1
|
Shembekar SS, Nikolaus P, Honnert U, Höring M, Attia A, Topp K, Lohmann B, Liebisch G, Bähler M. Regulation of mitochondrial cristae organization by Myo19, Miro1 and Miro2, and metaxin 3. J Cell Sci 2025; 138:JCS263637. [PMID: 39882711 PMCID: PMC11925395 DOI: 10.1242/jcs.263637] [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: 02/29/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025] Open
Abstract
The actin-based motor myosin-19 (Myo19) exerts force on mitochondrial membrane receptors Miro1/2, influencing endoplasmic reticulum (ER)-mitochondria contact sites and mitochondrial cristae structure. The mitochondrial intermembrane bridging (MIB) complex connects the outer and inner mitochondrial membranes at the cristae junction through the mitochondrial contact site and cristae organization system (MICOS). However, the interaction between Myo19, Miro1 and Miro2 (hereafter Miro1/2), and the MIB-MICOS complex in cristae regulation remains unclear. This study investigates the roles of Miro1/2 and metaxin 3 (Mtx3), a MIB complex component, in linking Myo19 to MIB-MICOS. We show that Miro1/2 interact with Myo19 and the MIB complex but not with Mtx3. Their mitochondrial membrane anchors are not essential for MIB interaction or cristae structure. However, Mtx3 is crucial for the connection between MIB-MICOS and the Myo19 and Miro1/2 proteins. Deleting Miro1/2 mimics the effects of Myo19 deficiency on ER-mitochondria contacts and cristae structure, whereas Mtx3 deletion does not. Notably, the loss of Myo19 and Miro1/2 alters mitochondrial lipid composition, reducing cardiolipin and its precursors, suggesting Myo19 and Miro1/2 influence cristae indirectly via lipid transfer at ER-mitochondria contact sites.
Collapse
Affiliation(s)
- Samruddhi S. Shembekar
- Institute of Integrative Cell Biology and Physiology, University of Münster, 48149 Münster, Germany
| | - Petra Nikolaus
- Institute of Integrative Cell Biology and Physiology, University of Münster, 48149 Münster, Germany
| | - Ulrike Honnert
- Institute of Integrative Cell Biology and Physiology, University of Münster, 48149 Münster, Germany
| | - Marcus Höring
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, 93053 Regensburg, Germany
| | - Aya Attia
- Institute of Integrative Cell Biology and Physiology, University of Münster, 48149 Münster, Germany
| | - Karin Topp
- Institute of Integrative Cell Biology and Physiology, University of Münster, 48149 Münster, Germany
| | - Birgit Lohmann
- Institute of Integrative Cell Biology and Physiology, University of Münster, 48149 Münster, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, 93053 Regensburg, Germany
| | - Martin Bähler
- Institute of Integrative Cell Biology and Physiology, University of Münster, 48149 Münster, Germany
| |
Collapse
|
2
|
Song L, Jiang W, Yu Y, Yu J, Zheng R. The mechanism of PLTP on sepsis-associated acute kidney injury: some hints. Crit Care 2025; 29:127. [PMID: 40114252 PMCID: PMC11924828 DOI: 10.1186/s13054-025-05333-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 02/22/2025] [Indexed: 03/22/2025] Open
Affiliation(s)
- Lin Song
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Wei Jiang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Yang Yu
- School of Laboratory Animal & Shandong Laboratory Animal Center, Shandong First Medical University & Shandong Academy of Medical Science, Jinan, Shandong, China
| | - Jiangquan Yu
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China
- Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, China
| | - Ruiqiang Zheng
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, China.
- Intensive Care Unit, Northern Jiangsu People's Hospital, Yangzhou, China.
| |
Collapse
|
3
|
Gao L, Nowakowska MB, Selby K, Przykopanski A, Chen B, Krüger M, Douillard FP, Lam KH, Chen P, Huang T, Minton NP, Dorner MB, Dorner BG, Rummel A, Lindström M, Jin R. Botulinum neurotoxins exploit host digestive proteases to boost their oral toxicity via activating OrfXs/P47. Nat Struct Mol Biol 2025:10.1038/s41594-024-01479-0. [PMID: 39838108 DOI: 10.1038/s41594-024-01479-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 12/18/2024] [Indexed: 01/23/2025]
Abstract
Botulinum neurotoxins (BoNTs) rank among the most potent toxins and many of them are produced by bacteria carrying the orfX gene cluster that also encodes four nontoxic proteins (OrfX1, OrfX2, OrfX3 and P47). The orfX gene cluster is also found in the genomes of many non-BoNT-producing bacteria, often alongside genes encoding oral insecticidal toxins. However, the functions of these OrfXs and P47 remain elusive. Here, we demonstrate that the combined action of all four components (OrfXs and P47) drastically boosts the oral toxicity of BoNT in mice, following proteolytic activation by digestive proteases that oral toxins regularly confront. In particular, OrfX2 adopts a self-inhibiting state, engaging with BoNT through another clostridial protein, nontoxic non-hemagglutinin (NTNH), only after proteolytic activation. Cryo-electron microscopy studies unveil that two molecules of protease-activated OrfX2 simultaneously associate with NTNH, a binding mode crucial for boosting BoNT oral toxicity. Collectively, these studies offer novel insights into the physiological functions and regulatory mechanisms of OrfXs and P47 of BoNTs, shedding light on the pathogenesis of other bacterial toxins associated with homologous OrfXs and P47.
Collapse
Affiliation(s)
- Linfeng Gao
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Maria Barbara Nowakowska
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Katja Selby
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | | | - Baohua Chen
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Maren Krüger
- Biological Toxins, Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - François Paul Douillard
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Kwok-Ho Lam
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Peng Chen
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Ting Huang
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA
| | - Nigel Peter Minton
- Clostridia Research Group, BBSRC/EPSRC Synthetic Biology Research Centre (SBRC), School of Life Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Martin Bernhard Dorner
- Biological Toxins, Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Brigitte Gertrud Dorner
- Biological Toxins, Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Berlin, Germany
| | - Andreas Rummel
- Institute for Toxicology, Hannover Medical School, Hannover, Germany
| | - Miia Lindström
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland.
| | - Rongsheng Jin
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
4
|
Egorkin NA, Dominnik EE, Raevskii RI, Kuklina DD, Varfolomeeva LA, Popov VO, Boyko KM, Sluchanko NN. Structural basis of selective beta-carotene binding by a soluble protein. Structure 2024; 32:2123-2133.e3. [PMID: 39383875 DOI: 10.1016/j.str.2024.09.014] [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: 07/09/2024] [Revised: 08/23/2024] [Accepted: 09/12/2024] [Indexed: 10/11/2024]
Abstract
β-carotene (BCR) is the most abundant carotenoid, a colorant, antioxidant, and provitamin A. The extreme hydrophobicity of this hydrocarbon requires special mechanisms for distribution in aqueous media, including water-soluble carotenoproteins. However, all known carotenoproteins prefer oxygenated carotenoids and bind BCR inefficiently. Here, we present the crystal structure of the BCR-binding protein (BBP) from gregarious male locusts, which is responsible for their vivid yellow body coloration, in complex with its natural ligand, BCR. BBP forms an antiparallel tubular homodimer with α/β-wrap folded monomers, each forming a hydrophobic 47 Å long, coaxial tunnel that opens outward and is occupied by one s-cisC6-C7, all-trans BCR molecule. In the BCR absence, BBP accepts a range of xanthophylls, with reduced efficiency depending on the position and number of oxygen atoms, but rejects lycopene. The structure captures a pigment complex with a Takeout 1 protein and inspires potential applications of BBP as a BCR solubilizer.
Collapse
Affiliation(s)
- Nikita A Egorkin
- A.N. Bach Institute of Biochemistry, Federal Research Centre of Biotechnology of the Russian Academy of Sciences, 33 Leninsky prospect, building 1, Moscow 119071, Russia; M.V. Lomonosov Moscow State University, School of Biology, 1 Lenin Hills, building 12, Moscow 119991, Russia
| | - Eva E Dominnik
- A.N. Bach Institute of Biochemistry, Federal Research Centre of Biotechnology of the Russian Academy of Sciences, 33 Leninsky prospect, building 1, Moscow 119071, Russia; M.V. Lomonosov Moscow State University, School of Chemistry, 1 Lenin Hills, building 3, Moscow 119991, Russia
| | - Roman I Raevskii
- A.N. Bach Institute of Biochemistry, Federal Research Centre of Biotechnology of the Russian Academy of Sciences, 33 Leninsky prospect, building 1, Moscow 119071, Russia
| | - Daria D Kuklina
- Moscow Institute of Physics and Technology, Institutski per. 9, Dolgoprudny 141700, Russia
| | - Larisa A Varfolomeeva
- A.N. Bach Institute of Biochemistry, Federal Research Centre of Biotechnology of the Russian Academy of Sciences, 33 Leninsky prospect, building 1, Moscow 119071, Russia
| | - Vladimir O Popov
- A.N. Bach Institute of Biochemistry, Federal Research Centre of Biotechnology of the Russian Academy of Sciences, 33 Leninsky prospect, building 1, Moscow 119071, Russia
| | - Konstantin M Boyko
- A.N. Bach Institute of Biochemistry, Federal Research Centre of Biotechnology of the Russian Academy of Sciences, 33 Leninsky prospect, building 1, Moscow 119071, Russia
| | - Nikolai N Sluchanko
- A.N. Bach Institute of Biochemistry, Federal Research Centre of Biotechnology of the Russian Academy of Sciences, 33 Leninsky prospect, building 1, Moscow 119071, Russia.
