasis in the larval and adult Drosophila brain. Drosophila Vps13 mutants are suitable to investigate the function of Vps13 in the brain, to identify genetic enhancers and suppressors and to screen for potential therapeutic targets for Chorea-Acanthocytosis.
Collapse
Affiliation(s)
- Jan J. Vonk
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Wondwossen M. Yeshaw
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Francesco Pinto
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anita I. E. Faber
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Liza L. Lahaye
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Bart Kanon
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Marianne van der Zwaag
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Raimundo Freire
- Unidad de Investigación, Hospital Universitario de Canarias, Instituto de Tecnologías Biomédicas, Ofra s/n, La Laguna, Tenerife, Spain
| | - Sven C. van IJzendoorn
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Nicola A. Grzeschik
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Ody C. M. Sibon
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
- * E-mail:
| |
Collapse
|
15
|
Lupo F, Tibaldi E, Matte A, Sharma AK, Brunati AM, Alper SL, Zancanaro C, Benati D, Siciliano A, Bertoldi M, Zonta F, Storch A, Walker RH, Danek A, Bader B, Hermann A, De Franceschi L. A new molecular link between defective autophagy and erythroid abnormalities in chorea-acanthocytosis. Blood 2016; 128:2976-2987. [PMID: 27742708 PMCID: PMC5179337 DOI: 10.1182/blood-2016-07-727321] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 09/24/2016] [Indexed: 01/04/2023] Open
Abstract
Chorea-acanthocytosis is one of the hereditary neurodegenerative disorders known as the neuroacanthocytoses. Chorea-acanthocytosis is characterized by circulating acanthocytes deficient in chorein, a protein of unknown function. We report here for the first time that chorea-acanthocytosis red cells are characterized by impaired autophagy, with cytoplasmic accumulation of active Lyn and of autophagy-related proteins Ulk1 and Atg7. In chorea-acanthocytosis erythrocytes, active Lyn is sequestered by HSP90-70 to form high-molecular-weight complexes that stabilize and protect Lyn from its proteasomal degradation, contributing to toxic Lyn accumulation. An interplay between accumulation of active Lyn and autophagy was found in chorea-acanthocytosis based on Lyn coimmunoprecipitation with Ulk1 and Atg7 and on the presence of Ulk1 in Lyn-containing high-molecular-weight complexes. In addition, chorein associated with Atg7 in healthy but not in chorea-acanthocytosis erythrocytes. Electron microscopy detected multivesicular bodies and membrane remnants only in circulating chorea-acanthocytosis red cells. In addition, reticulocyte-enriched chorea-acanthocytosis red cell fractions exhibited delayed clearance of mitochondria and lysosomes, further supporting the impairment of authophagic flux. Because autophagy is also important in erythropoiesis, we studied in vitro CD34+-derived erythroid precursors. In chorea-acanthocytosis, we found (1) dyserythropoiesis; (2) increased active Lyn; (3) accumulation of a marker of autophagic flux and autolysososme degradation; (4) accumlation of Lamp1, a lysosmal membrane protein, and LAMP1-positive aggregates; and (5) reduced clearance of lysosomes and mitochondria. Our results uncover in chorea-acanthocytosis erythroid cells an association between accumulation of active Lyn and impaired autophagy, suggesting a link between chorein and autophagic vesicle trafficking in erythroid maturation.
