51
|
Ruzickova E, Lichvarova M, Osickova A, Filipi K, Jurnecka D, Khaliq H, Espinosa-Vinals C, Pompach P, Masin J, Osicka R. Two pairs of back-to-back α-helices of Kingella kingae RtxA toxin are crucial for the formation of a membrane pore. Int J Biol Macromol 2024; 283:137604. [PMID: 39542310 DOI: 10.1016/j.ijbiomac.2024.137604] [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: 09/27/2024] [Revised: 11/01/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
The RtxA cytotoxin, a member of the RTX (Repeats in ToXin) family of pore-forming toxins, is the primary virulence factor of the paediatric facultative pathogen Kingella kingae. Although structure-function studies of RTX toxins have defined their characteristic domains and features, the exact membrane topology of RTX toxins remains unknown. Here, we used labelling of cell-bound RtxA with a membrane-impermeable, lysine-reactive reagent and subsequent detection of the labelled lysine residues by mass spectrometry, which revealed that most of the membrane-bound toxin is localised extracellularly. A trypsin protection assay with cell-bound RtxA demonstrated that five of seven transmembrane α-helices, predicted by various algorithms within the N-terminal half of the molecule, are irreversibly embedded in the membrane. Structure-function analysis showed that these α-helices, four of which are arranged as two pairs of back-to-back helices, are essential for the formation of an ion-conducting membrane pore. In contrast, the C-terminal half of RtxA is required for the interaction with the cell surface and for the irreversible insertion of the toxin into the membrane via acyl chains covalently linked to the molecule. These findings advance our understanding of the structure-function relationships of RtxA and enable us to propose a membrane topology model of the toxin.
Collapse
Affiliation(s)
- Eliska Ruzickova
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic; Faculty of Science, Charles University, Prague, Czech Republic
| | - Michaela Lichvarova
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic; Faculty of Science, Charles University, Prague, Czech Republic
| | - Adriana Osickova
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Katerina Filipi
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - David Jurnecka
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Humaira Khaliq
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic; University of Chemistry and Technology, Prague, Prague, Czech Republic
| | - Carlos Espinosa-Vinals
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic; University of Chemistry and Technology, Prague, Prague, Czech Republic
| | - Petr Pompach
- Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czech Republic
| | - Jiri Masin
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Radim Osicka
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
52
|
Okada K, Yachi K, Nguyen TAN, Kanno S, Yasuda S, Tadai H, Tateda C, Lee TH, Nguyen U, Inoue K, Tsuchida N, Ishihara T, Miyashima S, Hiruma K, Miwa K, Maekawa T, Notaguchi M, Saijo Y. Defense-related callose synthase PMR4 promotes root hair callose deposition and adaptation to phosphate deficiency in Arabidopsis thaliana. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2024; 120:2639-2655. [PMID: 39544094 DOI: 10.1111/tpj.17134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 10/10/2024] [Accepted: 10/26/2024] [Indexed: 11/17/2024]
Abstract
Plants acquire phosphorus (P) primarily as inorganic phosphate (Pi) from the soil. Under Pi deficiency, plants induce an array of physiological and morphological responses, termed phosphate starvation response (PSR), thereby increasing Pi acquisition and use efficiency. However, the mechanisms by which plants adapt to Pi deficiency remain to be elucidated. Here, we report that deposition of a β-1,3-glucan polymer called callose is induced in Arabidopsis thaliana root hairs under Pi deficiency, in a manner independent of PSR-regulating PHR1/PHL1 transcription factors and LPR1/LPR2 ferroxidases. Genetic studies revealed PMR4 (GSL5) callose synthase being required for the callose deposition in Pi-depleted root hairs. Loss of PMR4 also reduces Pi acquisition in shoots and plant growth under low Pi conditions. The defects are not recovered by simultaneous disruption of SID2, mediating defense-associated salicylic acid (SA) biosynthesis, excluding SA defense activation from the cause of the observed pmr4 phenotypes. Grafting experiments and characterization of plants expressing PMR4 specifically in root hair cells suggest that a PMR4 pool in the cell type contributes to shoot growth under Pi deficiency. Our findings thus suggest an important role for PMR4 in plant adaptation to Pi deficiency.
Collapse
Affiliation(s)
- Kentaro Okada
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Takayama-cho, Ikoma, 630-0192, Japan
- Bioscience and Biotechnology Center, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601, Japan
| | - Koei Yachi
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Takayama-cho, Ikoma, 630-0192, Japan
| | - Tan Anh Nhi Nguyen
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Takayama-cho, Ikoma, 630-0192, Japan
| | - Satomi Kanno
- Institute for Advanced Research, Nagoya University, Furo-cho, Chikusa, Nagoya, 464-8601, Japan
| | - Shigetaka Yasuda
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Takayama-cho, Ikoma, 630-0192, Japan
| | - Haruna Tadai
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Takayama-cho, Ikoma, 630-0192, Japan
| | - Chika Tateda
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Takayama-cho, Ikoma, 630-0192, Japan
| | - Tae-Hong Lee
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Takayama-cho, Ikoma, 630-0192, Japan
| | - Uyen Nguyen
- Institute for Plant Sciences, University of Cologne, Cologne, D-50674, Germany
| | - Kanako Inoue
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Takayama-cho, Ikoma, 630-0192, Japan
| | - Natsuki Tsuchida
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Takayama-cho, Ikoma, 630-0192, Japan
| | - Taiga Ishihara
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Takayama-cho, Ikoma, 630-0192, Japan
| | - Shunsuke Miyashima
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Takayama-cho, Ikoma, 630-0192, Japan
| | - Kei Hiruma
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Takayama-cho, Ikoma, 630-0192, Japan
| | - Kyoko Miwa
- Graduate School of Environmental Science, Hokkaido University, Sapporo, 060-0810, Japan
| | - Takaki Maekawa
- Institute for Plant Sciences, University of Cologne, Cologne, D-50674, Germany
- CEPLAS Cluster of Excellence on Plant Sciences at the University of Cologne, Cologne, D-50674, Germany
| | - Michitaka Notaguchi
- Bioscience and Biotechnology Center, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, 464-8601, Japan
| | - Yusuke Saijo
- Graduate School of Science and Technology, Nara Institute of Science and Technology, Takayama-cho, Ikoma, 630-0192, Japan
| |
Collapse
|
53
|
Chen LL, Naesström M, Halvorsen M, Fytagoridis A, Crowley SB, Mataix-Cols D, Rück C, Crowley JJ, Pascal D. Genomics of severe and treatment-resistant obsessive-compulsive disorder treated with deep brain stimulation: A preliminary investigation. Am J Med Genet B Neuropsychiatr Genet 2024; 195:e32983. [PMID: 38650085 PMCID: PMC11493841 DOI: 10.1002/ajmg.b.32983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 01/25/2024] [Accepted: 03/20/2024] [Indexed: 04/25/2024]
Abstract
Individuals with severe and treatment-resistant obsessive-compulsive disorder (trOCD) represent a small but severely disabled group of patients. Since trOCD cases eligible for deep brain stimulation (DBS) probably comprise the most severe end of the OCD spectrum, we hypothesize that they may be more likely to have a strong genetic contribution to their disorder. Therefore, while the worldwide population of DBS-treated cases may be small (~300), screening these individuals with modern genomic methods may accelerate gene discovery in OCD. As such, we have begun to collect DNA from trOCD cases who qualify for DBS, and here we report results from whole exome sequencing and microarray genotyping of our first five cases. All participants had previously received DBS in the bed nucleus of stria terminalis (BNST), with two patients responding to the surgery and one showing a partial response. Our analyses focused on gene-disruptive rare variants (GDRVs; rare, predicted-deleterious single-nucleotide variants or copy number variants overlapping protein-coding genes). Three of the five cases carried a GDRV, including a missense variant in the ion transporter domain of KCNB1, a deletion at 15q11.2, and a duplication at 15q26.1. The KCNB1 variant (hg19 chr20-47991077-C-T, NM_004975.3:c.1020G>A, p.Met340Ile) causes substitution of methionine for isoleucine in the trans-membrane region of neuronal potassium voltage-gated ion channel KV2.1. This KCNB1 substitution (Met340Ile) is located in a highly constrained region of the protein where other rare missense variants have previously been associated with neurodevelopmental disorders. The patient carrying the Met340Ile variant responded to DBS, which suggests that genetic factors could potentially be predictors of treatment response in DBS for OCD. In sum, we have established a protocol for recruiting and genomically characterizing trOCD cases. Preliminary results suggest that this will be an informative strategy for finding risk genes in OCD.
Collapse
Affiliation(s)
- Long Long Chen
- Department of Clinical Neuroscience, Centre for Psychiatry Research Karolinska Institutet, & Stockholm Health Care Services, Stockholm, Sweden
| | - Matilda Naesström
- Department of Clinical Sciences/Psychiatry, Umeå University, Umeå, Sweden
| | - Matthew Halvorsen
- Department of Clinical Neuroscience, Centre for Psychiatry Research Karolinska Institutet, & Stockholm Health Care Services, Stockholm, Sweden
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anders Fytagoridis
- Department of Neurosurgery, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | | | - David Mataix-Cols
- Department of Clinical Neuroscience, Centre for Psychiatry Research Karolinska Institutet, & Stockholm Health Care Services, Stockholm, Sweden
| | - Christian Rück
- Department of Clinical Neuroscience, Centre for Psychiatry Research Karolinska Institutet, & Stockholm Health Care Services, Stockholm, Sweden
| | - James J. Crowley
- Department of Clinical Neuroscience, Centre for Psychiatry Research Karolinska Institutet, & Stockholm Health Care Services, Stockholm, Sweden
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Diana Pascal
- Department of Clinical Neuroscience, Centre for Psychiatry Research Karolinska Institutet, & Stockholm Health Care Services, Stockholm, Sweden
| |
Collapse
|
54
|
Brauer BK, Chen Z, Beirow F, Li J, Meisinger D, Capriotti E, Schweizer M, Wagner L, Wienberg J, Hobohm L, Blume L, Qiao W, Narimatsu Y, Carette JE, Clausen H, Winter D, Braulke T, Jabs S, Voss M. GOLPH3 and GOLPH3L maintain Golgi localization of LYSET and a functional mannose 6-phosphate transport pathway. EMBO J 2024; 43:6264-6290. [PMID: 39587297 PMCID: PMC11649813 DOI: 10.1038/s44318-024-00305-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/27/2024] Open
Abstract
Glycosylation, which plays an important role in modifying lipids and sorting of proteins, is regulated by asymmetric intra-Golgi distribution and SPPL3-mediated cleavage of Golgi enzymes. We found that cells lacking LYSET/TMEM251, a retention factor for Golgi N-acetylglucosamine-1-phosphotransferase (GNPT), display SPPL3-dependent hypersecretion of the Golgi membrane protein B4GALT5. We demonstrate that in wild-type cells B4GALT5 is tagged with mannose 6-phosphate (M6P), a sorting tag typical of soluble lysosomal hydrolases. Hence, M6P-tagging of B4GALT5 may represent a novel degradative lysosomal pathway. We also observed B4GALT5 hypersecretion and prominent destabilization of LYSET-GNPT complexes, impaired M6P-tagging, and disturbed maturation and trafficking of lysosomal enzymes in multiple human cell lines lacking the COPI adaptors GOLPH3 and GOLPH3L. Mechanistically, we identified LYSET as a novel, atypical client of GOLPH3/GOLPH3L. Thus, by ensuring the cis-Golgi localization of the LYSET-GNPT complex and maintaining its Golgi polarity, GOLPH3/GOLPH3L is essential for the integrity of the M6P-tagging machinery and homeostasis of lysosomes.
Collapse
Affiliation(s)
- Berit K Brauer
- Institute of Biochemistry, Kiel University, Kiel, Germany
| | - Zilei Chen
- Institute of Clinical Molecular Biology, Kiel University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Felix Beirow
- Institute of Biochemistry, Kiel University, Kiel, Germany
| | - Jiaran Li
- Institute for Biochemistry and Molecular Biology, Medical Faculty, Rheinische Friedrich-Wilhelms-University of Bonn, Bonn, Germany
| | | | - Emanuela Capriotti
- Department of Osteology and Biomechanics, Cell Biology of Rare Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michaela Schweizer
- Morphology and Electron Microscopy, University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology (ZMNH), Hamburg, Germany
| | - Lea Wagner
- Institute of Biochemistry, Kiel University, Kiel, Germany
| | | | - Laura Hobohm
- Institute of Biochemistry, Kiel University, Kiel, Germany
| | - Lukas Blume
- Institute of Biochemistry, Kiel University, Kiel, Germany
- Institute of Cellular and Integrative Physiology, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Wenjie Qiao
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yoshiki Narimatsu
- Faculty of Health Sciences, Centre for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Jan E Carette
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
| | - Henrik Clausen
- Faculty of Health Sciences, Centre for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Dominic Winter
- Institute for Biochemistry and Molecular Biology, Medical Faculty, Rheinische Friedrich-Wilhelms-University of Bonn, Bonn, Germany
| | - Thomas Braulke
- Department of Osteology and Biomechanics, Cell Biology of Rare Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sabrina Jabs
- Institute of Clinical Molecular Biology, Kiel University and University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.
| | - Matthias Voss
- Institute of Biochemistry, Kiel University, Kiel, Germany.
| |
Collapse
|
55
|
Kovalchik KA, Hamelin DJ, Kubiniok P, Bourdin B, Mostefai F, Poujol R, Paré B, Simpson SM, Sidney J, Bonneil É, Courcelles M, Saini SK, Shahbazy M, Kapoor S, Rajesh V, Weitzen M, Grenier JC, Gharsallaoui B, Maréchal L, Wu Z, Savoie C, Sette A, Thibault P, Sirois I, Smith MA, Decaluwe H, Hussin JG, Lavallée-Adam M, Caron E. Machine learning-enhanced immunopeptidomics applied to T-cell epitope discovery for COVID-19 vaccines. Nat Commun 2024; 15:10316. [PMID: 39609459 PMCID: PMC11604954 DOI: 10.1038/s41467-024-54734-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 11/20/2024] [Indexed: 11/30/2024] Open
Abstract
Next-generation T-cell-directed vaccines for COVID-19 focus on establishing lasting T-cell immunity against current and emerging SARS-CoV-2 variants. Precise identification of conserved T-cell epitopes is critical for designing effective vaccines. Here we introduce a comprehensive computational framework incorporating a machine learning algorithm-MHCvalidator-to enhance mass spectrometry-based immunopeptidomics sensitivity. MHCvalidator identifies unique T-cell epitopes presented by the B7 supertype, including an epitope from a + 1-frameshift in a truncated Spike antigen, supported by ribosome profiling. Analysis of 100,512 COVID-19 patient proteomes shows Spike antigen truncation in 0.85% of cases, revealing frameshifted viral antigens at the population level. Our EpiTrack pipeline tracks global mutations of MHCvalidator-identified CD8 + T-cell epitopes from the BNT162b4 vaccine. While most vaccine epitopes remain globally conserved, an immunodominant A*01-associated epitope mutates in Delta and Omicron variants. This work highlights SARS-CoV-2 antigenic features and emphasizes the importance of continuous adaptation in T-cell vaccine development.
Collapse
Affiliation(s)
- Kevin A Kovalchik
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - David J Hamelin
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
- Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
- Mila-Quebec AI Institute, Montreal, QC, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Peter Kubiniok
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - Benoîte Bourdin
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - Fatima Mostefai
- Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
- Mila-Quebec AI Institute, Montreal, QC, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Raphaël Poujol
- Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada
| | - Bastien Paré
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - Shawn M Simpson
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Éric Bonneil
- Institute of Research in Immunology and Cancer, Montreal, QC, Canada
| | | | - Sunil Kumar Saini
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Mohammad Shahbazy
- Department of Biochemistry and Molecular Biology and Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - Saketh Kapoor
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Vigneshwar Rajesh
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Maya Weitzen
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | | | - Bayrem Gharsallaoui
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - Loïze Maréchal
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - Zhaoguan Wu
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - Christopher Savoie
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Pierre Thibault
- Institute of Research in Immunology and Cancer, Montreal, QC, Canada
- Department of Chemistry, Université de Montréal, Montreal, QC, Canada
| | - Isabelle Sirois
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - Martin A Smith
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Hélène Decaluwe
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
- Microbiology, Infectiology and Immunology Department, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
- Pediatric Immunology and Rheumatology Division, Department of Pediatrics, Université de Montréal, Montreal, QC, Canada
| | - Julie G Hussin
- Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada.
- Mila-Quebec AI Institute, Montreal, QC, Canada.
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.
| | - Mathieu Lavallée-Adam
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada.
| | - Etienne Caron
- CHU Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada.