| |
Collapse
|
5
|
Yu S, Jia H, Li Z, Ding S, Li F, Xu P, Tian Y, Ma L, Qian F, Li M, Zhang N, Wang H. Plasma levels of bactericidal/permeability-increasing protein correlate with systemic inflammation in acute coronary syndrome. Heliyon 2024; 10:e32470. [PMID: 38933945 PMCID: PMC11201113 DOI: 10.1016/j.heliyon.2024.e32470] [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: 03/10/2023] [Revised: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Background Neutrophils play important roles in atherosclerosis and atherothrombosis. Bactericidal/permeability-increasing protein (BPI) is mainly expressed in the granules of human neutrophils in response to inflammatory stress. This observational, cross-sectional study investigated the plasma level of BPI in patients with acute coronary syndrome (ACS) and its correlation with blood neutrophil counts and circulating inflammatory biomarkers. Methods A total of 367 patients who had acute chest pain and who were admitted to our hospital for coronary angiography (CAG) and/or percutaneous coronary intervention (PCI) from May 1, 2020 to August 31, 2020 were recruited. Among them, 256 had a cardiac troponin value above the 99th percentile upper reference limit and were diagnosed with ACS. The remaining patients (n = 111) were classified as non-ACS. The TIMI and GRACE scores were calculated at admission. The Gensini score based on CAG was used to determine atherosclerotic burden. Plasma levels of interleukin (IL)-1β, myeloperoxidase-DNA (MPO-DNA), high sensitivity C-reactive protein (hs-CRP), S100A8/A9, and BPI were measured using enzyme-linked immunosorbent assays. Correlations of plasma BPI levels with examination scores and levels of circulating inflammatory biomarkers were explored. Receiver operating characteristic (ROC) curve analysis was used to determine the diagnostic efficacy of BPI for ACS and myocardial infarction. Results Patients in the ACS group showed significantly higher plasma BPI levels compared to the non-ACS group (46.42 ± 16.61 vs. 16.23 ± 6.19 ng/mL, p < 0.05). Plasma levels of IL-1β, MPO-DNA, hs-CRP, and S100A8/A9 in the ACS group were also significantly higher than those in the non-ACS group (all p < 0.05). In addition, plasma BPI levels were positively correlated with the TIMI, GRACE, and Gensini scores (r = 0.176, p = 0.003; r = 0.320, p < 0.001; r = 0.263, p < 0.001, respectively) in patients with ACS. Plasma BPI levels were also positively correlated with blood neutrophil counts (r = 0.266, p < 0.001) and levels of circulating inflammatory biomarkers (IL-1β, r = 0.512; MPO-DNA, r = 0.452; hs-CRP, r = 0.554; S100A8/A9, r = 0.434; all p < 0.001) in patients with ACS. ROC curve analysis revealed that the diagnostic efficacy of BPI for ACS was not inferior to that of IL-1β, MPO-DNA, hs-CRP, S100A8/A9, or blood neutrophil counts. ROC analysis also showed that the diagnostic efficacy of BPI for myocardial infarction was not inferior to that of creatine kinase (CK)-MB or cardiac troponin I. Conclusion BPI is associated with systemic inflammation in ACS and may be involved in the process of atherosclerosis and atherothrombosis. The potential of BPI as a prognostic and diagnostic biomarker for ACS should be investigated in clinical settings.
Collapse
Affiliation(s)
- Shicheng Yu
- Department of Cardiology, Lu'an Hospital of Anhui Medical University, Lu'an, Anhui, 237000, PR China
| | - Haoxuan Jia
- Graduate School of Bengbu Medical College, Bengbu, Anhui, 233004, PR China
| | - Zheng Li
- Graduate School of Bengbu Medical College, Bengbu, Anhui, 233004, PR China
| | - Shengkai Ding
- Department of Cardiology, Lu'an Hospital of Anhui Medical University, Lu'an, Anhui, 237000, PR China
| | - Fengyun Li
- Department of Cardiology, Lu'an Hospital of Anhui Medical University, Lu'an, Anhui, 237000, PR China
| | - Pan Xu
- Department of Cardiology, Lu'an Hospital of Anhui Medical University, Lu'an, Anhui, 237000, PR China
| | - Yuan Tian
- Department of Cardiology, Lu'an Hospital of Anhui Medical University, Lu'an, Anhui, 237000, PR China
| | - Lingling Ma
- Department of Cardiology, Lu'an Hospital of Anhui Medical University, Lu'an, Anhui, 237000, PR China
| | - Fudong Qian
- Department of Cardiology, Lu'an Hospital of Anhui Medical University, Lu'an, Anhui, 237000, PR China
| | - Miaonan Li
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, 287 Changhuai Road, Bengbu, Anhui, 233004, PR China
| | - Nana Zhang
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, 287 Changhuai Road, Bengbu, Anhui, 233004, PR China
| | - Hongju Wang
- Department of Cardiology, The First Affiliated Hospital of Bengbu Medical College, 287 Changhuai Road, Bengbu, Anhui, 233004, PR China
| |
Collapse
|
6
|
Guzmán-Beltrán S, Juárez E, Cruz-Muñoz BL, Páez-Cisneros CA, Sarabia C, González Y. Bactericidal Permeability-Increasing Protein (BPI) Inhibits Mycobacterium tuberculosis Growth. Biomolecules 2024; 14:475. [PMID: 38672491 PMCID: PMC11048543 DOI: 10.3390/biom14040475] [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: 02/17/2024] [Revised: 04/02/2024] [Accepted: 04/05/2024] [Indexed: 04/28/2024] Open
Abstract
Bactericidal permeability-increasing protein (BPI) is a multifunctional cationic protein produced by neutrophils, eosinophils, fibroblasts, and macrophages with antibacterial anti-inflammatory properties. In the context of Gram-negative infection, BPI kills bacteria, neutralizes the endotoxic activity of lipopolysaccharides (LPSs), and, thus, avoids immune hyperactivation. Interestingly, BPI increases in patients with Gram-positive meningitis, interacts with lipopeptides and lipoteichoic acids of Gram-positive bacteria, and significantly enhances the immune response in peripheral blood mononuclear cells. We evaluated the antimycobacterial and immunoregulatory properties of BPI in human macrophages infected with Mycobacterium tuberculosis. Our results showed that recombinant BPI entered macrophages, significantly reduced the intracellular growth of M. tuberculosis, and inhibited the production of the proinflammatory cytokine tumor necrosis factor-alpha (TNF-α). Furthermore, BPI decreased bacterial growth directly in vitro. These data suggest that BPI has direct and indirect bactericidal effects inhibiting bacterial growth and potentiating the immune response in human macrophages and support that this new protein's broad-spectrum antibacterial activity has the potential for fighting tuberculosis.
Collapse
Affiliation(s)
- Silvia Guzmán-Beltrán
- Department of Microbiology, National Institute for Respiratory Diseases Ismael Cosio Villegas, Mexico City 14080, Mexico; (E.J.); (B.L.C.-M.); (C.A.P.-C.); (C.S.); (Y.G.)
| | | | | | | | | | | |
Collapse
|
7
|
Sun S, Zhao G, Jia M, Jiang Q, Li S, Wang H, Li W, Wang Y, Bian X, Zhao YG, Huang X, Yang G, Cai H, Pastor-Pareja JC, Ge L, Zhang C, Hu J. Stay in touch with the endoplasmic reticulum. SCIENCE CHINA. LIFE SCIENCES 2024; 67:230-257. [PMID: 38212460 DOI: 10.1007/s11427-023-2443-9] [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: 03/19/2023] [Accepted: 08/28/2023] [Indexed: 01/13/2024]
Abstract
The endoplasmic reticulum (ER), which is composed of a continuous network of tubules and sheets, forms the most widely distributed membrane system in eukaryotic cells. As a result, it engages a variety of organelles by establishing membrane contact sites (MCSs). These contacts regulate organelle positioning and remodeling, including fusion and fission, facilitate precise lipid exchange, and couple vital signaling events. Here, we systematically review recent advances and converging themes on ER-involved organellar contact. The molecular basis, cellular influence, and potential physiological functions for ER/nuclear envelope contacts with mitochondria, Golgi, endosomes, lysosomes, lipid droplets, autophagosomes, and plasma membrane are summarized.
Collapse
Affiliation(s)
- Sha Sun
- National Laboratory of Biomacromolecules, Institute of Biophysics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Gan Zhao
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Mingkang Jia
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Qing Jiang
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Shulin Li
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Haibin Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wenjing Li
- Laboratory of Computational Biology & Machine Intelligence, School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yunyun Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Xin Bian
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| | - Yan G Zhao
- Brain Research Center, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
| | - Xun Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Innovation Academy for Seed Design, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Ge Yang
- Laboratory of Computational Biology & Machine Intelligence, School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Huaqing Cai
- National Laboratory of Biomacromolecules, Institute of Biophysics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jose C Pastor-Pareja
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
- Institute of Neurosciences, Consejo Superior de Investigaciones Cientfflcas-Universidad Miguel Hernandez, San Juan de Alicante, 03550, Spain.
| | - Liang Ge
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| | - Chuanmao Zhang
- The Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, College of Life Sciences, Peking University, Beijing, 100871, China.
| | - Junjie Hu
- National Laboratory of Biomacromolecules, Institute of Biophysics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
8
|
Villegas G, Pereira MT, Love CR, Edery I. DAYWAKE implicates novel roles for circulating lipid-binding proteins as extracerebral regulators of daytime wake-sleep behavior. FEBS Lett 2024; 598:321-330. [PMID: 38112219 PMCID: PMC10922413 DOI: 10.1002/1873-3468.14789] [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: 11/07/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/21/2023]
Abstract
Sleep during the midday, commonly referred to as siesta, is a common trait of animals that mainly sleep during the night. Work using Drosophila led to the identification of the daywake (dyw) gene, found to have anti-siesta activity. Herein, we show that the DYW protein undergoes signal peptide-dependent secretion, is present in the circulatory system, and accumulates in multiple organs, but, surprisingly, it is not detected in the brain where wake-sleep centers are located. The abundance of DYW in adult flies is regulated by age, sex, temperature, and the splicing efficiency of a nearby thermosensitive intron. We suggest that DYW regulates daytime wake-sleep balance in an indirect, extracerebral manner, via a multi-organ network that interfaces with the circulatory system.