Collapse
Affiliation(s)
- Francesca Lupo
- Department of Medicine, University of Verona and Azienda ospedaliera Universitaria Integrata di Verona, Verona, Italy
| | - Elena Tibaldi
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Alessandro Matte
- Department of Medicine, University of Verona and Azienda ospedaliera Universitaria Integrata di Verona, Verona, Italy
| | - Alok K Sharma
- Division of Nephrology and Vascular Biology Research Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | | | - Seth L Alper
- Division of Nephrology and Vascular Biology Research Center, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA
| | - Carlo Zancanaro
- Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Donatella Benati
- Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Angela Siciliano
- Department of Medicine, University of Verona and Azienda ospedaliera Universitaria Integrata di Verona, Verona, Italy
| | - Mariarita Bertoldi
- Department of Neuroscience, Biomedicine and Movement, University of Verona, Verona, Italy
| | - Francesca Zonta
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Alexander Storch
- Center for Regenerative Therapies, and
- Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, Dresden, Germany
- Center for Neurodegenerative Diseases, Dresden, Germany
| | - Ruth H Walker
- Department of Neurology, James J. Peters VA Medical Center, Bronx, NY
- Mount Sinai School of Medicine, New York, NY; and
| | - Adrian Danek
- Department of Neurology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Benedikt Bader
- Department of Neurology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Andreas Hermann
- Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, Dresden, Germany
| | - Lucia De Franceschi
- Department of Medicine, University of Verona and Azienda ospedaliera Universitaria Integrata di Verona, Verona, Italy
| |
Collapse
|
16
|
Guerrier S, Plattner H, Richardson E, Dacks JB, Turkewitz AP. An evolutionary balance: conservation vs innovation in ciliate membrane trafficking. Traffic 2016; 18:18-28. [PMID: 27696651 DOI: 10.1111/tra.12450] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 09/20/2016] [Accepted: 09/20/2016] [Indexed: 12/15/2022]
Abstract
As most of eukaryotic diversity lies in single-celled protists, they represent unique opportunities to ask questions about the balance of conservation and innovation in cell biological features. Among free-living protists the ciliates offer ease of culturing, a rich array of experimental approaches, and versatile molecular tools, particularly in Tetrahymena thermophila and Paramecium tetraurelia. These attributes have been exploited by researchers to analyze a wealth of cellular structures in these large and complex cells. This mini-review focuses on 3 aspects of ciliate membrane dynamics, all linked with endolysosomal trafficking. First is nutrition based on phagocytosis and maturation of food vacuoles. Secondly, we discuss regulated exocytosis from vesicles that have features of both dense core secretory granules but also lysosome-related organelles. The third topic is the targeting, breakdown and resorption of parental nuclei in mating partners. For all 3 phenomena, it is clear that elements of the canonical membrane-trafficking system have been retained and in some cases repurposed. In addition, there is evidence that recently evolved, lineage-specific proteins provide determinants in these pathways.
Collapse
Affiliation(s)
| | - Helmut Plattner
- Department of Biology, University of Konstanz, Konstanz, Germany
| | | | - Joel B Dacks
- Department of Cell Biology, University of Alberta, Canada
| | - Aaron P Turkewitz
- Department of Molecular Genetics and Cell Biology, The University of Chicago, Chicago, Illinois
| |
Collapse
|
17
|
Mesquita A, Cardenal-Muñoz E, Dominguez E, Muñoz-Braceras S, Nuñez-Corcuera B, Phillips BA, Tábara LC, Xiong Q, Coria R, Eichinger L, Golstein P, King JS, Soldati T, Vincent O, Escalante R. Autophagy in Dictyostelium: Mechanisms, regulation and disease in a simple biomedical model. Autophagy 2016; 13:24-40. [PMID: 27715405 DOI: 10.1080/15548627.2016.1226737] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Autophagy is a fast-moving field with an enormous impact on human health and disease. Understanding the complexity of the mechanism and regulation of this process often benefits from the use of simple experimental models such as the social amoeba Dictyostelium discoideum. Since the publication of the first review describing the potential of D. discoideum in autophagy, significant advances have been made that demonstrate both the experimental advantages and interest in using this model. Since our previous review, research in D. discoideum has shed light on the mechanisms that regulate autophagosome formation and contributed significantly to the study of autophagy-related pathologies. Here, we review these advances, as well as the current techniques to monitor autophagy in D. discoideum. The comprehensive bioinformatics search of autophagic proteins that was a substantial part of the previous review has not been revisited here except for those aspects that challenged previous predictions such as the composition of the Atg1 complex. In recent years our understanding of, and ability to investigate, autophagy in D. discoideum has evolved significantly and will surely enable and accelerate future research using this model.