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
- Yale Center for Immuno-Oncology, Yale Center for Systems and Engineering Immunology, Yale Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
56
|
Lyons CL, Cowan E, Nilsson O, Mohar M, Peña-Martínez P, Eliasson L, Lagerstedt JO. Apolipoprotein A-I priming via SR-BI and ABCA1 receptor binding upregulates mitochondrial metabolism to promote insulin secretion in INS-1E cells. PLoS One 2024; 19:e0311039. [PMID: 39546458 PMCID: PMC11567530 DOI: 10.1371/journal.pone.0311039] [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: 07/08/2024] [Accepted: 09/11/2024] [Indexed: 11/17/2024] Open
Abstract
Apolipoprotein A-I (ApoA-I), the primary component of high-density lipoprotein (HDL) cholesterol primes β-cells to increase insulin secretion, however, the mechanisms involved are not fully defined. Here, we aimed to confirm ApoA-I receptors in β-cells and delineate ApoA-I-receptor pathways in β-cell insulin output. An LRC-TriCEPS experiment was performed using the INS-1E rat β-cell model and ApoA-I for unbiased identification of ApoA-I receptors. Identified targets, alongside ATP binding cassette transporter A1 (ABCA1) (included control) were silenced in the same cells, and insulin secretion (ELISA) and mitochondrial metabolism (seahorse) were assessed with/without ApoA-I priming. Human β-cell expression data was used to investigate ApoA-I receptor pathways in type 2 diabetes (T2D). Scavenger receptor B1 (SR-BI) and regulator of microtubule dynamics 1 were identified as ApoA-I targets. SR-BI or ABCA1 silencing abolished ApoA-I induced increases in insulin secretion. ApoA-I priming increased mitochondrial OXPHOS, however this was greatly attenuated with SR-BI or ABCA1 silencing. Supporting this, human β-cell expression data investigations found SR-BI and ABCA1 to be correlated with genes associated with mitochondrial pathways. In all, SR-BI and ABCA1 correlated with 73 and 3 genes differentially expressed in T2D, respectively. We confirm that SR-BI and ABCA1 are the primary β-cell ApoA-I receptors and demonstrate that ApoA-I priming enhances β-cell insulin secretion via the upregulation of mitochondrial metabolism through ApoA-I-SR-BI and ApoA-I-ABCA1 pathways. We propose that SR-BI relies on mitochondrial and exocytotic pathways, while ABCA1 depends solely on mitochondrial pathways. Our findings uncover new targets in ApoA-I β-cell mechanism for T2D therapies.
Collapse
Affiliation(s)
- Claire L. Lyons
- Department of Experimental Medical Sciences, Unit of Medical Protein Science, Lund University, Lund, Sweden
| | - Elaine Cowan
- Department of Clinical Sciences, Unit of Islet Cell Exocytosis, Lund University Diabetes Centre, Malmö, Sweden
| | - Oktawia Nilsson
- Department of Experimental Medical Sciences, Unit of Medical Protein Science, Lund University, Lund, Sweden
| | - Manca Mohar
- Department of Experimental Medical Sciences, Unit of Medical Protein Science, Lund University, Lund, Sweden
| | - Pablo Peña-Martínez
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Lena Eliasson
- Department of Clinical Sciences, Unit of Islet Cell Exocytosis, Lund University Diabetes Centre, Malmö, Sweden
| | - Jens O. Lagerstedt
- Department of Experimental Medical Sciences, Unit of Medical Protein Science, Lund University, Lund, Sweden
- Department of Clinical Sciences, Unit of Islet Cell Exocytosis, Lund University Diabetes Centre, Malmö, Sweden
| |
Collapse
|
57
|
Brenes AJ, Griesser E, Sinclair LV, Davidson L, Prescott AR, Singh F, Hogg EKJ, Espejo-Serrano C, Jiang H, Yoshikawa H, Platani M, Swedlow JR, Findlay GM, Cantrell DA, Lamond AI. Proteomic and functional comparison between human induced and embryonic stem cells. eLife 2024; 13:RP92025. [PMID: 39540879 PMCID: PMC11563575 DOI: 10.7554/elife.92025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) have great potential to be used as alternatives to embryonic stem cells (hESCs) in regenerative medicine and disease modelling. In this study, we characterise the proteomes of multiple hiPSC and hESC lines derived from independent donors and find that while they express a near-identical set of proteins, they show consistent quantitative differences in the abundance of a subset of proteins. hiPSCs have increased total protein content, while maintaining a comparable cell cycle profile to hESCs, with increased abundance of cytoplasmic and mitochondrial proteins required to sustain high growth rates, including nutrient transporters and metabolic proteins. Prominent changes detected in proteins involved in mitochondrial metabolism correlated with enhanced mitochondrial potential, shown using high-resolution respirometry. hiPSCs also produced higher levels of secreted proteins, including growth factors and proteins involved in the inhibition of the immune system. The data indicate that reprogramming of fibroblasts to hiPSCs produces important differences in cytoplasmic and mitochondrial proteins compared to hESCs, with consequences affecting growth and metabolism. This study improves our understanding of the molecular differences between hiPSCs and hESCs, with implications for potential risks and benefits for their use in future disease modelling and therapeutic applications.
Collapse
Affiliation(s)
- Alejandro J Brenes
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
- Cell Signalling & Immunology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
- Human Pluripotent Stem Cell Facility, School of Life Sciences, University of Dundee, Dow St, Dundee DD1 5EHDundeeUnited Kingdom
| | - Eva Griesser
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Linda V Sinclair
- Cell Signalling & Immunology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Lindsay Davidson
- Human Pluripotent Stem Cell Facility, School of Life Sciences, University of Dundee, Dow St, Dundee DD1 5EHDundeeUnited Kingdom
| | - Alan R Prescott
- Dundee Imaging Facility, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Francois Singh
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Elizabeth KJ Hogg
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Carmen Espejo-Serrano
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Hao Jiang
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Harunori Yoshikawa
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Melpomeni Platani
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Jason R Swedlow
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Greg M Findlay
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Doreen A Cantrell
- Cell Signalling & Immunology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Angus I Lamond
- Molecular, Cell and Developmental Biology, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| |
Collapse
|
58
|
Yoneno S, Yamamoto K, Tabata K, Shimizu-Motohashi Y, Tomita A, Hayashi T, Maki H, Sato N, Inoue K, Saitsu H, Komaki H. A novel heterozygous TMEM63A variant in a familial case with early onset nystagmus, severe hypomyelination, and a favorable adult prognosis. J Hum Genet 2024; 69:607-611. [PMID: 38951194 DOI: 10.1038/s10038-024-01268-z] [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: 05/17/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/03/2024]
Abstract
Heterozygous transmembrane protein 63A (TMEM63A) variants cause transient infantile hypomyelinating leukodystrophy-19, which features remarkable natural resolution of clinical and imaging findings during childhood. Previous reports have mainly described de novo variants lacking detailed familial cases. Herein, we describe the clinical course of familial cases with a TMEM63A variant. A 5-month-old girl presented with nystagmus, global hypotonia, and difficulty swallowing since birth. Brain magnetic resonance imaging at 1.5 and 5 months revealed diffuse hypomyelination. Her mother, maternal aunt, and grandfather had nystagmus and motor developmental delays in infancy, which resolved spontaneously during childhood. Compared with these cases, the proband's motor developmental delay was profound, and she was the only one with feeding difficulties, necessitating nasogastric tube feeding. Genetic testing revealed a heterozygous TMEM63A variant (NM_014698.3:c.1658G>A, p.(Gly553Asp)) in the proband and her family. This is the first three-generation familial report of a TMEM63A variant that provides insight into its history and heterogeneity.
Collapse
Affiliation(s)
- Shota Yoneno
- Department of Child Neurology, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Kaoru Yamamoto
- Department of Child Neurology, National Center of Neurology and Psychiatry, Tokyo, Japan.
| | - Kenshiro Tabata
- Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yuko Shimizu-Motohashi
- Department of Child Neurology, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Ayaka Tomita
- Department of Neonatology, Toho University Omori Medical Center, Tokyo, Japan
| | - Taiju Hayashi
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hiroyuki Maki
- Department of Radiology, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Noriko Sato
- Department of Radiology, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Ken Inoue
- Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Hirotomo Saitsu
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Hirofumi Komaki
- Department of Child Neurology, National Center of Neurology and Psychiatry, Tokyo, Japan
- Translational Medical Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
59
|
Čunátová K, Fernández‐Vizarra E. Pathological variants in nuclear genes causing mitochondrial complex III deficiency: An update. J Inherit Metab Dis 2024; 47:1278-1291. [PMID: 39053894 PMCID: PMC11586608 DOI: 10.1002/jimd.12751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/24/2024] [Accepted: 05/02/2024] [Indexed: 07/27/2024]
Abstract
Mitochondrial disorders are a group of clinically and biochemically heterogeneous genetic diseases within the group of inborn errors of metabolism. Primary mitochondrial diseases are mainly caused by defects in one or several components of the oxidative phosphorylation system (complexes I-V). Within these disorders, those associated with complex III deficiencies are the least common. However, thanks to a deeper knowledge about complex III biogenesis, improved clinical diagnosis and the implementation of next-generation sequencing techniques, the number of pathological variants identified in nuclear genes causing complex III deficiency has expanded significantly. This updated review summarizes the current knowledge concerning the genetic basis of complex III deficiency, and the main clinical features associated with these conditions.
Collapse
Affiliation(s)
- Kristýna Čunátová
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
- Veneto Institute of Molecular MedicinePadovaItaly
| | - Erika Fernández‐Vizarra
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
- Veneto Institute of Molecular MedicinePadovaItaly
| |
Collapse
|
60
|
Ahlberg E, Jenmalm MC, Karlsson A, Karlsson R, Tingö L. Proteome characterization of extracellular vesicles from human milk: Uncovering the surfaceome by a lipid-based protein immobilization technology. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70020. [PMID: 39512873 PMCID: PMC11541861 DOI: 10.1002/jex2.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/19/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024]
Abstract
Breast milk is an essential source of nutrition and hydration for the infant. In addition, this highly complex fluid is rich in extracellular vesicles (EVs). Here, we have applied a microfluidic technology, lipid-based protein immobilization (LPI) and liquid chromatography with tandem mass spectrometry (LC-MS/MS) to characterize the proteome of human milk EVs. Mature milk from six mothers was subjected to EV isolation by ultracentrifugation followed by size exclusion chromatography. Three of the samples were carefully characterized; suggesting a subset enriched by small EVs. The EVs were digested by trypsin in an LPI flow cell and in-solution digestion, giving rise to two fractions of peptides originating from the surface proteome (LPI fraction) or the complete proteome (in-solution digestion). LC-MS/MS recovered peptides corresponding to 582 proteins in the LPI fraction and 938 proteins in the in-solution digested samples; 400 of these proteins were uniquely found in the in-solution digested samples and were hence denoted "cargo proteome". GeneOntology overrepresentation analysis gave rise to distinctly different functional predictions of the EV surfaceome and the cargo proteome. The surfaceome tends to be overrepresented in functions and components of relevance for the immune system, while the cargo proteome primarily seems to be associated with EV biogenesis.
Collapse
Affiliation(s)
- Emelie Ahlberg
- Department of Biomedical and Clinical Sciences, Division of Inflammation and InfectionLinköping UniversityLinkopingSweden
| | - Maria C. Jenmalm
- Department of Biomedical and Clinical Sciences, Division of Inflammation and InfectionLinköping UniversityLinkopingSweden
| | | | - Roger Karlsson
- Nanoxis Consulting ABGothenburgSweden
- Department of Clinical MicrobiologySahlgrenska University HospitalGothenburgSweden
| | - Lina Tingö
- Department of Biomedical and Clinical Sciences, Division of Inflammation and InfectionLinköping UniversityLinkopingSweden
- School of Medical SciencesÖrebro UniversityOrebroSweden
| |
Collapse
|
61
|
Kölz C, Gaugaz FZ, Handin N, Schaeffeler E, Tremmel R, Winter S, Klein K, Zanger UM, Artursson P, Schwab M, Nies AT. In silico and biological analyses of missense variants of the human biliary efflux transporter ABCC2: effects of novel rare missense variants. Br J Pharmacol 2024; 181:4593-4609. [PMID: 39096023 DOI: 10.1111/bph.16508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/17/2024] [Accepted: 06/18/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND AND PURPOSE The ATP-dependent biliary efflux transporter ABCC2, also known as multidrug resistance protein 2 (MRP2), is essential for the cellular disposition and detoxification of various xenobiotics including drugs as well as endogenous metabolites. Common functionally relevant ABCC2 genetic variants significantly alter drug responses and contribute to side effects. The aim of this study was to determine functional consequences of rare variants identified in subjects with European ancestry using in silico tools and in vitro analyses. EXPERIMENTAL APPROACH Targeted next-generation sequencing of the ABCC2 gene was used to identify novel variants in European subjects (n = 143). Twenty-six in silico tools were used to predict functional consequences. For biological validation, transport assays were carried out with membrane vesicles prepared from cell lines overexpressing the newly identified ABCC2 variants and estradiol β-glucuronide and carboxydichlorofluorescein as the substrates. KEY RESULTS Three novel rare ABCC2 missense variants were identified (W227R, K402T, V489F). Twenty-five in silico tools predicted W227R as damaging and one as potentially damaging. Prediction of functional consequences was not possible for K402T and V489F and for the common linked variants V1188E/C1515Y. Characterisation in vitro showed increased function of W227R, V489F and V1188E/C1515Y for both substrates, whereas K402T function was only increased for carboxydichlorofluorescein. CONCLUSION AND IMPLICATIONS In silico tools were unable to accurately predict the substrate-dependent increase in function of ABCC2 missense variants. In vitro biological studies are required to accurately determine functional activity to avoid misleading consequences for drug therapy.
Collapse
Affiliation(s)
- Charlotte Kölz
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | | | - Niklas Handin
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Elke Schaeffeler
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Roman Tremmel
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tuebingen, Tuebingen, Germany
| | - Stefan Winter
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tuebingen, Tuebingen, Germany
| | - Kathrin Klein
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tuebingen, Tuebingen, Germany
| | - Ulrich M Zanger
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tuebingen, Tuebingen, Germany
| | - Per Artursson
- Department of Pharmacy, Uppsala University, Uppsala, Sweden
| | - Matthias Schwab
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
- Department of Clinical Pharmacology, Pharmacy and Biochemistry, University of Tuebingen, Tuebingen, Germany
| | - Anne T Nies
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tuebingen, Tuebingen, Germany
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| |
Collapse
|
62
|
Qin B, Fan B, Li Y, Wang Y, Shen B, Xia N. An endoplasmic reticulum localized acetyl-CoA transporter is required for efficient fatty acid synthesis in Toxoplasma gondii. Open Biol 2024; 14:240184. [PMID: 39532149 PMCID: PMC11557232 DOI: 10.1098/rsob.240184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/11/2024] [Accepted: 09/13/2024] [Indexed: 11/16/2024] Open
Abstract
Toxoplasma gondii is an obligate intracellular parasite that can infect humans and diverse animals. Fatty acids are critical for the growth and proliferation of T. gondii, which has at least two pathways to synthesize fatty acids, including the type II de novo synthesis pathway in the apicoplast and the elongation pathway in the endoplasmic reticulum (ER). Acetyl-CoA is the key substrate for both fatty acid synthesis pathways. In the apicoplast, acetyl-CoA is mainly provided by the pyruvate dehydrogenase complex. However, how the ER acquires acetyl-CoA is not fully understood. Here, we identified a putative acetyl-CoA transporter (TgAT1) that localized to the ER of T. gondii. Deletion of TgAT1 impaired parasite growth and invasion in vitro and attenuated tachyzoite virulence in vivo. Metabolic tracing using 13C-acetate found that loss of TgAT1 reduced the incorporation of 13C into certain fatty acids, suggesting reduced activities of elongation. Truncation of AT1 was previously reported to confer resistance to the antimalarial compound GNF179 in Plasmodium falciparum. Interestingly, GNF179 had much weaker inhibitory effect on Toxoplasma than on Plasmodium. In addition, deletion of AT1 did not affect the susceptibility of Toxoplasma to GNF179, suggesting that this compound might be taken up differently or has different inhibitory mechanisms in these parasites. Together, our data show that TgAT1 has important roles for parasite growth and fatty acid synthesis, but its disruption does not confer GNF179 resistance in T. gondii.
