1
|
Ye Y, Osmani AH, Liu ZR, Kern A, Wu JQ. Fission yeast GPI inositol deacylase Bst1 regulates ER-Golgi transport and functions in late stages of cytokinesis. Mol Biol Cell 2025; 36:ar27. [PMID: 39813093 DOI: 10.1091/mbc.e24-08-0375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025] Open
Abstract
The Munc13/UNC-13 family protein Ync13 is essential for septum integrity and cytokinesis in fission yeast. To further explore the mechanism of Ync13 functions, spontaneous suppressors of ync13 mutants, which can suppress the colony-formation defects and lysis phenotype of ync13 mutant cells, are isolated and characterized. One of the suppressor mutants, bst1-s27, shows defects in the cytokinetic contractile ring constriction, septation, and daughter cell separation, similar to bst1Δ mutant. Bst1, a predicted GPI inositol deacylase, was an uncharacterized protein in fission yeast. It localizes to ER and puncta structures in the cytoplasm. The Bst1 puncta overlaps frequently with Anp1, which is a marker of endoplasmic reticulum (ER)-Golgi transport, but rarely with trans-Golgi marker Sec72. The nuclear ER signal of Anp1 increases in bst1Δ mutant, whereas Sec72 localization shows no obvious changes. In addition, more cytoplasmic puncta structures of COPII subunits, Sec13 and Sec24, are observed in bst1Δ mutant, and acid phosphatase secretion is compromised without Bst1. Consistently, the division site targeting of the β-glucanase Eng1 and α-glucanase Agn1 is reduced in bst1Δ and bst1Δ ync13Δ mutant. Taken together, our results suggest that Bst1 regulates ER-Golgi transport and is involved in cytokinesis through regulating the secretion of glucanases.
Collapse
Affiliation(s)
- Yanfang Ye
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210
- College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Aysha H Osmani
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210
| | - Zhen-Ru Liu
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210
| | - Addie Kern
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210
| | - Jian-Qiu Wu
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
2
|
Seibel C, Pudewell S, Rafii P, Ettich J, Weitz HT, Lang A, Petzsch P, Köhrer K, Floss DM, Scheller J. Synthetic trimeric interleukin-6 receptor complexes with a STAT3 phosphorylation dominated activation profile. Cytokine 2024; 184:156766. [PMID: 39348731 DOI: 10.1016/j.cyto.2024.156766] [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: 08/07/2024] [Revised: 09/11/2024] [Accepted: 09/18/2024] [Indexed: 10/02/2024]
Abstract
In Interleukin (IL)-6 signalling, IL-6 site I binds to the IL-6 receptor (IL-6R) first, following by IL-6 site II interaction to domain 2/3 of gp130 to form premature trimeric IL-6:IL-6R:gp130 receptor complexes. Formation of the mature hexameric receptor complex is then facilitated by the inter-trimeric interaction of IL-6 site III with domain 1 of the opposing gp130. The two gp130-associated Janus kinases (JAKs) trans-phosphorylate when their spatiotemporal pairing is correct, which causes the activation of STAT, ERK, and AKT pathways in a balanced manner. Since the intracellular domain (ICD) of IL-6R is not needed for STAT/ERK/AKT phosphorylation, we investigated the conditions under which a chimeric IL-6RECD-gp130TMD/ICD receptor protein confers biological activity. For IL-6RECD-gp130TMD/ICD, the extracellular domain (ECD) of IL-6R was fused to the transmembrane domain (TMD) and ICD of gp130. Co-expression of IL-6RECD-gp130TMD/ICD with signalling-deficient gp130 variants did not induce IL-6 signalling, suggesting that the assembly of hexameric complexes failed to dimerize the IL-6R-associated JAKs correctly. By mimicking the premature trimeric receptor complex, IL-6-mediated dimerization of IL-6RECD-gp130TMD/ICD with the single-cytokine-binding variant gp130ΔD1 induced signalling. Of note, IL-6 signalling via these synthetic gp130ΔD1:IL-6RECD-gp130TMD/ICD complexes resulted predominantly in STAT3 phosphorylation. A STAT3-dominated profile was also observed after IL-6-induced signalling mediated by a JAK-deficient IL-6RECD-gp130TMD/ICDΔJAK variant in complex with the JAK-proficient but STAT/ERK/AKT-deficient gp130JAKΔICD variant. Our data showed that effective ERK/AKT signalling could not be executed after intracellular domain swapping from gp130 to the IL-6R. Taken together, the chimeric IL-6R/gp130 receptor may be helpful in the creation of customized synthetic IL-6 signalling.
Collapse
Affiliation(s)
- Christiane Seibel
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Silke Pudewell
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Puyan Rafii
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Julia Ettich
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Hendrik T Weitz
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Alexander Lang
- Cardiovascular Research Laboratory, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Patrick Petzsch
- Biological and Medical Research Center (BMFZ), Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Karl Köhrer
- Biological and Medical Research Center (BMFZ), Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Doreen M Floss
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Jürgen Scheller
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany.
| |
Collapse
|
3
|
Lyu Z, Chen J, Lyu J, Guo P, Liu J, Liu J, Zhang W. Spraying double-stranded RNA targets UDP-N-acetylglucosamine pyrophosphorylase in the control of Nilaparvata lugens. Int J Biol Macromol 2024; 271:132455. [PMID: 38795878 DOI: 10.1016/j.ijbiomac.2024.132455] [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: 01/17/2024] [Revised: 05/04/2024] [Accepted: 05/14/2024] [Indexed: 05/28/2024]
Abstract
The rice pest Nilaparvata lugens (the brown planthopper, BPH) has developed different levels of resistance to at least 11 chemical pesticides. RNAi technology has contributed to the development of environmentally friendly RNA biopesticides designed to reduce chemical use. Consequently, more precise targets need to be identified and characterized, and efficient dsRNA delivery methods are necessary for effective field pest control. In this study, a low off-target risk dsNlUAP fragment (166 bp) was designed in silico to minimize the potential adverse effects on non-target organisms. Knockdown of NlUAP via microinjection significantly decreased the content of UDP-N-acetylglucosamine and chitin, causing chitinous structural disorder and abnormal phenotypes in wing and body wall, reduced fertility, and resulted in pest mortality up to 100 %. Furthermore, dsNlUAP was loaded with ROPE@C, a chitosan-modified nanomaterial for spray application, which significantly downregulated the expression of NlUAP, led to 48.9 % pest mortality, and was confirmed to have no adverse effects on Cyrtorhinus lividipennis, an important natural enemy of BPH. These findings will contribute to the development of safer biopesticides for the control of N. lugens.
Collapse
Affiliation(s)
- Zihao Lyu
- State Key Laboratory of Biocontrol, School of Agriculture, Sun Yat-sen University, Guangzhou, China
| | - Jingxiang Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jun Lyu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Pingping Guo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jiahui Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jinhui Liu
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Wenqing Zhang
- State Key Laboratory of Biocontrol, School of Agriculture, Sun Yat-sen University, Guangzhou, China; State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
4
|
Mishra AK, Ye T, Banday S, Thakare RP, Su CTT, Pham NNH, Ali A, Kulshreshtha A, Chowdhury SR, Simone TM, Hu K, Zhu LJ, Eisenhaber B, Deibler SK, Simin K, Thompson PR, Kelliher MA, Eisenhaber F, Malonia SK, Green MR. Targeting the GPI transamidase subunit GPAA1 abrogates the CD24 immune checkpoint in ovarian cancer. Cell Rep 2024; 43:114041. [PMID: 38573857 DOI: 10.1016/j.celrep.2024.114041] [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: 10/26/2023] [Revised: 01/25/2024] [Accepted: 03/19/2024] [Indexed: 04/06/2024] Open
Abstract
CD24 is frequently overexpressed in ovarian cancer and promotes immune evasion by interacting with its receptor Siglec10, present on tumor-associated macrophages, providing a "don't eat me" signal that prevents targeting and phagocytosis by macrophages. Factors promoting CD24 expression could represent novel immunotherapeutic targets for ovarian cancer. Here, using a genome-wide CRISPR knockout screen, we identify GPAA1 (glycosylphosphatidylinositol anchor attachment 1), a factor that catalyzes the attachment of a glycosylphosphatidylinositol (GPI) lipid anchor to substrate proteins, as a positive regulator of CD24 cell surface expression. Genetic ablation of GPAA1 abolishes CD24 cell surface expression, enhances macrophage-mediated phagocytosis, and inhibits ovarian tumor growth in mice. GPAA1 shares structural similarities with aminopeptidases. Consequently, we show that bestatin, a clinically advanced aminopeptidase inhibitor, binds to GPAA1 and blocks GPI attachment, resulting in reduced CD24 cell surface expression, increased macrophage-mediated phagocytosis, and suppressed growth of ovarian tumors. Our study highlights the potential of targeting GPAA1 as an immunotherapeutic approach for CD24+ ovarian cancers.
Collapse
Affiliation(s)
- Alok K Mishra
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| | - Tianyi Ye
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Shahid Banday
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Ritesh P Thakare
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Chinh Tran-To Su
- Bioinformatics Institute (BII), Agency for Science, Technology, and Research (A(∗)STAR), 30 Biopolis Street, Matrix, #07-01, Singapore 138671, Singapore
| | - Ngoc N H Pham
- Faculty of Biology and Biotechnology, University of Science, Vietnam National University, 227 Nguyen Van Cu Street, District 5, Ho Chi Minh City, Vietnam
| | - Amjad Ali
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Ankur Kulshreshtha
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Shreya Roy Chowdhury
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Tessa M Simone
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Kai Hu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Lihua Julie Zhu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Program in Molecular Medicine and Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Birgit Eisenhaber
- Bioinformatics Institute (BII), Agency for Science, Technology, and Research (A(∗)STAR), 30 Biopolis Street, Matrix, #07-01, Singapore 138671, Singapore; Lausitz Advanced Scientific Applications (LASA) gGmbH, Straße der Einheit 2-24, 02943 Weißwasser, Germany
| | - Sara K Deibler
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Karl Simin
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Paul R Thompson
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Michelle A Kelliher
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Frank Eisenhaber
- Bioinformatics Institute (BII), Agency for Science, Technology, and Research (A(∗)STAR), 30 Biopolis Street, Matrix, #07-01, Singapore 138671, Singapore; Lausitz Advanced Scientific Applications (LASA) gGmbH, Straße der Einheit 2-24, 02943 Weißwasser, Germany; School of Biological Sciences, Nanyang Technological University (NTU), 60 Nanyang Drive, Singapore 637551, Singapore.
| | - Sunil K Malonia
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA.
| | - Michael R Green
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
5
|
Kol I, Rishiq A, Cohen M, Kahlon S, Pick O, Dassa L, Stein N, Bar-On Y, Wolf DG, Seidel E, Mandelboim O. CLPTM1L is a GPI-anchoring pathway component targeted by HCMV. J Cell Biol 2023; 222:e202207104. [PMID: 37389656 PMCID: PMC10316631 DOI: 10.1083/jcb.202207104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 04/03/2023] [Accepted: 05/19/2023] [Indexed: 07/01/2023] Open
Abstract
The GPI-anchoring pathway plays important roles in normal development and immune modulation. MHC Class I Polypeptide-related Sequence A (MICA) is a stress-induced ligand, downregulated by human cytomegalovirus (HCMV) to escape immune recognition. Its most prevalent allele, MICA*008, is GPI-anchored via an uncharacterized pathway. Here, we identify cleft lip and palate transmembrane protein 1-like protein (CLPTM1L) as a GPI-anchoring pathway component and show that during infection, the HCMV protein US9 downregulates MICA*008 via CLPTM1L. We show that the expression of some GPI-anchored proteins (CD109, CD59, and MELTF)-but not others (ULBP2, ULBP3)-is CLPTM1L-dependent, and further show that like MICA*008, MELTF is downregulated by US9 via CLPTM1L during infection. Mechanistically, we suggest that CLPTM1L's function depends on its interaction with a free form of PIG-T, normally a part of the GPI transamidase complex. We suggest that US9 inhibits this interaction and thereby downregulates the expression of CLPTM1L-dependent proteins. Altogether, we report on a new GPI-anchoring pathway component that is targeted by HCMV.