Collapse
Affiliation(s)
- Gabriel Villegas
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA
- Department of Molecular Biology and Biochemistry, Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA
| | - Mathew T Pereira
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA
| | - Cameron R Love
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA
| | - Isaac Edery
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA
- Department of Molecular Biology and Biochemistry, Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA
| |
Collapse
|
9
|
Dabravolski S, Orekhov NA, Melnichenko A, Sukhorukov VN, Popov MA, Orekhov A. Cholesteryl Ester Transfer Protein (CETP) Variations in Relation to Lipid Profiles and Cardiovascular Diseases: An Update. Curr Pharm Des 2024; 30:742-756. [PMID: 38425105 DOI: 10.2174/0113816128284695240219093612] [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: 10/26/2023] [Revised: 01/02/2024] [Accepted: 01/11/2024] [Indexed: 03/02/2024]
Abstract
Lipid metabolism plays an essential role in the pathogenesis of cardiovascular and metabolic diseases. Cholesteryl ester transfer protein (CETP) is a crucial glycoprotein involved in lipid metabolism by transferring cholesteryl esters (CE) and triglycerides (TG) between plasma lipoproteins. CETP activity results in reduced HDL-C and increased VLDL- and LDL-C concentrations, thus increasing the risk of cardiovascular and metabolic diseases. In this review, we discuss the structure of CETP and its mechanism of action. Furthermore, we focus on recent experiments on animal CETP-expressing models, deciphering the regulation and functions of CETP in various genetic backgrounds and interaction with different external factors. Finally, we discuss recent publications revealing the association of CETP single nucleotide polymorphisms (SNPs) with the risk of cardiovascular and metabolic diseases, lifestyle factors, diet and therapeutic interventions. While CETP SNPs can be used as effective diagnostic markers, diet, lifestyle, gender and ethnic specificity should also be considered for effective treatment.
Collapse
Affiliation(s)
- Siarhei Dabravolski
- Department of Biotechnology Engineering, ORT Braude College, Braude Academic College of Engineering, Karmiel, Israel
| | - Nikolay A Orekhov
- Laboratory of Angiopatology, Research Institute of General Pathology and Pathophysiology, The Russian Academy of Medical Sciences, Moscow, Russian Federation
| | - Alexandra Melnichenko
- Laboratory of Angiopatology, Research Institute of General Pathology and Pathophysiology, The Russian Academy of Medical Sciences, Moscow, Russian Federation
| | - Vasily N Sukhorukov
- Laboratory of Angiopatology, Research Institute of General Pathology and Pathophysiology, The Russian Academy of Medical Sciences, Moscow, Russian Federation
| | - Mikhail A Popov
- Laboratory of Angiopatology, Research Institute of General Pathology and Pathophysiology, The Russian Academy of Medical Sciences, Moscow, Russian Federation
| | - Alexander Orekhov
- Laboratory of Angiopatology, Research Institute of General Pathology and Pathophysiology, The Russian Academy of Medical Sciences, Moscow, Russian Federation
| |
Collapse
|
10
|
Košenina S, Stenmark P. Crystal structure of the OrfX1-OrfX3 complex from the PMP1 neurotoxin gene cluster. FEBS Lett 2023; 597:515-523. [PMID: 36403098 DOI: 10.1002/1873-3468.14542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/10/2022] [Indexed: 11/21/2022]
Abstract
Paraclostridial mosquitocidal protein 1 (PMP1) is a member of the clostridial neurotoxin (CNT) family, which includes botulinum and tetanus neurotoxins. PMP1 has unique selectivity for anopheline mosquitos and is the only known member of the family that targets insects. PMP1 is encoded in an orfX gene cluster, which in addition to the toxin, consists of OrfX1, OrfX2, OrfX3, P47 and NTNH, which have been shown to aid in PMP1 toxicity. We here show that OrfX1 and OrfX3 form a complex and present its structure at 2.7 Å. The OrfX1-OrfX3 complex mimics the structure of full-length OrfX2 and belongs to the lipid-binding TULIP protein superfamily. With this report, the structures of all proteins encoded in the orfX gene cluster of CNTs are now determined.
Collapse
Affiliation(s)
- Sara Košenina
- Department of Biochemistry and Biophysics, Stockholm University, Sweden
| | - Pål Stenmark
- Department of Biochemistry and Biophysics, Stockholm University, Sweden
| |
Collapse
|
11
|
Gao L, Lam KH, Liu S, Przykopanski A, Lübke J, Qi R, Krüger M, Nowakowska MB, Selby K, Douillard FP, Dorner MB, Perry K, Lindström M, Dorner BG, Rummel A, Jin R. Crystal structures of OrfX1, OrfX2 and the OrfX1-OrfX3 complex from the orfX gene cluster of botulinum neurotoxin E1. FEBS Lett 2023; 597:524-537. [PMID: 36653893 PMCID: PMC10019085 DOI: 10.1002/1873-3468.14576] [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: 12/07/2022] [Accepted: 12/12/2022] [Indexed: 01/20/2023]
Abstract
Botulinum neurotoxins (BoNTs) are among the most lethal toxins known to humans, comprising seven established serotypes termed BoNT/A-G encoded in two types of gene clusters (ha and orfX) in BoNT-producing clostridia. The ha cluster encodes four non-toxic neurotoxin-associated proteins (NAPs) that assemble with BoNTs to protect and enhance their oral toxicity. However, the structure and function of the orfX-type NAPs remain largely unknown. Here, we report the crystal structures for OrfX1, OrfX2, and an OrfX1-OrfX3 complex, which are encoded in the orfX cluster of a BoNT/E1-producing Clostridium botulinum strain associated with human foodborne botulism. These structures lay the foundation for future studies on the potential roles of OrfX proteins in oral intoxication and pathogenesis of BoNTs.
Collapse
Affiliation(s)
- Linfeng Gao
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Kwok-ho Lam
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Shun Liu
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Adina Przykopanski
- Institute of Toxicology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Johanna Lübke
- Institute of Toxicology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Ruifeng Qi
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| | - Maren Krüger
- Biological Toxins, Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Seestrasse 10, 13353 Berlin, Germany
| | - Maria B. Nowakowska
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, P. O. Box 66, 00014 University of Helsinki, Finland
| | - Katja Selby
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, P. O. Box 66, 00014 University of Helsinki, Finland
| | - François P. Douillard
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, P. O. Box 66, 00014 University of Helsinki, Finland
| | - Martin B. Dorner
- Biological Toxins, Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Seestrasse 10, 13353 Berlin, Germany
| | - Kay Perry
- NE-CAT and Department of Chemistry and Chemical Biology, Cornell University, Argonne National Laboratory, Argonne, IL 60439, USA
| | - Miia Lindström
- Department of Food Hygiene and Environmental Health, Faculty of Veterinary Medicine, P. O. Box 66, 00014 University of Helsinki, Finland
| | - Brigitte G. Dorner
- Biological Toxins, Centre for Biological Threats and Special Pathogens, Robert Koch Institute, Seestrasse 10, 13353 Berlin, Germany
| | - Andreas Rummel
- Institute of Toxicology, Hannover Medical School, Carl-Neuberg-Strasse 1, 30625 Hannover, Germany
| | - Rongsheng Jin
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
12
|
Fujimoto S, Tashiro S, Tamura Y. Complementation Assay Using Fusion of Split-GFP and TurboID (CsFiND) Enables Simultaneous Visualization and Proximity Labeling of Organelle Contact Sites in Yeast. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2023; 6:25152564231153621. [PMID: 37366411 PMCID: PMC10243572 DOI: 10.1177/25152564231153621] [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] [Indexed: 06/28/2023]
Abstract
Numerous studies have revealed that organelle membrane contact sites (MCSs) play important roles in diverse cellular events, including the transport of lipids and ions between connected organelles. To understand MCS functions, it is essential to uncover proteins that accumulate at MCSs. Here, we develop a complementation assay system termed CsFiND (Complementation assay using Fusion of split-GFP and TurboID) for the simultaneous visualization of MCSs and identification of MCS-localized proteins. We express the CsFiND proteins on the endoplasmic reticulum and mitochondrial outer membrane in yeast to verify the reliability of CsFiND as a tool for identifying MCS-localized proteins.
Collapse
Affiliation(s)
| | - Shinya Tashiro
- Faculty of Science, Yamagata University, Yamagata, Japan
| | - Yasushi Tamura
- Faculty of Science, Yamagata University, Yamagata, Japan
| |
Collapse
|
13
|
Clausmeyer L, Fröhlich F. Mechanisms of Nonvesicular Ceramide Transport. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2023; 6:25152564231208250. [PMID: 37859671 PMCID: PMC10583516 DOI: 10.1177/25152564231208250] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/29/2023] [Accepted: 09/29/2023] [Indexed: 10/21/2023]
Abstract
Ceramides, as key components of cellular membranes, play essential roles in various cellular processes, including apoptosis, cell proliferation, and cell signaling. Ceramides are the precursors of all complex sphingolipids in eukaryotic cells. They are synthesized in the endoplasmic reticulum and are further processed at the Golgi apparatus. Therefore, ceramides have to be transported between these two organelles. In mammalian cells, the ceramide transfer protein forms a contact site between the ER and the trans-Golgi region and transports ceramide utilizing its steroidogenic acute regulatory protein-related lipid transfer domain. In yeast, multiple mechanisms of nonvesicular ceramide transport have been described. This involves the nuclear-vacuolar junction protein Nvj2, the yeast tricalbin proteins, and the lipocalin-like protein Svf1. This review aims to provide a comprehensive overview of nonvesicular ceramide transport mechanisms and their relevance in cellular physiology. We will highlight the physiological and pathological consequences of perturbations in nonvesicular ceramide transport and discuss future challenges in identifying and analyzing ceramide transfer proteins.
Collapse
Affiliation(s)
- Lena Clausmeyer
- Department of Biology/Chemistry, Bioanalytical Chemistry Section, Osnabrück University, Osnabrück, Germany
| | - Florian Fröhlich
- Department of Biology/Chemistry, Bioanalytical Chemistry Section, Osnabrück University, Osnabrück, Germany
- Center of Cellular Nanoanalytics Osnabrück (CellNanOs), Osnabrück University, Osnabrück, Germany
| |
Collapse
|
14
|
Dittmer NT, Hiromasa Y, Kanost MR. Proteomic analysis of pharate pupal molting fluid from the tobacco hornworm, Manduca sexta. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2022; 149:103844. [PMID: 36115517 PMCID: PMC9875806 DOI: 10.1016/j.ibmb.2022.103844] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 08/30/2022] [Accepted: 09/09/2022] [Indexed: 06/15/2023]
Abstract
The insect cuticle is a key component of their success, being important for protection, communication, locomotion, and support. Conversely, as an exoskeleton, it also limits the size of the insect and must be periodically molted and a new one synthesized, to permit growth. To achieve this, the insect secretes a solution of chitinases, proteases and other proteins, known collectively as molting fluid, during each molting process to break down and recycle components of the old cuticle. Previous research has focused on the degradative enzymes in molting fluid and offered some characterization of their biochemical properties. However, identification of the specific proteins involved remained to be determined. We have used 2D SDS-PAGE and LC/MS-based proteomic analysis to identify proteins in the molting fluid of the tobacco hornworm, Manduca sexta, undergoing the larval to pupal molt. We categorized these proteins based on their proposed functions including chitin metabolism, proteases, peptidases, and immunity. This analysis complements previous reported work on M. sexta molting fluid and identifies candidate genes for enzymes involved in cuticle remodeling. Proteins classified as having an immune function highlight potential for molting fluid to act as an immune barrier to prevent infections during the cuticle degradation and ecdysis processes. Several proteins known to function in melanin synthesis as an immune response in hemolymph were present in molting fluid. We demonstrated that the bacterium Micrococcus luteus and the entomopathogenic fungus Beauveria bassiana can stimulate activation of phenoloxidase in molting fluid, indicating that the recognition proteins, protease cascade, and prophenoloxidase needed for melanin synthesis are present as a defense against infection during cuticle degradation. This analysis offers insights for proteins that may be important not only for molting in M. sexta but for insects in general.