Collapse
Affiliation(s)
- Ana Mesquita
- a Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM) , Madrid , Spain.,b University of Cincinnati College of Medicine , Cincinnati , OH , USA
| | - Elena Cardenal-Muñoz
- c Départment de Biochimie , Faculté des Sciences, Université de Genève , Switzerland
| | - Eunice Dominguez
- a Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM) , Madrid , Spain.,d Departamento de Genética Molecular , Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Mexico City , México
| | - Sandra Muñoz-Braceras
- a Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM) , Madrid , Spain
| | | | - Ben A Phillips
- e Department of Biomedical Sciences , University of Sheffield , UK
| | - Luis C Tábara
- a Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM) , Madrid , Spain
| | - Qiuhong Xiong
- f Center for Biochemistry, Medical Faculty, University of Cologne , Cologne , Germany
| | - Roberto Coria
- d Departamento de Genética Molecular , Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Mexico City , México
| | - Ludwig Eichinger
- f Center for Biochemistry, Medical Faculty, University of Cologne , Cologne , Germany
| | - Pierre Golstein
- g Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2 , Inserm, U1104, CNRS UMR7280, Marseille , France
| | - Jason S King
- e Department of Biomedical Sciences , University of Sheffield , UK
| | - Thierry Soldati
- c Départment de Biochimie , Faculté des Sciences, Université de Genève , Switzerland
| | - Olivier Vincent
- a Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM) , Madrid , Spain
| | - Ricardo Escalante
- a Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM) , Madrid , Spain
| |
Collapse
|
18
|
Mehta ZB, Fine N, Pullen TJ, Cane MC, Hu M, Chabosseau P, Meur G, Velayos-Baeza A, Monaco AP, Marselli L, Marchetti P, Rutter GA. Changes in the expression of the type 2 diabetes-associated gene VPS13C in the β-cell are associated with glucose intolerance in humans and mice. Am J Physiol Endocrinol Metab 2016; 311:E488-507. [PMID: 27329800 PMCID: PMC5005967 DOI: 10.1152/ajpendo.00074.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 06/20/2016] [Indexed: 12/31/2022]
Abstract
Single nucleotide polymorphisms (SNPs) close to the VPS13C, C2CD4A and C2CD4B genes on chromosome 15q are associated with impaired fasting glucose and increased risk of type 2 diabetes. eQTL analysis revealed an association between possession of risk (C) alleles at a previously implicated causal SNP, rs7163757, and lowered VPS13C and C2CD4A levels in islets from female (n = 40, P < 0.041) but not from male subjects. Explored using promoter-reporter assays in β-cells and other cell lines, the risk variant at rs7163757 lowered enhancer activity. Mice deleted for Vps13c selectively in the β-cell were generated by crossing animals bearing a floxed allele at exon 1 to mice expressing Cre recombinase under Ins1 promoter control (Ins1Cre). Whereas Vps13c(fl/fl):Ins1Cre (βVps13cKO) mice displayed normal weight gain compared with control littermates, deletion of Vps13c had little effect on glucose tolerance. Pancreatic histology revealed no significant change in β-cell mass in KO mice vs. controls, and glucose-stimulated insulin secretion from isolated islets was not altered in vitro between control and βVps13cKO mice. However, a tendency was observed in female null mice for lower insulin levels and β-cell function (HOMA-B) in vivo. Furthermore, glucose-stimulated increases in intracellular free Ca(2+) were significantly increased in islets from female KO mice, suggesting impaired Ca(2+) sensitivity of the secretory machinery. The present data thus provide evidence for a limited role for changes in VPS13C expression in conferring altered disease risk at this locus, particularly in females, and suggest that C2CD4A may also be involved.