Collapse
Affiliation(s)
- Inbal Kol
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel Canada, Hadassah—Hebrew University Medical Center, Jerusalem, Israel
| | - Ahmed Rishiq
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel Canada, Hadassah—Hebrew University Medical Center, Jerusalem, Israel
| | - Mevaseret Cohen
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel Canada, Hadassah—Hebrew University Medical Center, Jerusalem, Israel
| | - Shira Kahlon
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel Canada, Hadassah—Hebrew University Medical Center, Jerusalem, Israel
| | - Ophir Pick
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel Canada, Hadassah—Hebrew University Medical Center, Jerusalem, Israel
| | - Liat Dassa
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel Canada, Hadassah—Hebrew University Medical Center, Jerusalem, Israel
| | - Natan Stein
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel Canada, Hadassah—Hebrew University Medical Center, Jerusalem, Israel
| | - Yotam Bar-On
- Department of Immunology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Dana G. Wolf
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel Canada, Hadassah—Hebrew University Medical Center, Jerusalem, Israel
- Clinical Virology Unit, Hadassah Hebrew University Medical Center, Jerusalem, Israel
| | - Einat Seidel
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel Canada, Hadassah—Hebrew University Medical Center, Jerusalem, Israel
| | - Ofer Mandelboim
- The Concern Foundation Laboratories at the Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel Canada, Hadassah—Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
6
|
Ben Ayed I, Jallouli O, Murakami Y, Souissi A, Mallouli S, Bouzid A, Kamoun F, Elloumi I, Frikha F, Tlili A, Weckhuysen S, Kinoshita T, Triki CC, Masmoudi S. Case report: Functional analysis of the p.Arg507Trp variant of the PIGT gene supporting the moderate epilepsy phenotype of mutations in the C-terminal region. Front Neurol 2023; 14:1092887. [PMID: 36970549 PMCID: PMC10034188 DOI: 10.3389/fneur.2023.1092887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/01/2023] [Indexed: 03/29/2023] Open
Abstract
Pathogenic germline variants in the PIGT gene are associated with the "multiple congenital anomalies-hypotonia-seizures syndrome 3" (MCAHS3) phenotype. So far, fifty patients have been reported, most of whom suffer from intractable epilepsy. Recently, a comprehensive analysis of a cohort of 26 patients with PIGT variants has broadened the phenotypical spectrum and indicated that both p.Asn527Ser and p.Val528Met are associated with a milder epilepsy phenotype and less severe outcomes. Since all reported patients are of Caucasian/Polish origin and most harbor the same variant (p.Val528Met), the ability to draw definitive conclusions regarding the genotype-phenotype correlation remains limited. We report a new case with a homozygous variant p.Arg507Trp in the PIGT gene, detected on clinical exome sequencing. The North African patient in question displays a predominantly neurological phenotype with global developmental delay, hypotonia, brain abnormalities, and well-controlled epileptic seizures. Homozygous and heterozygous variants in codon 507 have been reported to cause PIGT deficiency without biochemical confirmation. In this study, FACS analysis of knockout HEK293 cells that had been transfected with wild-type or mutant cDNA constructs demonstrated that the p.Arg507Trp variant leads to mildly reduced activity. Our result confirm the pathogenicity of this variant and strengthen recently reported evidence on the genotype-phenotype correlation of the PIGT variant.
Collapse
Affiliation(s)
- Ikhlas Ben Ayed
- Laboratory of Molecular and Cellular Screening Processes (LPCMC), Center of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
- Medical Genetics Department, University Hedi Chaker Hospital of Sfax, Sfax, Tunisia
| | - Olfa Jallouli
- Child Neurology Department, University Hedi Chaker Hospital of Sfax, Sfax, Tunisia
- Research Laboratory “Neuropédiatrie” LR19ES15, Sfax University, Sfax, Tunisia
| | - Yoshiko Murakami
- Laboratory of Immunoglycobiology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Amal Souissi
- Laboratory of Molecular and Cellular Screening Processes (LPCMC), Center of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Salma Mallouli
- Child Neurology Department, University Hedi Chaker Hospital of Sfax, Sfax, Tunisia
- Research Laboratory “Neuropédiatrie” LR19ES15, Sfax University, Sfax, Tunisia
| | - Amal Bouzid
- Laboratory of Molecular and Cellular Screening Processes (LPCMC), Center of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Fatma Kamoun
- Child Neurology Department, University Hedi Chaker Hospital of Sfax, Sfax, Tunisia
- Research Laboratory “Neuropédiatrie” LR19ES15, Sfax University, Sfax, Tunisia
| | - Ines Elloumi
- Laboratory of Molecular and Cellular Screening Processes (LPCMC), Center of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Fakher Frikha
- Laboratory of Molecular and Cellular Screening Processes (LPCMC), Center of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Abdelaziz Tlili
- Department of Applied Biology, College of Sciences, University of Sharjah, Sharjah, United Arab Emirates
- Human Genetics and Stem Cell Laboratory, Research Institute of Sciences and Engineering, University of Sharjah, Sharjah, United Arab Emirates
| | | | - Taroh Kinoshita
- Laboratory of Immunoglycobiology, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
- Center for Infectious Disease Education and Research, Osaka University, Suita, Japan
| | - Chahnez Charfi Triki
- Child Neurology Department, University Hedi Chaker Hospital of Sfax, Sfax, Tunisia
- Research Laboratory “Neuropédiatrie” LR19ES15, Sfax University, Sfax, Tunisia
| | - Saber Masmoudi
- Laboratory of Molecular and Cellular Screening Processes (LPCMC), Center of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
- *Correspondence: Saber Masmoudi
| |
Collapse
|
7
|
Ku YS, Cheng SS, Cheung MY, Law CH, Lam HM. The Re-Localization of Proteins to or Away from Membranes as an Effective Strategy for Regulating Stress Tolerance in Plants. MEMBRANES 2022; 12:membranes12121261. [PMID: 36557168 PMCID: PMC9788111 DOI: 10.3390/membranes12121261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 06/12/2023]
Abstract
The membranes of plant cells are dynamic structures composed of phospholipids and proteins. Proteins harboring phospholipid-binding domains or lipid ligands can localize to membranes. Stress perception can alter the subcellular localization of these proteins dynamically, causing them to either associate with or detach from membranes. The mechanisms behind the re-localization involve changes in the lipidation state of the proteins and interactions with membrane-associated biomolecules. The functional significance of such re-localization includes the regulation of molecular transport, cell integrity, protein folding, signaling, and gene expression. In this review, proteins that re-localize to or away from membranes upon abiotic and biotic stresses will be discussed in terms of the mechanisms involved and the functional significance of their re-localization. Knowledge of the re-localization mechanisms will facilitate research on increasing plant stress adaptability, while the study on re-localization of proteins upon stresses will further our understanding of stress adaptation strategies in plants.
Collapse
|
8
|
Siavrienė E, Maldžienė Ž, Mikštienė V, Petraitytė G, Rančelis T, Dapkūnas J, Burnytė B, Benušienė E, Sasnauskienė A, Grikinienė J, Griškevičiūtė E, Utkus A, Preikšaitienė E. PIGN-Related Disease in Two Lithuanian Families: A Report of Two Novel Pathogenic Variants, Molecular and Clinical Characterisation. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:1526. [PMID: 36363484 PMCID: PMC9693321 DOI: 10.3390/medicina58111526] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/19/2022] [Accepted: 10/24/2022] [Indexed: 11/05/2022]
Abstract
Background and Objectives: Pathogenic variants of PIGN are a known cause of multiple congenital anomalies-hypotonia-seizures syndrome 1 (MCAHS1). Many affected individuals have clinical features overlapping with Fryns syndrome and are mainly characterised by developmental delay, congenital anomalies, hypotonia, seizures, and specific minor facial anomalies. This study investigates the clinical and molecular data of three individuals from two unrelated families, the clinical features of which were consistent with a diagnosis of MCAHS1. Materials and Methods: Next-generation sequencing (NGS) technology was used to identify the changes in the DNA sequence. Sanger sequencing of gDNA of probands and their parents was used for validation and segregation analysis. Bioinformatics tools were used to investigate the consequences of pathogenic or likely pathogenic PIGN variants at the protein sequence and structure level. Results: The analysis of NGS data and segregation analysis revealed a compound heterozygous NM_176787.5:c.[1942G>T];[1247_1251del] PIGN genotype in family 1 and NG_033144.1(NM_176787.5):c.[932T>G];[1674+1G>C] PIGN genotype in family 2. In silico, c.1942G>T (p.(Glu648Ter)), c.1247_1251del (p.(Glu416GlyfsTer22)), and c.1674+1G>C (p.(Glu525AspfsTer68)) variants are predicted to result in a premature termination codon that leads to truncated and functionally disrupted protein causing the phenotype of MCAHS1 in the affected individuals. Conclusions: PIGN-related disease represents a wide spectrum of phenotypic features, making clinical diagnosis inaccurate and complicated. The genetic testing of every individual with this phenotype provides new insights into the origin and development of the disease.
Collapse
Affiliation(s)
- Evelina Siavrienė
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, 08410 Vilnius, Lithuania
| | - Živilė Maldžienė
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, 08410 Vilnius, Lithuania
| | - Violeta Mikštienė
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, 08410 Vilnius, Lithuania
| | - Gunda Petraitytė
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, 08410 Vilnius, Lithuania
| | - Tautvydas Rančelis
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, 08410 Vilnius, Lithuania
| | - Justas Dapkūnas
- Department of Bioinformatics, Institute of Biotechnology, Life Sciences Center, Vilnius University, 10257 Vilnius, Lithuania
| | - Birutė Burnytė
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, 08410 Vilnius, Lithuania
| | - Eglė Benušienė
- Centre for Medical Genetics, Vilnius University Hospital Santaros Klinikos, 08410 Vilnius, Lithuania
| | - Aušra Sasnauskienė
- Department of Biochemistry and Molecular Biology, Institute of Biosciences, Life Sciences Centre, Vilnius University, 10257 Vilnius, Lithuania
| | - Jurgita Grikinienė
- Centre of Pediatrics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | | | - Algirdas Utkus
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, 08410 Vilnius, Lithuania
| | - Eglė Preikšaitienė
- Department of Human and Medical Genetics, Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, 08410 Vilnius, Lithuania
| |
Collapse
|
9
|
Jiang LH, Mu LL, Jin L, Anjum AA, Li GQ. Silencing uridine diphosphate N-acetylglucosamine pyrophosphorylase gene impairs larval development in Henosepilachna vigintioctopunctata. PEST MANAGEMENT SCIENCE 2022; 78:3894-3902. [PMID: 34523212 DOI: 10.1002/ps.6643] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/29/2021] [Accepted: 09/15/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Uridine diphosphate-N-acetylglucosamine (UDP-GlcNAc) diphosphorylase (UAP) catalyzes the formation of UDP-GlcNAc, the precursor for the production of chitin in ectodermally derived epidermal cells and midgut, for GlcNAcylation of proteins and for generation of glycosyl-phosphatidyl-inositol anchors in all tissues in Drosophila melanogaster. RESULTS Here, we identified a putative HvUAP gene in Henosepilachna vigintioctopunctata. Knockdown of HvUAP at the second-, third- and fourth-instar stages impaired larval development. Most resultant HvUAP hypomorphs showed arrested development at the third-, fourth-instar larval or prepupal stages, and became paralyzed, depending on the age when treated. Some HvUAP-silenced larvae had weak and soft scoli. A portion of HvUAP-depleted beetles formed misshapen pupae. No HvUAP RNA interference pupae successfully emerged as adults. Dissection and microscopic observation revealed that knockdown of HvUAP affected gut growth and food ingestion, reduced cuticle thickness, and negatively affected the formation of newly generated cuticle layers during ecdysis. Furthermore, HvUAP deficiency inhibited development of the tracheal respiratory system and thinned tracheal taenidia. CONCLUSION The phenotypical defects in HvUAP hypomorphs suggest that HvUAP is involved in the production of chitin. Moreover, our findings will enable the development of a double-stranded RNA-based pesticide to control H. vigintioctopunctata. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Lin-Hong Jiang
- Agriculture Ministry Key Laboratory of Integrated Pest Management on Crops in East China/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Li-Li Mu
- Agriculture Ministry Key Laboratory of Integrated Pest Management on Crops in East China/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Lin Jin
- Agriculture Ministry Key Laboratory of Integrated Pest Management on Crops in East China/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Ahmad A Anjum
- Agriculture Ministry Key Laboratory of Integrated Pest Management on Crops in East China/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| | - Guo-Qing Li
- Agriculture Ministry Key Laboratory of Integrated Pest Management on Crops in East China/State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Department of Entomology, College of Plant Protection, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
10
|
Molecular Characterization of UDP- N-Acetylglucosamine Pyrophosphorylase and Its Role in the Growth and Development of the White-Backed Planthopper Sogatella furcifera (Hemiptera: Delphacidae). Genes (Basel) 2022; 13:genes13081340. [PMID: 35893078 PMCID: PMC9332809 DOI: 10.3390/genes13081340] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/15/2022] [Accepted: 07/23/2022] [Indexed: 02/01/2023] Open
Abstract
UDP-N-acetylglucosamine pyrophosphorylase (UAP) is a key enzyme in the chitin biosynthesis pathway of insects. Here, we described the gene SfUAP in the white-backed planthopper Sogatella furcifera (Horváth) with an open reading frame of 1470 bp. Quantitative real-time polymerase chain reaction (qPCR) suggested that SfUAP exhibits a different developmental expression pattern and a higher expression after molting. The highest expression of SfUAP was observed in the integument tissues of adults, whereas head tissues showed negligible expression. RNAi-based gene silencing decreased the mRNA transcript levels in S. furcifera nymphs injected with double-stranded RNA of SfUAP. Finally, SfUAP silencing led to 84% mortality and malformed phenotypes in nymphs. Thus, our results can help better understand the role of SfUAP in S. furcifera.
Collapse
|
11
|
Ness TJ, Gamage DG, Ekanayaka SA, Hendrickson TL. A Soluble, Minimalistic Glycosylphosphatidylinositol Transamidase (GPI-T) Retains Transamidation Activity. Biochemistry 2022; 61:1273-1285. [PMID: 35730892 DOI: 10.1021/acs.biochem.2c00196] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Glycosylphosphatidylinositol (GPI) anchoring of proteins is a eukaryotic, post-translational modification catalyzed by GPI transamidase (GPI-T). The Saccharomyces cerevisiae GPI-T is composed of five membrane-bound subunits: Gpi8, Gaa1, Gpi16, Gpi17, and Gab1. GPI-T has been recalcitrant to in vitro structure and function studies because of its complexity and membrane-solubility. Furthermore, a reliable, quantitative, in vitro assay for this important post-translational modification has remained elusive despite its discovery more than three decades ago.Three recent reports describe the structure of GPI-T from S. cerevisiae and humans, shedding critical light on this important enzyme and offering insight into the functions of its different subunits. Here, we present the purification and characterization of a truncated soluble GPI-T heterotrimer complex (Gpi823-306, Gaa150-343, and Gpi1620-551) without transmembrane domains. Using this simplified heterotrimer, we report the first quantitative method to measure GPI-T activity in vitro and demonstrate that this soluble, minimalistic GPI-T retains transamidase activity. These results contribute to our understanding of how this enzyme is organized and functions, and provide a method to screen potential GPI-T inhibitors.