Collapse
Affiliation(s)
- Neal T Dittmer
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA.
| | - Yasuaki Hiromasa
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA
| | - Michael R Kanost
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA
| |
Collapse
|
15
|
Moreau T, Gautron J, Hincke MT, Monget P, Réhault-Godbert S, Guyot N. Antimicrobial Proteins and Peptides in Avian Eggshell: Structural Diversity and Potential Roles in Biomineralization. Front Immunol 2022; 13:946428. [PMID: 35967448 PMCID: PMC9363672 DOI: 10.3389/fimmu.2022.946428] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 06/22/2022] [Indexed: 11/21/2022] Open
Abstract
The calcitic avian eggshell provides physical protection for the embryo during its development, but also regulates water and gaseous exchange, and is a calcium source for bone mineralization. The calcified eggshell has been extensively investigated in the chicken. It is characterized by an inventory of more than 900 matrix proteins. In addition to proteins involved in shell mineralization and regulation of its microstructure, the shell also contains numerous antimicrobial proteins and peptides (AMPPs) including lectin-like proteins, Bacterial Permeability Increasing/Lipopolysaccharide Binding Protein/PLUNC family proteins, defensins, antiproteases, and chelators, which contribute to the innate immune protection of the egg. In parallel, some of these proteins are thought to be crucial determinants of the eggshell texture and its resulting mechanical properties. During the progressive solubilization of the inner mineralized eggshell during embryonic development (to provide calcium to the embryo), some antimicrobials may be released simultaneously to reinforce egg defense and protect the egg from contamination by external pathogens, through a weakened eggshell. This review provides a comprehensive overview of the diversity of avian eggshell AMPPs, their three-dimensional structures and their mechanism of antimicrobial activity. The published chicken eggshell proteome databases are integrated for a comprehensive inventory of its AMPPs. Their biochemical features, potential dual function as antimicrobials and as regulators of eggshell biomineralization, and their phylogenetic evolution will be described and discussed with regard to their three-dimensional structural characteristics. Finally, the repertoire of chicken eggshell AMPPs are compared to orthologs identified in other avian and non-avian eggshells. This approach sheds light on the similarities and differences exhibited by AMPPs, depending on bird species, and leads to a better understanding of their sequential or dual role in biomineralization and innate immunity.
Collapse
Affiliation(s)
- Thierry Moreau
- INRAE, Université de Tours, BOA, Nouzilly, France
- *Correspondence: Nicolas Guyot, ; Thierry Moreau,
| | - Joël Gautron
- INRAE, Université de Tours, BOA, Nouzilly, France
| | - Maxwell T. Hincke
- Department of Innovation in Medical Education, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Philippe Monget
- INRAE, CNRS, IFCE, Université de Tours, PRC, Nouzilly, France
| | | | - Nicolas Guyot
- INRAE, Université de Tours, BOA, Nouzilly, France
- *Correspondence: Nicolas Guyot, ; Thierry Moreau,
| |
Collapse
|
16
|
Hanna MG, Suen PH, Wu Y, Reinisch KM, De Camilli P. SHIP164 is a chorein motif lipid transfer protein that controls endosome-Golgi membrane traffic. J Cell Biol 2022; 221:e202111018. [PMID: 35499567 PMCID: PMC9067936 DOI: 10.1083/jcb.202111018] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 03/07/2022] [Accepted: 04/08/2022] [Indexed: 02/03/2023] Open
Abstract
Cellular membranes differ in protein and lipid composition as well as in the protein-lipid ratio. Thus, progression of membranous organelles along traffic routes requires mechanisms to control bilayer lipid chemistry and their abundance relative to proteins. The recent structural and functional characterization of VPS13-family proteins has suggested a mechanism through which lipids can be transferred in bulk from one membrane to another at membrane contact sites, and thus independently of vesicular traffic. Here, we show that SHIP164 (UHRF1BP1L) shares structural and lipid transfer properties with these proteins and is localized on a subpopulation of vesicle clusters in the early endocytic pathway whose membrane cargo includes the cation-independent mannose-6-phosphate receptor (MPR). Loss of SHIP164 disrupts retrograde traffic of these organelles to the Golgi complex. Our findings raise the possibility that bulk transfer of lipids to endocytic membranes may play a role in their traffic.
Collapse
Affiliation(s)
- Michael G. Hanna
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT
| | - Patreece H. Suen
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
| | - Yumei Wu
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT
| | - Karin M. Reinisch
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD
| | - Pietro De Camilli
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT
- Kavli Institue for Neuroscience, Yale University School of Medicine, New Haven, CT
- Aligning Science Across Parkinson’s Collaborative Research Network, Chevy Chase, MD
| |
Collapse
|
17
|
Qian T, Li C, Liu F, Xu K, Wan C, Liu Y, Yu H. Arabidopsis synaptotagmin 1 mediates lipid transport in a lipid composition-dependent manner. Traffic 2022; 23:346-356. [PMID: 35451158 DOI: 10.1111/tra.12844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 04/11/2022] [Accepted: 04/15/2022] [Indexed: 11/29/2022]
Abstract
The endoplasmic reticulum (ER) - plasma membrane (PM) contact sites (EPCSs) are structurally conserved in eukaryotes. The Arabidopsis ER-anchored synaptotagmin 1 (SYT1), enriched in EPCSs, plays a critical role in plant abiotic stress tolerance. It has become clear that SYT1 interacts with PM to mediate ER-PM connectivity. However, whether SYT1 performs additional functions at EPCSs remains unknown. Here, we reported that SYT1 efficiently transfers phospholipids between membranes. The lipid transfer activity of SYT1 is highly dependent on PI(4,5)P2 , a signal lipid accumulated at the PM under abiotic stress. Mechanically, while SYT1 transfers lipids fundamentally through the synaptotagmin-like mitochondrial-lipid-binding protein (SMP) domain, the efficient lipid transport requires the C2A domain-mediated membrane tethering. Interestingly, we observed that Ca2+ could stimulate SYT1-mediated lipid transport. In addition to PI(4,5)P2 , the Ca2+ activation requires the phosphatidylserine, another negatively charged lipid on the opposed membrane. Together, our studies identified Arabidopsis SYT1 as a lipid transfer protein at EPCSs and demonstrated it takes conserved as well as divergent mechanisms with other extend-synaptotagmins. The critical role of lipid composition and Ca2+ reveals SYT1-mediated lipid transport is highly regulated by signals in response to abiotic stresses.
Collapse
Affiliation(s)
- Tiantian Qian
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Chenlu Li
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Furong Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Kai Xu
- Jiangsu Key Laboratory for Microbes and Functional Genomics, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Chun Wan
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, USA
| | - Yinghui Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Haijia Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
18
|
Reconstitution and biochemical studies of extended synaptotagmin-mediated lipid transport. Methods Enzymol 2022; 675:33-62. [DOI: 10.1016/bs.mie.2022.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Egea PF. Mechanisms of Non-Vesicular Exchange of Lipids at Membrane Contact Sites: Of Shuttles, Tunnels and, Funnels. Front Cell Dev Biol 2021; 9:784367. [PMID: 34912813 PMCID: PMC8667587 DOI: 10.3389/fcell.2021.784367] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
Eukaryotic cells are characterized by their exquisite compartmentalization resulting from a cornucopia of membrane-bound organelles. Each of these compartments hosts a flurry of biochemical reactions and supports biological functions such as genome storage, membrane protein and lipid biosynthesis/degradation and ATP synthesis, all essential to cellular life. Acting as hubs for the transfer of matter and signals between organelles and throughout the cell, membrane contacts sites (MCSs), sites of close apposition between membranes from different organelles, are essential to cellular homeostasis. One of the now well-acknowledged function of MCSs involves the non-vesicular trafficking of lipids; its characterization answered one long-standing question of eukaryotic cell biology revealing how some organelles receive and distribute their membrane lipids in absence of vesicular trafficking. The endoplasmic reticulum (ER) in synergy with the mitochondria, stands as the nexus for the biosynthesis and distribution of phospholipids (PLs) throughout the cell by contacting nearly all other organelle types. MCSs create and maintain lipid fluxes and gradients essential to the functional asymmetry and polarity of biological membranes throughout the cell. Membrane apposition is mediated by proteinaceous tethers some of which function as lipid transfer proteins (LTPs). We summarize here the current state of mechanistic knowledge of some of the major classes of LTPs and tethers based on the available atomic to near-atomic resolution structures of several "model" MCSs from yeast but also in Metazoans; we describe different models of lipid transfer at MCSs and analyze the determinants of their specificity and directionality. Each of these systems illustrate fundamental principles and mechanisms for the non-vesicular exchange of lipids between eukaryotic membrane-bound organelles essential to a wide range of cellular processes such as at PL biosynthesis and distribution, lipid storage, autophagy and organelle biogenesis.