Collapse
Affiliation(s)
- Zenobia B Mehta
- Section of Cell Biology and Functional Genomics, Imperial College London, London, United Kingdom
| | - Nicholas Fine
- Section of Cell Biology and Functional Genomics, Imperial College London, London, United Kingdom
| | - Timothy J Pullen
- Section of Cell Biology and Functional Genomics, Imperial College London, London, United Kingdom
| | - Matthew C Cane
- Section of Cell Biology and Functional Genomics, Imperial College London, London, United Kingdom
| | - Ming Hu
- Section of Cell Biology and Functional Genomics, Imperial College London, London, United Kingdom
| | - Pauline Chabosseau
- Section of Cell Biology and Functional Genomics, Imperial College London, London, United Kingdom
| | - Gargi Meur
- Section of Cell Biology and Functional Genomics, Imperial College London, London, United Kingdom
| | | | - Anthony P Monaco
- Wellcome Trust Centre for Human Genetics, Oxford, United Kingdom; and
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Imperial College London, London, United Kingdom;
| |
Collapse
|
19
|
Sasaki N, Nakamura M, Kodama A, Urata Y, Shiokawa N, Hayashi T, Sano A. Chorein interacts with α‐tubulin and histone deacetylase 6, and overexpression preserves cell viability during nutrient deprivation in human embryonic kidney 293 cells. FASEB J 2016; 30:3726-3732. [DOI: 10.1096/fj.201500191rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 07/18/2016] [Indexed: 11/11/2022]
Affiliation(s)
- Natsuki Sasaki
- Department of PsychiatryKagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Masayuki Nakamura
- Department of PsychiatryKagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Akiko Kodama
- Department of PsychiatryKagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Yuka Urata
- Department of PsychiatryKagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Nari Shiokawa
- Department of PsychiatryKagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Takehiro Hayashi
- Department of PsychiatryKagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| | - Akira Sano
- Department of PsychiatryKagoshima University Graduate School of Medical and Dental Sciences Kagoshima Japan
| |
Collapse
|
20
|
Wideman JG, Muñoz-Gómez SA. The evolution of ERMIONE in mitochondrial biogenesis and lipid homeostasis: An evolutionary view from comparative cell biology. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:900-912. [PMID: 26825688 DOI: 10.1016/j.bbalip.2016.01.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 01/19/2016] [Accepted: 01/25/2016] [Indexed: 12/17/2022]
Abstract
The ER-mitochondria organizing network (ERMIONE) in Saccharomyces cerevisiae is involved in maintaining mitochondrial morphology and lipid homeostasis. ERMES and MICOS are two scaffolding complexes of ERMIONE that contribute to these processes. ERMES is ancient but has been lost in several lineages including animals, plants, and SAR (stramenopiles, alveolates and rhizaria). On the other hand, MICOS is ancient and has remained present in all organisms bearing mitochondrial cristae. The ERMIONE precursor evolved in the α-proteobacterial ancestor of mitochondria which had the central subunit of MICOS, Mic60. The subsequent evolution of ERMIONE and its interactors in eukaryotes reflects the integrative co-evolution of mitochondria and their hosts and the adaptive paths that some lineages have followed in their specialization to certain environments. By approaching the ERMIONE from a perspective of comparative evolutionary cell biology, we hope to shed light on not only its evolutionary history, but also how ERMIONE components may function in organisms other than S. cerevisiae. This article is part of a Special Issue entitled: The cellular lipid landscape edited by Tim P. Levine and Anant K. Menon.