Collapse
Affiliation(s)
- Biyun Qin
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province, People’s Republic of China
| | - Bolin Fan
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province, People’s Republic of China
| | - Yazhou Li
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province, People’s Republic of China
| | - Yidan Wang
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province, People’s Republic of China
| | - Bang Shen
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province, People’s Republic of China
- Key Laboratory of Preventive Veterinary Medicine in Hubei Province, Wuhan, Hubei Province, People’s Republic of China
- Hubei Hongshan Laboratory, Wuhan, Hubei Province, People’s Republic of China
- Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Shenzhen, Guangdong Province, People’s Republic of China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, Guangdong Province, People’s Republic of China
| | - Ningbo Xia
- National Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei Province, People’s Republic of China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong Province, People’s Republic of China
| |
Collapse
|
63
|
Karagöl A, Karagöl T, Li M, Zhang S. Inhibitory Potential of the Truncated Isoforms on Glutamate Transporter Oligomerization Identified by Computational Analysis of Gene-Centric Isoform Maps. Pharm Res 2024; 41:2173-2187. [PMID: 39487385 PMCID: PMC11599315 DOI: 10.1007/s11095-024-03786-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 10/14/2024] [Indexed: 11/04/2024]
Abstract
OBJECTIVE Glutamate transporters play a key role in central nervous system physiology by maintaining excitatory neurotransmitter homeostasis. Biological assemblies of the transporters, consisting of cyclic homotrimers, emerge as a crucial aspect of glutamate transporter modulation. Hence targeting heteromerization promises an effective approach for modulator design. On the other hand, the dynamic nature of transcription allows for the generation of transporter isoforms in structurally distinct manners. METHODS The potential isoforms were identified through the analysis of computationally generated gene-centric isoform maps. The conserved features of isoform sequences were revealed by computational chemistry methods and subsequent structural analysis of AlphaFold2 predictions. Truncated isoforms were further subjected to a wide range of docking analyses, 50ns molecular dynamics simulations, and evolutionary coupling analyses. RESULTS Energetic landscapes of isoform-canonical transporter complexes suggested an inhibitory potential of truncated isoforms on glutamate transporter bio-assembly. Moreover, isoforms that mimic the trimerization domain (in particular, TM2 helices) exhibited stronger interactions with canonical transporters, underscoring the role of transmembrane helices in isoform interactions. Additionally, self-assembly dynamics observed in truncated isoforms mimicking canonical TM5 helices indicate a potential protective role against unwanted interactions with canonical transporters. CONCLUSION Our computational studies on glutamate transporters offer insights into the roles of alternative splicing on protein interactions and identifies potential drug targets for physiological or pathological processes.
Collapse
Affiliation(s)
- Alper Karagöl
- Istanbul University Istanbul Medical Faculty, Istanbul, Turkey
| | - Taner Karagöl
- Istanbul University Istanbul Medical Faculty, Istanbul, Turkey
| | - Mengke Li
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Shuguang Zhang
- Laboratory of Molecular Architecture, Media Lab, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA.
| |
Collapse
|
64
|
Kato F, Bandou R, Yamaguchi Y, Inouye K, Inouye M. Characterization of a membrane toxin-antitoxin system, tsaAT, from Staphylococcus aureus. FEBS J 2024; 291:5015-5036. [PMID: 39356479 DOI: 10.1111/febs.17289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 08/01/2024] [Accepted: 09/23/2024] [Indexed: 10/03/2024]
Abstract
Bacterial toxin-antitoxin (TA) systems consist of a toxin that inhibits essential cellular processes, such as DNA replication, transcription, translation, or ATP synthesis, and an antitoxin neutralizing their cognate toxin. These systems have roles in programmed cell death, defense against phage, and the formation of persister cells. Here, we characterized the previously identified Staphylococcus aureus TA system, tsaAT, which consists of two putative membrane proteins: TsaT and TsaA. Expression of the TsaT toxin caused cell death and disrupted membrane integrity, whereas TsaA did not show any toxicity and neutralized the toxicity of TsaT. Furthermore, subcellular fractionation analysis demonstrated that both TsaA and TsaT localized to the cytoplasmic membrane of S. aureus expressing either or both 3xFLAG-tagged TsaA and 3xFLAG-tagged TsaT. Taken together, these results demonstrate that the TsaAT TA system consists of two membrane proteins, TsaA and TsaT, where TsaT disrupts membrane integrity, ultimately leading to cell death. Although sequence analyses showed that the tsaA and tsaT genes were conserved among Staphylococcus species, amino acid substitutions between TsaT orthologs highlighted the critical role of the 6th residue for its toxicity. Further amino acid substitutions indicated that the glutamic acid residue at position 63 in the TsaA antitoxin and the cluster of five lysine residues in the TsaT toxin are involved in TsaA's neutralization reaction. This study is the first to describe a bacterial TA system wherein both toxin and antitoxin are membrane proteins. These findings contribute to our understanding of S. aureus TA systems and, more generally, give new insight into highly diverse bacterial TA systems.
Collapse
Affiliation(s)
- Fuminori Kato
- Graduate School of Biomedical and Health Sciences, Hiroshima University, Japan
| | - Risa Bandou
- Faculty of Dentistry, Hiroshima University, Japan
| | - Yoshihiro Yamaguchi
- Department of Biology, Graduate School of Sciences, Osaka Metropolitan University, Japan
| | - Keiko Inouye
- Department of Biochemistry and Molecular Biology, Center for Advanced Biotechnology and Medicine, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Masayori Inouye
- Department of Biochemistry and Molecular Biology, Center for Advanced Biotechnology and Medicine, Rutgers-Robert Wood Johnson Medical School, Piscataway, NJ, USA
| |
Collapse
|
65
|
Thomy J, Schvarcz CR, McBeain KA, Edwards KF, Steward GF. Eukaryotic viruses encode the ribosomal protein eL40. NPJ VIRUSES 2024; 2:51. [PMID: 39464202 PMCID: PMC11499249 DOI: 10.1038/s44298-024-00060-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/20/2024] [Indexed: 10/29/2024]
Abstract
Viruses in the phylum Nucleocytoviricota are large, complex and have an exceptionally diverse metabolic repertoire. Some encode hundreds of products involved in the translation of mRNA into protein, but none was known to encode any of the proteins in ribosomes, the central engines of translation. With the discovery of the eL40 gene in FloV-SA2, we report the first example of a eukaryotic virus encoding a ribosomal protein and show that this gene is also present and expressed in other uncultivated marine giant viruses. FloV-SA2 also encodes a "group II" viral rhodopsin, a viral light-activated protein of unknown function previously only reported in metagenomes. FloV-SA2 is thus a valuable model system for investigating new mechanisms by which viruses manipulate eukaryotic cell metabolism.
Collapse
Affiliation(s)
- Julie Thomy
- Daniel K. Inouye Center for Microbial Oceanography: Research and Education, School of Ocean and Earth Science and Technology (SOEST), University of Hawaiʻi at Mānoa, Honolulu, HI USA
- Department of Oceanography, School of Ocean and Earth Science and Technology (SOEST), University of Hawaiʻi at Mānoa, Honolulu, HI USA
| | - Christopher R. Schvarcz
- Daniel K. Inouye Center for Microbial Oceanography: Research and Education, School of Ocean and Earth Science and Technology (SOEST), University of Hawaiʻi at Mānoa, Honolulu, HI USA
- Department of Oceanography, School of Ocean and Earth Science and Technology (SOEST), University of Hawaiʻi at Mānoa, Honolulu, HI USA
| | - Kelsey A. McBeain
- Daniel K. Inouye Center for Microbial Oceanography: Research and Education, School of Ocean and Earth Science and Technology (SOEST), University of Hawaiʻi at Mānoa, Honolulu, HI USA
- Department of Oceanography, School of Ocean and Earth Science and Technology (SOEST), University of Hawaiʻi at Mānoa, Honolulu, HI USA
| | - Kyle F. Edwards
- Department of Oceanography, School of Ocean and Earth Science and Technology (SOEST), University of Hawaiʻi at Mānoa, Honolulu, HI USA
| | - Grieg F. Steward
- Daniel K. Inouye Center for Microbial Oceanography: Research and Education, School of Ocean and Earth Science and Technology (SOEST), University of Hawaiʻi at Mānoa, Honolulu, HI USA
- Department of Oceanography, School of Ocean and Earth Science and Technology (SOEST), University of Hawaiʻi at Mānoa, Honolulu, HI USA
| |
Collapse
|
66
|
Reeves AE, Vilen Z, Fuentecilla TR, Parker CG, Huang ML. Charting the Dynamic Trophoblast Plasma Membrane Identifies LYN As a Functional Regulator of Syncytialization. ACS Chem Biol 2024; 19:2220-2231. [PMID: 39289808 PMCID: PMC11863573 DOI: 10.1021/acschembio.4c00443] [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] [Indexed: 09/19/2024]
Abstract
The differentiation of placental cytotrophoblasts (CTBs) into the syncytiotrophoblast (STB) layer results in a significant remodeling of the plasma membrane proteome. Here, we use a peroxidase-catalyzed proximity labeling strategy to map the dynamic plasma membrane proteomes of CTBs and STBs. Coupled with mass-spectrometry-based proteomics, we identify hundreds of plasma membrane proteins and observe relative changes in protein abundance throughout differentiation, including the upregulation of the plasma-membrane-localized nonreceptor tyrosine kinase LYN. We show that both siRNA-mediated knockdown and small molecule inhibition of LYN kinase function impairs CTB fusion and reduces the expression of syncytialization markers, presenting a function for LYN outside of its canonical role in immunological signaling. Our results demonstrate the use of the proximity labeling platform to discover functional regulators within the plasma membrane and provide new avenues to regulate trophoblast differentiation.
Collapse
Affiliation(s)
- Abigail E Reeves
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N. Torrey Pines Rd., La Jolla, California 92037, United States
- Department of Chemistry, Scripps Research, 10550 N. Torrey Pines Rd., La Jolla, California 92037, United States
| | - Zak Vilen
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N. Torrey Pines Rd., La Jolla, California 92037, United States
- Department of Chemistry, Scripps Research, 10550 N. Torrey Pines Rd., La Jolla, California 92037, United States
| | - Trinity R Fuentecilla
- Department of Chemistry, Scripps Research, 10550 N. Torrey Pines Rd., La Jolla, California 92037, United States
| | - Christopher G Parker
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N. Torrey Pines Rd., La Jolla, California 92037, United States
- Department of Chemistry, Scripps Research, 10550 N. Torrey Pines Rd., La Jolla, California 92037, United States
| | - Mia L Huang
- Skaggs Graduate School of Chemical and Biological Sciences, Scripps Research, 10550 N. Torrey Pines Rd., La Jolla, California 92037, United States
- Department of Chemistry, Scripps Research, 10550 N. Torrey Pines Rd., La Jolla, California 92037, United States
| |
Collapse
|
67
|
Bhowmick S, Viveros RP, Latoscha A, Commichau FM, Wrede C, Al-Bassam MM, Tschowri N. Cell shape and division septa positioning in filamentous Streptomyces require a functional cell wall glycopolymer ligase CglA. mBio 2024; 15:e0149224. [PMID: 39248520 PMCID: PMC11481543 DOI: 10.1128/mbio.01492-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 08/01/2024] [Indexed: 09/10/2024] Open
Abstract
The cell wall of monoderm bacteria consists of peptidoglycan and glycopolymers in roughly equal proportions and is crucial for cellular integrity, cell shape, and bacterial vitality. Despite the immense value of Streptomyces in biotechnology and medicine as antibiotic producers, we know very little about their cell wall biogenesis, composition, and functions. Here, we have identified the LCP-LytR_C domain protein CglA (Vnz_13690) as a key glycopolymer ligase, which specifically localizes in zones of cell wall biosynthesis in S. venezuelae. Reduced amount of glycopolymers in the cglA mutant results in enlarged vegetative hyphae and failures in FtsZ-rings formation and positioning. Consequently, division septa are misplaced leading to the formation of aberrant cell compartments, misshaped spores, and reduced cell vitality. In addition, we report our discovery that c-di-AMP signaling and decoration of the cell wall with glycopolymers are physiologically linked in Streptomyces since the deletion of cglA restores growth of the S. venezuelae disA mutant at high salt. Altogether, we have identified and characterized CglA as a novel component of cell wall biogenesis in Streptomyces, which is required for cell shape maintenance and cellular vitality in filamentous, multicellular bacteria.IMPORTANCEStreptomyces are our key producers of antibitiotics and other bioactive molecules and are, therefore, of high value for medicine and biotechnology. They proliferate by apical extension and branching of hyphae and undergo complex cell differentiation from filaments to spores during their life cycle. For both, growth and sporulation, coordinated cell wall biogenesis is crucial. However, our knowledge about cell wall biosynthesis, functions, and architecture in Streptomyces and in other Actinomycetota is still very limited. Here, we identify CglA as the key enzyme needed for the attachment of glycopolymers to the cell wall of S. venezuelae. We demonstrate that defects in the cell wall glycopolymer content result in loss of cell shape in these filamentous bacteria and show that division-competent FtsZ-rings cannot assemble properly and fail to be positioned correctly. As a consequence, cell septa placement is disturbed leading to the formation of misshaped spores with reduced viability.
Collapse
Affiliation(s)
- Sukanya Bhowmick
- Institute of Microbiology, Leibniz Universität Hannover, Hannover, Germany
| | - Ruth P. Viveros
- Institute of Microbiology, Leibniz Universität Hannover, Hannover, Germany
| | - Andreas Latoscha
- Institute of Biology/Microbiology, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Fabian M. Commichau
- Institute of Biology, FG Molecular Microbiology 190 h, Universität Hohenheim, Stuttgart, Germany
| | - Christoph Wrede
- Institute of Functional and Applied Anatomy, Research Core Unit Electron Microscopy, Hannover Medical School, Hannover, Germany
| | | | - Natalia Tschowri
- Institute of Microbiology, Leibniz Universität Hannover, Hannover, Germany
| |
Collapse
|
68
|
Holtz A, Van Weyenbergh J, Hong SL, Cuypers L, O'Toole Á, Dudas G, Gerdol M, Potter BI, Ntoumi F, Mapanguy CCM, Vanmechelen B, Wawina-Bokalanga T, Van Holm B, Menezes SM, Soubotko K, Van Pottelbergh G, Wollants E, Vermeersch P, Jacob AS, Maes B, Obbels D, Matheeussen V, Martens G, Gras J, Verhasselt B, Laffut W, Vael C, Goegebuer T, van der Kant R, Rousseau F, Schymkowitz J, Serrano L, Delgado J, Wenseleers T, Bours V, André E, Suchard MA, Rambaut A, Dellicour S, Maes P, Durkin K, Baele G. Emergence of the B.1.214.2 SARS-CoV-2 lineage with an Omicron-like spike insertion and a unique upper airway immune signature. BMC Infect Dis 2024; 24:1139. [PMID: 39390446 PMCID: PMC11468156 DOI: 10.1186/s12879-024-09967-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/20/2024] [Indexed: 10/12/2024] Open
Abstract
We investigate the emergence, mutation profile, and dissemination of SARS-CoV-2 lineage B.1.214.2, first identified in Belgium in January 2021. This variant, featuring a 3-amino acid insertion in the spike protein similar to the Omicron variant, was speculated to enhance transmissibility or immune evasion. Initially detected in international travelers, it substantially transmitted in Central Africa, Belgium, Switzerland, and France, peaking in April 2021. Our travel-aware phylogeographic analysis, incorporating travel history, estimated the origin to the Republic of the Congo, with primary European entry through France and Belgium, and multiple smaller introductions during the epidemic. We correlate its spread with human travel patterns and air passenger data. Further, upon reviewing national reports of SARS-CoV-2 outbreaks in Belgian nursing homes, we found this strain caused moderately severe outcomes (8.7% case fatality ratio). A distinct nasopharyngeal immune response was observed in elderly patients, characterized by 80% unique signatures, higher B- and T-cell activation, increased type I IFN signaling, and reduced NK, Th17, and complement system activation, compared to similar outbreaks. This unique immune response may explain the variant's epidemiological behavior and underscores the need for nasal vaccine strategies against emerging variants.