Collapse
Affiliation(s)
- Pascal F. Egea
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, CA, United States
| |
Collapse
|
20
|
Yu S, Li M, Li Z, Xu P, Yao Z, Qian S, Qian F, Gao D, Wang H. Positive correlations between plasma BPI level and MPO-DNA and S100A8/A9 in myocardial infarction. Platelets 2021; 33:603-611. [PMID: 34387532 DOI: 10.1080/09537104.2021.1962836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Bactericidal/permeability-increasing protein (BPI) exhibits a number of important characteristics. RNA-seq analysis revealed that the BPI expression was increased in platelets of (non)ST-elevated myocardial infarction (NSTEMI/STEMI) patients. Activated platelets can induce NETosis which may be accompanied by the release of myeloperoxidase-DNA (MPO-DNA) and S100A8/A9. This study investigated the plasma BPI levels in myocardial infarction patients and its correlation with MPO-DNA and S100A8/A9. This prospective study recruited 80 control individuals, as well as 63 NSTEMI and 59 STEMI patients who were admitted to the First Affiliated Hospital of Bengbu Medical College for coronary angiography (CAG) and/or percutaneous coronary intervention (PCI) between May 1, 2020 and August 31, 2020. Demographic and clinical characteristics, clinical indicators, hs-CRP, IL-1β, MPO-DNA (a circulated marker of NETs), circulating levels of S100A8/A9 and BPI were measured from each individual. The severity of coronary lesions was evaluated by the Gensini score, based on the results of the CAG. Pearson's or spearman's correlation was used to examine the correlation between BPI and the above-mentioned parameters, as well as the severity of coronary artery disease. Linear regression analysis was applied to identify the independent predictive factors of BPI. Received operating characteristic (ROC) curve analysis was used to evaluate the diagnostic efficacy of plasma BPI for MI. The plasma BPI levels increased by 8.76 times in the STEMI group and 5.38 times in the NSTEMI group compared to the control group. The plasma level of hs-CRP and IL-1β in both STEMI and NSTEMI groups were also significantly higher than the control group. In addition, the plasma levels of MPO-DNA and S100A8/A9 in the STEMI and NSTEMI groups were significantly higher than the control group. Plasma levels of BPI were positively correlated with IL-1β, hs-CRP, MPO-DNA and S100A8/A9. The correlation between BPI and the severity of coronary artery disease was also significant. The optimal cutoff value of plasma BPI was 35.1705 ng/ml for MI patients from the ROC curve analysis. Plasma BPI levels are increased in myocardial infarction patients and positively correlated with MPO-DNA and S100A8/A9. Plasma BPI level may serve as a potential biomarker of myocardial infarction.
Collapse
Affiliation(s)
- Shicheng Yu
- Anhui Provincial Hospital, Cheeloo College Of Medicine, Shandong University, Jinan, Shandong P.R. China.,Department Of Cardiology, Lu'an People's Hospital, Lu'an, Anhui P.R. China
| | - Miaonan Li
- Department Of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui P.R. China
| | - Zheng Li
- Department Of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui P.R. China
| | - Pan Xu
- Department Of Cardiology, Lu'an People's Hospital, Lu'an, Anhui P.R. China
| | - Zhuoya Yao
- Department Of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui P.R. China
| | - Shaohuan Qian
- Department Of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui P.R. China
| | - Fudong Qian
- Department Of Cardiology, Lu'an People's Hospital, Lu'an, Anhui P.R. China
| | - Dasheng Gao
- Department Of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui P.R. China
| | - Hongju Wang
- Anhui Provincial Hospital, Cheeloo College Of Medicine, Shandong University, Jinan, Shandong P.R. China.,Department Of Cardiology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui P.R. China
| |
Collapse
|
21
|
Tamura Y, Kawano S, Endo T. Lipid homeostasis in mitochondria. Biol Chem 2021; 401:821-833. [PMID: 32229651 DOI: 10.1515/hsz-2020-0121] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/10/2020] [Indexed: 12/13/2022]
Abstract
Mitochondria are surrounded by the two membranes, the outer and inner membranes, whose lipid compositions are optimized for proper functions and structural organizations of mitochondria. Although a part of mitochondrial lipids including their characteristic lipids, phosphatidylethanolamine and cardiolipin, are synthesized within mitochondria, their precursor lipids and other lipids are transported from other organelles, mainly the ER. Mitochondrially synthesized lipids are re-distributed within mitochondria and to other organelles, as well. Recent studies pointed to the important roles of inter-organelle contact sites in lipid trafficking between different organelle membranes. Identification of Ups/PRELI proteins as lipid transfer proteins shuttling between the mitochondrial outer and inner membranes established a part of the molecular and structural basis of the still elusive intra-mitochondrial lipid trafficking.
Collapse
Affiliation(s)
- Yasushi Tamura
- Faculty of Science, Yamagata University, 1-4-12, Kojirakawa-machi, Yamagata, Yamagata 990-8560, Japan
| | - Shin Kawano
- Faculty of Life Sciences, Kyoto Sangyo University, Kamigamo-motoyama, Kita-ku, Kyoto 603-8555, Japan
| | - Toshiya Endo
- Faculty of Life Sciences, Kyoto Sangyo University, Kamigamo-motoyama, Kita-ku, Kyoto 603-8555, Japan.,Institute for Protein Dynamics, Kyoto Sangyo University, Kamigamo-motoyama, Kita-ku, Kyoto 603-8555, Japan
| |
Collapse
|
22
|
Le Roy N, Stapane L, Gautron J, Hincke MT. Evolution of the Avian Eggshell Biomineralization Protein Toolkit - New Insights From Multi-Omics. Front Genet 2021; 12:672433. [PMID: 34046059 PMCID: PMC8144736 DOI: 10.3389/fgene.2021.672433] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/08/2021] [Indexed: 11/13/2022] Open
Abstract
The avian eggshell is a remarkable biomineral, which is essential for avian reproduction; its properties permit embryonic development in the desiccating terrestrial environment, and moreover, are critically important to preserve unfertilized egg quality for human consumption. This calcium carbonate (CaCO3) bioceramic is made of 95% calcite and 3.5% organic matrix; it protects the egg contents against microbial penetration and mechanical damage, allows gaseous exchange, and provides calcium for development of the embryonic skeleton. In vertebrates, eggshell occurs in the Sauropsida and in a lesser extent in Mammalia taxa; avian eggshell calcification is one of the fastest known CaCO3 biomineralization processes, and results in a material with excellent mechanical properties. Thus, its study has triggered a strong interest from the researcher community. The investigation of eggshell biomineralization in birds over the past decades has led to detailed characterization of its protein and mineral constituents. Recently, our understanding of this process has been significantly improved using high-throughput technologies (i.e., proteomics, transcriptomics, genomics, and bioinformatics). Presently, more or less complete eggshell proteomes are available for nine birds, and therefore, key proteins that comprise the eggshell biomineralization toolkit are beginning to be identified. In this article, we review current knowledge on organic matrix components from calcified eggshell. We use these data to analyze the evolution of selected matrix proteins and underline their role in the biological toolkit required for eggshell calcification in avian species. Amongst the panel of eggshell-associated proteins, key functional domains are present such as calcium-binding, vesicle-binding and protein-binding. These technical advances, combined with progress in mineral ultrastructure analyses, have opened the way for new hypotheses of mineral nucleation and crystal growth in formation of the avian eggshell, including transfer of amorphous CaCO3 in vesicles from uterine cells to the eggshell mineralization site. The enrichment of multi-omics datasets for bird species is critical to understand the evolutionary context for development of CaCO3 biomineralization in metazoans, leading to the acquisition of the robust eggshell in birds (and formerly dinosaurs).
Collapse
Affiliation(s)
| | | | | | - Maxwell T Hincke
- Department of Innovation in Medical Education, University of Ottawa, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
23
|
Qian T, Li C, He R, Wan C, Liu Y, Yu H. Calcium-dependent and -independent lipid transfer mediated by tricalbins in yeast. J Biol Chem 2021; 296:100729. [PMID: 33933446 PMCID: PMC8163979 DOI: 10.1016/j.jbc.2021.100729] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/23/2021] [Accepted: 04/28/2021] [Indexed: 11/12/2022] Open
Abstract
Membrane contact sites (MCSs) formed between the endoplasmic reticulum (ER) and the plasma membrane (PM) provide a platform for nonvesicular lipid exchange. The ER-anchored tricalbins (Tcb1, Tcb2, and Tcb3) are critical tethering factors at ER–PM MCSs in yeast. Tricalbins possess a synaptotagmin-like mitochondrial-lipid-binding protein (SMP) domain and multiple Ca2+-binding C2 domains. Although tricalbins have been suggested to be involved in lipid exchange at the ER–PM MCSs, it remains unclear whether they directly mediate lipid transport. Here, using in vitro lipid transfer assays, we discovered that tricalbins are capable of transferring phospholipids between membranes. Unexpectedly, while its lipid transfer activity was markedly elevated by Ca2+, Tcb3 constitutively transferred lipids even in the absence of Ca2+. The stimulatory activity of Ca2+ on Tcb3 required intact Ca2+-binding sites on both the C2C and C2D domains of Tcb3, while Ca2+-independent lipid transport was mediated by the SMP domain that transferred lipids via direct interactions with phosphatidylserine and other negatively charged lipid molecules. These findings establish tricalbins as lipid transfer proteins, and reveal Ca2+-dependent and -independent lipid transfer activities mediated by these tricalbins, providing new insights into their mechanism in maintaining PM integrity at ER–PM MCSs.
Collapse
Affiliation(s)
- Tiantian Qian
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Chenlu Li
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Ruyue He
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Chun Wan
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado, USA
| | - Yinghui Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China.
| | - Haijia Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China.
| |
Collapse
|
24
|
Hakoupian M, Ferino E, Jickling GC, Amini H, Stamova B, Ander BP, Alomar N, Sharp FR, Zhan X. Bacterial lipopolysaccharide is associated with stroke. Sci Rep 2021; 11:6570. [PMID: 33753837 PMCID: PMC7985504 DOI: 10.1038/s41598-021-86083-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 03/08/2021] [Indexed: 01/22/2023] Open
Abstract
We aimed to determine if plasma levels of bacterial lipopolysaccharide (LPS) and lipoteichoic acid (LTA) are associated with different causes of stroke and correlate with C-reactive protein (CRP), LPS-binding protein (LBP), and the NIH stroke scale (NIHSS). Ischemic stroke (cardioembolic (CE), large artery atherosclerosis (LAA), small vessel occlusion (SVO)), intracerebral hemorrhage (ICH), transient ischemic attack (TIA) and control subjects were compared (n = 205). Plasma LPS, LTA, CRP, and LBP levels were quantified by ELISA. LPS and CRP levels were elevated in ischemic strokes (CE, LAA, SVO) and ICH compared to controls. LBP levels were elevated in ischemic strokes (CE, LAA) and ICH. LTA levels were increased in SVO stroke compared to TIA but not controls. LPS levels correlated with CRP and LBP levels in stroke and TIA. LPS, LBP and CRP levels positively correlated with the NIHSS and WBC count but negatively correlated with total cholesterol. Plasma LPS and LBP associate with major causes of ischemic stroke and with ICH, whereas LPS/LBP do not associate with TIAs. LTA only associated with SVO stroke. LPS positively correlated with CRP, LBP, and WBC but negatively correlated with cholesterol. Higher LPS levels were associated with worse stroke outcomes.