Collapse
Affiliation(s)
- Travis J Ness
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, United States
| | - Dilani G Gamage
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, United States
| | - Sandamali A Ekanayaka
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, United States
| | - Tamara L Hendrickson
- Department of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, United States
| |
Collapse
|
12
|
Glycosylphosphatidylinositol Anchor Biosynthesis Pathway-Related Protein GPI7 Is Required for the Vegetative Growth and Pathogenicity of Colletotrichum graminicola. Int J Mol Sci 2022; 23:ijms23062985. [PMID: 35328406 PMCID: PMC8949851 DOI: 10.3390/ijms23062985] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 01/04/2023] Open
Abstract
Glycosylphosphatidylinositol (GPI) anchoring is a common post-translational modification in eukaryotic cells and has been demonstrated to have a wide range of biological functions, such as signal transduction, cellular adhesion, protein transport, immune response, and maintaining cell wall integrity. More than 25 proteins have been proven to participate in the GPI anchor synthesis pathway which occurs in the cytoplasmic and the luminal face of the ER membrane. However, the essential proteins of the GPI anchor synthesis pathway are still less characterized in maize pathogen Colletotrichum graminicola. In the present study, we analyzed the biological function of the GPI anchor synthesis pathway-related gene, CgGPI7, that encodes an ethanolamine phosphate transferase, which is localized in ER. The vegetative growth and conidia development of the ΔCgGPI7 mutant was significantly impaired in C. graminicola. and qRT-PCR results showed that the transcriptional level of CgGPI7 was specifically induced in the initial infection stage and that the pathogenicity of ΔCgGPI7 mutant was also significantly decreased compared with the wild type. Furthermore, the ΔCgGPI7 mutant displayed more sensitivity to cell wall stresses, suggesting that CgGPI7 may play a role in the cell wall integrity of C. graminicola. Cell wall synthesis-associated genes were also quantified in the ΔCgGPI7 mutant, and the results showed that chitin and β-1,3-glucans synthesis genes were significantly up-regulated in ΔCgGPI7 mutants. Our results suggested that CgGPI7 is required for vegetative growth and pathogenicity and might depend on the cell wall integrity of C. graminicola.
Collapse
|
13
|
Structure of human glycosylphosphatidylinositol transamidase. Nat Struct Mol Biol 2022; 29:203-209. [DOI: 10.1038/s41594-022-00726-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/11/2022] [Indexed: 01/31/2023]
|
14
|
Julius BT, McCubbin TJ, Mertz RA, Baert N, Knoblauch J, Grant DG, Conner K, Bihmidine S, Chomet P, Wagner R, Woessner J, Grote K, Peevers J, Slewinski TL, McCann MC, Carpita NC, Knoblauch M, Braun DM. Maize Brittle Stalk2-Like3, encoding a COBRA protein, functions in cell wall formation and carbohydrate partitioning. THE PLANT CELL 2021; 33:3348-3366. [PMID: 34323976 PMCID: PMC8505866 DOI: 10.1093/plcell/koab193] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/16/2021] [Indexed: 05/14/2023]
Abstract
Carbohydrate partitioning from leaves to sink tissues is essential for plant growth and development. The maize (Zea mays) recessive carbohydrate partitioning defective28 (cpd28) and cpd47 mutants exhibit leaf chlorosis and accumulation of starch and soluble sugars. Transport studies with 14C-sucrose (Suc) found drastically decreased export from mature leaves in cpd28 and cpd47 mutants relative to wild-type siblings. Consistent with decreased Suc export, cpd28 mutants exhibited decreased phloem pressure in mature leaves, and altered phloem cell wall ultrastructure in immature and mature leaves. We identified the causative mutations in the Brittle Stalk2-Like3 (Bk2L3) gene, a member of the COBRA family, which is involved in cell wall development across angiosperms. None of the previously characterized COBRA genes are reported to affect carbohydrate export. Consistent with other characterized COBRA members, the BK2L3 protein localized to the plasma membrane, and the mutants condition a dwarf phenotype in dark-grown shoots and primary roots, as well as the loss of anisotropic cell elongation in the root elongation zone. Likewise, both mutants exhibit a significant cellulose deficiency in mature leaves. Therefore, Bk2L3 functions in tissue growth and cell wall development, and this work elucidates a unique connection between cellulose deposition in the phloem and whole-plant carbohydrate partitioning.
Collapse
Affiliation(s)
- Benjamin T Julius
- Divisions of Plant Science and Technology, Biological Sciences, Interdisciplinary Plant Group, and the Missouri Maize Center, University of Missouri, Columbia, Missouri 65211, USA
- Bayer Crop Science, Chesterfield, Missouri 63017, USA
| | - Tyler J McCubbin
- Divisions of Plant Science and Technology, Biological Sciences, Interdisciplinary Plant Group, and the Missouri Maize Center, University of Missouri, Columbia, Missouri 65211, USA
| | - Rachel A Mertz
- Divisions of Plant Science and Technology, Biological Sciences, Interdisciplinary Plant Group, and the Missouri Maize Center, University of Missouri, Columbia, Missouri 65211, USA
- Present address: Inari Agriculture, West Lafayette, Indiana 47906, USA
| | - Nick Baert
- Divisions of Plant Science and Technology, Biological Sciences, Interdisciplinary Plant Group, and the Missouri Maize Center, University of Missouri, Columbia, Missouri 65211, USA
| | - Jan Knoblauch
- School of Biological Sciences, Washington State University, Pullman, Washington 99164, USA
| | - DeAna G Grant
- Electron Microscopy Core Facility, University of Missouri, Columbia, Missouri 65211, USA
| | - Kyle Conner
- Divisions of Plant Science and Technology, Biological Sciences, Interdisciplinary Plant Group, and the Missouri Maize Center, University of Missouri, Columbia, Missouri 65211, USA
| | - Saadia Bihmidine
- Divisions of Plant Science and Technology, Biological Sciences, Interdisciplinary Plant Group, and the Missouri Maize Center, University of Missouri, Columbia, Missouri 65211, USA
| | - Paul Chomet
- NRGene Inc., 8910 University Center Lane, San Diego, California 92122, USA
| | - Ruth Wagner
- Bayer Crop Science, Chesterfield, Missouri 63017, USA
| | - Jeff Woessner
- Bayer Crop Science, Chesterfield, Missouri 63017, USA
| | - Karen Grote
- Bayer Crop Science, Chesterfield, Missouri 63017, USA
| | | | | | - Maureen C McCann
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907, USA
- Purdue Center for Plant Biology, Purdue University, West Lafayette, Indiana 47907, USA
| | - Nicholas C Carpita
- Department of Biological Sciences, Purdue University, West Lafayette, Indiana 47907, USA
- Purdue Center for Plant Biology, Purdue University, West Lafayette, Indiana 47907, USA
- Department of Botany and Plant Pathology, Purdue University, West Lafayette, Indiana 47907, USA
| | - Michael Knoblauch
- School of Biological Sciences, Washington State University, Pullman, Washington 99164, USA
| | - David M Braun
- Divisions of Plant Science and Technology, Biological Sciences, Interdisciplinary Plant Group, and the Missouri Maize Center, University of Missouri, Columbia, Missouri 65211, USA
- Author for correspondence:
| |
Collapse
|
15
|
Functional Analysis of the GPI Transamidase Complex by Screening for Amino Acid Mutations in Each Subunit. Molecules 2021; 26:molecules26185462. [PMID: 34576938 PMCID: PMC8465894 DOI: 10.3390/molecules26185462] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/01/2021] [Accepted: 09/01/2021] [Indexed: 12/20/2022] Open
Abstract
Glycosylphosphatidylinositol (GPI) anchor modification is a posttranslational modification of proteins that has been conserved in eukaryotes. The biosynthesis and transfer of GPI to proteins are carried out in the endoplasmic reticulum. Attachment of GPI to proteins is mediated by the GPI-transamidase (GPI-TA) complex, which recognizes and cleaves the C-terminal GPI attachment signal of precursor proteins. Then, GPI is transferred to the newly exposed C-terminus of the proteins. GPI-TA consists of five subunits: PIGK, GPAA1, PIGT, PIGS, and PIGU, and the absence of any subunit leads to the loss of activity. Here, we analyzed functionally important residues of the five subunits of GPI-TA by comparing conserved sequences among homologous proteins. In addition, we optimized the purification method for analyzing the structure of GPI-TA. Using purified GPI-TA, preliminary single particle images were obtained. Our results provide guidance for the structural and functional analysis of GPI-TA.
Collapse
|
16
|
First insights into the honey bee (Apis mellifera) brain lipidome and its neonicotinoid-induced alterations associated with reduced self-grooming behavior. J Adv Res 2021; 37:75-89. [PMID: 35499051 PMCID: PMC9039751 DOI: 10.1016/j.jare.2021.08.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/21/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022] Open
Abstract
First bee brain characterization shows distinctive low plasmalogens and high alkyl-ether levels. PC 20:3e/15:0, PC 16:0/18:3, PA 18:0/24:1 increased by the highest dose of clothianidin. Levels of CL 18:3/18:1/14:0/22:6, TG 6:0/11:2/18:1 and eLPE 18:0e were linked to intense grooming. Membrane lipids, like PC 18:1e/20:3, ePC 8:1e/20:3, and pPE 16:1p/24:1 were up-regulated by clothianidin. Clothianidin exposure up-regulated genes linked to GPI-anchor biosynthesis pathway. Lipids can be used as biomarkers to assess the effect of neurotoxins on behaviors.
Introduction Honey bees (Apis mellifera) play key roles in food production performing complex behaviors, like self-grooming to remove parasites. However, the lipids of their central nervous system have not been examined, even though they likely play a crucial role in the performance of cognitive process to perform intricate behaviors. Lipidomics has greatly advanced our understanding of neuropathologies in mammals and could provide the same for honey bees. Objectives The objectives of this study were to characterize the brain lipidome of adult honey bees and to assess the effect of clothianidin (a neurotoxic insecticide) on the brain lipid composition, gene expression, and performance of self-grooming behavior under controlled conditions (cage experiments). Methods After seven days of exposure to oral sublethal doses of clothianidin, the bees were assessed for self-grooming behavior; their brains were dissected to analyze the lipidome using an untargeted lipidomics approach and to perform a high throughput RNAseq analysis. Results Compared to all other organisms, healthy bee brain lipidomes contain unusually high levels of alkyl-ether linked (plasmanyl) phospholipids (51.42%) and low levels of plasmalogens (plasmenyl phospholipids; 3.46%). This could make it more susceptible to the effects of toxins in the environment. A positive correlation between CL 18:3/18:1/14:0/22:6, TG 6:0/11:2/18:1, LPE 18:0e and intense self-grooming was found. Sublethal doses of a neonicotinoid altered PC 20:3e/15:0, PC 16:0/18:3, PA 18:0/24:1, and TG 18:1/18:1/18/1 levels, and affected gene expression linked to GPI-anchor biosynthesis pathway and energy metabolism that may be partially responsible for the altered lipid composition. Conclusion This study showed that lipidomics can reveal honey bee neuropathologies associated with reduced grooming behavior due to sublethal neonicotinoid exposure. The ease of use, unusual brain lipidome as well as characterized behaviors that are affected by the environment make honey bees a promising model organism for studying the neurolipidome and associations with neurobehavioral disorders.
Collapse
|
17
|
Evolutionary Overview of Molecular Interactions and Enzymatic Activities in the Yeast Cell Walls. Int J Mol Sci 2020; 21:ijms21238996. [PMID: 33256216 PMCID: PMC7730094 DOI: 10.3390/ijms21238996] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/23/2020] [Accepted: 11/25/2020] [Indexed: 11/25/2022] Open
Abstract
Fungal cell walls are composed of a polysaccharide network that serves as a scaffold in which different glycoproteins are embedded. Investigation of fungal cell walls, besides simple identification and characterization of the main cell wall building blocks, covers the pathways and regulations of synthesis of each individual component of the wall and biochemical reactions by which they are cross-linked and remodeled in response to different growth phase and environmental signals. In this review, a survey of composition and organization of so far identified and characterized cell wall components of different yeast genera including Saccharomyces, Candida, Kluyveromyces, Yarrowia, and Schizosaccharomyces are presented with the focus on their cell wall proteomes.