Collapse
Affiliation(s)
- Andrew Holtz
- Lyssavirus Epidemiology and Neuropathology Unit, Institut Pasteur, Université Paris Cité, Paris, France.
| | - Johan Van Weyenbergh
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium.
| | - Samuel L Hong
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Lize Cuypers
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
- Department of Laboratory Medicine, University Hospitals Leuven, National Reference Centre for Respiratory Pathogens, Leuven, Belgium
| | - Áine O'Toole
- Institute of Ecology and Evolution, University of Edinburgh, Edinburgh, UK
| | - Gytis Dudas
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Marco Gerdol
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Barney I Potter
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Francine Ntoumi
- Fondation Congolaise Pour La Recherche Médicale, Brazzaville, Republic of Congo
- Institute for Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Claujens Chastel Mfoutou Mapanguy
- Fondation Congolaise Pour La Recherche Médicale, Brazzaville, Republic of Congo
- Faculty of Sciences and Techniques, University Marien Ngouabi, Brazzaville, Republic of Congo
| | - Bert Vanmechelen
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Tony Wawina-Bokalanga
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Bram Van Holm
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Soraya Maria Menezes
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | | | | | - Elke Wollants
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Pieter Vermeersch
- Department of Laboratory Medicine, University Hospitals Leuven, National Reference Centre for Respiratory Pathogens, Leuven, Belgium
| | - Ann-Sophie Jacob
- Department of Laboratory Medicine, University Hospitals Leuven, National Reference Centre for Respiratory Pathogens, Leuven, Belgium
| | - Brigitte Maes
- Laboratory for Molecular Diagnostics, Jessa Hospital, Hasselt, Belgium
- Hasselt University, Hasselt, Belgium
- Limburg Clinical Research Center, Hasselt, Belgium
| | | | - Veerle Matheeussen
- Department of Laboratory Medicine, Antwerp University Hospital (UZA), Edegem, Belgium
- Laboratory of Medical Biochemistry and Laboratory of Medical Microbiology, University of Antwerp, Wilrijk, Belgium
| | - Geert Martens
- Department of Laboratory Medicine, AZ Delta General Hospital, Roeselare, Belgium
| | - Jérémie Gras
- Institut de Pathologie Et de Génétique, Gosselies, Belgium
| | - Bruno Verhasselt
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Wim Laffut
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Carl Vael
- Department of Laboratory Medicine, KLINA General Hospital, Brasschaat, AZ, Belgium
| | | | - Rob van der Kant
- Switch Laboratory, VIB Center for Brain and Disease Research and Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Frederic Rousseau
- Switch Laboratory, VIB Center for Brain and Disease Research and Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Joost Schymkowitz
- Switch Laboratory, VIB Center for Brain and Disease Research and Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Luis Serrano
- Center for Genomic Regulation, Barcelona Institute for Science and Technology, 08003, Barcelona, Spain
- Universitat Pompeu Fabra, 08002, Barcelona, Spain
- Institució Catalana de Recerca I Estudis Avançats, 08010, Barcelona, Spain
| | - Javier Delgado
- Center for Genomic Regulation, Barcelona Institute for Science and Technology, 08003, Barcelona, Spain
| | | | - Vincent Bours
- Department of Medical Genetics, CHU Liege, Liege, Belgium
| | - Emmanuel André
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Marc A Suchard
- Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, CA, USA
| | - Andrew Rambaut
- Institute of Ecology and Evolution, University of Edinburgh, Edinburgh, UK
| | - Simon Dellicour
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
- Université Libre de Bruxelles, Brussels, Belgium
| | - Piet Maes
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| | - Keith Durkin
- Laboratory of Human Genetics, GIGA Research Institute, Liège, Belgium
| | - Guy Baele
- Department of Microbiology, Immunology and Transplantation, Rega Institute, KU Leuven, Leuven, Belgium
| |
Collapse
|
69
|
Santana PA, Forero JC, Guzmán F, Gaete S, Acosta F, Mercado LA, Álvarez CA. Detection and Localization of IL-8 and CXCR1 in Rainbow Trout Larvae in Response to Pseudomonas aeruginosa Lipopolysaccharide. Animals (Basel) 2024; 14:2878. [PMID: 39409827 PMCID: PMC11475925 DOI: 10.3390/ani14192878] [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: 08/14/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
The salmonid industry faces challenges due to the susceptibility of fish to opportunistic pathogens, particularly in early developmental stages. Understanding the immunological capacity during these stages is crucial for developing effective disease control strategies. IL-8R, a member of the G-protein-coupled receptor family, acts as a receptor for Interleukin 8 (IL-8). The binding of IL-8 to IL-8R plays a major role in the pathophysiology of a wide spectrum of inflammatory conditions. This study focused on the immune response capacity of rainbow trout (Oncorhynchus mykiss) larvae by analyzing IL-8/CXCR1 response to lipopolysaccharide (LPS) from Pseudomonas aeruginosa. Previous research demonstrated that LPS from P. aeruginosa acts as a potent immunostimulant in teleost, enhancing pro-inflammatory cytokines. The methodology included in silico analysis and the synthesis and characterization of an omCXCR1-derived epitope peptide, which was used to produce omCXCR1-specific anti98 serum in mice. The research revealed that rainbow trout larvae 19 days post-hatching (dph) exhibited pronounced immune responses post-stimulation with 1 µg/mL of LPS. This was evidenced by the upregulated protein expression of IL-8 and omCXCR1 in trout larvae 2 and 8 h after LPS challenge, as analyzed by ELISA and immunohistochemistry. Furthermore, fluorescence microscopy successfully revealed the colocalization of IL-8 and its receptor in cells from mucosal tissues after LPS challenge in larvae 19 dph. These findings underscore the efficacy of LPS immersion as a method to activate the innate immune system in trout larvae. Furthermore, we propose IL-8 and its receptor as molecular markers for evaluating immunostimulation in the early developmental stages of salmonids.
Collapse
Affiliation(s)
- Paula A. Santana
- Instituto de Ciencias Aplicadas, Facultad de Ingeniería, Universidad Autónoma de Chile, Santiago 8910060, Chile;
| | - Juan C. Forero
- Laboratorio de Bioingeniería de Tejidos e Innovación Odontológica, Facultad de Odontología, Universidad de Valparaíso, Valparaíso 2360004, Chile;
- Cátedra de Ciencias Básicas, Escuela de Odontología, Facultad de Odontología, Universidad de Valparaíso, Valparaíso 2360004, Chile
| | - Fanny Guzmán
- Núcleo Biotecnología Curauma, Pontificia Universidad Católica de Valparaíso, Valparaíso 2340025, Chile;
| | - Sandra Gaete
- Laboratorio de Diagnóstico de COVID-19, Unidad de Detección y Análisis, Universidad de O’Higgins, Rancagua 2841959, Chile;
| | - Félix Acosta
- Grupo de Investigación en Acuicultura (GIA), Instituto Universitario Ecoaqua, Universidad de Las Palmas de Gran Canaria, Islas Canarias, 35214 Taliarte, Spain;
| | - Luis A. Mercado
- Instituto de Biología, Pontificia Universidad Católica de Valparaíso, Valparaíso 2340025, Chile
| | - Claudio A. Álvarez
- Laboratorio de Cultivo de Peces, Departamento de Acuicultura, Universidad Católica del Norte, Coquimbo 1781421, Chile
- Laboratorio de Fisiología y Genética Marina (FIGEMA), Centro de Estudios Avanzados en Zonas Áridas (CEAZA), Coquimbo 1781421, Chile
| |
Collapse
|
70
|
Kim CS, Sayler AL, Dean H, Ruel NM, Hammond JR. Functional comparison of human and murine equilibrative nucleobase transporter 1. PLoS One 2024; 19:e0311519. [PMID: 39361655 PMCID: PMC11449324 DOI: 10.1371/journal.pone.0311519] [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: 07/05/2024] [Accepted: 09/19/2024] [Indexed: 10/05/2024] Open
Abstract
6-Mercaptopurine (6-MP) maintenance therapy is the mainstay for various types of leukemia and inflammatory bowel disease. 6-MP is associated with numerous adverse effects including gastrointestinal intolerance, myelotoxicity, and hepatotoxicity. This can lead to therapy discontinuation which is associated with a higher risk of relapse. Drug transporter expression is a known factor contributing to patient variability in drug response and toxicity. We have established that the SLC43A3-encoded equilibrative nucleobase transporter 1 (ENBT1) mediates the transport of 6-MP into human lymphocytes and human embryonic kidney 293 (HEK293) cell lines transfected with SLC43A3. ENBT1 is known to be expressed in the gastrointestinal tract, bone marrow, and the liver. However, the relationship between ENBT1 and 6-MP-associated adverse events, and its pharmacokinetics, is unknown. To validate the use of mouse models (e.g. slc43a3-null mice) for exploring this relationship, we assessed the functional similarities between human and murine ENBT1 using HEK293 cells transfected with the respective SLC43A3/slc43a3 constructs, and the leukemia cell lines MOLT-4 (human) and L1210 (murine). Based on in silico analyses of structural similarities between transporters, we hypothesized that human and murine ENBT1 will have similar 6-MP transport/inhibition kinetics and a similar impact on 6-MP-induced cytotoxicity. We show herein that mslc43a3-encoded mouse ENBT1 transports both [3H]6-MP and [3H]adenine with kinetics similar to those of hSLC43A3-encoded human ENBT1. Both are also similarly distributed in mouse and human tissues. Therefore, data obtained from mouse models where ENBT1 is disrupted or modified may provide clinically relevant insights on its roles in modulating the actions of 6-MP.
Collapse
Affiliation(s)
- Chan S. Kim
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Aaron L. Sayler
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Hannah Dean
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Nicholas M. Ruel
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - James R. Hammond
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
71
|
Lespine A, Blancfuney C, Prichard R, Alberich M. P-glycoproteins in anthelmintic safety, efficacy, and resistance. Trends Parasitol 2024; 40:896-913. [PMID: 39168719 DOI: 10.1016/j.pt.2024.07.008] [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: 06/04/2024] [Revised: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 08/23/2024]
Abstract
P-glycoprotein (PGP) is a pivotal transmembrane transporter governing the cellular flux of diverse substances shielding mammals from toxics. It can thwart the effectiveness of medicines such as ivermectin (IVM) and other macrocyclic lactone (ML) anthelmintics, undermining therapeutic efforts. We analyze the role of PGPs in limiting the toxicity of these drugs in hosts, and their potential contribution to anthelmintic resistance in nematodes. Targeting nematode PGPs to increase drug sensitivity to MLs seems interesting, but is hampered by the lack of selective inhibitors. The nuclear hormone receptor (NHR)-8 should be seriously considered as a target because it upregulates multiple PGPs involved in anthelmintic resistance and it is specific to nematodes. This would advance our understanding of host-pathogen dynamics and foster innovative therapeutic strategies.
Collapse
Affiliation(s)
- Anne Lespine
- INTHERES, Université de Toulouse, INRAE, ENVT, Toulouse, France.
| | | | - Roger Prichard
- Institute of Parasitology, McGill University, Ste Anne-de-Bellevue, Canada
| | | |
Collapse
|
72
|
Papouskova K, Zimmermannova O, Sychrova H. Distinct regions of its first intracellular loop contribute to the proper localization, transport activity and substrate-affinity adjustment of the main yeast K + importer Trk1. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184369. [PMID: 38969203 DOI: 10.1016/j.bbamem.2024.184369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 06/04/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024]
Abstract
Trk1 is the main K+ importer of Saccharomyces cerevisiae. Its proper functioning enables yeast cells to grow in environments with micromolar amounts of K+. Although the structure of Trk1 has not been experimentally determined, the transporter is predicted to be composed of four MPM (transmembrane segment - pore loop - transmembrane segment) motifs which are connected by intracellular loops. Of those, in particular the first loop (IL1) is unique in its length; it forms more than half of the entire protein. The deletion of the majority of IL1 does not abolish the transport activity of Trk1. However IL1 is thought to be involved in the modulation of the transporter's functioning. In this work, we prepared a series of internally shortened versions of Trk1 that lacked various parts of IL1, and we studied their properties in S. cerevisiae cells without chromosomal copies of TRK genes. Using this approach, we were able to determine that both N- and C-border regions of IL1 are necessary for the proper localization of Trk1. Moreover, the N-border part of IL1 is also important for the functioning of Trk1, as its absence resulted in a decrease in the transporter's substrate affinity. In addition, in the internal part of IL1, we newly identified a stretch of amino-acid residues that are indispensable for retaining the transporter's maximum velocity, and another region whose deletion affected the ability of Trk1 to adjust its affinity in response to external levels of K+.
Collapse
Affiliation(s)
- Klara Papouskova
- Laboratory of Membrane Transport, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague 4, Czech Republic.
| | - Olga Zimmermannova
- Laboratory of Membrane Transport, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague 4, Czech Republic.
| | - Hana Sychrova
- Laboratory of Membrane Transport, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200 Prague 4, Czech Republic.
| |
Collapse
|
73
|
Karagöl A, Karagöl T, Zhang S. Molecular Dynamic Simulations Reveal that Water-Soluble QTY-Variants of Glutamate Transporters EAA1, EAA2 and EAA3 Retain the Conformational Characteristics of Native Transporters. Pharm Res 2024; 41:1965-1977. [PMID: 39322794 PMCID: PMC11530497 DOI: 10.1007/s11095-024-03769-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/13/2024] [Indexed: 09/27/2024]
Abstract
OBJECTIVE Glutamate transporters play a crucial role in neurotransmitter homeostasis, but studying their structure and function is challenging due to their membrane-bound nature. This study aims to investigate whether water-soluble QTY-variants of glutamate transporters EAA1, EAA2 and EAA3 retain the conformational characteristics and dynamics of native membrane-bound transporters. METHODS Molecular dynamics simulations and comparative genomics were used to analyze the structural dynamics of both native transporters and their QTY-variants. Native transporters were simulated in lipid bilayers, while QTY-variants were simulated in aqueous solution. Lipid distortions, relative solvent accessibilities, and conformational changes were examined. Evolutionary conservation profiles were correlated with structural dynamics. Statistical analyses included multivariate analysis to account for confounding variables. RESULTS QTY-variants exhibited similar residue-wise conformational dynamics to their native counterparts, with correlation coefficients of 0.73 and 0.56 for EAA1 and EAA3, respectively (p < 0.001). Hydrophobic interactions of native helices correlated with water interactions of QTY- helices (rs = 0.4753, p < 0.001 for EAA1). QTY-variants underwent conformational changes resembling the outward-to-inward transition of native transporters. CONCLUSIONS Water-soluble QTY-variants retain key structural properties of native glutamate transporters and mimic aspects of native lipid interactions, including conformational flexibility. This research provides valuable insights into the conformational changes and molecular mechanisms of glutamate transport, potentially offering a new approach for studying membrane protein dynamics and drug interactions.
Collapse
Affiliation(s)
- Alper Karagöl
- Istanbul University Istanbul Medical Faculty, Istanbul, Turkey
| | - Taner Karagöl
- Istanbul University Istanbul Medical Faculty, Istanbul, Turkey
| | - Shuguang Zhang
- Laboratory of Molecular Architecture, Media Lab, Massachusetts Institute of Technology, Massachusetts Avenue, Cambridge, MA, 02139, USA.
| |
Collapse
|
74
|
Fabri LM, Moraes CM, Garçon DP, McNamara JC, Faria SC, Leone FA. Primary amino acid sequences of decapod (Na +, K +)-ATPase provide evolutionary insights into osmoregulatory mechanisms. Comp Biochem Physiol A Mol Integr Physiol 2024; 296:111696. [PMID: 39004301 DOI: 10.1016/j.cbpa.2024.111696] [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: 04/29/2024] [Revised: 07/08/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
Decapod Crustacea exhibit a marine origin, but many taxa have occupied environments ranging from brackish to fresh water and terrestrial habitats, overcoming their inherent osmotic challenges. Osmotic and ionic regulation is achieved by the gill epithelia, driven by two active ATP-hydrolyzing ion transporters, the basal (Na+, K+)-ATPase and the apical V(H+)-ATPase. The kinetic characteristic of gill (Na+, K+)-ATPase and the mRNA expression of its α subunit have been widely studied in various decapod species under different salinity challenges. However, the evolution of the primary structure has not been explored, especially considering the functional modifications associated with decapod phylogeny. Here, we proposed a model for the topology of the decapod α subunit, identifying the sites and motifs involved in its function and regulation, as well as the patterns of its evolution assuming a decapod phylogeny. We also examined both the amino acid substitutions and their functional implications within the context of biochemical and physiological adaptation. The α-subunit of decapod crustaceans shows greater conservation (∼94% identity) compared to the β-subunit (∼40%). While the binding sites for ATP and modulators are conserved in the decapod enzyme, the residues involved in the α-β interaction are only partially conserved. In the phylogenetic context of the complete sequence of (Na+, K+)-ATPase α-subunit, most substitutions appear to be characteristic of the entire group, with specific changes for different subgroups, especially among brachyuran crabs. Interestingly, there was no consistent separation of α-subunit partial sequences related to habitat, suggesting that the convergent evolution for freshwater or terrestrial modes of life is not correlated with similar changes in the enzyme's primary amino acid sequence.