Collapse
Affiliation(s)
- Marisa Hakoupian
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Eva Ferino
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Glen C Jickling
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA.,Division of Neurology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Hajar Amini
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Boryana Stamova
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Bradley P Ander
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Noor Alomar
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Frank R Sharp
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA
| | - Xinhua Zhan
- Department of Neurology, University of California Davis School of Medicine, Sacramento, CA, USA. .,Department of Neurology and MIND Institute, University of California Davis Medical Center, 2805 50th Street, Sacramento, CA, 95817, USA.
| |
Collapse
|
25
|
Hewlett B, Singh NP, Vannier C, Galli T. ER-PM Contact Sites - SNARING Actors in Emerging Functions. Front Cell Dev Biol 2021; 9:635518. [PMID: 33681218 PMCID: PMC7928305 DOI: 10.3389/fcell.2021.635518] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/21/2021] [Indexed: 11/13/2022] Open
Abstract
The compartmentalisation achieved by confining cytoplasm into membrane-enclosed organelles in eukaryotic cells is essential for maintaining vital functions including ATP production, synthetic and degradative pathways. While intracellular organelles are highly specialised in these functions, the restricting membranes also impede exchange of molecules responsible for the synchronised and responsive cellular activities. The initial identification of contact sites between the ER and plasma membrane (PM) provided a potential candidate structure for communication between organelles without mixing by fusion. Over the past decades, research has revealed a far broader picture of the events. Membrane contact sites (MCSs) have been recognized as increasingly important actors in cell differentiation, plasticity and maintenance, and, upon dysfunction, responsible for pathological conditions such as cancer and neurodegenerative diseases. Present in multiple organelles and cell types, MCSs promote transport of lipids and Ca2+ homoeostasis, with a range of associated protein families. Interestingly, each MCS displays a unique molecular signature, adapted to organelle functions. This review will explore the literature describing the molecular components and interactions taking place at ER-PM contact sites, their functions, and implications in eukaryotic cells, particularly neurons, with emphasis on lipid transfer proteins and emerging function of SNAREs.
Collapse
Affiliation(s)
- Bailey Hewlett
- INSERM U1266, Institut de Psychiatrie et Neurosciences de Paris, Université de Paris, Paris, France
| | - Neha Pratap Singh
- INSERM U1266, Institut de Psychiatrie et Neurosciences de Paris, Université de Paris, Paris, France
| | - Christian Vannier
- INSERM U1266, Institut de Psychiatrie et Neurosciences de Paris, Université de Paris, Paris, France
| | - Thierry Galli
- INSERM U1266, Institut de Psychiatrie et Neurosciences de Paris, Université de Paris, Paris, France.,GHU PARIS Psychiatrie and Neurosciences, Paris, France
| |
Collapse
|
26
|
Li C, Qian T, He R, Wan C, Liu Y, Yu H. Endoplasmic Reticulum-Plasma Membrane Contact Sites: Regulators, Mechanisms, and Physiological Functions. Front Cell Dev Biol 2021; 9:627700. [PMID: 33614657 PMCID: PMC7889955 DOI: 10.3389/fcell.2021.627700] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/13/2021] [Indexed: 12/13/2022] Open
Abstract
The endoplasmic reticulum (ER) forms direct membrane contact sites with the plasma membrane (PM) in eukaryotic cells. These ER-PM contact sites play essential roles in lipid homeostasis, ion dynamics, and cell signaling, which are carried out by protein-protein or protein-lipid interactions. Distinct tethering factors dynamically control the architecture of ER-PM junctions in response to intracellular signals or external stimuli. The physiological roles of ER-PM contact sites are dependent on a variety of regulators that individually or cooperatively perform functions in diverse cellular processes. This review focuses on proteins functioning at ER-PM contact sites and highlights the recent progress in their mechanisms and physiological roles.
Collapse
Affiliation(s)
- Chenlu Li
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Tiantian Qian
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Ruyue He
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Chun Wan
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, CO, United States
| | - Yinghui Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Haijia Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
27
|
Gigon L, Yousefi S, Karaulov A, Simon HU. Mechanisms of toxicity mediated by neutrophil and eosinophil granule proteins. Allergol Int 2021; 70:30-38. [PMID: 33277190 DOI: 10.1016/j.alit.2020.11.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022] Open
Abstract
Neutrophils and eosinophils are granulocytes which are characterized by the presence of granules in the cytoplasm. Granules provide a safe storage site for granule proteins that play important roles in the immune function of granulocytes. Upon granulocytes activation, diverse proteins are released from the granules into the extracellular space and contribute to the fight against infections. In this article, we describe granule proteins of both neutrophils and eosinophils able to kill pathogens and review their anticipated mechanism of antimicrobial toxicity. It should be noted that an excess of granules protein release can lead to tissue damage of the host resulting in chronic inflammation and organ dysfunction.
Collapse
|
28
|
Shirane M. Lipid Transfer-Dependent Endosome Maturation Mediated by Protrudin and PDZD8 in Neurons. Front Cell Dev Biol 2020; 8:615600. [PMID: 33385000 PMCID: PMC7769939 DOI: 10.3389/fcell.2020.615600] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 11/23/2020] [Indexed: 12/16/2022] Open
Abstract
Endosome maturation refers to the conversion of early endosomes (EEs) to late endosomes (LEs) for subsequent fusion with lysosomes. It is an incremental process that involves a combination of endosome fusion and fission and which occurs at contact sites between endosomes and the endoplasmic reticulum (ER), with knowledge of the underlying mechanisms having increased greatly in recent years. Protrudin is an ER-resident protein that was originally shown to regulate neurite formation by promoting endosome trafficking, whereas PDZD8 is a mammalian paralog of a subunit of the yeast ERMES (ER-mitochondrial encounter structure) complex that possesses lipid transfer activity. A complex of protrudin and PDZD8 was recently found to promote endosome maturation by mediating lipid transfer at ER-endosome membrane contact sites. This review focuses on the roles of the protrudin-PDZD8 complex in tethering of endosomes to the ER, in mediating lipid transfer at such contact sites, and in regulating endosome dynamics, especially in neuronal cells. It also addresses the physiological contribution of endosome maturation mediated by this complex to neuronal polarity and integrity.
Collapse
Affiliation(s)
- Michiko Shirane
- Department of Molecular Biology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Japan
| |
Collapse
|
29
|
Lipopolysaccharide Binding Protein and Bactericidal/Permeability-Increasing Protein as Biomarkers for Invasive Pulmonary Aspergillosis. J Fungi (Basel) 2020; 6:jof6040304. [PMID: 33233831 PMCID: PMC7712449 DOI: 10.3390/jof6040304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/18/2022] Open
Abstract
Early diagnosis of invasive pulmonary aspergillosis (IPA) is crucial to prevent lethal disease in immunocompromized hosts. So far, lipopolysaccharide binding protein (LBP) and bactericidal/permeability-increasing protein (BPI) levels have not been evaluated as biomarkers for IPA. IL-8, previously introduced as a biomarker for IPA, was also included in this study. Bronchoalveolar lavage fluid (BALF) of IPA patients and control patients with non-infectious lung disease was collected according to clinical indications. Measurements in BALF displayed significantly higher levels of LBP (p < 0.0001), BPI (p = 0.0002) and IL-8 (p < 0.0001) in IPA compared to control patients. Receiver operating characteristic curve analysis revealed higher AUC for LBP (0.98, 95% CI 0.95–1.00) than BPI (0.84, 95% CI 0.70–0.97; p = 0.0301). Although not significantly different, AUC of IL-8 (0.93, 95% CI 0.85–1.00) also tended to be higher than AUC for BPI (p = 0.0624). When the subgroup of non-hematological patients was analyzed, test performance of LBP (AUC 0.99, 95% CI 0.97–1.00), BPI (AUC 0.97, 95% CI 0.91–1.00) and IL-8 (AUC 0.96, 95% CI: 0.90–1.00) converged. In conclusion, LBP and—to a lesser extend—BPI displayed high AUCs that were comparable to those of IL-8 for diagnosis of IPA in BALF. Further investigations are worthwhile, especially in non-hematological patients in whom sensitive biomarkers for IPA are lacking.
Collapse
|
30
|
Protrudin and PDZD8 contribute to neuronal integrity by promoting lipid extraction required for endosome maturation. Nat Commun 2020; 11:4576. [PMID: 32917905 PMCID: PMC7486383 DOI: 10.1038/s41467-020-18413-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/21/2020] [Indexed: 01/08/2023] Open
Abstract
Endosome maturation depends on membrane contact sites (MCSs) formed between endoplasmic reticulum (ER) and endolysosomes (LyLEs). The mechanism underlying lipid supply for this process and its pathophysiological relevance remains unclear, however. Here, we identify PDZD8-the mammalian ortholog of a yeast ERMES subunit-as a protein that interacts with protrudin, which is located at ER-LyLE MCSs. Protrudin and PDZD8 promote the formation of ER-LyLE MCSs, and PDZD8 shows the ability to extract various lipids from the ER. Overexpression of both protrudin and PDZD8 in HeLa cells, as well as their depletion in mouse primary neurons, impairs endosomal homeostasis by inducing the formation of abnormal large vacuoles reminiscent of those apparent in spastin- or REEP1-deficient neurons. The protrudin-PDZD8 system is also essential for the establishment of neuronal polarity. Our results suggest that protrudin and PDZD8 cooperatively promote endosome maturation by mediating ER-LyLE tethering and lipid extraction at MCSs, thereby maintaining neuronal polarity and integrity.