Collapse
Affiliation(s)
| | - Sergio A Muñoz-Gómez
- Centre for Comparative Genomics and Evolutionary Bioinformatics, Department of Biochemistry and Molecular Biology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| |
Collapse
|
21
|
Plattner H. Signalling in ciliates: long- and short-range signals and molecular determinants for cellular dynamics. Biol Rev Camb Philos Soc 2015; 92:60-107. [PMID: 26487631 DOI: 10.1111/brv.12218] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 07/28/2015] [Accepted: 08/21/2015] [Indexed: 12/30/2022]
Abstract
In ciliates, unicellular representatives of the bikont branch of evolution, inter- and intracellular signalling pathways have been analysed mainly in Paramecium tetraurelia, Paramecium multimicronucleatum and Tetrahymena thermophila and in part also in Euplotes raikovi. Electrophysiology of ciliary activity in Paramecium spp. is a most successful example. Established signalling mechanisms include plasmalemmal ion channels, recently established intracellular Ca2+ -release channels, as well as signalling by cyclic nucleotides and Ca2+ . Ca2+ -binding proteins (calmodulin, centrin) and Ca2+ -activated enzymes (kinases, phosphatases) are involved. Many organelles are endowed with specific molecules cooperating in signalling for intracellular transport and targeted delivery. Among them are recently specified soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs), monomeric GTPases, H+ -ATPase/pump, actin, etc. Little specification is available for some key signal transducers including mechanosensitive Ca2+ -channels, exocyst complexes and Ca2+ -sensor proteins for vesicle-vesicle/membrane interactions. The existence of heterotrimeric G-proteins and of G-protein-coupled receptors is still under considerable debate. Serine/threonine kinases dominate by far over tyrosine kinases (some predicted by phosphoproteomic analyses). Besides short-range signalling, long-range signalling also exists, e.g. as firmly installed microtubular transport rails within epigenetically determined patterns, thus facilitating targeted vesicle delivery. By envisaging widely different phenomena of signalling and subcellular dynamics, it will be shown (i) that important pathways of signalling and cellular dynamics are established already in ciliates, (ii) that some mechanisms diverge from higher eukaryotes and (iii) that considerable uncertainties still exist about some essential aspects of signalling.
Collapse
Affiliation(s)
- Helmut Plattner
- Department of Biology, University of Konstanz, PO Box M625, 78457, Konstanz, Germany
| |
Collapse
|
22
|
Muñoz-Braceras S, Calvo R, Escalante R. TipC and the chorea-acanthocytosis protein VPS13A regulate autophagy inDictyosteliumand human HeLa cells. Autophagy 2015; 11:918-27. [DOI: 10.1080/15548627.2015.1034413] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
23
|
Walker RH. Untangling the Thorns: Advances in the Neuroacanthocytosis Syndromes. J Mov Disord 2015; 8:41-54. [PMID: 26090076 PMCID: PMC4460540 DOI: 10.14802/jmd.15009] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 04/27/2015] [Accepted: 04/28/2015] [Indexed: 12/25/2022] Open
Abstract
There have been significant advances in neuroacanthocytosis (NA) syndromes in the past 20 years, however, confusion still exists regarding the precise nature of these disorders and the correct nomenclature. This article seeks to clarify these issues and to summarise the recent literature in the field. The four key NA syndromes are described here-chorea-acanthocytosis, McLeod syndrome, Huntington's disease-like 2, and pantothenate kinase- associated neurodegeneration. In the first two, acanthocytosis is a frequent, although not invariable, finding; in the second two, it occurs in approximately 10% of patients. Degeneration affecting the basal ganglia is the key neuropathologic finding, thus the clinical presentations can be remarkably similar. The characteristic phenotype comprises a variety of movement disorders, including chorea, dystonia, and parkinsonism, and also psychiatric and cognitive symptoms attributable to basal ganglia dysfunction. The age of onset, inheritance patterns, and ethnic background differ in each condition, providing diagnostic clues. Other investigations, including routine blood testing and neuroimaging can be informative. Genetic diagnosis, if available, provides a definitive diagnosis, and is important for genetic counseling, and hopefully molecular therapies in the future. In this article I provide a historical perspective on each NA syndrome. The first 3 disorders, chorea-acanthocytosis, McLeod syndrome, Huntington's disease-like 2, are discussed in detail, with a comprehensive review of the literature to date for each, while pantothenate kinase-associated neurodegeneration is presented in summary, as this disorder has recently been reviewed in this journal. Therapy for all of these diseases is, at present, purely symptomatic.