Collapse
Affiliation(s)
- Leonardo M Fabri
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Cintya M Moraes
- Departamento de Bioquímica e Imunologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - John C McNamara
- Departamento de Biologia Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil; Centro de Biologia Marinha, Universidade de São Paulo, São Sebastião, São Paulo, Brazil
| | - Samuel C Faria
- Centro de Biologia Marinha, Universidade de São Paulo, São Sebastião, São Paulo, Brazil
| | - Francisco A Leone
- Departamento de Química, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
75
|
Pernecker M, Ciarimboli G. Regulation of renal organic cation transporters. FEBS Lett 2024; 598:2328-2347. [PMID: 38831380 DOI: 10.1002/1873-3468.14943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/30/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024]
Abstract
Transporters for organic cations (OCs) facilitate exchange of positively charged molecules through the plasma membrane. Substrates for these transporters encompass neurotransmitters, metabolic byproducts, drugs, and xenobiotics. Consequently, these transporters actively contribute to the regulation of neurotransmission, cellular penetration and elimination process for metabolic products, drugs, and xenobiotics. Therefore, these transporters have significant physiological, pharmacological, and toxicological implications. In cells of renal proximal tubules, the vectorial secretion pathways for OCs involve expression of organic cation transporters (OCTs) and multidrug and toxin extrusion proteins (MATEs) on basolateral and apical membrane domains, respectively. This review provides an overview of documented regulatory mechanisms governing OCTs and MATEs. Additionally, regulation of these transporters under various pathological conditions is summarized. The expression and functionality of OCTs and MATEs are subject to diverse pre- and post-translational modifications, providing insights into their regulation in various pathological conditions. Typically, in diseases, downregulation of transporter expression is observed, probably as a protective mechanism to prevent additional damage to kidney tissue. This regulation may be attributed to the intricate network of modifications these transporters undergo, shedding light on their dynamic responses in pathological contexts.
Collapse
Affiliation(s)
- Moritz Pernecker
- Experimental Nephrology, Department of Internal Medicine D, University Hospital Münster, Germany
| | - Giuliano Ciarimboli
- Experimental Nephrology, Department of Internal Medicine D, University Hospital Münster, Germany
| |
Collapse
|
76
|
Iyyappan S, Rather MA, Ahmad I, Ahmad I. Comparative mitochondrial genomics analysis of selected species of Schizothoracinae sub family to explore the differences at mitochondrial DNA level. Comput Biol Chem 2024; 112:108165. [PMID: 39106606 DOI: 10.1016/j.compbiolchem.2024.108165] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/09/2024]
Abstract
A comprehensive analysis of the whole mitochondrial genomes of the Schizothoracinae subfamily of the family Cyprinidae has been revealed for the first time. The species analyzed include Schizothorax niger, Schizothorax esocinus, Schizothorax labiatus and Schizothorax plagoistomus. The total mitochondrial DNA (mtDNA) length was determined to be 16585 bp, 16583 bp, 16582 bp and 16576 bp, respectively with 13 protein-coding genes, 2 rRNA genes, 22 tRNA genes and 2 non-coding area genes. The combined mean base compositions of the four species were as follows: A: 29.91 % T: 25.47 % G: 17.65 % C 27.01 %. The range of the GC content is 45-44 %, respectively. All protein coding genes (PCGs) commenced with the typical ATG codon, except for the cytochrome c oxidase subunit 1 (COX1) gene with GTG. The analysis of vital amino acid biosynthesis genes (COX1, ATPase 6, ATPase 8) in four different species revealed no significant differences. All 13 PCGs had Ka/Ks ratios that were all lesser than one, demonstrating purifying selection on those molecules. These tRNA genes were predicted to fold into the typical cloverleaf secondary structures with normal base pairing and ranged in size from 66 to 75 nucleotides. Additionally, the phylogenetic tree analysis revealed that S. esocinus species that was most alike to S. labiatus. This study provides critical data for phylogenetic analysis of the Schizothoracinae subfamily, which will help to resolve taxonomic difficulties and identify evolutionary links. Detailed mtDNA data are an invaluable resource for studying genetic diversity, population structure, and gene flow. Understanding genetic makeup can help inform conservation plans, identify unique populations, and track genetic variation to ensure effective preservation.
Collapse
Affiliation(s)
- S Iyyappan
- Division of Fish Genetics and Biotechnology, Faculty of Fisheries, Rangil, Ganderbal, Sher-e-Kashmir University of Agricultural Science and Technology, Kashmir 190006, India
| | - Mohd Ashraf Rather
- Division of Fish Genetics and Biotechnology, Faculty of Fisheries, Rangil, Ganderbal, Sher-e-Kashmir University of Agricultural Science and Technology, Kashmir 190006, India.
| | - Ishtiyaq Ahmad
- Division of Fish Genetics and Biotechnology, Faculty of Fisheries, Rangil, Ganderbal, Sher-e-Kashmir University of Agricultural Science and Technology, Kashmir 190006, India
| | - Irfan Ahmad
- Division of Fish Genetics and Biotechnology, Faculty of Fisheries, Rangil, Ganderbal, Sher-e-Kashmir University of Agricultural Science and Technology, Kashmir 190006, India
| |
Collapse
|
77
|
Sharma GK, Sharma R, Joshi K, Qureshi S, Mathur S, Sinha S, Chatterjee S, Nunia V. Advancing microbial diagnostics: a universal phylogeny guided computational algorithm to find unique sequences for precise microorganism detection. Brief Bioinform 2024; 25:bbae545. [PMID: 39441245 PMCID: PMC11497845 DOI: 10.1093/bib/bbae545] [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: 01/06/2024] [Revised: 09/21/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024] Open
Abstract
Sequences derived from organisms sharing common evolutionary origins exhibit similarity, while unique sequences, absent in related organisms, act as good diagnostic marker candidates. However, the approach focused on identifying dissimilar regions among closely-related organisms poses challenges as it requires complex multiple sequence alignments, making computation and parsing difficult. To address this, we have developed a biologically inspired universal NAUniSeq algorithm to find the unique sequences for microorganism diagnosis by traveling through the phylogeny of life. Mapping through a phylogenetic tree ensures a low number of cross-contamination and false positives. We have downloaded complete taxonomy data from Taxadb database and sequence data from National Center for Biotechnology Information Reference Sequence Database (NCBI-Refseq) and, with the help of NetworkX, created a phylogenetic tree. Sequences were assigned over the graph nodes, k-mers were created for target and non-target nodes and search was performed over the graph using the depth first search algorithm. In a memory efficient alternative NoSQL approach, we created a collection of Refseq sequences in MongoDB database using tax-id and path of FASTA files. We queried the MongoDB collection for the target and non-target sequences. In both the approaches, we used an alignment free sliding window k-mer-based procedure that quickly compares k-mers of target and non-target sequences and returns unique sequences that are not present in the non-target. We have validated our algorithm with target nodes Mycobacterium tuberculosis, Neisseria gonorrhoeae, and Monkeypox and generated unique sequences. This universal algorithm is a powerful tool for generating diagnostic sequences, enabling the accurate identification of microbial strains with high phylogenetic precision.
Collapse
Affiliation(s)
- Gulshan Kumar Sharma
- Malaviya National Institute of Technology, Jawahar Lal Nehru Marg, Jhalana Gram, Malviya Nagar, Jaipur, Rajasthan 302017, India
| | - Rakesh Sharma
- Centre for Converging Technologies, University of Rajasthan, Jawahar Lal Nehru Marg, Talvandi, Jaipur, Rajasthan 302004, India
| | - Kavita Joshi
- Department of Zoology, University of Rajasthan, Jawahar Lal Nehru Marg, Talvandi, Jaipur, Rajasthan 302004, India
| | - Sameer Qureshi
- Department of Zoology, University of Rajasthan, Jawahar Lal Nehru Marg, Talvandi, Jaipur, Rajasthan 302004, India
| | - Shubhita Mathur
- Department of Zoology, University of Rajasthan, Jawahar Lal Nehru Marg, Talvandi, Jaipur, Rajasthan 302004, India
| | - Sharad Sinha
- Department of Mathematics, University of Rajasthan, Jawahar Lal Nehru Marg, Talvandi, Jaipur, Rajasthan 302004, India
| | - Samit Chatterjee
- Department of Zoology, University of Rajasthan, Jawahar Lal Nehru Marg, Talvandi, Jaipur, Rajasthan 302004, India
| | - Vandana Nunia
- Department of Zoology, University of Rajasthan, Jawahar Lal Nehru Marg, Talvandi, Jaipur, Rajasthan 302004, India
| |
Collapse
|
78
|
Desai M, Sun B. Protein Visualizer 2.0: Intuitive and Interactive Visualization of Protein Topology and Co/Post-Translational Modifications. ACS Chem Biol 2024; 19:1930-1934. [PMID: 39250597 DOI: 10.1021/acschembio.4c00485] [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: 09/11/2024]
Abstract
Topology and post-translational modifications (PTMs) are critical features in studying the structures and functions of proteins. Several popular and detailed protein visualization algorithms are available for such studies, such as PyMOL, NCBI's iCn3D, UniProt PTM viewing, and Protter. However, none of the studies depict the special relationships between key structural features such as N-glycosylation, disulfide bonds, potential N-glycosylation sites (free sequons), potential disulfide bonds (free cysteine sites), and protein topology in a way conducive to detailed analyses. We introduce Protein Visualizer 2.0 (https://sfu-sun-lab.github.io/protein-visualizer-2.0/), a web-based tool that visualizes these features and the topology of all human proteins. This tool allows users to readily assess potential conflicts among predicted protein topology and known co/post-translational modifications. This tool also helps to reveal hidden relationships among displayed structural features.
Collapse
Affiliation(s)
- Manthan Desai
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
- Department of Computing Science, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Bingyun Sun
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| |
Collapse
|
79
|
Nadel CM, Pokhrel S, Wucherer K, Oehler A, Thwin AC, Basu K, Callahan MD, Southworth DR, Mordes DA, Craik CS, Gestwicki JE. Phosphorylation of tau at a single residue inhibits binding to the E3 ubiquitin ligase, CHIP. Nat Commun 2024; 15:7972. [PMID: 39266525 PMCID: PMC11393453 DOI: 10.1038/s41467-024-52075-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 08/23/2024] [Indexed: 09/14/2024] Open
Abstract
Microtubule-associated protein tau (MAPT/tau) accumulates in a family of neurodegenerative diseases, including Alzheimer's disease (AD). In disease, tau is aberrantly modified by post-translational modifications (PTMs), including hyper-phosphorylation. However, it is often unclear which of these PTMs contribute to tau's accumulation or what mechanisms might be involved. To explore these questions, we focus on a cleaved proteoform of tau (tauC3), which selectively accumulates in AD and was recently shown to be degraded by its direct binding to the E3 ubiquitin ligase, CHIP. Here, we find that phosphorylation of tauC3 at a single residue, pS416, is sufficient to weaken its interaction with CHIP. A co-crystal structure of CHIP bound to the C-terminus of tauC3 reveals the mechanism of this clash, allowing design of a mutation (CHIPD134A) that partially restores binding and turnover of pS416 tauC3. We confirm that, in our models, pS416 is produced by the known AD-associated kinase, MARK2/Par-1b, providing a potential link to disease. In further support of this idea, an antibody against pS416 co-localizes with tauC3 in degenerative neurons within the hippocampus of AD patients. Together, these studies suggest a molecular mechanism for how phosphorylation at a discrete site contributes to accumulation of a tau proteoform.
Collapse
Affiliation(s)
- Cory M Nadel
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, 94158, USA
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Saugat Pokhrel
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, 94158, USA
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Kristin Wucherer
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Abby Oehler
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Aye C Thwin
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, 94158, USA
- Department of Biochemistry & Biophysics, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Koli Basu
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Matthew D Callahan
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, 94158, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Daniel R Southworth
- Institute for Neurodegenerative Diseases, University of California San Francisco, San Francisco, CA, 94158, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Daniel A Mordes
- Department of Biochemistry & Biophysics, University of California San Francisco, San Francisco, CA, 94158, USA
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Charles S Craik
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Jason E Gestwicki
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, 94158, USA.
- Department of Pathology, University of California San Francisco, San Francisco, CA, 94158, USA.
| |
Collapse
|
80
|
Maurya P, Kumar M, Jain R, Thaer Abdulhameed Almuqdadi H, Singh H, Gupta A, Arenz C, Gaur NA, Singh S. Expression of Plasmodium major facilitator superfamily protein in transporters - Δ Candida identifies a drug transporter. Future Microbiol 2024; 19:1293-1307. [PMID: 39235058 PMCID: PMC11485967 DOI: 10.1080/17460913.2024.2389750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
Aim: To assess the functional relevance of a putative Major Facilitator Superfamily protein (PF3D7_0210300; 'PfMFSDT') as a drug transporter, using Candida glabrata for orthologous protein expression.Methods: Complementary Determining Sequence encoding PfMFSDT was integrated into the genome of genetically engineered C. glabrata strain MSY8 via homologous recombination, followed by assessing its functional relevance as a drug transporter.Results & conclusion: The modified C. glabrata strain exhibited plasma membrane localization of PfMFSDT and characteristics of an Major Facilitator Superfamily transporter, conferring resistance to antifungals, ketoconazole and itraconazole. The nanomolar inhibitory effects of the drugs on the intra-erythrocytic growth of Plasmodium falciparum highlight their antimalarial properties. This study proposes PfMFSDT as a drug transporter, expanding the repertoire of the currently known antimalarial 'resistome'.
Collapse
Affiliation(s)
- Preeti Maurya
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Mohit Kumar
- Yeast Biofuel Group, International Centre for Genetic Engineering & Biotechnology, New Delhi, 110067, India
| | - Ravi Jain
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Haider Thaer Abdulhameed Almuqdadi
- Medicinal Chemistry Laboratory, Department of Bioscience, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
- Department of Chemistry, College of Science, Al-Nahrain University, Baghdad, Iraq
| | - Harshita Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
- Institut für Chemie, Humboldt-Universität zu Berlin, Berlin, 12489, Germany
| | - Aashima Gupta
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Christoph Arenz
- Institut für Chemie, Humboldt-Universität zu Berlin, Berlin, 12489, Germany
| | - Naseem A Gaur
- Yeast Biofuel Group, International Centre for Genetic Engineering & Biotechnology, New Delhi, 110067, India
| | - Shailja Singh
- Special Centre for Molecular Medicine, Jawaharlal Nehru University, New Delhi, 110067, India
| |
Collapse
|
81
|
Twort VG, Laine VN, Field KA, Whiting-Fawcett F, Ito F, Reiman M, Bartonicka T, Fritze M, Ilyukha VA, Belkin VV, Khizhkin EA, Reeder DM, Fukui D, Jiang TL, Lilley TM. Signals of positive selection in genomes of palearctic Myotis-bats coexisting with a fungal pathogen. BMC Genomics 2024; 25:828. [PMID: 39227786 PMCID: PMC11370307 DOI: 10.1186/s12864-024-10722-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 08/19/2024] [Indexed: 09/05/2024] Open
Abstract
Disease can act as a driving force in shaping genetic makeup across populations, even species, if the impacts influence a particularly sensitive part of their life cycles. White-nose disease is caused by a fungal pathogen infecting bats during hibernation. The mycosis has caused massive population declines of susceptible species in North America, particularly in the genus Myotis. However, Myotis bats appear to tolerate infection in Eurasia, where the fungal pathogen has co-evolved with its bat hosts for an extended period of time. Therefore, with susceptible and tolerant populations, the fungal disease provides a unique opportunity to tease apart factors contributing to tolerance at a genomic level to and gain an understanding of the evolution of non-harmful in host-parasite interactions. To investigate if the fungal disease has caused adaptation on a genomic level in Eurasian bat species, we adopted both whole-genome sequencing approaches and a literature search to compile a set of 300 genes from which to investigate signals of positive selection in genomes of 11 Eurasian bats at the codon-level. Our results indicate significant positive selection in 38 genes, many of which have a marked role in responses to infection. Our findings suggest that white-nose syndrome may have applied a significant selective pressure on Eurasian Myotis-bats in the past, which can contribute their survival in co-existence with the pathogen. Our findings provide an insight on the selective pressure pathogens afflict on their hosts using methodology that can be adapted to other host-pathogen study systems.