Collapse
|
31
|
Dupas S, Neiers F, Granon E, Rougeux E, Dupont S, Beney L, Bousquet F, Shaik HA, Briand L, Wojtasek H, Charles JP. Collisional mechanism of ligand release by Bombyxmori JHBP, a member of the TULIP / Takeout family of lipid transporters. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2020; 117:103293. [PMID: 31809784 DOI: 10.1016/j.ibmb.2019.103293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/29/2019] [Accepted: 12/01/2019] [Indexed: 06/10/2023]
Abstract
Juvenile hormones (JHs) regulate important processes in insects, such as postembryonic development and reproduction. In the hemolymph of Lepidoptera, these lipophilic sesquiterpenic hormones are transported from their site of synthesis to target tissues by high affinity carriers, the juvenile hormone binding proteins (JHBPs). Lepidopteran JHBPs belong to a recently uncovered, yet very ancient family of proteins sharing a common lipid fold (TULIP domain) and involved in shuttling various lipid ligands. One important, but poorly understood aspect of JHs action, is the mechanism of hormone transfer to or through the plasma membranes of target cells. Since many membrane-active peptides and proteins, such as the pore-forming bacterial toxins, are activated by low pH or interaction with phospholipid membranes, we have examined the effect of these factors on JH binding by JHBPs. The affinity of Bombyx mori and Manduca sexta JHBPs for JH III was determined by the DCC assay, equilibrium dialysis, and isothermal titration calorimetry, and found to be greatly reduced at low pH, in agreement with previous observations. Loss of binding was accompanied by changes in fluorescence and near-UV CD spectra, indicating significant changes in protein structure in the environment of aromatic residues. The apparent dissociation rate constant (koff) of the JHBP-JH III complex was greater at acidic pH, suggesting that low pH favors ligand release by opening of the binding pocket. The affinity of recombinant B. mori JHBP (rBmJHBP) was also decreased in the presence of anionic phospholipid vesicles. Measurements of steady-state fluorescence anisotropy with the lipophilic probe TMA-DPH demonstrated that rBmJHBP specifically interacts with anionic membranes. These results suggest the existence of a collisional mechanism for ligand release that may be important for delivery of JHs to the target cells, and could be relevant to the function of related members of this emerging family of lipid-transport proteins.
Collapse
Affiliation(s)
- Stéphane Dupas
- Université de Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, UMR 6265 CNRS, UMR 1324 INRA, 6, Bd Gabriel, F-21000, Dijon, France
| | - Fabrice Neiers
- Université de Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, UMR 6265 CNRS, UMR 1324 INRA, 6, Bd Gabriel, F-21000, Dijon, France
| | - Emma Granon
- Université de Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, UMR 6265 CNRS, UMR 1324 INRA, 6, Bd Gabriel, F-21000, Dijon, France
| | - Erwan Rougeux
- Université de Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, UMR 6265 CNRS, UMR 1324 INRA, 6, Bd Gabriel, F-21000, Dijon, France
| | - Sébastien Dupont
- Université de Bourgogne Franche-Comté, AgroSup Dijon, PAM UMR A 02.102, F-21000, Dijon, France
| | - Laurent Beney
- Université de Bourgogne Franche-Comté, AgroSup Dijon, PAM UMR A 02.102, F-21000, Dijon, France
| | - François Bousquet
- Université de Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, UMR 6265 CNRS, UMR 1324 INRA, 6, Bd Gabriel, F-21000, Dijon, France
| | - Haq Abdul Shaik
- Université de Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, UMR 6265 CNRS, UMR 1324 INRA, 6, Bd Gabriel, F-21000, Dijon, France
| | - Loic Briand
- Université de Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, UMR 6265 CNRS, UMR 1324 INRA, 6, Bd Gabriel, F-21000, Dijon, France
| | - Hubert Wojtasek
- Institute of Chemistry, Opole University, Ul. Oleska 48, 45-052, Opole, Poland.
| | - Jean-Philippe Charles
- Université de Bourgogne Franche-Comté, Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, UMR 6265 CNRS, UMR 1324 INRA, 6, Bd Gabriel, F-21000, Dijon, France.
| |
Collapse
|
32
|
Oliveira HCF, Raposo HF. Cholesteryl Ester Transfer Protein and Lipid Metabolism and Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:15-25. [PMID: 32705591 DOI: 10.1007/978-981-15-6082-8_2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
In this chapter, we present the major advances in CETP research since the detection, isolation, and characterization of its activity in the plasma of humans and several species. Since CETP is a major modulator of HDL plasma levels, the clinical importance of CETP activity was recognized very early. We describe the participation of CETP in reverse cholesterol transport, conflicting results in animal and human genetic studies, possible new functions of CETP, and the results of the main clinical trials on CETP inhibition. Despite major setbacks in clinical trials, the hypothesis that CETP inhibitors are anti-atherogenic in humans is still being tested.
Collapse
Affiliation(s)
- Helena C F Oliveira
- Department of Structural and Functional Biology, Biology Institute, State University of Campinas, Campinas, SP, Brazil.
| | - Helena F Raposo
- Department of Structural and Functional Biology, Biology Institute, State University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
33
|
Yu Y, Song G. Lipopolysaccharide-Binding Protein and Bactericidal/Permeability-Increasing Protein in Lipid Metabolism and Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:27-35. [PMID: 32705592 DOI: 10.1007/978-981-15-6082-8_3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Lipopolysaccharide-binding protein (LBP) and bactericidal/permeability-increasing protein (BPI) are the main members of BPI-like family based on the similar protein structure and conserved gene homology. Both LBP and BPI participate in lipid metabolism and thereby involve in pathogenesis of certain cardiovascular diseases. This chapter describes four aspects: (1) the loci of BPI and LBP in genome, (2) the characteristics of the cDNAs and expression patterns of LBP and BPI, (3) the structures and functions of LBP and BPI, and (4) the LBP and BPI in lipid metabolism and cardiovascular research.
Collapse
Affiliation(s)
- Yang Yu
- Institute of Atherosclerosis, Shandong First Medical University, Shandong, China.
| | - Guohua Song
- Institute of Atherosclerosis, Shandong First Medical University, Shandong, China
| |
Collapse
|
34
|
Pemberton JG, Kim YJ, Balla T. Integrated regulation of the phosphatidylinositol cycle and phosphoinositide-driven lipid transport at ER-PM contact sites. Traffic 2019; 21:200-219. [PMID: 31650663 DOI: 10.1111/tra.12709] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/02/2019] [Accepted: 10/16/2019] [Indexed: 12/20/2022]
Abstract
Among the structural phospholipids that form the bulk of eukaryotic cell membranes, phosphatidylinositol (PtdIns) is unique in that it also serves as the common precursor for low-abundance regulatory lipids, collectively referred to as polyphosphoinositides (PPIn). The metabolic turnover of PPIn species has received immense attention because of the essential functions of these lipids as universal regulators of membrane biology and their dysregulation in numerous human pathologies. The diverse functions of PPIn lipids occur, in part, by orchestrating the spatial organization and conformational dynamics of peripheral or integral membrane proteins within defined subcellular compartments. The emerging role of stable contact sites between adjacent membranes as specialized platforms for the coordinate control of ion exchange, cytoskeletal dynamics, and lipid transport has also revealed important new roles for PPIn species. In this review, we highlight the importance of membrane contact sites formed between the endoplasmic reticulum (ER) and plasma membrane (PM) for the integrated regulation of PPIn metabolism within the PM. Special emphasis will be placed on non-vesicular lipid transport during control of the PtdIns biosynthetic cycle as well as toward balancing the turnover of the signaling PPIn species that define PM identity.
Collapse
Affiliation(s)
- Joshua G Pemberton
- Section on Molecular Signal Transduction, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Bethesda, Maryland
| | - Yeun Ju Kim
- Section on Molecular Signal Transduction, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Bethesda, Maryland
| | - Tamas Balla
- Section on Molecular Signal Transduction, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
35
|
PDZD8 mediates a Rab7-dependent interaction of the ER with late endosomes and lysosomes. Proc Natl Acad Sci U S A 2019; 116:22619-22623. [PMID: 31636202 DOI: 10.1073/pnas.1913509116] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Contacts between the endoplasmic reticulum (ER) and other membranes are hot spots for protein-mediated lipid transport between the 2 adjacent bilayers. Compiling a molecular inventory of lipid transport proteins present at these sites is a premise to the elucidation of their function. Here we show that PDZD8, an intrinsic membrane protein of the ER with a lipid transport module of the SMP domain family, concentrates at contacts between the ER and late endosomes/lysosomes, where it interacts with GTP-Rab7. These findings suggest that PDZD8 may cooperate with other proteins that function at the ER-endo/lysosome interface in coordinating endocytic flow with lipid transport between endocytic membranes and the ER.
Collapse
|
36
|
A programmable DNA-origami platform for studying lipid transfer between bilayers. Nat Chem Biol 2019; 15:830-837. [PMID: 31320758 PMCID: PMC6650167 DOI: 10.1038/s41589-019-0325-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 06/14/2019] [Indexed: 02/07/2023]
Abstract
Non-vesicular lipid transport between bilayers at membrane contact sites plays important physiological roles. Mechanistic insight into the action of lipid transport proteins localized at these sites (bridge/tunnel versus shuttle models) requires a determination of the distance between bilayers at which this transport can occur. Here, we developed DNA-origami nanostructures to organize size-defined liposomes at precise distances and used them to study lipid transfer by the SMP domain of E-Syt1. Pairs of DNA-ring-templated donor and acceptor liposomes were docked through DNA pillars, which determined their distance. The SMP domain was anchored to donor liposomes via an unstructured linker and lipid transfer was assessed via a FRET-based assay. We show that lipid transfer can occur over distances that exceed the length of SMP dimer, compatible with a shuttle model. The DNA nanostructures developed here can be adapted to study other processes occurring where two membranes are closely apposed to each other.