Collapse
Affiliation(s)
- Ruth H. Walker
- Department of Neurology, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA
- Department of Neurology, Mount Sinai School of Medicine, New York, NY, USA
| |
Collapse
|
24
|
Annexin A2 promotes phagophore assembly by enhancing Atg16L⁺ vesicle biogenesis and homotypic fusion. Nat Commun 2015; 6:5856. [PMID: 25597631 PMCID: PMC4299943 DOI: 10.1038/ncomms6856] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 11/13/2014] [Indexed: 12/22/2022] Open
Abstract
Plasma membrane budding of Atg-16L-positive vesicles represents a very early event in the generation of the phagophore and in the process of macroautophagy. Here we show that the membrane curvature-inducing protein annexin A2 contributes to the formation of these vesicles and their fusion to form phagophores. Ultrastructural, proteomic and FACS analyses of Atg16L-positive vesicles reveal that 30% of Atg16L-positive vesicles are also annexin A2-positive. Lipidomic analysis of annexin A2-deficient mouse cells indicates that this protein plays a role in recruiting phosphatidylserine and phosphatidylinositides to Atg16L-positive vesicles. Absence of annexin A2 reduces both vesicle formation and homotypic Atg16L vesicle fusion. Ultimately, a reduction in LC3 flux and dampening of macroautophagy are observed in dendritic cells from Anxa2−/− mice. Together, our analyses highlight the importance of annexin A2 in vesiculation of a population of Atg16L-positive structures from the plasma membrane, and in their homotypic fusion to form phagophore structures. The earliest steps in autophagy are thought to include the budding of Atg16L-containing vesicles from the plasma membrane and their homotypic fusion to form a phagophore. Morozova et al. reveal a role for the membrane curvature-inducing protein Annexin A2 in the formation and fusion of these vesicles.
Collapse
|
25
|
Variation in the salivary proteomes of differentially virulent greenbug (Schizaphis graminum Rondani) biotypes. J Proteomics 2013; 105:186-203. [PMID: 24355481 DOI: 10.1016/j.jprot.2013.12.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 11/27/2013] [Accepted: 12/09/2013] [Indexed: 01/02/2023]
Abstract
UNLABELLED Greenbug (Schizaphis graminum Rondani) biotypes are classified by their differential virulence to wheat, barley, and sorghum varieties possessing greenbug resistance genes. Virulent greenbug biotypes exert phytotoxic effects upon their hosts during feeding, directly inducing physiological and metabolic alterations and accompanying foliar damage. Comparative analyses of the salivary proteomes of four differentially virulent greenbug biotypes C, E, G, and H showed significant proteomic divergence between biotypes. Thirty-two proteins were identified by LC-MS/MS; the most prevalent of which were three glucose dehydrogenase paralogs (GDH), lipophorin, complementary sex determiner, three proteins of unknown function, carbonic anhydrase, fibroblast growth factor receptor, and abnormal oocyte (ABO). Seven nucleotide-binding proteins were identified, including ABO which is involved in mRNA splicing. Quantitative variation among greenbug biotypes was detected in six proteins; two GDH paralogs, carbonic anhydrase, ABO, and two proteins of unknown function. Our findings reveal that the greenbug salivary proteome differs according to biotype and diverges substantially from those reported for other aphids. The proteomic profiles of greenbug biotypes suggest that interactions between aphid salivary proteins and the plant host result in suppression of plant defenses and cellular transport, and may manipulate transcriptional regulation in the plant host, ultimately allowing the aphid to maintain phloem ingestion. BIOLOGICAL SIGNIFICANCE Greenbug (Schizaphis graminum Rondani, GB) is a major phytotoxic aphid pest of wheat, sorghum, and barley. Unlike non-phytotoxic aphids, GB directly damages its host, causing uniformly characteristic symptoms leading to host death. As saliva is the primary interface between the aphid and its plant host, saliva is also the primary aphid biotypic determinant, and differences in biotypic virulence are the result of biotypic variations in salivary content. This study analyzed the exuded saliva of four distinct Greenbug biotypes with a range of virulence to crop lines containing greenbug resistance traits in order to identify differences between salivary proteins of the examined biotypes. Our analyses confirmed that the salivary proteomes of the examined greenbug biotypes differ widely, identified 32 proteins of the greenbug salivary proteome, and found significant proteomic variation between six identified salivary proteins. The proteomic variation identified herein is likely the basis of biotypic virulence, and the proteins identified can serve as the basis for functional studies into both greenbug-induced phytotoxic damage and into the molecular basis of virulence in specific GB biotypes. This article is part of a Special Issue entitled: SI: Proteomics of non-model organisms.