Collapse
Affiliation(s)
- V G Twort
- Finnish Museum of Natural History, BatLab Finland, University of Helsinki, Helsinki, Finland
| | - V N Laine
- Finnish Museum of Natural History, BatLab Finland, University of Helsinki, Helsinki, Finland
| | - K A Field
- Department of Biology, Bucknell University, Lewisburg, PA, USA
| | - F Whiting-Fawcett
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - F Ito
- Finnish Museum of Natural History, BatLab Finland, University of Helsinki, Helsinki, Finland
| | - M Reiman
- Finnish Museum of Natural History, BatLab Finland, University of Helsinki, Helsinki, Finland
| | - T Bartonicka
- Dept. Botany and Zoology, Faculty of Science, Masaryk University, Kotlarska 2, Brno, 611 37, Czech Republic
| | - M Fritze
- Zoological Institute and Museum, University of Greifswald, Greifswald, Germany
- German Bat Observatory, Berlin, Germany
- Competence Center for Bat Conservation Saxony Anhalt, in the South Harz Karst Landscape Biosphere Reserve, Südharz, Germany
| | - V A Ilyukha
- Papanin Institute for Biology of Inland Waters, Russian Academy of Sciences, Borok, Russia
| | - V V Belkin
- Institute of Biology, Karelian Research Centre, Russian Academy of Sciences, Petrozavodsk, Russia
| | - E A Khizhkin
- Institute of Biology, Karelian Research Centre, Russian Academy of Sciences, Petrozavodsk, Russia
| | - D M Reeder
- Department of Biology, Bucknell University, Lewisburg, PA, USA
| | - D Fukui
- Graduate School of Agricultural and Life Sciences, The University of Tokyo Fuji Iyashinomori Woodland Study Center, The University of Tokyo, Yamanakako, Japan
| | - T L Jiang
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China
| | - T M Lilley
- Finnish Museum of Natural History, BatLab Finland, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
82
|
İnak E, De Rouck S, Koç-İnak N, Erdem E, Rüstemoğlu M, Dermauw W, Van Leeuwen T. Identification and CRISPR-Cas9 validation of a novel β-adrenergic-like octopamine receptor mutation associated with amitraz resistance in Varroa destructor. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2024; 204:106080. [PMID: 39277393 DOI: 10.1016/j.pestbp.2024.106080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/07/2024] [Indexed: 09/17/2024]
Abstract
Varroa destructor is widely recognized as a significant contributor to colony collapse disorder. Chemical acaricides, such as amitraz, have been extensively used for Varroa control due to their selectivity within beehives. However, the increasing number of cases of amitraz resistance across global V. destructor populations poses a significant challenge. In this study, we conducted a comprehensive molecular screening of the β-adrenergic-like octopamine receptor (Octβ2R), the target-site of amitraz, across 66 Turkish and 63 Belgian V. destructor populations. Although previously reported amitraz resistance mutations were not detected, the screening revealed a novel Y337F mutation located within transmembrane 7 (TM7) of Octβ2R in Turkish Varroa populations. Notably, this mutation was identified in the last residue of the highly conserved NPxxY motif associated with the activation of G-protein coupled receptors (GPCR). Among the 66 Varroa samples from Türkiye, twenty harbored the Y337F mutation, with eight samples exhibiting fixation of the mutation. Subsequent bioassays revealed over 8-fold resistance to amitraz in populations that contain the Y337F mutation. Genotyping of mites after exposure to 10 mg a.i./l amitraz demonstrated that all surviving mites were homozygous for the Y337F mutation, whereas dead mites carried susceptible alleles, providing genetic linkage between mutation and phenotype. Further, we used CRISPR-Cas9 editing to introduce the Y337F mutation in the orthologous Octβ2R of the model organism Tetranychus urticae. Crispants exhibited over threefold resistance to amitraz. In conclusion, this study identified and validated a novel amitraz resistance mutation. Additional research is required to further evaluate the phenotypic strength of Y337F in the context of operational resistance with current treatment strategies.
Collapse
Affiliation(s)
- Emre İnak
- Laboratory of Agrozoology, Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium; Department of Plant Protection, Faculty of Agriculture, Ankara University, Diskapi, 06110, Ankara, Türkiye.
| | - Sander De Rouck
- Laboratory of Agrozoology, Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium.
| | - Nafiye Koç-İnak
- Department of Parasitology, Faculty of Veterinary Medicine, Ankara University, Altindag, 06070, Ankara, Türkiye.
| | - Esengül Erdem
- Plant Protection Department, Faculty of Agriculture, Şırnak University, Şırnak, Türkiye.
| | - Mustafa Rüstemoğlu
- Plant Protection Department, Faculty of Agriculture, Şırnak University, Şırnak, Türkiye
| | - Wannes Dermauw
- Laboratory of Agrozoology, Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium.
| | - Thomas Van Leeuwen
- Laboratory of Agrozoology, Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium.
| |
Collapse
|
83
|
Dulon D, de Monvel JB, Plion B, Mallet A, Petit C, Condamine S, Bouleau Y, Safieddine S. A free intravesicular C-terminal of otoferlin is essential for synaptic vesicle docking and fusion at auditory inner hair cell ribbon synapses. Prog Neurobiol 2024; 240:102658. [PMID: 39103114 DOI: 10.1016/j.pneurobio.2024.102658] [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: 03/21/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 08/07/2024]
Abstract
Our understanding of how otoferlin, the major calcium sensor in inner hair cells (IHCs) synaptic transmission, contributes to the overall dynamics of synaptic vesicle (SV) trafficking remains limited. To address this question, we generated a knock-in mouse model expressing an otoferlin-GFP protein, where GFP was fused to its C-terminal transmembrane domain. Similar to the wild type protein, the GFP-tagged otoferlin showed normal expression and was associated with IHC SV. Surprisingly, while the heterozygote Otof+/GFP mice exhibited a normal hearing function, homozygote OtofGFP/GFP mice were profoundly deaf attributed to severe reduction in SV exocytosis. Fluorescence recovery after photobleaching revealed a markedly increased mobile fraction of the otof-GFP-associated SV in Otof GFP/GFP IHCs. Correspondingly, 3D-electron tomographic of the ribbon synapses indicated a reduced density of SV attached to the ribbon active zone. Collectively, these results indicate that otoferlin requires a free intravesicular C-terminal end for normal SV docking and fusion.
Collapse
Affiliation(s)
- Didier Dulon
- Institut Pasteur, Université Paris Cité, Inserm U06, Institut de l'Audition, Paris, France; Bordeaux Neurocampus, Université de Bordeaux, Bordeaux 33076, France.
| | | | - Baptiste Plion
- Institut Pasteur, Université Paris Cité, Inserm U06, Institut de l'Audition, Paris, France
| | - Adeline Mallet
- Institut Pasteur, Université Paris Cité, Inserm U06, Institut de l'Audition, Paris, France
| | - Christine Petit
- Institut Pasteur, Université Paris Cité, Inserm U06, Institut de l'Audition, Paris, France
| | - Steven Condamine
- Institut Pasteur, Université Paris Cité, Inserm U06, Institut de l'Audition, Paris, France; Bordeaux Neurocampus, Université de Bordeaux, Bordeaux 33076, France
| | - Yohan Bouleau
- Institut Pasteur, Université Paris Cité, Inserm U06, Institut de l'Audition, Paris, France; Bordeaux Neurocampus, Université de Bordeaux, Bordeaux 33076, France
| | - Saaid Safieddine
- Institut Pasteur, Université Paris Cité, Inserm U06, Institut de l'Audition, Paris, France; Centre National de la Recherche Scientifique, Paris, France.
| |
Collapse
|
84
|
Durante-Rodríguez G, de Francisco-Polanco S, García JL, Díaz E. Characterization of a MHYT domain-coupled transcriptional regulator that responds to carbon monoxide. Nucleic Acids Res 2024; 52:8849-8860. [PMID: 38966994 PMCID: PMC11347149 DOI: 10.1093/nar/gkae575] [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: 03/19/2024] [Revised: 06/11/2024] [Accepted: 06/25/2024] [Indexed: 07/06/2024] Open
Abstract
The MHYT domain, identified over two decades ago for its potential to detect diatomic gases like CO, O2 or NO, has awaited experimental validation as a protein sensory domain. Here, we characterize the MHYT domain-containing transcriptional regulator CoxC, which governs the expression of the cox genes responsible for aerobic CO oxidation in the carboxidotrophic bacterium Afipia carboxidovorans OM5. The C-terminal LytTR-type DNA-binding domain of CoxC binds to an operator region consisting of three direct repeats sequences overlapping the -35 box at the target PcoxB promoter, which is consistent with the role of CoxC as a specific transcriptional repressor of the cox genes. Notably, the N-terminal transmembrane MHYT domain endows CoxC with the ability to sense CO as an effector molecule, as demonstrated by the relief of CoxC-mediated repression and binding to the PcoxB promoter upon CO exposure. Furthermore, copper serves as the essential divalent cation for the interaction of CO with CoxC, thereby confirming previous hypothesis regarding the role of copper in the gas-sensing mechanism of MHYT domains. CoxC represents the prototype of a novel subfamily of single-component LytTR transcriptional regulators, characterized by the fusion of a DNA-binding domain with a membrane-bound MHYT sensor domain.
Collapse
Affiliation(s)
- Gonzalo Durante-Rodríguez
- Department of Microbial and Plant Biotechnology, Centro de Investigaciones Biológicas Margarita Salas-CSIC, Calle Ramiro de Maeztu, 9, 28040 Madrid. Spain
| | - Sofía de Francisco-Polanco
- Department of Microbial and Plant Biotechnology, Centro de Investigaciones Biológicas Margarita Salas-CSIC, Calle Ramiro de Maeztu, 9, 28040 Madrid. Spain
| | - José Luis García
- Department of Microbial and Plant Biotechnology, Centro de Investigaciones Biológicas Margarita Salas-CSIC, Calle Ramiro de Maeztu, 9, 28040 Madrid. Spain
| | - Eduardo Díaz
- Department of Microbial and Plant Biotechnology, Centro de Investigaciones Biológicas Margarita Salas-CSIC, Calle Ramiro de Maeztu, 9, 28040 Madrid. Spain
| |
Collapse
|
85
|
Zeng W, Wang Y, Gao R, Wen H, Yu M. Unlocking the Reverse Targeting Mechanisms of Cannabidiol: Unveiling New Therapeutic Avenues. J Med Chem 2024; 67:14574-14585. [PMID: 39092992 DOI: 10.1021/acs.jmedchem.4c01353] [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: 08/04/2024]
Abstract
Cannabidiol (CBD) and Δ9-tetrahydrocannabinol (THC), the main components of Cannabis sativa plants, have attracted a significant amount of attention due to their biological activities. This study identified GPR18 as the target of partial agonist CBD activating the p42/p44 MAPK pathway leading to migration of endometrial epithelial cells. Induced fit docking (IFD) showed that the affinity of THC for GPR18 is higher than that of CBD, and molecular dynamics (MD) simulations showed that CBD-GPR18 complexes at 130/200 ns might have stable conformations, potentially activating GPR18 by changing the distances of key residues in its active pocket. In contrast, THC maintains "metastable" conformations, generating a "shrinking space" leading to full agonism of THC by adding mechanical constraints in GPR18's active pocket. Steered molecular dynamics (SMD) revealed GPR18's active pocket was influenced more by CBD's partial agonism compared with THC. This combined IFD-MD-SMD method may be used to explain the mechanism of activation of partial or full agonists of GPR18.
Collapse
Affiliation(s)
- Wen Zeng
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
- Key Laboratory of Medical Molecule Science and Pharmaceutical Engineering, Yangtze Delta Region Academy of Beijing Institute of Technology, Jiaxing 314019, China
| | - Yifei Wang
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Rui Gao
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| | - Hongliang Wen
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
- Key Laboratory of Medical Molecule Science and Pharmaceutical Engineering, Yangtze Delta Region Academy of Beijing Institute of Technology, Jiaxing 314019, China
| | - Mingjia Yu
- School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
86
|
Moreira D, Blaz J, Kim E, Eme L. A gene-rich mitochondrion with a unique ancestral protein transport system. Curr Biol 2024; 34:3812-3819.e3. [PMID: 39084221 DOI: 10.1016/j.cub.2024.07.017] [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: 11/26/2023] [Revised: 05/03/2024] [Accepted: 07/02/2024] [Indexed: 08/02/2024]
Abstract
Mitochondria originated from an ancient endosymbiosis involving an alphaproteobacterium.1,2,3 Over time, these organelles reduced their gene content massively, with most genes being transferred to the host nucleus before the last eukaryotic common ancestor (LECA).4 This process has yielded varying gene compositions in modern mitogenomes, including the complete loss of this organellar genome in some extreme cases.5,6,7,8,9,10,11,12,13,14 At the other end of the spectrum, jakobids harbor the most gene-rich mitogenomes, encoding 60-66 proteins.8 Here, we introduce the mitogenome of Mantamonas sphyraenae, a protist from the deep-branching CRuMs supergroup.15,16 Remarkably, it boasts the most gene-rich mitogenome outside of jakobids, by housing 91 genes, including 62 protein-coding ones. These include rare homologs of the four subunits of the bacterial-type cytochrome c maturation system I (CcmA, CcmB, CcmC, and CcmF) alongside a unique ribosomal protein S6. During the early evolution of mitochondria, gene transfer from the proto-mitochondrial endosymbiont to the nucleus became possible thanks to systems facilitating the transport of proteins synthesized in the host cytoplasm back to the mitochondrion. In addition to the universally found eukaryotic protein import systems, jakobid mitogenomes were reported to uniquely encode the SecY transmembrane protein of the Sec general secretory pathway, whose evolutionary origin was however unclear. The Mantamonas mitogenome not only encodes SecY but also SecA, SecE, and SecG, making it the sole eukaryote known to house a complete mitochondrial Sec translocation system. Furthermore, our phylogenetic and comparative genomic analyses provide compelling evidence for the alphaproteobacterial origin of this system, establishing its presence in LECA.
Collapse
Affiliation(s)
- David Moreira
- Unité d'Ecologie Systématique et Evolution, CNRS, Université Paris-Saclay, AgroParisTech, 91190 Gif-sur-Yvette, France.
| | - Jazmin Blaz
- Unité d'Ecologie Systématique et Evolution, CNRS, Université Paris-Saclay, AgroParisTech, 91190 Gif-sur-Yvette, France
| | - Eunsoo Kim
- Division of EcoScience, Ewha Womans University, Seoul, South Korea; Division of Invertebrate Zoology, American Museum of Natural History, New York, NY, USA
| | - Laura Eme
- Unité d'Ecologie Systématique et Evolution, CNRS, Université Paris-Saclay, AgroParisTech, 91190 Gif-sur-Yvette, France.
| |
Collapse
|
87
|
Ramos M, Martín-García R, Curto MÁ, Gómez-Delgado L, Moreno MB, Sato M, Portales E, Osumi M, Rincón SA, Pérez P, Ribas JC, Cortés JC. Fission yeast Bgs1 glucan synthase participates in the control of growth polarity and membrane traffic. iScience 2024; 27:110477. [PMID: 39156640 PMCID: PMC11326927 DOI: 10.1016/j.isci.2024.110477] [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: 01/25/2024] [Revised: 02/14/2024] [Accepted: 07/05/2024] [Indexed: 08/20/2024] Open
Abstract
Rod-shaped fission yeast grows through cell wall expansion at poles and septum, synthesized by essential glucan synthases. Bgs1 synthesizes the linear β(1,3)glucan of primary septum at cytokinesis. Linear β(1,3)glucan is also present in the wall poles, suggesting additional Bgs1 roles in growth polarity. Our study reveals an essential collaboration between Bgs1 and Tea1-Tea4, but not other polarity factors, in controlling growth polarity. Simultaneous absence of Bgs1 function and Tea1-Tea4 causes complete loss of growth polarity, spread of other glucan synthases, and spherical cell formation, indicating this defect is specifically due to linear β(1,3)glucan absence. Furthermore, linear β(1,3)glucan absence induces actin patches delocalization and sterols spread, which are ultimately responsible for the growth polarity loss without Tea1-Tea4. This suggests strong similarities in Bgs1 functions controlling actin structures during cytokinesis and polarized growth. Collectively, our findings unveil that cell wall β(1,3)glucan regulates polarized growth, like the equivalent extracellular matrix in neuronal cells.