Collapse
|
18
|
Su CTT, Sinha S, Eisenhaber B, Eisenhaber F. Structural modelling of the lumenal domain of human GPAA1, the metallo-peptide synthetase subunit of the transamidase complex, reveals zinc-binding mode and two flaps surrounding the active site. Biol Direct 2020; 15:14. [PMID: 32993792 PMCID: PMC7522609 DOI: 10.1186/s13062-020-00266-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/30/2020] [Indexed: 02/01/2023] Open
Abstract
Background The transamidase complex is a molecular machine in the endoplasmic reticulum of eukaryotes that attaches a glycosylphosphatidylinositol (GPI) lipid anchor to substrate proteins after cleaving a C-terminal propeptide with a defined sequence signal. Its five subunits are very hydrophobic; thus, solubility, heterologous expression and complex reconstruction are difficult. Therefore, theoretical approaches are currently the main source of insight into details of 3D structure and of the catalytic process. Results In this work, we generated model 3D structures of the lumenal domain of human GPAA1, the M28-type metallo-peptide-synthetase subunit of the transamidase, including zinc ion and model substrate positions. In comparative molecular dynamics (MD) simulations of M28-type structures and our GPAA1 models, we estimated the metal ion binding energies with evolutionary conserved amino acid residues in the catalytic cleft. We find that canonical zinc binding sites 2 and 3 are strongest binders for Zn1 and, where a second zinc is available, sites 2 and 4 for Zn2. Zinc interaction of site 5 with Zn1 enhances upon substrate binding in structures with only one zinc. Whereas a previously studied glutaminyl cyclase structure, the best known homologue to GPAA1, binds only one zinc ion at the catalytic site, GPAA1 can sterically accommodate two. The M28-type metallopeptidases segregate into two independent branches with regard to one/two zinc ion binding modality in a phylogenetic tree where the GPAA1 family is closer to the joint origin of both groups. For GPAA1 models, MD studies revealed two large loops (flaps) surrounding the active site being involved in an anti-correlated, breathing-like dynamics. Conclusions In the light of combined sequence-analytic and phylogenetic arguments as well as 3D structural modelling results, GPAA1 is most likely a single zinc ion metallopeptidase. Two large flaps environ the catalytic site restricting access to large substrates. Reviewers This article was reviewed by Thomas Dandekar (MD) and Michael Gromiha.
Collapse
Affiliation(s)
- Chinh Tran-To Su
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, # 07-01, Matrix, Singapore, 138671, Singapore
| | - Swati Sinha
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, # 07-01, Matrix, Singapore, 138671, Singapore
| | - Birgit Eisenhaber
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, # 07-01, Matrix, Singapore, 138671, Singapore.
| | - Frank Eisenhaber
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, # 07-01, Matrix, Singapore, 138671, Singapore. .,School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore.
| |
Collapse
|
19
|
Bayat A, Knaus A, Pendziwiat M, Afenjar A, Barakat TS, Bosch F, Callewaert B, Calvas P, Ceulemans B, Chassaing N, Depienne C, Endziniene M, Ferreira CR, Moura de Souza CF, Freihuber C, Ganesan S, Gataullina S, Guerrini R, Guerrot A, Hansen L, Jezela‐Stanek A, Karsenty C, Kievit A, Kooy FR, Korff CM, Kragh Hansen J, Larsen M, Layet V, Lesca G, McBride KL, Meuwissen M, Mignot C, Montomoli M, Moore H, Naudion S, Nava C, Nougues M, Parrini E, Pastore M, Schelhaas JH, Skinner S, Szczałuba K, Thomas A, Thomassen M, Tranebjærg L, Slegtenhorst M, Wolfe LA, Lal D, Gardella E, Bomme Ousager L, Brünger T, Helbig I, Krawitz P, Møller RS. Lessons learned from 40 novel
PIGA
patients and a review of the literature. Epilepsia 2020; 61:1142-1155. [DOI: 10.1111/epi.16545] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 01/08/2023]
Affiliation(s)
- Allan Bayat
- Institute for Regional Health Services University of Southern Denmark Odense Denmark
- Department of Epilepsy Genetics and Personalized Medicine Danish Epilepsy Center Dianalund Denmark
| | - Alexej Knaus
- Institute for Genomic Statistics and Bioinformatics University Hospital Bonn Rheinische Friedrich‐Wilhelms‐University Bonn Bonn Germany
| | - Manuela Pendziwiat
- Department of Neuropediatrics University Medical Center Schleswig‐Holstein Christian Albrechts University Kiel Germany
| | - Alexandra Afenjar
- CRMR Congenital Malformations and Diseases of the Cerebellum and Rare Causes of Intellectual Disabilities Department of Genetics Sorbonne University, AP‐HP, Trousseau Hospital Paris France
| | - Tahsin Stefan Barakat
- Department of Clinical Genetics Erasmus MC, University Medical Center Rotterdam the Netherlands
| | | | - Bert Callewaert
- Center for Medical Genetics Ghent University Hospital Ghent Belgium
- Department of Biomolecular Medicine Ghent University Ghent Belgium
| | - Patrick Calvas
- UMR1056 INSERM‐Université de Toulouse, Department of Genetics University Hospital of Toulouse Toulouse France
| | - Berten Ceulemans
- Department of Pediatric Neurology University Hospital and University of Antwerp Antwerp Belgium
| | - Nicolas Chassaing
- UMR1056 INSERM‐Université de Toulouse, Department of Genetics University Hospital of Toulouse Toulouse France
| | - Christel Depienne
- Institute of Human Genetics University Hospital Essen University of Duisburg‐Essen Essen Germany
- UMR S1127, Inserm U1127, CNRS UMR 7225 Institute of brain and spinal cord Sorbonne University Paris France
| | - Milda Endziniene
- Neurology Department Medical Academy Lithuanian University of Health Sciences Kaunas Lithuania
| | - Carlos R. Ferreira
- Medical Genomics and Metabolic Genetics Branch National Human Genome Research Institute, National Institutes of Health Bethesda MarylandUSA
| | | | - Cécile Freihuber
- Department of Pediatric Neurology AP‐HP, GHUEP Armand Trousseau University Hospital Paris France
- GRC ConCer‐LD Sorbonne University, UPMC University of Paris 06 Paris France
| | - Shiva Ganesan
- Division of Neurology Children’s Hospital of Philadelphia Philadelphia PennsylvaniaUSA
- Epilepsy NeuroGenetics Initiative Children's Hospital of Philadelphia Philadelphia PennsylvaniaUSA
- Department of Biomedical and Health Informatics Children’s Hospital of Philadelphia Philadelphia PennsylvaniaUSA
| | - Svetlana Gataullina
- Sleep Disorders Center AP‐HP, Antoine‐Béclère Hospital Clamart France
- Department of Pediatrics and Neonatal Intensive Care André Grégoire Hospital Montreuil France
| | - Renzo Guerrini
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories Department of Neuroscience, A. Meyer Children's Hospital University of Florence Florence Italy
| | - Anne‐Marie Guerrot
- Department of Genetics and Reference Center for Developmental Disorders Normandy Center for Genomic and Personalized Medicine Normandy University, UNIROUEN Inserm U1245 and Rouen University Hospital Rouen France
| | - Lars Hansen
- Department of Cellular and Molecular Medicine Faculty of Health Science Copenhagen Center for Glycomics Copenhagen Denmark
| | - Aleksandra Jezela‐Stanek
- Department of Genetics and Clinical Immunology National Institute of Tuberculosis and Lung Diseases Warsaw Poland
| | - Caroline Karsenty
- Neuropediatrics Department University Hospital of Toulouse Toulouse France
| | - Anneke Kievit
- Department of Clinical Genetics Erasmus MC, University Medical Center Rotterdam the Netherlands
| | - Frank R. Kooy
- Department of Medical Genetics University of Antwerp Antwerp Belgium
| | - Christian M. Korff
- Pediatric Neurology Unit Department of the Woman, Child, and Adolescent University Hospitals Geneva Geneva Switzerland
| | | | - Martin Larsen
- Department of Clinical Genetics Odense University Hospital Odense Denmark
- Human Genetics Department of Clinical Research University of Southern Denmark Odense Denmark
| | - Valérie Layet
- Department of Genetics Du Havre Hospital Le Havre France
| | - Gaetan Lesca
- Department of Medical Genetics Lyon University Hospital Lyon France
- Institut Neuromyogene University Claude Bernard Lyon 1, Lyon University Lyon France
| | - Kim L. McBride
- Division of Genetic and Genomic Medicine Nationwide Children's Hospital Columbus OhioUSA
- Center for Cardiovascular Research Nationwide Children's Hospital Columbus OhioUSA
- Department of Pediatrics Ohio State University Columbus OhioUSA
| | - Marije Meuwissen
- Department of Medical Genetics University of Antwerp Antwerp Belgium
| | - Cyril Mignot
- APHP Department of Genetics Pitié‐Salpêtrière Hospital Reference Center for Rare Causes of Intellectual Disabilities Paris France
- Department of Genetics Inserm U1127, CNRS UMR 7225 Institute for brain and spinal cord ICM, AP‐HP De la Pitié Salpêtrière Hospital, Sorbonne University Paris France
| | - Martino Montomoli
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories Department of Neuroscience, A. Meyer Children's Hospital University of Florence Florence Italy
| | - Hannah Moore
- Greenwood Genetic Center Greenwood South CarolinaUSA
| | - Sophie Naudion
- Department of Genetics, University of Bordeaux Bordeaux France
| | - Caroline Nava
- Department of Genetics Inserm U1127, CNRS UMR 7225 Institute for brain and spinal cord ICM, AP‐HP De la Pitié Salpêtrière Hospital, Sorbonne University Paris France
| | | | - Elena Parrini
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories Department of Neuroscience, A. Meyer Children's Hospital University of Florence Florence Italy
| | - Matthew Pastore
- Division of Genetic and Genomic Medicine Nationwide Children's Hospital Columbus OhioUSA
- Department of Pediatrics Ohio State University Columbus OhioUSA
| | | | | | | | - Ashley Thomas
- Department of Neurology University of Alabama at Birmingham Birmingham AlabamaUSA
| | - Mads Thomassen
- Department of Clinical Genetics Odense University Hospital Odense Denmark
- Human Genetics Department of Clinical Research University of Southern Denmark Odense Denmark
| | - Lisbeth Tranebjærg
- Department of Clinical Genetics Rigshospitalet/Kennedy Center Glostrup Denmark
- Institute of Clinical Medicine University of Copenhagen Copenhagen Denmark
| | - Marjon Slegtenhorst
- Department of Clinical Genetics Erasmus MC, University Medical Center Rotterdam the Netherlands
| | - Lynne A. Wolfe
- Undiagnosed Diseases Program, Common Fund National Institutes of Health Bethesda MarylandUSA
- Section of Human Biochemical Genetics National Human Genome Research Institute Bethesda MarylandUSA
| | - Dennis Lal
- Cologne Center for Genomics University Hospital Cologne, University of Cologne Cologne Germany
- Stanley Center for Psychiatric Research Broad Institute of Massachusetts Institute of Technology and Harvard Cambridge MassachusettsUSA
- Analytic and Translational Genetics Unit Massachusetts General Hospital Boston MassachusettsUSA
- Epilepsy Center Neurological Institute Cleveland Clinic Cleveland OhioUSA
- Genomic Medicine Institute Lerner Research Institute Cleveland Clinic Cleveland OhioUSA
| | - Elena Gardella
- Institute for Regional Health Services University of Southern Denmark Odense Denmark
- Department of Epilepsy Genetics and Personalized Medicine Danish Epilepsy Center Dianalund Denmark
- Department of Clinical Neurophysiology Danish Epilepsy Center Dianalund Denmark
| | - Lilian Bomme Ousager
- Department of Clinical Genetics Odense University Hospital Odense Denmark
- Human Genetics Department of Clinical Research University of Southern Denmark Odense Denmark
| | - Tobias Brünger
- Cologne Center for Genomics University Hospital Cologne, University of Cologne Cologne Germany
| | - Ingo Helbig
- Department of Neuropediatrics University Medical Center Schleswig‐Holstein Christian Albrechts University Kiel Germany
- Division of Neurology Children’s Hospital of Philadelphia Philadelphia PennsylvaniaUSA
- Epilepsy NeuroGenetics Initiative Children's Hospital of Philadelphia Philadelphia PennsylvaniaUSA
- Department of Biomedical and Health Informatics Children’s Hospital of Philadelphia Philadelphia PennsylvaniaUSA
- Department of Neurology University of Pennsylvania, Perelman School of Medicine Philadelphia PennsylvaniaUSA
| | - Peter Krawitz
- Institute for Genomic Statistics and Bioinformatics University Hospital Bonn Rheinische Friedrich‐Wilhelms‐University Bonn Bonn Germany
| | - Rikke S. Møller
- Institute for Regional Health Services University of Southern Denmark Odense Denmark
- Department of Epilepsy Genetics and Personalized Medicine Danish Epilepsy Center Dianalund Denmark
| |
Collapse
|
20
|
Yu J, Murthy V, Liu SL. Relating GPI-Anchored Ly6 Proteins uPAR and CD59 to Viral Infection. Viruses 2019; 11:E1060. [PMID: 31739586 PMCID: PMC6893729 DOI: 10.3390/v11111060] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 11/10/2019] [Accepted: 11/12/2019] [Indexed: 12/30/2022] Open
Abstract
The Ly6 (lymphocyte antigen-6)/uPAR (urokinase-type plasminogen activator receptor) superfamily protein is a group of molecules that share limited sequence homology but conserved three-fingered structures. Despite diverse cellular functions, such as in regulating host immunity, cell adhesion, and migration, the physiological roles of these factors in vivo remain poorly characterized. Notably, increasing research has focused on the interplays between Ly6/uPAR proteins and viral pathogens, the results of which have provided new insight into viral entry and virus-host interactions. While LY6E (lymphocyte antigen 6 family member E), one key member of the Ly6E/uPAR-family proteins, has been extensively studied, other members have not been well characterized. Here, we summarize current knowledge of Ly6/uPAR proteins related to viral infection, with a focus on uPAR and CD59. Our goal is to provide an up-to-date view of the Ly6/uPAR-family proteins and associated virus-host interaction and viral pathogenesis.
Collapse
Affiliation(s)
- Jingyou Yu
- Center for Retrovirus Research, The Ohio State University, Columbus, OH 43210, USA; (J.Y.); (V.M.)
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA
| | - Vaibhav Murthy
- Center for Retrovirus Research, The Ohio State University, Columbus, OH 43210, USA; (J.Y.); (V.M.)