Collapse
|
26
|
Shiokawa N, Nakamura M, Sameshima M, Deguchi A, Hayashi T, Sasaki N, Sano A. Chorein, the protein responsible for chorea-acanthocytosis, interacts with β-adducin and β-actin. Biochem Biophys Res Commun 2013; 441:96-101. [PMID: 24129186 DOI: 10.1016/j.bbrc.2013.10.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 10/05/2013] [Indexed: 10/26/2022]
Abstract
Chorea-acanthocytosis (ChAc) is an autosomal, recessive hereditary disease characterized by striatal neurodegeneration and acanthocytosis, and caused by loss of function mutations in the vacuolar protein sorting 13 homolog A (VPS13A) gene. VPS13A encodes chorein whose physiological function at the molecular level is poorly understood. In this study, we show that chorein interacts with β-adducin and β-actin. We first compare protein expression in human erythrocyte membranes using proteomic analysis. Protein levels of β-adducin isoform 1 and β-actin are markedly decreased in erythrocyte membranes from a ChAc patient. Subsequent co-immunoprecipitation (co-IP) and reverse co-IP assays using extracts from chorein-overexpressing human embryonic kidney 293 (HEK293) cells, shows that β-adducin (isoforms 1 and 2) and β-actin interact with chorein. Immunocytochemical analysis using chorein-overexpressing HEK293 cells demonstrates co-localization of chorein with β-adducin and β-actin. In addition, immunoreactivity of β-adducin isoform 1 is significantly decreased in the striatum of gene-targeted ChAc-model mice. Adducin and actin are membrane cytoskeletal proteins, involved in synaptic function. Expression of β-adducin is restricted to the brain and hematopoietic tissues, corresponding to the main pathological lesions of ChAc, and thereby implicating β-adducin and β-actin in ChAc pathogenesis.
Collapse
Affiliation(s)
- Nari Shiokawa
- Department of Psychiatry, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan
| | | | | | | | | | | | | |
Collapse
|
27
|
Prohaska R, Sibon OC, Rudnicki DD, Danek A, Hayflick SJ, Verhaag EM, Jan J V, Margolis RL, Walker RH. Brain, blood, and iron: perspectives on the roles of erythrocytes and iron in neurodegeneration. Neurobiol Dis 2012; 46:607-24. [PMID: 22426390 PMCID: PMC3352961 DOI: 10.1016/j.nbd.2012.03.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 01/17/2012] [Accepted: 03/01/2012] [Indexed: 12/20/2022] Open
Abstract
The terms "neuroacanthocytosis" (NA) and "neurodegeneration with brain iron accumulation" (NBIA) both refer to groups of genetically heterogeneous disorders, classified together due to similarities of their phenotypic or pathological findings. Even collectively, the disorders that comprise these sets are exceedingly rare and challenging to study. The NBIA disorders are defined by their appearance on brain magnetic resonance imaging, with iron deposition in the basal ganglia. Clinical features vary, but most include a movement disorder. New causative genes are being rapidly identified; however, the mechanisms by which mutations cause iron accumulation and neurodegeneration are not well understood. NA syndromes are also characterized by a progressive movement disorder, accompanied by cognitive and psychiatric features, resulting from mutations in a number of genes whose roles are also basically unknown. An overlapping feature of the two groups, NBIA and NA, is the occurrence of acanthocytes, spiky red cells with a poorly-understood membrane dysfunction. In this review we summarise recent developments in this field, specifically insights into cellular mechanisms and from animal models. Cell membrane research may shed light upon the significance of the erythrocyte abnormality, and upon possible connections between the two sets of disorders. Shared pathophysiologic mechanisms may lead to progress in the understanding of other types of neurodegeneration.