Collapse
Affiliation(s)
- Mariona Ramos
- Instituto de Biología Funcional y Genómica, Consejo Superior de Investigaciones Científicas (CSIC) and Universidad de Salamanca, Salamanca, Spain
| | - Rebeca Martín-García
- Instituto de Biología Funcional y Genómica, Consejo Superior de Investigaciones Científicas (CSIC) and Universidad de Salamanca, Salamanca, Spain
| | - M. Ángeles Curto
- Instituto de Biología Funcional y Genómica, Consejo Superior de Investigaciones Científicas (CSIC) and Universidad de Salamanca, Salamanca, Spain
| | - Laura Gómez-Delgado
- Instituto de Biología Funcional y Genómica, Consejo Superior de Investigaciones Científicas (CSIC) and Universidad de Salamanca, Salamanca, Spain
| | - M. Belén Moreno
- Instituto de Biología Funcional y Genómica, Consejo Superior de Investigaciones Científicas (CSIC) and Universidad de Salamanca, Salamanca, Spain
| | - Mamiko Sato
- Laboratory of Electron Microscopy and Bio-imaging Center, Japan Women’s University, 2-8-1 Mejirodai, Bunkyo-ku, Tokyo, Japan
| | - Elvira Portales
- Instituto de Biología Funcional y Genómica, Consejo Superior de Investigaciones Científicas (CSIC) and Universidad de Salamanca, Salamanca, Spain
| | - Masako Osumi
- Laboratory of Electron Microscopy and Bio-imaging Center, Japan Women’s University, 2-8-1 Mejirodai, Bunkyo-ku, Tokyo, Japan
- Integrated Imaging Research Support (IIRS), Villa Royal Hirakawa 103, 1-7-5 Hirakawa-cho, Chiyoda-ku, Tokyo, Japan
| | - Sergio A. Rincón
- Instituto de Biología Funcional y Genómica, Consejo Superior de Investigaciones Científicas (CSIC) and Universidad de Salamanca, Salamanca, Spain
| | - Pilar Pérez
- Instituto de Biología Funcional y Genómica, Consejo Superior de Investigaciones Científicas (CSIC) and Universidad de Salamanca, Salamanca, Spain
| | - Juan C. Ribas
- Instituto de Biología Funcional y Genómica, Consejo Superior de Investigaciones Científicas (CSIC) and Universidad de Salamanca, Salamanca, Spain
| | - Juan C.G. Cortés
- Instituto de Biología Funcional y Genómica, Consejo Superior de Investigaciones Científicas (CSIC) and Universidad de Salamanca, Salamanca, Spain
| |
Collapse
|
88
|
Voges TS, Lim EB, MacKenzie A, Mudler K, DeSouza R, Onyejekwe NE, Johnston SD. Phenotypic assessment of Cox10 variants and their implications for Leigh Syndrome. BMC Res Notes 2024; 17:228. [PMID: 39152498 PMCID: PMC11328382 DOI: 10.1186/s13104-024-06879-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 07/30/2024] [Indexed: 08/19/2024] Open
Abstract
OBJECTIVES Cox10 is an enzyme required for the activity of cytochrome c oxidase. Humans who lack at least one functional copy of Cox10 have a form of Leigh Syndrome, a genetic disease that is usually fatal in infancy. As more human genomes are sequenced, new alleles are being discovered; whether or not these alleles encode functional proteins remains unclear. Thus, we set out to measure the phenotypes of many human Cox10 variants by expressing them in yeast cells. RESULTS We successfully expressed the reference sequence and 25 variants of human Cox10 in yeast. We quantitated the ability of these variants to support growth on nonfermentable media and directly measured cytochrome c oxidase activity. 11 of these Cox10 variants supported approximately half or more the cytochrome c oxidase activity compared to the reference sequence. All of the strains containing those 11 variants also grew robustly using a nonfermentable carbon source. Cells expressing the other variants showed low cytochrome c oxidase activity and failed to grow on nonfermentable media.
Collapse
Affiliation(s)
- Thomas-Shadi Voges
- Department of Biology, North Central College, Naperville, IL, USA
- Department of Physiology and Biophysics, University of Illinois, Chicago, Chicago, IL, USA
| | - Eun Bi Lim
- Department of Biology, North Central College, Naperville, IL, USA
- Department of Microbiology and Immunology, Loyola University of Chicago, Maywood, IL, USA
| | | | - Kyle Mudler
- Department of Biology, North Central College, Naperville, IL, USA
| | - Rebecca DeSouza
- Department of Biology, North Central College, Naperville, IL, USA
| | | | | |
Collapse
|
89
|
Sailer AL, Jevtic Z, Stoll B, Wörtz J, Sharma K, Urlaub H, Dyall-Smith M, Pfeiffer F, Marchfelder A, Lenz C. Iron starvation results in up-regulation of a probable Haloferax volcanii siderophore transporter. Front Microbiol 2024; 15:1422844. [PMID: 39206359 PMCID: PMC11349517 DOI: 10.3389/fmicb.2024.1422844] [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: 04/24/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
The response of the haloarchaeal model organism Haloferax volcanii to iron starvation was analyzed at the proteome level by data-independent acquisition mass spectrometry. Cells grown in minimal medium with normal iron levels were compared to those grown under low iron conditions, with samples being separated into membrane and cytoplasmic fractions in order to focus on import/export processes which are frequently associated with metal homeostasis. Iron starvation not only caused a severe retardation of growth but also altered the levels of many proteins. Using a comprehensive annotated spectral library and data-independent acquisition mass spectrometry (DIA-MS), we found that iron starvation resulted in significant changes to both the membrane and the soluble proteomes of Hfx. volcanii. The most affected protein is the RND family permease HVO_A0467, which is 44-fold enriched in cells grown under iron starvation. The gene HVO_A0467 can be deleted suggesting that it is not essential under standard conditions. Compared to wild type cells the deletion strain shows only slight changes in growth and cell morphologies show no differences. Molecular docking predictions indicated that HVO_A0467 may be an exporter of the siderophore schizokinen for which a potential biosynthesis cluster is encoded in the Hfx. volcanii genome. Together, these findings confirm the importance of iron for archaeal cells and suggest HVO_0467 as a siderophore exporter.
Collapse
Affiliation(s)
| | - Zivojin Jevtic
- Department of Biomedicine, University Children’s Hospital, University of Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | | | - Kundan Sharma
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Bioanalytics Group, Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Mike Dyall-Smith
- Computational Systems Biochemistry, Max Planck Institute for Biochemistry, Martinsried, Germany
- Veterinary Biosciences, Faculty of Science, Melbourne Veterinary School, University of Melbourne, Parkville, VIC, Australia
| | - Friedhelm Pfeiffer
- Biology II, Ulm University, Ulm, Germany
- Computational Systems Biochemistry, Max Planck Institute for Biochemistry, Martinsried, Germany
| | | | - Christof Lenz
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Bioanalytics Group, Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
90
|
Chang G, Aroge FA, Venkateshappa R, Claydon TW, Sun B. Development of an Absolute Quantification Method for hERG Using PRM with Single Isotopologue in-Sample Calibration. ACS OMEGA 2024; 9:33972-33982. [PMID: 39130540 PMCID: PMC11308013 DOI: 10.1021/acsomega.4c04541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 08/13/2024]
Abstract
The human ether-à-go-go-related gene (KCNH2)-encoded protein hERG constitutes the α subunit of the Kv11.1 channel and contributes to the I kr current, which plays an important role in the cardiac action potential. Genetically and xenobiotically triggered malfunctions of hERG can cause arrhythmia. The expression of hERG in various study systems was assessed mainly as the fold change relative to the corresponding control. Here, we developed a simple and sensitive quantitation method using targeted mass spectrometry, i.e., the parallel reaction monitoring approach, to measure the absolute quantity of hERG in copy number. Such measurements do not require controls, and the obtained values can be compared with similar results for any other protein. To effectively avoid matrix effects, we used the heavy-match-light (HML) in-sample calibration approach that requires only a single isotopologue to achieve copy-number quantitation. No significant difference was observed in the results obtained by HML and by the classic standard addition in-sample calibration approach. Using four proteotypic peptides, we quantified the average number of copies of hERG in the HEK293T heterologous expression system as 3.6 ± 0.5 × 106 copies/cell, i.e., 1 million copies/cell for the fully assembled Kv11.1 channel.
Collapse
Affiliation(s)
- Ge Chang
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A1S6, Canada
| | - Fabusuyi A. Aroge
- School
of Mechatronic Systems Engineering, Simon
Fraser University, Surrey, British Columbia V3T0A3, Canada
| | - Ravichandra Venkateshappa
- Department
of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia V5A1S6, Canada
| | - Tom W. Claydon
- Department
of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia V5A1S6, Canada
| | - Bingyun Sun
- Department
of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A1S6, Canada
| |
Collapse
|
91
|
Berg Luecke L, Mesidor R, Littrell J, Carpenter M, Wojtkiewicz M, Gundry RL. Veneer Is a Webtool for Rapid, Standardized, and Transparent Interpretation, Annotation, and Reporting of Mammalian Cell Surface N-Glycocapture Data. J Proteome Res 2024; 23:3235-3248. [PMID: 38412263 PMCID: PMC11301670 DOI: 10.1021/acs.jproteome.3c00800] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 01/23/2024] [Accepted: 02/12/2024] [Indexed: 02/29/2024]
Abstract
Currently, no consensus exists regarding criteria required to designate a protein within a proteomic data set as a cell surface protein. Most published proteomic studies rely on varied ontology annotations or computational predictions instead of experimental evidence when attributing protein localization. Consequently, standardized approaches for analyzing and reporting cell surface proteome data sets would increase confidence in localization claims and promote data use by other researchers. Recently, we developed Veneer, a web-based bioinformatic tool that analyzes results from cell surface N-glycocapture workflows─the most popular cell surface proteomics method used to date that generates experimental evidence of subcellular location. Veneer assigns protein localization based on defined experimental and bioinformatic evidence. In this study, we updated the criteria and process for assigning protein localization and added new functionality to Veneer. Results of Veneer analysis of 587 cell surface N-glycocapture data sets from 32 published studies demonstrate the importance of applying defined criteria when analyzing cell surface proteomics data sets and exemplify how Veneer can be used to assess experimental quality and facilitate data extraction for informing future biological studies and annotating public repositories.
Collapse
Affiliation(s)
- Linda Berg Luecke
- CardiOmics
Program, Center for Heart and Vascular Research and Department of
Cellular and Integrative Physiology, University
of Nebraska Medical Center, Omaha, Nebraska 68198, United States
- Department
of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Roneldine Mesidor
- CardiOmics
Program, Center for Heart and Vascular Research and Department of
Cellular and Integrative Physiology, University
of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Jack Littrell
- CardiOmics
Program, Center for Heart and Vascular Research and Department of
Cellular and Integrative Physiology, University
of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Morgan Carpenter
- CardiOmics
Program, Center for Heart and Vascular Research and Department of
Cellular and Integrative Physiology, University
of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Melinda Wojtkiewicz
- CardiOmics
Program, Center for Heart and Vascular Research and Department of
Cellular and Integrative Physiology, University
of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| | - Rebekah L. Gundry
- CardiOmics
Program, Center for Heart and Vascular Research and Department of
Cellular and Integrative Physiology, University
of Nebraska Medical Center, Omaha, Nebraska 68198, United States
| |
Collapse
|
92
|
Tamayo-Trujillo R, Paz-Cruz E, Cadena-Ullauri S, Guevara-Ramirez P, Ruiz-Pozo VA, Ibarra-Castillo R, Laso-Bayas JL, Zambrano AK. Exploring Atrial Fibrillation: Understanding the Complex Relation Between Lifestyle and Genetic Factors. J Med Cases 2024; 15:186-194. [PMID: 39091575 PMCID: PMC11287905 DOI: 10.14740/jmc4250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 06/21/2024] [Indexed: 08/04/2024] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide across diverse ethnic groups. Among these, atrial fibrillation (AF) stands as one of the most prevalent types of arrhythmias and the primary cause of stroke. Risk factors associated with AF include alcohol consumption, aging, high blood pressure, hypertension, inflammation, and genetic factors. A family history of CVD could indicate an increased risk. Consequently, genetic, and genomic testing should be performed to identify the molecular etiology of CVDs and assess at-risk patients. It is important to note that CVDs are the results of the complex interplay of genes and environmental factors, including ethnicity. In this case, the proband's clinic story includes a history of smoking abuse for 10 years (10 cigarettes per day), obesity, hypertension, and an associated familial history. These risk factors, along with genetic variants, could trigger the early onset of AF. In recent years, genetic and genomic studies have significantly advanced our understanding of CVD etiology, given that next-generation sequencing (NGS) allows for the identification of genetic variants that could contribute to these pathologies. Furthermore, NGS facilitates early diagnosis, personalized pharmacological approaches, and identification of novel biomarkers. Thus, NGS is a valuable tool in CVD management. However, such studies are limited in Ecuador, a low- and middle-income country. Several challenges contribute to this gap, encompassing economic, infrastructural, and educational obstacles. Notably, the cost of genetic and genomic studies may also pose a barrier, restricting access to a portion of the population. In this case report, we present a 56-year-old Ecuadorian woman, who has been diagnosed with AF; however, after performing NGS no disease-associated variants were found, despite having strong clinical signs and symptoms. In summary, this case report contributes valuable insights into the complex interplay between genetic and lifestyle factors in the development and management of AF. The case report aims to underscore the potential impact of genetic variants on disease risk, even when classified as variants of uncertain significance, and the importance of an integral approach to patient care that includes genetic screening, lifestyle interventions, and tailored pharmacological treatment.
Collapse
Affiliation(s)
- Rafael Tamayo-Trujillo
- Centro de Investigacion Genetica y Genomica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
- These authors contributed equally to this work and share first authorship
| | - Elius Paz-Cruz
- Centro de Investigacion Genetica y Genomica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
- These authors contributed equally to this work and share first authorship
| | - Santiago Cadena-Ullauri
- Centro de Investigacion Genetica y Genomica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
- These authors contributed equally to this work and share first authorship
| | - Patricia Guevara-Ramirez
- Centro de Investigacion Genetica y Genomica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
- These authors contributed equally to this work and share first authorship
| | - Viviana A. Ruiz-Pozo
- Centro de Investigacion Genetica y Genomica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | | | | | - Ana Karina Zambrano
- Centro de Investigacion Genetica y Genomica, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| |
Collapse
|
93
|
Wang Z, Yang J, Yang L, Zhong Y, Wang P. Characteristics of a pseudolysogenic phage vB_YpM_HQ103 infecting Yersinia pestis. Virus Res 2024; 346:199395. [PMID: 38782263 PMCID: PMC11177050 DOI: 10.1016/j.virusres.2024.199395] [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: 01/15/2024] [Revised: 05/09/2024] [Accepted: 05/11/2024] [Indexed: 05/25/2024]
Abstract
The plague, caused by Yersinia pestis, is a natural focal disease and the presence of Y. pestis in the environment is a critical ecological concern worldwide. The role of Y. pestis phages in the ecological life cycle of the plague is crucial. Previously, a temperature-sensitive phage named vB_YpM_HQ103 was isolated from plague foci in Yunnan province, China. Upon infecting the EV76 strain of Y. pestis, vB_YpM_HQ103 exhibits lysogenic behavior at 21 °C and lytic behavior at 37 °C. Various methods including continuous passage lysogenic tests, in vitro lysis tests, comparative genomic assays, fluorescence quantitative PCR and receptor identification tests were employed to demonstrate that the lysogenic life cycle of this phage is applicable to wild Y. pestis strains; its lysogeny is pseudolysogenic (carrying but not integrating), allowing it to replicate and proliferate within Y. pestis. Furthermore, we have identified the outer membrane protein OmpA of Y. pestis as the receptor for phage infection. In conclusion, our research provides insight into the characteristics and receptors of a novel Y. pestis phage infection with a pseudolysogenic cycle. The findings of this study enhance our understanding of Y. pestis phages and plague microecology, offering valuable insights for future studies on the conservation and genetic evolution of Y. pestis in nature.
Collapse
Affiliation(s)
- Zijian Wang
- Yunnan Key Laboratory for Zoonosis Control and Prevention, Yunnan Institute for Endemic Disease Control and Prevention, Dali, 671000, China
| | - Jiao Yang
- Yunnan Key Laboratory for Zoonosis Control and Prevention, Yunnan Institute for Endemic Disease Control and Prevention, Dali, 671000, China
| | - Lihua Yang
- Yunnan Key Laboratory for Zoonosis Control and Prevention, Yunnan Institute for Endemic Disease Control and Prevention, Dali, 671000, China
| | - Youhong Zhong
- Yunnan Key Laboratory for Zoonosis Control and Prevention, Yunnan Institute for Endemic Disease Control and Prevention, Dali, 671000, China
| | - Peng Wang
- Yunnan Key Laboratory for Zoonosis Control and Prevention, Yunnan Institute for Endemic Disease Control and Prevention, Dali, 671000, China.
| |
Collapse
|
94
|
Ha K, Mundt-Machado N, Bisignano P, Pinedo A, Raleigh DR, Loeb G, Reiter JF, Cao E, Delling M. Cilia-enriched oxysterol 7β,27-DHC is required for polycystin ion channel activation. Nat Commun 2024; 15:6468. [PMID: 39085216 PMCID: PMC11291729 DOI: 10.1038/s41467-024-50318-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 06/28/2024] [Indexed: 08/02/2024] Open
Abstract
Polycystin-1 (PC-1) and PC-2 form a heteromeric ion channel complex that is abundantly expressed in primary cilia of renal epithelial cells. This complex functions as a non-selective cation channel, and mutations within the polycystin complex cause autosomal dominant polycystic kidney disease (ADPKD). The spatial and temporal regulation of the polycystin complex within the ciliary membrane remains poorly understood. Using both whole-cell and ciliary patch-clamp recordings, we identify a cilia-enriched oxysterol, 7β,27-dihydroxycholesterol (DHC), that serves as a necessary activator of the polycystin complex. We further identify an oxysterol-binding pocket within PC-2 and showed that mutations within this binding pocket disrupt 7β,27-DHC-dependent polycystin activation. Pharmacologic and genetic inhibition of oxysterol synthesis reduces channel activity in primary cilia. In summary, our findings reveal a regulator of the polycystin complex. This oxysterol-binding pocket in PC-2 may provide a specific target for potential ADPKD therapeutics.