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA
| | - Shan-Lu Liu
- Center for Retrovirus Research, The Ohio State University, Columbus, OH 43210, USA; (J.Y.); (V.M.)
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210, USA
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA
- Viruses and Emerging Pathogens Program, Infectious Diseases Institute, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
21
|
Patel PK, Free SJ. The Genetics and Biochemistry of Cell Wall Structure and Synthesis in Neurospora crassa, a Model Filamentous Fungus. Front Microbiol 2019; 10:2294. [PMID: 31649638 PMCID: PMC6796803 DOI: 10.3389/fmicb.2019.02294] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/20/2019] [Indexed: 01/25/2023] Open
Abstract
This review discusses the wealth of information available for the N. crassa cell wall. The basic organization and structure of the cell wall is presented and how the wall changes during the N. crassa life cycle is discussed. Over forty cell wall glycoproteins have been identified by proteomic analyses. Genetic and biochemical studies have identified many of the key enzymes needed for cell wall biogenesis, and the roles these enzymes play in cell wall biogenesis are discussed. The review includes a discussion of how the major cell wall components (chitin, β-1,3-glucan, mixed β-1,3-/ β-1,4- glucans, glycoproteins, and melanin) are synthesized and incorporated into the cell wall. We present a four-step model for how cell wall glycoproteins are covalently incorporated into the cell wall. In N. crassa, the covalent incorporation of cell wall glycoproteins into the wall occurs through a glycosidic linkage between lichenin (a mixed β-1,3-/β-1,4- glucan) and a "processed" galactomannan that has been attached to the glycoprotein N-linked oligosaccharides. The first step is the addition of the galactomannan to the N-linked oligosaccharide. Mutants affected in galactomannan formation are unable to incorporate glycoproteins into their cell walls. The second step is carried out by the enzymes from the GH76 family of α-1,6-mannanases, which cleave the galactomannan to generate a processed galactomannan. The model suggests that the third and fourth steps are carried out by members of the GH72 family of glucanosyltransferases. In the third step the glucanosyltransferases cleave lichenin and generate enzyme/substrate intermediates in which the lichenin is covalently attached to the active site of the glucanosyltransferases. In the final step, the glucanosyltransferases attach the lichenin onto the processed galactomannans, which creates new glycosidic bonds and effectively incorporates the glycoproteins into the cross-linked cell wall glucan/chitin matrix.
Collapse
Affiliation(s)
| | - Stephen J. Free
- Department of Biological Sciences, SUNY University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
22
|
Abstract
Metabolic pathways integrate to support tissue homeostasis and to prompt changes in cell phenotype. In particular, the heart consumes relatively large amounts of substrate not only to regenerate ATP for contraction but also to sustain biosynthetic reactions for replacement of cellular building blocks. Metabolic pathways also control intracellular redox state, and metabolic intermediates and end products provide signals that prompt changes in enzymatic activity and gene expression. Mounting evidence suggests that the changes in cardiac metabolism that occur during development, exercise, and pregnancy as well as with pathological stress (eg, myocardial infarction, pressure overload) are causative in cardiac remodeling. Metabolism-mediated changes in gene expression, metabolite signaling, and the channeling of glucose-derived carbon toward anabolic pathways seem critical for physiological growth of the heart, and metabolic inefficiency and loss of coordinated anabolic activity are emerging as proximal causes of pathological remodeling. This review integrates knowledge of different forms of cardiac remodeling to develop general models of how relationships between catabolic and anabolic glucose metabolism may fortify cardiac health or promote (mal)adaptive myocardial remodeling. Adoption of conceptual frameworks based in relational biology may enable further understanding of how metabolism regulates cardiac structure and function.
Collapse
Affiliation(s)
- Andrew A Gibb
- From the Center for Translational Medicine, Lewis Katz School of Medicine, Temple University, Philadelphia, PA (A.A.G.)
| | - Bradford G Hill
- the Department of Medicine, Institute of Molecular Cardiology, Diabetes and Obesity Center, University of Louisville School of Medicine, KY (B.G.H.).
| |
Collapse
|
23
|
Metabolic Labeling and Structural Analysis of Glycosylphosphatidylinositols from Parasitic Protozoa. Methods Mol Biol 2019. [PMID: 31256378 DOI: 10.1007/978-1-4939-9055-9_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Glycosylphosphatidylinositol (GPI) is a complex glycolipid structure that acts as a membrane anchor for many cell-surface proteins of eukaryotes. GPI-anchored proteins are particularly abundant in protozoa and represent the major carbohydrate modification of many cell-surface parasite proteins. A minimal GPI-anchor precursor consists of core glycan (ethanolamine-PO4-Manα1-2Manα1-6Manα1-4GlcNH2) linked to the 6-position of the D-myo-inositol ring of phosphatidylinositol. Although the GPI core glycan is conserved in all organisms, many differences in additional modifications to GPI structures and biosynthetic pathways have been reported. The preassembled GPI-anchor precursor is post-translationally transferred to a variety of membrane proteins in the lumen of the endoplasmic reticulum in a transamidase-like reaction during which a C-terminal GPI attachment signal is released. Increasing evidence shows that a significant proportion of the synthesized GPIs are not used for protein anchoring, particularly in protozoa in which a large amount of free GPIs are being displayed at the cell surface. The characteristics of GPI biosynthesis are currently being explored for the development of parasite-specific inhibitors. Especially this pathway, at least for Trypanosoma brucei, has been validated as a drug target. Furthermore, thanks to an increase of new innovative strategies to produce pure synthetic carbohydrates, a novel era in the use of GPIs in diagnostic, anti-GPI antibody production, as well as parasitic protozoa GPI-based vaccine approach is developing fast.
Collapse
|
24
|
Komath SS, Singh SL, Pratyusha VA, Sah SK. Generating anchors only to lose them: The unusual story of glycosylphosphatidylinositol anchor biosynthesis and remodeling in yeast and fungi. IUBMB Life 2019; 70:355-383. [PMID: 29679465 DOI: 10.1002/iub.1734] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 02/16/2018] [Accepted: 02/22/2018] [Indexed: 02/06/2023]
Abstract
Glycosylphosphatidylinositol (GPI)-anchored proteins (GPI-APs) are present ubiquitously at the cell surface in all eukaryotes. They play a crucial role in the interaction of the cell with its external environment, allowing the cell to receive signals, respond to challenges, and mediate adhesion. In yeast and fungi, they also participate in the structural integrity of the cell wall and are often essential for survival. Roughly four decades after the discovery of the first GPI-APs, this review provides an overview of the insights gained from studies of the GPI biosynthetic pathway and the future challenges in the field. In particular, we focus on the biosynthetic pathway in Saccharomyces cerevisiae, which has for long been studied as a model organism. Where available, we also provide information about the GPI biosynthetic steps in other yeast/ fungi. Although the core structure of the GPI anchor is conserved across organisms, several variations are built into the biosynthetic pathway. The present Review specifically highlights these variations and their implications. There is growing evidence to suggest that several phenotypes are common to GPI deficiency and should be expected in GPI biosynthetic mutants. However, it appears that several phenotypes are unique to a specific step in the pathway and may even be species-specific. These could suggest the points at which the GPI biosynthetic pathway intersects with other important cellular pathways and could be points of regulation. They could be of particular significance in the study of pathogenic fungi and in identification of new and specific antifungal drugs/ drug targets. © 2018 IUBMB Life, 70(5):355-383, 2018.
Collapse
Affiliation(s)
| | - Sneh Lata Singh
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | | | - Sudisht Kumar Sah
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
25
|
Cui A, Li Y, Zhou X, Wang L, Luo E. Characterization of Plasmodium berghei Homologues of T-cell Immunomodulatory Protein as a New Potential Candidate for Protecting against Experimental Cerebral Malaria. THE KOREAN JOURNAL OF PARASITOLOGY 2019; 57:101-115. [PMID: 31104402 PMCID: PMC6526220 DOI: 10.3347/kjp.2019.57.2.101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 04/14/2019] [Accepted: 04/16/2019] [Indexed: 11/27/2022]
Abstract
The pathogenesis of cerebral malaria is biologically complex and involves multi-factorial mechanisms such as microvascular congestion, immunopathology by the pro-inflammatory cytokine and endothelial dysfunction. Recent data have suggested that a pleiotropic T-cell immunomodulatory protein (TIP) could effectively mediate inflammatory cytokines of mammalian immune response against acute graft-versus-host disease in animal models. In this study, we identified a conserved homologue of TIP in Plasmodium berghei (PbTIP) as a membrane protein in Plasmodium asexual stage. Compared with PBS control group, the pathology of experimental cerebral malaria (ECM) in rPbTIP intravenous injection (i.v.) group was alleviated by the downregulation of pro-inflammatory responses, and rPbTIP i.v. group elicited an expansion of regulatory T-cell response. Therefore, rPbTIP i.v. group displayed less severe brain pathology and feverish mice in rPbTIP i.v. group died from ECM. This study suggested that PbTIP may be a novel promising target to alleviate the severity of ECM.
Collapse
Affiliation(s)
- Ai Cui
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Yucen Li
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Xia Zhou
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Lin Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Enjie Luo
- Department of Pathogen Biology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning 110122, P.R. China
| |
Collapse
|
26
|
Liu X, Zhang J, Zhu KY. Chitin in Arthropods: Biosynthesis, Modification, and Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1142:169-207. [PMID: 31102247 DOI: 10.1007/978-981-13-7318-3_9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Chitin is a structural constituent of extracellular matrices including the cuticle of the exoskeleton and the peritrophic matrix (PM) of the midgut in arthropods. Chitin chains are synthesized through multiple biochemical reactions, organized in several hierarchical levels and associated with various proteins that give their unique physicochemical characteristics of the cuticle and PM. Because, arthropod growth and morphogenesis are dependent on the capability of remodeling chitin-containing structures, chitin biosynthesis and degradation are highly regulated, allowing ecdysis and regeneration of the cuticle and PM. Over the past 20 years, much progress has been made in understanding the physiological functions of chitinous matrices. In this chapter, we mainly discussed the biochemical processes of chitin biosynthesis, modification and degradation, and various enzymes involved in these processes. We also discussed cuticular proteins and PM proteins, which largely determine the physicochemical properties of the cuticle and PM. Although rapid advances in genomics, proteomics, RNA interference, and other technologies have considerably facilitated our research in chitin biosynthesis, modification, and metabolism in recent years, many aspects of these processes are still partially understood. Further research is needed in understanding how the structural organization of chitin synthase in plasma membrane accommodate chitin biosynthesis, transport of chitin chain across the plasma membrane, and release of the chitin chain from the enzyme. Other research is also needed in elucidating the roles of chitin deacetylases in chitin organization and the mechanism controlling the formation of different types of chitin in arthropods.
Collapse
Affiliation(s)
- Xiaojian Liu
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Jianzhen Zhang
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi, 030006, China.
| | - Kun Yan Zhu
- Department of Entomology, Kansas State University, 123 Waters Hall, Manhattan, KS, 66506, USA.
| |
Collapse
|
27
|
The Glycosylphosphatidylinositol Transamidase Complex Subunit PbGPI16 of Plasmodium berghei Is Important for Inducing Experimental Cerebral Malaria. Infect Immun 2018; 86:IAI.00929-17. [PMID: 29784863 DOI: 10.1128/iai.00929-17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 05/18/2018] [Indexed: 02/07/2023] Open
Abstract
In animal models of experimental cerebral malaria (ECM), the glycosylphosphatidylinositols (GPIs) and GPI anchors are the major factors that induce nuclear factor kappa B (NF-κB) activation and proinflammatory responses, which contribute to malaria pathogenesis. GPIs and GPI anchors are transported to the cell surface via a process called GPI transamidation, which involves the GPI transamidase (GPI-T) complex. In this study, we showed that GPI16, one of the GPI-T subunits, is highly conserved among Plasmodium species. Genetic knockout of pbgpi16 (Δpbgpi16) in the rodent malaria parasite Plasmodium berghei strain ANKA led to a significant reduction of the amounts of GPIs in the membranes of merozoites, as well as surface display of several GPI-anchored merozoite surface proteins. Compared with the wild-type parasites, Δpbgpi16 parasites in C57BL/6 mice caused much less NF-κB activation and elicited a substantially attenuated T helper type 1 response. As a result, Δpbgpi16 mutant-infected mice displayed much less severe brain pathology, and considerably fewer Δpbgpi16 mutant-infected mice died from ECM. This study corroborated the GPI toxin as a significant inducer of ECM and further suggested that vaccines against parasite GPIs may be a promising strategy to limit the severity of malaria.
Collapse
|
28
|
Eisenhaber B, Sinha S, Wong WC, Eisenhaber F. Function of a membrane-embedded domain evolutionarily multiplied in the GPI lipid anchor pathway proteins PIG-B, PIG-M, PIG-U, PIG-W, PIG-V, and PIG-Z. Cell Cycle 2018; 17:874-880. [PMID: 29764287 PMCID: PMC6056205 DOI: 10.1080/15384101.2018.1456294] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Distant homology relationships among proteins with many transmembrane regions (TMs) are difficult to detect as they are clouded by the TMs’ hydrophobic compositional bias and mutational divergence in connecting loops. In the case of several GPI lipid anchor biosynthesis pathway components, the hidden evolutionary signal can be revealed with dissectHMMER, a sequence similarity search tool focusing on fold-critical, high complexity sequence segments. We find that a sequence module with 10 TMs in PIG-W, described as acyl transferase, is homologous to PIG-U, a transamidase subunit without characterized molecular function, and to mannosyltransferases PIG-B, PIG-M, PIG-V and PIG-Z. We conclude that this new, membrane-embedded domain named BindGPILA functions as the unit for recognizing, binding and stabilizing the GPI lipid anchor in a modification-competent form as this appears the only functional aspect shared among all proteins. Thus, PIG-U's likely molecular function is shuttling/presenting the anchor in a productive conformation to the transamidase complex.