Collapse
Affiliation(s)
- Rainer Prohaska
- Max F. Perutz Laboratories, Medical University of Vienna, Vienna, Austria
| | - Ody C.M. Sibon
- Section of Radiation & Stress Cell Biology, Department of Cell Biology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Dobrila D. Rudnicki
- Department of Psychiatry, Division of Neurobiology, Laboratory of Genetic Neurobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Adrian Danek
- Neurologische Klinik und Poliklinik, Ludwig-Maximilians-Universität, Munich, Germany
| | - Susan J. Hayflick
- Departments of Molecular & Medical Genetics, Pediatrics and Neurology, Oregon Health & Science University, Portland OR USA
| | - Esther M. Verhaag
- Section of Radiation & Stress Cell Biology, Department of Cell Biology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Vonk Jan J
- Section of Radiation & Stress Cell Biology, Department of Cell Biology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Russell L. Margolis
- Department of Psychiatry, Division of Neurobiology, Laboratory of Genetic Neurobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology and Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ruth H. Walker
- Departments of Neurology, James J. Peters Veterans Affairs Medical Center, Bronx, NY, USA and Mount Sinai School of Medicine, New York, NY USA
| |
Collapse
|
28
|
Park JS, Neiman AM. VPS13 regulates membrane morphogenesis during sporulation in Saccharomyces cerevisiae. J Cell Sci 2012; 125:3004-11. [PMID: 22442115 DOI: 10.1242/jcs.105114] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The hereditary disorders chorea acanthocytosis and Cohen syndrome are caused by mutations in different members of a family of genes that are orthologs of yeast VPS13. In vegetatively growing yeast, VPS13 is involved in the delivery of proteins to the vacuole. During sporulation, VPS13 is important for formation of the prospore membrane that encapsulates the daughter nuclei to give rise to spores. We report that VPS13 is required for multiple aspects of prospore membrane morphogenesis. VPS13 (1) promotes expansion of the prospore membrane through regulation of phosphatidylinositol phosphates, which in turn activate the phospholipase D, Spo14; (2) is required for a late step in cytokinesis that gives rise to spores; and (3) regulates a membrane-bending activity that generates intralumenal vesicles. These results demonstrate that Vps13 plays a broader role in membrane biology than previously known, which could have important implications for the functions of VPS13 orthologs in humans.
Collapse
Affiliation(s)
- Jae-Sook Park
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, New York 11794-5215, USA
| | | |
Collapse
|
29
|
Vacuolar protein sorting protein 13A, TtVPS13A, localizes to the tetrahymena thermophila phagosome membrane and is required for efficient phagocytosis. EUKARYOTIC CELL 2011; 10:1207-18. [PMID: 21764909 DOI: 10.1128/ec.05089-11] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Vacuolar protein sorting 13 (VPS13) proteins have been studied in a number of organisms, and mutations in VPS13 genes have been implicated in two human genetic disorders, but the function of these proteins is poorly understood. The TtVPS13A protein was previously identified in a mass spectrometry analysis of the Tetrahymena thermophila phagosome proteome (M. E. Jacobs et al., Eukaryot. Cell 5:1990-2000, 2006), suggesting that it is involved in phagocytosis. In this study, we analyzed the structure of the macronuclear TtVPS13A gene, which was found to be composed of 17 exons spanning 12.5 kb and was predicted to encode a protein of 3,475 amino acids (aa). A strain expressing a TtVPS13A-green fluorescent protein (GFP) fusion protein was constructed, and the protein was found to associate with the phagosome membrane during the entire cycle of phagocytosis. In addition, Tetrahymena cells with a TtVPS13A knockout mutation displayed impaired phagocytosis. Specifically, they grew slowly under conditions where phagocytosis is essential, they formed few phagosomes, and the digestion of phagosomal contents was delayed compared to wild-type cells. Overall, these results provide evidence that the TtVPS13A protein is required for efficient phagocytosis.
Collapse
|