Collapse
Affiliation(s)
- Kodaji Ha
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Nadine Mundt-Machado
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Paola Bisignano
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Aide Pinedo
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - David R Raleigh
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
| | - Gabriel Loeb
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Jeremy F Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Erhu Cao
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Markus Delling
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
95
|
Batista F, Moreira RS, Filho VB, Moura H, Wagner G, Miletti LC. Shotgun proteomics of detergent-solubilized proteins from Trypanosoma evansi. J Proteomics 2024; 304:105231. [PMID: 38906247 DOI: 10.1016/j.jprot.2024.105231] [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: 09/13/2023] [Revised: 05/28/2024] [Accepted: 06/18/2024] [Indexed: 06/23/2024]
Abstract
Trypanosoma evansi, the causative agent of surra, is the most prevalent pathogenic salivarian trypanosome and affects the majority of domesticated and wild animals in endemic regions. This work aimed to analyze detergent-solubilized T. evansi proteins and identify potential diagnostic biomarkers for surra. Triton X-114-extracted membrane-enriched proteins (MEP) of T. evansi bloodstream forms were analyzed using a gel-free technique (LC-ESI-MS/MS). 247 proteins were identified following the MS analysis of three biological and technical replicates. Two of these proteins were predicted to have a GPI-anchor, 100 (40%) were predicted to have transmembrane domains, and 166 (67%) were predicted to be membrane-bound based on at least one of six features: location (WolfPSORT, DeepLoc-2.0, Protcomp-9.0), transmembrane, GPI, and gene ontology. It was predicted that 76 (30%) of proteins had membrane evidence. Typical membrane proteins for each organelle were identified, among them ISG families (64, 65, and 75 kDa), flagellar calcium-binding protein, 24 kDa calflagin, syntaxins and oligosaccharyltransferase some of which had previously been studied in other trypanosomatids. T. evansi lacks singletons and exclusive orthologous groups, whereas three distinct epitopes have been identified. Data are available via ProteomeXchange with identifier PXD040594. SIGNIFICANCE: Trypanosoma evansi is a highly prevalent parasite that induces a pathological condition known as "surra" in various species of ungulates across five continents. The infection gives rise to symptoms that are not pathognomonic, thereby posing challenges in its diagnosis and leading to substantial economic losses in the livestock industry. A significant challenge arises from the absence of a diagnostic test capable of distinguishing between Trypanosoma equiperdum and T. evansi, both of which are implicated in equine diseases. Therefore, there is a pressing need to conduct research on the biochemistry of the parasite in order to identify proteins that could potentially serve as targets for differential diagnosis or therapeutic interventions.
Collapse
Affiliation(s)
- Franciane Batista
- Laboratório de Hemoparasitas e Vetores, Centro de Ciências Agroveterinárias (CAV), Universidade do Estado de Santa Catarina (UDESC), Av. Luís de Camões, 2090, Conta Dinheiro, Lages, SC 88520-000, Brazil
| | - Renato Simões Moreira
- Laboratório de Hemoparasitas e Vetores, Centro de Ciências Agroveterinárias (CAV), Universidade do Estado de Santa Catarina (UDESC), Av. Luís de Camões, 2090, Conta Dinheiro, Lages, SC 88520-000, Brazil; Instituto Federal de Santa Catarina (IFSC), Campus Gaspar, R. Adriano Kormann, 510 - Bela Vista, Gaspar, SC 89111-009, Brazil
| | - Vilmar Benetti Filho
- Laboratório de Bioinformática, Universidade Federal de Santa Catarina, Campus João David Ferreira Lima. Setor F, Bloco A, Sala 318. Caixa postal 476, Trindade, Florianópolis, SC 88040-970, Brazil
| | - Hércules Moura
- Biological Mass Spectrometry Laboratory, Centers for Disease Control and Prevention, Atlanta, USA
| | - Glauber Wagner
- Laboratório de Bioinformática, Universidade Federal de Santa Catarina, Campus João David Ferreira Lima. Setor F, Bloco A, Sala 318. Caixa postal 476, Trindade, Florianópolis, SC 88040-970, Brazil
| | - Luiz Claudio Miletti
- Laboratório de Hemoparasitas e Vetores, Centro de Ciências Agroveterinárias (CAV), Universidade do Estado de Santa Catarina (UDESC), Av. Luís de Camões, 2090, Conta Dinheiro, Lages, SC 88520-000, Brazil.
| |
Collapse
|
96
|
Chekesa B, Singh H, Gonzalez-Juarbe N, Vashee S, Wiscovitch-Russo R, Dupont CL, Girma M, Kerro O, Gumi B, Ameni G. Pangenome and genomic signatures linked to the dominance of the lineage-4 of Mycobacterium tuberculosis isolated from extrapulmonary tuberculosis patients in western Ethiopia. PLoS One 2024; 19:e0304060. [PMID: 39052555 PMCID: PMC11271921 DOI: 10.1371/journal.pone.0304060] [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: 03/06/2024] [Accepted: 05/06/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND The lineage 4 (L4) of Mycobacterium tuberculosis (MTB) is not only globally prevalent but also locally dominant, surpassing other lineages, with lineage 2 (L2) following in prevalence. Despite its widespread occurrence, factors influencing the expansion of L4 and its sub-lineages remain poorly understood both at local and global levels. Therefore, this study aimed to conduct a pan-genome and identify genomic signatures linked to the elevated prevalence of L4 sublineages among extrapulmonary TB (EPTB) patients in western Ethiopia. METHODS A cross-sectional study was conducted at an institutional level involving confirmed cases of extrapulmonary tuberculosis (EPTB) patients from August 5, 2018, to December 30, 2019. A total of 75 MTB genomes, classified under lineage 4 (L4), were used for conducting pan-genome and genome-wide association study (GWAS) analyses. After a quality check, variants were identified using MTBseq, and genomes were de novo assembled using SPAdes. Gene prediction and annotation were performed using Prokka. The pan-genome was constructed using GET_HOMOLOGUES, and its functional analysis was carried out with the Bacterial Pan-Genome Analysis tool (BPGA). For GWAS analysis, Scoary was employed with Benjamini-Hochberg correction, with a significance threshold set at p-value ≤ 0.05. RESULTS The analysis revealed a total of 3,270 core genes, predominantly associated with orthologous groups (COG) functions, notably in the categories of '[R] General function prediction only' and '[I] Lipid transport and metabolism'. Conversely, functions related to '[N] Cell motility' and '[Q] Secondary metabolites biosynthesis, transport, and catabolism' were primarily linked to unique and accessory genes. The pan-genome of MTB L4 was found to be open. Furthermore, the GWAS study identified genomic signatures linked to the prevalence of sublineages L4.6.3 and L4.2.2.2. CONCLUSIONS Apart from host and environmental factors, the sublineage of L4 employs distinct virulence factors for successful dissemination in western Ethiopia. Given that the functions of these newly identified genes are not well understood, it is advisable to experimentally validate their roles, particularly in the successful transmission of specific L4 sublineages over others.
Collapse
Affiliation(s)
- Basha Chekesa
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
- Collage of Natural and Computational Science, Wallaga University, Nekemte, Ethiopia
| | - Harinder Singh
- J. Craig Venter Institute, Rockville, Maryland, United States of America
| | | | - Sanjay Vashee
- J. Craig Venter Institute, Rockville, Maryland, United States of America
| | | | | | - Musse Girma
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Oudessa Kerro
- Institute of Agriculture, The University of Tennessee, Tennessee, Knoxville, United States of America
| | - Balako Gumi
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
| | - Gobena Ameni
- Aklilu Lemma Institute of Pathobiology, Addis Ababa University, Addis Ababa, Ethiopia
- College of Agriculture and Veterinary Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
97
|
Serson WR, Gishini MFS, Stupar RM, Stec AO, Armstrong PR, Hildebrand D. Identification and Candidate Gene Evaluation of a Large Fast Neutron-Induced Deletion Associated with a High-Oil Phenotype in Soybean Seeds. Genes (Basel) 2024; 15:892. [PMID: 39062671 PMCID: PMC11276498 DOI: 10.3390/genes15070892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 06/29/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
Since the dawn of agriculture, crops have been genetically altered for desirable characteristics. This has included the selection of natural and induced mutants. Increasing the production of plant oils such as soybean (Glycine max) oil as a renewable resource for food and fuel is valuable. Successful breeding for higher oil levels in soybeans, however, usually results in reduced seed protein. A soybean fast neutron population was screened for oil content, and three high oil mutants with minimal reductions in protein levels were found. Three backcross F2 populations derived from these mutants exhibited segregation for seed oil content. DNA was pooled from the high-oil and normal-oil plants within each population and assessed by comparative genomic hybridization. A deletion encompassing 20 gene models on chromosome 14 was found to co-segregate with the high-oil trait in two of the three populations. Eighteen genes in the deleted region have known functions that appear unrelated to oil biosynthesis and accumulation pathways, while one of the unknown genes (Glyma.14G101900) may contribute to the regulation of lipid droplet formation. This high-oil trait can facilitate the breeding of high-oil soybeans without protein reduction, resulting in higher meal protein levels.
Collapse
Affiliation(s)
- William R. Serson
- Department of Biology, Penn State University, Lehigh Valley, Center Valley, PA 18034, USA
| | | | - Robert M. Stupar
- Department of Agronomy and Plant Genetics, University of Minnesota, Saint Paul, MN 55108, USA; (R.M.S.); (A.O.S.)
| | - Adrian O. Stec
- Department of Agronomy and Plant Genetics, University of Minnesota, Saint Paul, MN 55108, USA; (R.M.S.); (A.O.S.)
| | - Paul R. Armstrong
- United States Department of Agriculture-Agricultural Research Service, Manhattan, KS 66502, USA
| | - David Hildebrand
- Department of Plant and Soil Sciences, University of Kentucky, Lexington, KY 40546, USA;
| |
Collapse
|
98
|
Castañeda S, Adeniyi-Ipadeola G, Wu Y, Suarez-Reyes C, Jain A, Ramírez JD, Weatherhead JE. Characterizing Excretory-Secretory Products Proteome Across Larval Development Stages in Ascaris suum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.03.601870. [PMID: 39005370 PMCID: PMC11245028 DOI: 10.1101/2024.07.03.601870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Introduction Ascaris lumbricoides and Ascaris suum are parasitic nematodes that primarily infest the small intestines of humans and pigs, respectively. Ascariasis poses a significant threat to human health and swine health. Understanding Ascaris larval development is crucial for developing novel therapeutic interventions that will prevent ascariasis in both humans and pigs. This study aimed to characterize the excretory-secretory (ES) proteome of different Ascaris suum larval stages (L3-egg, L3-lung, L3-trachea) to identify potential targets for intervention to prevent Ascaris -induced global morbidity. Methods Stage-specific larvae were isolated, cultured in vitro and ES-product was collected. Third-stage Ascaris larvae (L3) were isolated from embryonated eggs (L3-egg), isolated from the lungs of Balb/c mice infected with Ascaris suum eggs at day 8 post infection (L3-lungs) and isolated from the trachea of Balb/c mice infected with Ascaris suum eggs at day 12 post infection (L3-trachea). ES products were obtained by culturing larvae. Proteomic analysis was conducted using liquid chromatography-tandem mass spectrometry (LC-MS/MS) and bioinformatic tools including MaxQuant, Perseus, and Andromeda, following a detailed protocol available on GitHub. The analysis encompassed peptide identification, scoring, and quantification against an organism-specific database, with subsequent quality control, correlation assessment, and differential abundance determination using the Amica algorithm. Results A total of 58 unique proteins were identified in the ES products. Fourteen proteins were common across all stages, while others were stage-specific. Principal component analysis revealed distinct protein profiles for each stage, suggesting qualitatively different proteomes. Gene ontology analysis indicated stage-specific GO enrichment of specific protein classes, such as nuclear proteins in L3-egg ES products and metabolic enzymes in L3-lung and L3-trachea ES products. Discussion This study revealed stage-specific differences in the composition of Ascaris ES products. Further investigation into the functional roles of these proteins and their interactions with host cells is crucial for developing novel therapeutic and diagnostic strategies against ascariasis.
Collapse
|
99
|
Yanagi KS, Jochim B, Kunjo SO, Breen P, Ruvkun G, Lehrbach N. Mutations in nucleotide metabolism genes bypass proteasome defects in png-1/NGLY1-deficient Caenorhabditis elegans. PLoS Biol 2024; 22:e3002720. [PMID: 38991033 PMCID: PMC11265709 DOI: 10.1371/journal.pbio.3002720] [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: 12/17/2023] [Revised: 07/23/2024] [Accepted: 06/21/2024] [Indexed: 07/13/2024] Open
Abstract
The conserved SKN-1A/Nrf1 transcription factor regulates the expression of proteasome subunit genes and is essential for maintenance of adequate proteasome function in animal development, aging, and stress responses. Unusual among transcription factors, SKN-1A/Nrf1 is a glycoprotein synthesized in the endoplasmic reticulum (ER). N-glycosylated SKN-1A/Nrf1 exits the ER and is deglycosylated in the cytosol by the PNG-1/NGLY1 peptide:N-glycanase. Deglycosylation edits the protein sequence of SKN-1A/Nrf1 by converting N-glycosylated asparagine residues to aspartate, which is necessary for SKN-1A/Nrf1 transcriptional activation of proteasome subunit genes. Homozygous loss-of-function mutations in the peptide:N-glycanase (NGLY1) gene cause NGLY1 deficiency, a congenital disorder of deglycosylation. There are no effective treatments for NGLY1 deficiency. Since SKN-1A/Nrf1 is a major client of NGLY1, the resulting proteasome deficit contributes to NGLY1 disease. We sought to identify targets for mitigation of proteasome dysfunction in NGLY1 deficiency that might indicate new avenues for treatment. We isolated mutations that suppress the sensitivity to proteasome inhibitors caused by inactivation of the NGLY1 ortholog PNG-1 in Caenorhabditis elegans. We identified multiple suppressor mutations affecting 3 conserved genes: rsks-1, tald-1, and ent-4. We show that the suppressors act through a SKN-1/Nrf-independent mechanism and confer proteostasis benefits consistent with amelioration of proteasome dysfunction. ent-4 encodes an intestinal nucleoside/nucleotide transporter, and we show that restriction of nucleotide availability is beneficial, whereas a nucleotide-rich diet exacerbates proteasome dysfunction in PNG-1/NGLY1-deficient C. elegans. Our findings suggest that dietary or pharmacological interventions altering nucleotide availability have the potential to mitigate proteasome insufficiency in NGLY1 deficiency and other diseases associated with proteasome dysfunction.
Collapse
Affiliation(s)
- Katherine S. Yanagi
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Briar Jochim
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Sheikh Omar Kunjo
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| | - Peter Breen
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
| | - Gary Ruvkun
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts, United States of America
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Nicolas Lehrbach
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, United States of America
| |
Collapse
|
100
|
Toyoda Y, Matsuo H, Takada T. Functional characterization of variants in human ABCC11, an axillary osmidrosis risk factor. Hum Cell 2024; 37:1070-1079. [PMID: 38750405 DOI: 10.1007/s13577-024-01074-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 05/03/2024] [Indexed: 06/24/2024]
Abstract
Human ATP-binding cassette transporter C11 (ABCC11) is a membrane protein exhibiting ATP-dependent transport activity for a variety of lipophilic anions including endogenous substances and xenobiotics such as anti-cancer agents. Accumulating evidence indicates that ABCC11 wild type is responsible for the high-secretion phenotypes in human apocrine glands including wet type of earwax and the risk of axillary osmidrosis. Also, a less-functional variant of ABCC11 was reportedly associated with a risk for drug-induced toxicity in humans. Thus, functional change in ABCC11 may affect individual's constitution and drug toxicity, which led us to reason that functional validation of genetic variations in ABCC11 should be of importance. Therefore, in addition to p.G180R (a well-characterized non-functional variant of ABCC11), we studied cellular expression and function of 10 variants of ABCC11. In this study, ABCC11 function was evaluated as an ATP-dependent transport of radio labeled-dehydroepiandrosterone sulfate using ABCC11-expressing plasma membrane vesicles. Except for p.G180R, other 10 variants were maturated as an N-linked glycoprotein and expressed on the plasma membrane. We found that six variants impaired the net cellular function of ABCC11. Among them, p.R630W was most influential. Including this identification of a significantly-dysfunctional variant, our findings will extend our understanding of genetic variations and biochemical features of ABCC11 protein.
Collapse
Affiliation(s)
- Yu Toyoda
- Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan.
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| |
Collapse
|