Collapse
Affiliation(s)
- Birgit Eisenhaber
- a Bioinformatics Institute, Agency for Science , Technology and Research (A*STAR) , 30 Biopolis Street, #07-01 Matrix, Singapore 138671 , Republic of Singapore
| | - Swati Sinha
- a Bioinformatics Institute, Agency for Science , Technology and Research (A*STAR) , 30 Biopolis Street, #07-01 Matrix, Singapore 138671 , Republic of Singapore
| | - Wing-Cheong Wong
- a Bioinformatics Institute, Agency for Science , Technology and Research (A*STAR) , 30 Biopolis Street, #07-01 Matrix, Singapore 138671 , Republic of Singapore
| | - Frank Eisenhaber
- a Bioinformatics Institute, Agency for Science , Technology and Research (A*STAR) , 30 Biopolis Street, #07-01 Matrix, Singapore 138671 , Republic of Singapore.,b School of Computer Engineering , Nanyang Technological University (NTU) , 50 Nanyang Drive, Singapore 637553 , Republic of Singapore
| |
Collapse
|
29
|
Enk J, Levi A, Weisblum Y, Yamin R, Charpak-Amikam Y, Wolf DG, Mandelboim O. HSV1 MicroRNA Modulation of GPI Anchoring and Downstream Immune Evasion. Cell Rep 2017; 17:949-956. [PMID: 27760325 PMCID: PMC5081403 DOI: 10.1016/j.celrep.2016.09.077] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 06/28/2016] [Accepted: 09/22/2016] [Indexed: 12/24/2022] Open
Abstract
Herpes simplex virus 1 (HSV1) is a ubiquitous human pathogen that utilizes variable mechanisms to evade immune surveillance. The glycosylphosphatidylinositol (GPI) anchoring pathway is a multistep process in which a myriad of different proteins are covalently attached to a GPI moiety to be presented on the cell surface. Among the different GPI-anchored proteins there are many with immunological importance. We present evidence that the HSV1-encoded miR H8 directly targets PIGT, a member of the protein complex that covalently attaches proteins to GPI in the final step of GPI anchoring. This results in a membrane down-modulation of several different immune-related, GPI-anchored proteins, including ligands for natural killer-activating receptors and the prominent viral restriction factor tetherin. Thus, we suggest that by utilizing just one of dozens of miRNAs encoded by HSV1, the virus can counteract the host immune response at several key points. HSV1 miR H8 targets PIGT of the GPI anchoring pathway Expression of the anti-viral protein tetherin is reduced and viral spread enhanced Expression of GPI-anchored activating NK cell ligands is reduced Recognition and elimination by NK cells decrease
Collapse
Affiliation(s)
- Jonatan Enk
- The Lautenberg Center of General and Tumor Immunology, IMRIC, The Hebrew University Faculty of Medicine, Jerusalem 91120, Israel
| | - Assi Levi
- Photodermatosis Clinic and Laser Unit, Dermatology Department, Rabin Medical Center, Petah Tikva 4941492, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Yiska Weisblum
- Clinical Virology Unit, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel; Department of Biochemistry and the Chanock Center for Virology, IMRIC, The Hebrew University Faculty of Medicine, Jerusalem 91120, Israel
| | - Rachel Yamin
- The Lautenberg Center of General and Tumor Immunology, IMRIC, The Hebrew University Faculty of Medicine, Jerusalem 91120, Israel
| | - Yoav Charpak-Amikam
- The Lautenberg Center of General and Tumor Immunology, IMRIC, The Hebrew University Faculty of Medicine, Jerusalem 91120, Israel
| | - Dana G Wolf
- Clinical Virology Unit, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Ofer Mandelboim
- The Lautenberg Center of General and Tumor Immunology, IMRIC, The Hebrew University Faculty of Medicine, Jerusalem 91120, Israel.
| |
Collapse
|
30
|
The soluble domains of Gpi8 and Gaa1, two subunits of glycosylphosphatidylinositol transamidase (GPI-T), assemble into a complex. Arch Biochem Biophys 2017; 633:58-67. [DOI: 10.1016/j.abb.2017.09.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/06/2017] [Accepted: 09/07/2017] [Indexed: 11/23/2022]
|
31
|
Disulfide Bond Formation and N-Glycosylation Modulate Protein-Protein Interactions in GPI-Transamidase (GPIT). Sci Rep 2017; 8:45912. [PMID: 28374821 PMCID: PMC5379210 DOI: 10.1038/srep45912] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/06/2017] [Indexed: 12/28/2022] Open
Abstract
Glycosylphosphatidylinositol (GPI) transamidase (GPIT), the enzyme that attaches GPI anchors to proteins as they enter the lumen of the endoplasmic reticulum, is a membrane-bound hetero-pentameric complex consisting of Gpi8, Gpi16, Gaa1, Gpi17 and Gab1. Here, we expressed and purified the luminal domain of Saccharomyces cerevisiae (S. cerevisiae) Gpi8 using different expression systems, and examined its interaction with insect cell expressed luminal domain of S. cerevisiae Gpi16. We found that the N-terminal caspase-like domain of Gpi8 forms a disulfide-linked dimer, which is strengthened by N-glycosylation. The non-core domain of Gpi8 following the caspase-like domain inhibits this dimerization. In contrast to the previously reported disulfide linkage between Gpi8 and Gpi16 in human and trypanosome GPIT, our data show that the luminal domains of S. cerevisiae Gpi8 and S. cerevisiae Gpi16 do not interact directly, nor do they form a disulfide bond in the intact S. cerevisiae GPIT. Our data suggest that subunit interactions within the GPIT complex from different species may vary, a feature that should be taken into account in future structural and functional studies.
Collapse
|
32
|
Abe Y, Inoue H, Ashida H, Maeda Y, Kinoshita T, Kitada S. Glycan region of GPI anchored-protein is required for cytocidal oligomerization of an anticancer parasporin-2, Cry46Aa1 protein, from Bacillus thuringiensis strain A1547. J Invertebr Pathol 2016; 142:71-81. [PMID: 27863961 DOI: 10.1016/j.jip.2016.11.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 11/04/2016] [Accepted: 11/14/2016] [Indexed: 11/25/2022]
Abstract
Parasporin-2 (PS2), alternatively named Cry46Aa1, an anticancer protein derived from Bacillus thuringiensis strain A1547, causes specific cell damage via PS2 oligomerization in the cell membrane. Although PS2 requires glycosylphosphatidylinositol (GPI)-anchored proteins for its cytocidal action, their precise role is unknown. Here, we report that the glycan of GPI induces PS2 oligomerization, which causes cell death. Cytotoxicity, cell-binding and oligomerization of the toxin were not observed in GPI-anchored protein-deficient Chinese hamster ovary cells. Expression and protease-treatment analyses showed that the actions of the toxin were dependent on the glycan core, not the polypeptide moiety, of GPI-anchored proteins. However, surface expression of some GPI-anchored proteins is observed in PS2-insensitive cells. These data suggest that GPI-anchored proteins do not determine the target specificity, but instead function as a kind of coreceptor, in the cytocidal action of PS2.
Collapse
Affiliation(s)
- Yuich Abe
- Department of Chemistry, Faculty of Science, Kyushu University, Fukuoka 812-8581, Japan
| | - Hiroshi Inoue
- Department of Chemistry, Faculty of Science, Kyushu University, Fukuoka 812-8581, Japan
| | - Hisashi Ashida
- Department of Immunoregulation, Research Institute for Microbial Diseases, Osaka University, Osaka, Osaka 565-0871, Japan
| | - Yusuke Maeda
- Department of Immunoregulation, Research Institute for Microbial Diseases, Osaka University, Osaka, Osaka 565-0871, Japan
| | - Taroh Kinoshita
- Department of Immunoregulation, Research Institute for Microbial Diseases, Osaka University, Osaka, Osaka 565-0871, Japan
| | - Sakae Kitada
- Department of Chemistry, Faculty of Science, Kyushu University, Fukuoka 812-8581, Japan; Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Iizuka, Fukuoka 820-8502, Japan.
| |
Collapse
|
33
|
Oliveira-Garcia E, Deising HB. The Glycosylphosphatidylinositol Anchor Biosynthesis Genes GPI12, GAA1, and GPI8 Are Essential for Cell-Wall Integrity and Pathogenicity of the Maize Anthracnose Fungus Colletotrichum graminicola. MOLECULAR PLANT-MICROBE INTERACTIONS : MPMI 2016; 29:889-901. [PMID: 27937175 DOI: 10.1094/mpmi-09-16-0175-r] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Glycosylphosphatidylinositol (GPI) anchoring of proteins is one of the most common posttranslational modifications of proteins in eukaryotic cells and is important for associating proteins with the cell surface. In fungi, GPI-anchored proteins play essential roles in cross-linking of β-glucan cell-wall polymers and cell-wall rigidity. GPI-anchor synthesis is successively performed at the cytoplasmic and the luminal face of the ER membrane and involves approximately 25 proteins. While mutagenesis of auxiliary genes of this pathway suggested roles of GPI-anchored proteins in hyphal growth and virulence, essential genes of this pathway have not been characterized. Taking advantage of RNA interference (RNAi) we analyzed the function of the three essential genes GPI12, GAA1 and GPI8, encoding a cytoplasmic N-acetylglucosaminylphosphatidylinositol deacetylase, a metallo-peptide-synthetase and a cystein protease, the latter two representing catalytic components of the GPI transamidase complex. RNAi strains showed drastic cell-wall defects, resulting in exploding infection cells on the plant surface and severe distortion of in planta-differentiated infection hyphae, including formation of intrahyphal hyphae. Reduction of transcript abundance of the genes analyzed resulted in nonpathogenicity. We show here for the first time that the GPI synthesis genes GPI12, GAA1, and GPI8 are indispensable for vegetative development and pathogenicity of the causal agent of maize anthracnose, Colletotrichum graminicola.
Collapse
Affiliation(s)
- Ely Oliveira-Garcia
- 1 Martin-Luther-Universität Halle-Wittenberg, Naturwissenschaftliche Fakultät III, Institut für Agrar- und Ernährungswissenschaften, Phytopathologie und Pflanzenschutz, and
| | - Holger B Deising
- 1 Martin-Luther-Universität Halle-Wittenberg, Naturwissenschaftliche Fakultät III, Institut für Agrar- und Ernährungswissenschaften, Phytopathologie und Pflanzenschutz, and
- 2 Interdisziplinäres Zentrum für Nutzpflanzenforschung; Betty-Heimann-Str. 3. D-06120 Halle/Saale, Germany
| |
Collapse
|
34
|
Mukai Y, Takahashi D, Inoue K, Sugita H, Kikegawa T, Etchuya K. Secondary Structure of GPI Attachment Signal Region Monitored by Circular Dichroism. CHEM LETT 2016. [DOI: 10.1246/cl.160551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
35
|
Lombard J. The multiple evolutionary origins of the eukaryotic N-glycosylation pathway. Biol Direct 2016; 11:36. [PMID: 27492357 PMCID: PMC4973528 DOI: 10.1186/s13062-016-0137-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 07/26/2016] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND The N-glycosylation is an essential protein modification taking place in the membranes of the endoplasmic reticulum (ER) in eukaryotes and the plasma membranes in archaea. It shares mechanistic similarities based on the use of polyisoprenol lipid carriers with other glycosylation pathways involved in the synthesis of bacterial cell wall components (e.g. peptidoglycan and teichoic acids). Here, a phylogenomic analysis was carried out to examine the validity of rival hypotheses suggesting alternative archaeal or bacterial origins to the eukaryotic N-glycosylation pathway. RESULTS The comparison of several polyisoprenol-based glycosylation pathways from the three domains of life shows that most of the implicated proteins belong to a limited number of superfamilies. The N-glycosylation pathway enzymes are ancestral to the eukaryotes, but their origins are mixed: Alg7, Dpm and maybe also one gene of the glycosyltransferase 1 (GT1) superfamily and Stt3 have proteoarchaeal (TACK superphylum) origins; alg2/alg11 may have resulted from the duplication of the original GT1 gene; the lumen glycosyltransferases were probably co-opted and multiplied through several gene duplications during eukaryogenesis; Alg13/Alg14 are more similar to their bacterial homologues; and Alg1, Alg5 and a putative flippase have unknown origins. CONCLUSIONS The origin of the eukaryotic N-glycosylation pathway is not unique and less straightforward than previously thought: some basic components likely have proteoarchaeal origins, but the pathway was extensively developed before the eukaryotic diversification through multiple gene duplications, protein co-options, neofunctionalizations and even possible horizontal gene transfers from bacteria. These results may have important implications for our understanding of the ER evolution and eukaryogenesis. REVIEWERS This article was reviewed by Pr. Patrick Forterre and Dr. Sergei Mekhedov (nominated by Editorial Board member Michael Galperin).
Collapse
Affiliation(s)
- Jonathan Lombard
- National Evolutionary Synthesis Center, 2024 W. Main Street Suite A200, Durham, NC, 27705, USA.
- Biosciences, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, EX4 4QD, UK.
| |
Collapse
|
36
|
Extent of pre-translational regulation for the control of nucleocytoplasmic protein localization. BMC Genomics 2016; 17:472. [PMID: 27342569 PMCID: PMC4919871 DOI: 10.1186/s12864-016-2854-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 06/22/2016] [Indexed: 11/18/2022] Open
Abstract
Background Appropriate protein subcellular localization is essential for proper cellular function. Central to the regulation of protein localization are protein targeting motifs, stretches of amino acids serving as guides for protein entry in a specific cellular compartment. While the use of protein targeting motifs is modulated in a post-translational manner, mainly by protein conformational changes and post-translational modifications, the presence of these motifs in proteins can also be regulated in a pre-translational manner. Here, we investigate the extent of pre-translational regulation of the main signals controlling nucleo-cytoplasmic traffic: the nuclear localization signal (NLS) and the nuclear export signal (NES). Results Motif databases and manual curation of the literature allowed the identification of 175 experimentally validated NLSs and 120 experimentally validated NESs in human. Following mapping onto annotated transcripts, these motifs were found to be modular, most (73 % for NLS and 88 % for NES) being encoded entirely in only one exon. The presence of a majority of these motifs is regulated in an alternative manner at the transcript level (61 % for NLS and 72 % for NES) while the remaining motifs are present in all coding isoforms of their encoding gene. NLSs and NESs are pre-translationally regulated using four main mechanisms: alternative transcription/translation initiation, alternative translation termination, alternative splicing of the exon encoding the motif and frameshift, the first two being by far the most prevalent mechanisms. Quantitative analysis of the presence of these motifs using RNA-seq data indicates that inclusion of these motifs can be regulated in a tissue-specific and a combinatorial manner, can be altered in disease states in a directed way and that alternative inclusion of these motifs is often used by proteins with diverse interactors and roles in diverse pathways, such as kinases. Conclusions The pre-translational regulation of the inclusion of protein targeting motifs is a prominent and tightly-regulated mechanism that adds another layer in the control of protein subcellular localization. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2854-4) contains supplementary material, which is available to authorized users.
Collapse
|
37
|
The Recipe for Protein Sequence-Based Function Prediction and Its Implementation in the ANNOTATOR Software Environment. Methods Mol Biol 2016; 1415:477-506. [PMID: 27115649 DOI: 10.1007/978-1-4939-3572-7_25] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023]
|
38
|
Masuishi Y, Kimura Y, Arakawa N, Hirano H. Identification of glycosylphosphatidylinositol-anchored proteins and ω-sites using TiO2-based affinity purification followed by hydrogen fluoride treatment. J Proteomics 2016; 139:77-83. [PMID: 26972028 DOI: 10.1016/j.jprot.2016.03.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 03/01/2016] [Accepted: 03/02/2016] [Indexed: 11/19/2022]
Abstract
UNLABELLED Glycosylphosphatidylinositol anchored proteins (GPI-APs) in the outer leaflet of the membrane microdomains, commonly referred to as lipid rafts, play important roles in many biological processes such as signal transduction, cell adhesion, protein trafficking, and antigen presentation. From a topological viewpoint, elucidating the presence and localization of GPI-anchor modification sites (ω-sites) is important for the study of the biophysical properties and anchoring mechanisms of these proteins. However, very few reports have actually identified ω-sites of GPI-APs. To enable large-scale site-specific analysis of GPI anchoring, we developed a method for identification of ω-sites by mass spectrometry by combining titanium dioxide-based affinity purification and hydrogen fluoride treatment. This method was able to identify ~3-fold more GPI-APs than our previous method: the new technique identified a total of 73 ω-sites derived from 49 GPI-APs. In 13 of the 49 GPI-APs identified, the GPI-anchor attached to multiple amino acids in the C-terminal site, yielding a variety of different protein species. This method allows us to simultaneously identify many GPI-AP protein species with different ω-sites. We also demonstrated the C-terminal GPI anchor attachment signal peptide, based on information about the GPI anchor binding sites of 49 GPI-APs. Thus, our results provide evidence for new insight into the GPI-anchored proteome and the role of GPI anchoring. BIOLOGICAL SIGNIFICANCE GPI-anchored proteins (GPI-APs) are localized to the outer leaflet of the plasma membranes. Because the GPI anchor is a complex structure, the identification of GPI-anchored peptides by mass spectrometry has always been considered difficult. To improve the feasibility of large-scale site-specific analysis of GPI anchoring, we developed a method for identification of GPI-anchored peptides by combining titanium dioxide-based affinity purification with hydrogen fluoride treatment. Using this novel technique, we identified a total of 73 ω-sites derived from 49 GPI-APs. These data may help us to develop a comprehensive understanding of the GPI-anchored proteome and the role of GPI anchoring. Moreover, this method could be used to discover GPI-APs as candidate biomarkers.
Collapse
Affiliation(s)
- Yusuke Masuishi
- Graduate School of Nanobioscience, Yokohama City University, Fukuura 3-9, 8 Kanazawa, Yokohama 236-0004, Japan; Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, 8 Kanazawa, Yokohama 236-0004, Japan
| | - Yayoi Kimura
- Graduate School of Nanobioscience, Yokohama City University, Fukuura 3-9, 8 Kanazawa, Yokohama 236-0004, Japan; Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, 8 Kanazawa, Yokohama 236-0004, Japan
| | - Noriaki Arakawa
- Graduate School of Nanobioscience, Yokohama City University, Fukuura 3-9, 8 Kanazawa, Yokohama 236-0004, Japan; Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, 8 Kanazawa, Yokohama 236-0004, Japan
| | - Hisashi Hirano
- Graduate School of Nanobioscience, Yokohama City University, Fukuura 3-9, 8 Kanazawa, Yokohama 236-0004, Japan; Advanced Medical Research Center, Yokohama City University, Fukuura 3-9, 8 Kanazawa, Yokohama 236-0004, Japan.
| |
Collapse
|
39
|
Abstract
Protein glycosylation is an essential co- and post-translational modification of secretory and membrane proteins in all eukaryotes. The initial steps of N-glycosylation and N-glycan processing are highly conserved between plants, mammals and yeast. In contrast, late N-glycan maturation steps in the Golgi differ significantly in plants giving rise to complex N-glycans with β1,2-linked xylose, core α1,3-linked fucose and Lewis A-type structures. While the essential role of N-glycan modifications on distinct mammalian glycoproteins is already well documented, we have only begun to decipher the biological function of this ubiquitous protein modification in different plant species. In this review, I focus on the biosynthesis and function of different protein N-linked glycans in plants. Special emphasis is given on glycan-mediated quality control processes in the ER and on the biological role of characteristic complex N-glycan structures.
Collapse
Affiliation(s)
- Richard Strasser
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Vienna, Muthgasse 18, 1190 Vienna, Austria
| |
Collapse
|
40
|
Yu HZ, Wen DF, Wang WL, Geng L, Zhang Y, Xu JP. Identification of Genes Putatively Involved in Chitin Metabolism and Insecticide Detoxification in the Rice Leaf Folder (Cnaphalocrocis medinalis) Larvae through Transcriptomic Analysis. Int J Mol Sci 2015; 16:21873-96. [PMID: 26378520 PMCID: PMC4613286 DOI: 10.3390/ijms160921873] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 08/01/2015] [Accepted: 08/25/2015] [Indexed: 01/15/2023] Open
Abstract
The rice leaf roller (Cnaphalocrocis medinalis) is one of the most destructive agricultural pests. Due to its migratory behavior, it is difficult to control worldwide. To date, little is known about major genes of C. medinalis involved in chitin metabolism and insecticide detoxification. In order to obtain a comprehensive genome dataset of C. medinalis, we conducted de novo transcriptome sequencing which focused on the major feeding stage of fourth-instar larvae, and our work revealed useful information on chitin metabolism and insecticide detoxification and target genes of C. medinalis. We acquired 29,367,797 Illumina reads and assembled these reads into 63,174 unigenes with an average length of 753 bp. Among these unigenes, 31,810 were annotated against the National Center for Biotechnology Information non-redundant (NCBI nr) protein database, resulting in 24,246, 8669 and 18,176 assigned to Swiss-Prot, clusters of orthologous group (COG), and gene ontology (GO), respectively. We were able to map 10,043 unigenes into 285 pathways using the Kyoto Encyclopedia of Genes and Genomes Pathway database (KEGG). Specifically, 16 genes, including five chitin deacetylases, two chitin synthases, five chitinases and four other related enzymes, were identified to be putatively involved in chitin biosynthesis and degradation, whereas 360 genes, including cytochrome P450s, glutathione S-transferases, esterases, and acetylcholinesterases, were found to be potentially involved in insecticide detoxification or as insecticide targets. The reliability of the transcriptome data was determined by reverse transcription quantitative PCR (RT-qPCR) for the selected genes. Our data serves as a new and valuable sequence resource for genomic studies on C. medinalis. The findings should improve our understanding of C. medinalis genetics and contribute to management of this important agricultural pest.
Collapse
Affiliation(s)
- Hai-Zhong Yu
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China.
| | - De-Fu Wen
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China.
| | - Wan-Lin Wang
- Rice Research Institute, Anhui Academy of Agricultural Sciences, Hefei 230031, China.
| | - Lei Geng
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China.
| | - Yan Zhang
- Institute of Sericulture, Anhui Academy of Agricultural Sciences, Hefei 230061, China.
| | - Jia-Ping Xu
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China.
| |
Collapse
|
41
|
Ruhela D, Kamthan M, Saha P, Majumdar SS, Datta K, Abdin MZ, Datta A. In vivo role of Candida albicans β-hexosaminidase (HEX1) in carbon scavenging. Microbiologyopen 2015; 4:730-42. [PMID: 26177944 PMCID: PMC4618606 DOI: 10.1002/mbo3.274] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 05/25/2015] [Accepted: 05/26/2015] [Indexed: 12/16/2022] Open
Abstract
The capability to utilize of N-acetylglucosamine (GlcNAc) as a carbon source is an important virulence attribute of Candida albicans. But there is a lack of information about the in vivo source of GlcNAc for the pathogen within the host environment. Here, we have characterized the GlcNAc-inducible β-hexosaminidase gene (HEX1) of C. albicans showing a role in carbon scavenging. In contrast to earlier studies, we have reported HEX1 to be a nonessential gene as shown by homozygous trisomy test. Virulence study in the systemic mouse murine model showed that Δhex1 strain is significantly less virulent in comparison to the wild-type strain. Moreover, Δhex1 strain also showed a higher susceptibility to peritoneal macrophages. In an attempt to determine possible substrates of Hex1, hyaluronic acid (HA) was treated with purified Hex1 enzyme. A significant release of GlcNAc was observed by gas chromatography-mass spectrometry analysis analysis suggesting HA degradation. Interestingly, immunohistochemistry analysis showed significant accumulation of HA in the mice kidney infected with the wild-type strain of C. albicans. Northern blot analysis showed that C. albicans HEX1 is expressed during mice renal colonization. Thus, C. albicans can obtain GlcNAc during organ colonization by secreting Hex1 via degradation of host HA.
Collapse
Affiliation(s)
- Deepa Ruhela
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Mohan Kamthan
- Indian Institute of Toxicology Research, Lucknow, 226001, India
| | - Paramita Saha
- Biochemistry Laboratory, School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Subeer S Majumdar
- Division of Cellular Endocrinology, National Institute of Immunology, New Delhi, India
| | - Kasturi Datta
- Biochemistry Laboratory, School of Environmental Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Malik Zainul Abdin
- Department of Biotechnology, Faculty of Science, Jamia Hamdard University, New Delhi, 110062, India
| | - Asis Datta
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India
| |
Collapse
|
42
|
Yadav A, Singh SL, Yadav B, Komath SS. Saccharomyces cerevisiae Gpi2, an accessory subunit of the enzyme catalyzing the first step of glycosylphosphatidylinositol (GPI) anchor biosynthesis, selectively complements some of the functions of its homolog in Candida albicans. Glycoconj J 2014; 31:497-507. [PMID: 25117514 DOI: 10.1007/s10719-014-9536-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/21/2014] [Accepted: 06/23/2014] [Indexed: 11/28/2022]
Abstract
GPI2 encodes for one of the six accessory subunits of the GPI-N-acetylglucosaminyltransferase (GPI-GnT) complex that catalyzes the first step of GPI biosynthesis in S. cerevisiae and C. albicans. It has been previously reported in S. cerevisiae that this subunit physically interacts with and negatively modulates Ras signaling. On the other hand, studies from our lab have shown that the homologous subunit in C. albicans is a positive modulator of Ras signaling. Are the functions of this subunit therefore strictly species dependent? We present here functional complementation studies on GPI2 from S. cerevisiae and C. albicans that were carried out to address this issue. Expression of CaGPI2 in a ScGPI2 conditional lethal mutant could not restore its growth defects. Likewise, ScGPI2 overexpression in a CaGPI2 heterozygous mutant could not restore its deficient GPI-GnT activity or reverse defects in its cell wall integrity and could only poorly restore filamentation. However, interestingly, ScGPI2 could restore lanosterol demethylase (CaERG11) levels and reverse azole resistance of the CaGPI2 heterozygote. It appeared to do this by regulating levels of another GPI-GnT subunit, CaGPI19, which we have previously shown to be involved in cross-talk with CaERG11. Thus, the effect of CaGPI2 on sterol biosynthesis in C. albicans is independent of its interaction with the GPI-GnT complex and Ras signaling pathways. In addition, the interaction of Gpi2 with other subunits of the GPI-GnT complex as well as with Ras signaling appears to have evolved differently in the two organisms.
Collapse
Affiliation(s)
- Anshuman Yadav
- Sneha Sudha Komath, School of Life Sciences, Jawaharlal Nehru University, New Delhi, 110067, India
| | | | | | | |
Collapse
|
43
|
Eisenhaber B, Eisenhaber S, Kwang TY, Grüber G, Eisenhaber F. Transamidase subunit GAA1/GPAA1 is a M28 family metallo-peptide-synthetase that catalyzes the peptide bond formation between the substrate protein's omega-site and the GPI lipid anchor's phosphoethanolamine. Cell Cycle 2014; 13:1912-7. [PMID: 24743167 PMCID: PMC4111754 DOI: 10.4161/cc.28761] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The transamidase subunit GAA1/GPAA1 is predicted to be the enzyme that catalyzes the attachment of the glycosylphosphatidyl (GPI) lipid anchor to the carbonyl intermediate of the substrate protein at the ω-site. Its ~300-amino acid residue lumenal domain is a M28 family metallo-peptide-synthetase with an α/β hydrolase fold, including a central 8-strand β-sheet and a single metal (most likely zinc) ion coordinated by 3 conserved polar residues. Phosphoethanolamine is used as an adaptor to make the non-peptide GPI lipid anchor look chemically similar to the N terminus of a peptide.
Collapse
Affiliation(s)
- Birgit Eisenhaber
- Bioinformatics Institute (BII); A*STAR; Singapore, Republic of Singapore
| | - Stephan Eisenhaber
- Department of Physical Chemistry; University of Vienna; Wien/Vienna, Republic of Austria
| | - Toh Yew Kwang
- Bioinformatics Institute (BII); A*STAR; Singapore, Republic of Singapore
| | - Gerhard Grüber
- Bioinformatics Institute (BII); A*STAR; Singapore, Republic of Singapore; Nanyang Technological University; School of Biological Sciences; Singapore, Republic of Singapore
| | - Frank Eisenhaber
- Bioinformatics Institute (BII); A*STAR; Singapore, Republic of Singapore; Department of Biological Sciences (DBS); National University of Singapore (NUS); Singapore, Republic of Singapore; School of Computer Engineering (SCE); Nanyang Technological University (NTU); Singapore, Republic of Singapore
| |
Collapse
|
44
|
Yadav B, Bhatnagar S, Ahmad MF, Jain P, Pratyusha VA, Kumar P, Komath SS. First step of glycosylphosphatidylinositol (GPI) biosynthesis cross-talks with ergosterol biosynthesis and Ras signaling in Candida albicans. J Biol Chem 2013; 289:3365-82. [PMID: 24356967 DOI: 10.1074/jbc.m113.528802] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Candida albicans is a leading cause of fungal infections worldwide. It has several glycosylphosphatidylinositol (GPI)-anchored virulence factors. Inhibiting GPI biosynthesis attenuates its virulence. Building on our previous work, we explore the interaction of GPI biosynthesis in C. albicans with ergosterol biosynthesis and hyphal morphogenesis. This study is also the first report of transcriptional co-regulation existing between two subunits of the multisubunit enzyme complex, GPI-N-acetylglucosaminyltransferase (GPI-GnT), involved in the first step of GPI anchor biosynthesis in eukaryotes. Using mutational analysis, we show that the accessory subunits, GPI2 and GPI19, of GPI-GnT exhibit opposite effects on ergosterol biosynthesis and Ras signaling (which determines hyphal morphogenesis). This is because the two subunits negatively regulate one another; GPI19 mutants show up-regulation of GPI2, whereas GPI2 mutants show up-regulation of GPI19. Two different models were examined as follows. First, the two GPI-GnT subunits independently interact with ergosterol biosynthesis and Ras signaling. Second, the two subunits mutually regulate one another and thereby regulate sterol levels and Ras signaling. Analysis of double mutants of these subunits indicates that GPI19 controls ergosterol biosynthesis through ERG11 levels, whereas GPI2 determines the filamentation by cross-talk with Ras1 signaling. Taken together, this suggests that the first step of GPI biosynthesis talks to and regulates two very important pathways in C. albicans. This could have implications for designing new antifungal strategies.
Collapse
Affiliation(s)
- Bhawna Yadav
- From the School of Life Sciences, Jawaharlal Nehru University, New Delhi 110 067, India
| | | | | | | | | | | | | |
Collapse
|
45
|
Recent progress in synthetic and biological studies of GPI anchors and GPI-anchored proteins. Curr Opin Chem Biol 2013; 17:1006-13. [PMID: 24128440 DOI: 10.1016/j.cbpa.2013.09.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 09/22/2013] [Accepted: 09/24/2013] [Indexed: 10/26/2022]
Abstract
Covalent attachment of glycosylphosphatidylinositols (GPIs) to the protein C-terminus is one of the most common posttranslational modifications in eukaryotic cells. In addition to anchoring surface proteins to the cell membrane, GPIs also have many other important biological functions, determined by their unique structure and property. This account has reviewed the recent progress made in disclosing GPI and GPI-anchored protein biosynthesis, in the chemical and chemoenzymatic synthesis of GPIs and GPI-anchored proteins, and in understanding the conformation, organization, and distribution of GPIs in the lipid membrane.
Collapse
|
46
|
Gamage DG, Hendrickson TL. GPI Transamidase and GPI anchored proteins: Oncogenes and biomarkers for cancer. Crit Rev Biochem Mol Biol 2013; 48:446-64. [DOI: 10.3109/10409238.2013.831024] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
47
|
Liu X, Li F, Li D, Ma E, Zhang W, Zhu KY, Zhang J. Molecular and functional analysis of UDP-N-acetylglucosamine Pyrophosphorylases from the Migratory Locust, Locusta migratoria. PLoS One 2013; 8:e71970. [PMID: 23977188 PMCID: PMC3747057 DOI: 10.1371/journal.pone.0071970] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 07/06/2013] [Indexed: 01/25/2023] Open
Abstract
UDP-N-acetylglucosamine pyrophosphorylases (UAP) function in the formation of extracellular matrix by producing N-acetylglucosamine (GlcNAc) residues needed for chitin biosynthesis and protein glycosylation. Herein, we report two UAP cDNA's derived from two different genes (LmUAP1 and LmUAP2) in the migratory locust Locusta migratoria. Both the cDNA and their deduced amino acid sequences showed about 70% identities between the two genes. Phylogenetic analysis suggests that LmUAP1 and LmUAP2 derive from a relatively recent gene duplication event. Both LmUAP1 and LmUAP2 were widely expressed in all the major tissues besides chitin-containing tissues. However, the two genes exhibited different developmental expression patterns. High expression of LmUAP1 was detected during early embryogenesis, then decreased greatly, and slowly increased before egg hatch. During nymphal development, the highest expression of LmUAP1 appeared just after molting but declined in each inter-molting period and then increased before molting to the next stage, whereas LmUAP2 was more consistently expressed throughout all these stages. When the early second- and fifth-instar nymphs (1-day-old) were injected with LmUAP1 double-stranded RNA (dsRNA), 100% mortality was observed 2 days after the injection. When the middle second- and fifth-instar nymphs (3- to 4-day-old) were injected with LmUAP1 dsRNA, 100% mortality was observed during their next molting process. In contrast, when the insects at the same stages were injected with LmUAP2 dsRNA, these insects were able to develop normally and molt to the next stage successfully. It is presumed that the lethality caused by RNAi of LmUAP1 is due to reduced chitin biosynthesis of the integument and midgut, whereas LmUAP2 is not essential for locust development at least in nymph stage. This study is expected to help better understand different functions of UAP1 and UAP2 in the locust and other insect species.
Collapse
Affiliation(s)
- Xiaojian Liu
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi, People’s Republic of China
| | - Feng Li
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi, People’s Republic of China
| | - Daqi Li
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi, People’s Republic of China
| | - Enbo Ma
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi, People’s Republic of China
| | - Wenqing Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, People’s Republic of China
| | - Kun Yan Zhu
- Department of Entomology, Kansas State University, Manhattan, Kansas, United States of America
- * E-mail: (KYZ); (JZ)
| | - Jianzhen Zhang
- Research Institute of Applied Biology, Shanxi University, Taiyuan, Shanxi, People’s Republic of China
- * E-mail: (KYZ); (JZ)
| |
Collapse
|
48
|
Identification and functional analysis of Trypanosoma cruzi genes that encode proteins of the glycosylphosphatidylinositol biosynthetic pathway. PLoS Negl Trop Dis 2013; 7:e2369. [PMID: 23951384 PMCID: PMC3738449 DOI: 10.1371/journal.pntd.0002369] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 07/01/2013] [Indexed: 12/03/2022] Open
Abstract
Background Trypanosoma cruzi is a protist parasite that causes Chagas disease. Several proteins that are essential for parasite virulence and involved in host immune responses are anchored to the membrane through glycosylphosphatidylinositol (GPI) molecules. In addition, T. cruzi GPI anchors have immunostimulatory activities, including the ability to stimulate the synthesis of cytokines by innate immune cells. Therefore, T. cruzi genes related to GPI anchor biosynthesis constitute potential new targets for the development of better therapies against Chagas disease. Methodology/Principal Findings In silico analysis of the T. cruzi genome resulted in the identification of 18 genes encoding proteins of the GPI biosynthetic pathway as well as the inositolphosphorylceramide (IPC) synthase gene. Expression of GFP fusions of some of these proteins in T. cruzi epimastigotes showed that they localize in the endoplasmic reticulum (ER). Expression analyses of two genes indicated that they are constitutively expressed in all stages of the parasite life cycle. T. cruzi genes TcDPM1, TcGPI10 and TcGPI12 complement conditional yeast mutants in GPI biosynthesis. Attempts to generate T. cruzi knockouts for three genes were unsuccessful, suggesting that GPI may be an essential component of the parasite. Regarding TcGPI8, which encodes the catalytic subunit of the transamidase complex, although we were able to generate single allele knockout mutants, attempts to disrupt both alleles failed, resulting instead in parasites that have undergone genomic recombination and maintained at least one active copy of the gene. Conclusions/Significance Analyses of T. cruzi sequences encoding components of the GPI biosynthetic pathway indicated that they are essential genes involved in key aspects of host-parasite interactions. Complementation assays of yeast mutants with these T. cruzi genes resulted in yeast cell lines that can now be employed in high throughput screenings of drugs against this parasite. Chagas disease, considered one of the most neglected tropical diseases, is caused by the blood-borne parasite Trypanosoma cruzi and currently affects about 8 million people in Latin America. T. cruzi can be transmitted by insect vectors, blood transfusion, organ transplantation and mother-to-baby as well as through ingestion of contaminated food. Although T. cruzi causes life-long infections that can result in serious damage to the heart, the two drugs currently available to treat Chagas disease, benznidazole and nifurtimox, which have been used for more than 40 years, have proven efficacy only during the acute phase of the disease. Thus, there is an urgent need to develop new drugs that are more targeted, less toxic, and more effective against this parasite. Here we described the characterization of T. cruzi genes involved in the biosynthesis of GPI anchors, a molecule responsible for holding different types of glycoproteins on the parasite membrane. Since GPI anchored proteins are essential molecules T. cruzi uses during infection, besides helping understand how this parasite interacts with its host, this work may contribute to the development of better therapies against Chagas disease.
Collapse
|
49
|
Abstract
Every living cell is covered with a dense and complex array of covalently attached sugars or sugar chains. The majority of these glycans are linked to proteins via the so-called glycosylation process. Protein glycosylation is found in all three domains of life: Eukarya, Bacteria and Archaea. However, on the basis of the limit in analytic tools for glycobiology and genetics in Archaea, only in the last few years has research on archaeal glycosylation pathways started mainly in the Euryarchaeota Haloferax volcanii, Methanocaldococcus maripaludis and Methanococcus voltae. Recently, major steps of the crenarchaeal glycosylation process of the thermoacidophilic archaeon Sulfolobus acidocaldarius have been described. The present review summarizes the proposed N-glycosylation pathway of S. acidocaldarius, describing the phenotypes of the mutants disrupted in N-glycan biosynthesis as well as giving insights into the archaeal O-linked and glycosylphosphatidylinositol anchor glycosylation process.
Collapse
|
50
|
Low-resolution structure of the soluble domain GPAA1 (yGPAA170-247) of the glycosylphosphatidylinositol transamidase subunit GPAA1 from Saccharomyces cerevisiae. Biosci Rep 2013; 33:e00033. [PMID: 23458223 PMCID: PMC3610296 DOI: 10.1042/bsr20120107] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The GPI (glycosylphosphatidylinositol) transamidase complex catalyses the attachment of GPI anchors to eukaryotic proteins in the lumen of ER (endoplasmic reticulum). The Saccharomyces cerevisiae GPI transamidase complex consists of the subunits yPIG-K (Gpi8p), yPIG-S (Gpi17p), yPIG-T (Gpi16p), yPIG-U (CDC91/GAB1) and yGPAA1. We present the production of the two recombinant proteins yGPAA170–247 and yGPAA170–339 of the luminal domain of S. cerevisiae GPAA1, covering the amino acids 70–247 and 70–339 respectively. The secondary structural content of the stable and monodisperse yGPAA170–247 has been determined to be 28% α-helix and 27% β-sheet. SAXS (small-angle X-ray scattering) data showed that yGPAA170–247 has an Rg (radius of gyration) of 2.72±0.025 nm and Dmax (maximum dimension) of 9.14 nm. These data enabled the determination of the two domain low-resolution solution structure of yGPAA170–247. The large elliptical shape of yGPAA170–247 is connected via a short stalk to the smaller hook-like domain of 0.8 nm in length and 3.5 nm in width. The topological arrangement of yGPAA170–247 will be discussed together with the recently determined low-resolution structures of yPIG-K24–337 and yPIG-S38–467 from S. cerevisiae in the GPI transamidase complex.
Collapse
|