Collapse
|
37
|
Zhang H, Burrows J, Card GL, Attwood G, Wheeler TT, Arcus VL. The three dimensional structure of Bovine Salivary Protein 30b (BSP30b) and its interaction with specific rumen bacteria. PLoS One 2019; 14:e0206709. [PMID: 30978191 PMCID: PMC6461236 DOI: 10.1371/journal.pone.0206709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 03/26/2019] [Indexed: 11/18/2022] Open
Abstract
Bovine Salivary Protein 30b (BSP30b) is a member of the tubular lipid-binding (TULIP) superfamily that includes the human bactericidal/permeability-increasing proteins (BPI), lipopolysaccharide binding proteins (LBP) and palate, lung, and nasal epithelium carcinoma-associated proteins (PLUNC). BSP30b is most closely related to the PLUNC family and is predominantly found in bovine saliva. There are four BSP30 isoforms (BSP30a-d) and collectively, they are the most abundant protein component of bovine saliva. The PLUNC family members are proposed to be lipid binding proteins, although in most cases their lipid ligands are unknown. Here, we present the X-ray crystal structure of BSP30b at 2.0 Å resolution. We used a double methionine mutant and Se-Met SAD phasing to solve the structure. The structure adopts a curved cylindrical form with a hydrophobic channel formed by an α/β wrap, which is consistent with the TULIP superfamily. The structure of BSP30b in complex with oleic acid is also presented where the ligand is accommodated within the hydrophobic channel. The electron density for oleic acid suggests that the ligand is only partially occupied in the binding site implying that oleic acid may not be the preferred ligand. GFP-tagged BSP30b binds to the surface of olive oil droplets, as observed under fluorescent microscopy, and acts as a surfactant consistent with its association with decreased susceptibility to bloat in cattle. Bacteria extracted directly from bovine rumen contents indicate that the GFP_BSP30b fusion protein binds to a small number of selected bacterial species in vivo. These results suggest that BSP30b may bind to bacterial lipids from specific species and that this abundant protein may have important biological roles via interacting with rumen bacteria during feeding and rumination.
Collapse
Affiliation(s)
- Heng Zhang
- School of Science, University of Waikato, Hamilton, New Zealand
| | - Judith Burrows
- School of Science, University of Waikato, Hamilton, New Zealand
| | - Graeme L. Card
- Stanford Synchrotron Radiation Lightsource, Menlo Park, California, United States of America
| | - Graeme Attwood
- AgResearch Grasslands, Tennent Drive, Palmerston North, New Zealand
| | - Tom T. Wheeler
- Cawthron Research Institute, The Wood, Nelson, New Zealand
| | - Vickery L. Arcus
- School of Science, University of Waikato, Hamilton, New Zealand
- * E-mail:
| |
Collapse
|
38
|
Lipid transporter TMEM24/C2CD2L is a Ca 2+-regulated component of ER-plasma membrane contacts in mammalian neurons. Proc Natl Acad Sci U S A 2019; 116:5775-5784. [PMID: 30819882 DOI: 10.1073/pnas.1820156116] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Close appositions between the endoplasmic reticulum (ER) and the plasma membrane (PM) are a general feature of all cells and are abundant in neurons. A function of these appositions is lipid transport between the two adjacent bilayers via tethering proteins that also contain lipid transport modules. However, little is known about the properties and dynamics of these proteins in neurons. Here we focused on TMEM24/C2CD2L, an ER-localized SMP domain containing phospholipid transporter expressed at high levels in the brain, previously shown to be a component of ER-PM contacts in pancreatic β-cells. TMEM24 is enriched in neurons versus glial cells and its levels increase in parallel with neuronal differentiation. It populates ER-PM contacts in resting neurons, but elevations of cytosolic Ca2+ mediated by experimental manipulations or spontaneous activity induce its transient redistribution throughout the entire ER. Dissociation of TMEM24 from the plasma membrane is mediated by phosphorylation of an array of sites in the C-terminal region of the protein. These sites are only partially conserved in C2CD2, the paralogue of TMEM24 primarily expressed in nonneuronal tissues, which correspondingly display a much lower sensitivity to Ca2+ elevations. ER-PM contacts in neurons are also sites where Kv2 (the major delayed rectifier K+ channels in brain) and other PM and ER ion channels are concentrated, raising the possibility of a regulatory feedback mechanism between neuronal excitability and lipid exchange between the ER and the PM.
Collapse
|
39
|
Tamura Y, Kawano S, Endo T. Organelle contact zones as sites for lipid transfer. J Biochem 2019; 165:115-123. [PMID: 30371789 DOI: 10.1093/jb/mvy088] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 10/26/2018] [Indexed: 01/06/2023] Open
Abstract
Since the 1950s, electron microscopic observations have suggested the existence of special regions where the distinct organelle membranes are closely apposed to each other, yet their molecular basis and functions have not been examined for a long time. Recent studies using yeast as a model organism identified multiple organelle-membrane tethering sites/factors, such as ERMES (ER-mitochondria encounter structure), NVJ (Nuclear-vacuole junction), vCLAMP (Vacuole and mitochondria patch) and MICOS (Mitochondrial contact site). Among them, ERMES is the best-characterized contact-site protein complex, which was found to function as not only an organelle-tethering factor but also a phospholipid transfer protein complex. In this review, we will discuss recent advances in the characterization of ERMES and other organelle contact zones, vCLAMP, NVJ and MICOS in yeast.
Collapse
Affiliation(s)
- Yasushi Tamura
- Department of Material and Biological Chemistry, Faculty of Science, Yamagata University, 1-4-12 Kojirakawa-machi, Yamagata, Japan
| | - Shin Kawano
- Faculty of Life Sciences, Kyoto Sangyo University, Kamigamo-motoyama, Kita-ku, Kyoto, Japan.,Research Center for Protein Dynamics, Kyoto Sangyo University, Kamigamo-motoyama, Kita-ku, Kyoto, Japan
| | - Toshiya Endo
- Faculty of Life Sciences, Kyoto Sangyo University, Kamigamo-motoyama, Kita-ku, Kyoto, Japan.,Research Center for Protein Dynamics, Kyoto Sangyo University, Kamigamo-motoyama, Kita-ku, Kyoto, Japan
| |
Collapse
|
40
|
Jappe U, Schwager C, Schromm AB, González Roldán N, Stein K, Heine H, Duda KA. Lipophilic Allergens, Different Modes of Allergen-Lipid Interaction and Their Impact on Asthma and Allergy. Front Immunol 2019; 10:122. [PMID: 30837983 PMCID: PMC6382701 DOI: 10.3389/fimmu.2019.00122] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 01/15/2019] [Indexed: 12/12/2022] Open
Abstract
Molecular allergology research has provided valuable information on the structure and function of single allergenic molecules. There are several allergens in food and inhalant allergen sources that are able to interact with lipid ligands via different structural features: hydrophobic pockets, hydrophobic cavities, or specialized domains. For only a few of these allergens information on their associated ligands is already available. Several of the allergens are clinically relevant, so that it is highly probable that the individual structural features with which they interact with lipids have a direct effect on their allergenic potential, and thus on allergy development. There is some evidence for a protective effect of lipids delaying the enzymatic digestion of the peanut (Arachis hypogaea) allergen Ara h 8 (hydrophobic pocket), probably allowing this molecule to get to the intestinal immune system intact (sensitization). Oleosins from different food allergen sources are part of lipid storage organelles and potential marker allergens for the severity of the allergic reaction. House dust mite (HDM), is more often associated with allergic asthma than other sources of inhalant allergens. In particular, lipid-associated allergens from Dermatophagoides pteronyssinus which are Der p 2, Der p 5, Der p 7, Der p 13, Der p 14, and Der p 21 have been reported to be associated with severe allergic reactions and respiratory symptoms such as asthma. The exact mechanism of interaction of these allergens with lipids still has to be elucidated. Apart from single allergens glycolipids have been shown to directly induce allergic inflammation. Several-in parts conflicting-data exist on the lipid (and allergen) and toll-like receptor interactions. For only few single allergens mechanistic studies were performed on their interaction with the air-liquid interface of the lungs, in particular with the surfactant components SP-A and SP-D. The increasing knowledge on protein-lipid-interaction for lipophilic and hydrophobic food and inhalant allergens on the basis of their particular structure, of their capacity to be integral part of membranes (like the oleosins), and their ability to interact with membranes, surfactant components, and transport lipids (like the lipid transfer proteins) are essential to eventually clarify allergy and asthma development.
Collapse
Affiliation(s)
- Uta Jappe
- Division of Clinical and Molecular Allergology, Research Center Borstel, Leibniz Lung Center, Airway Research Center North, German Center for Lung Research, Borstel, Germany
- Interdisciplinary Allergy Outpatient Clinic, Department of Pneumology, University of Luebeck, Borstel, Germany
| | - Christian Schwager
- Division of Clinical and Molecular Allergology, Research Center Borstel, Leibniz Lung Center, Airway Research Center North, German Center for Lung Research, Borstel, Germany
| | - Andra B. Schromm
- Division of Immunobiophysics, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Nestor González Roldán
- Junior Research Group of Allergobiochemistry, Research Center Borstel, Leibniz Lung Center, Airway Research Center North, German Center for Lung Research, Borstel, Germany
| | - Karina Stein
- Division of Innate Immunity, Research Center Borstel, Leibniz Lung Center, Airway Research Center North, German Center for Lung Research, Borstel, Germany
| | - Holger Heine
- Division of Innate Immunity, Research Center Borstel, Leibniz Lung Center, Airway Research Center North, German Center for Lung Research, Borstel, Germany
| | - Katarzyna A. Duda
- Junior Research Group of Allergobiochemistry, Research Center Borstel, Leibniz Lung Center, Airway Research Center North, German Center for Lung Research, Borstel, Germany
| |
Collapse
|
41
|
Bülow S, Zeller L, Werner M, Toelge M, Holzinger J, Entzian C, Schubert T, Waldow F, Gisch N, Hammerschmidt S, Gessner A. Bactericidal/Permeability-Increasing Protein Is an Enhancer of Bacterial Lipoprotein Recognition. Front Immunol 2018; 9:2768. [PMID: 30581431 PMCID: PMC6293271 DOI: 10.3389/fimmu.2018.02768] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 11/12/2018] [Indexed: 12/12/2022] Open
Abstract
Adequate perception of immunologically important pathogen-associated molecular patterns like lipopolysaccharide and bacterial lipoproteins is essential for efficient innate and adaptive immune responses. In the context of Gram-negative infection, bactericidal/permeability-increasing protein (BPI) neutralizes endotoxic activity of lipopolysaccharides, and thus prohibits hyperactivation. So far, no immunological function of BPI has been described in Gram-positive infections. Here, we show a significant elevation of BPI in Gram-positive meningitis and, surprisingly, a positive correlation between BPI and pro-inflammatory markers like TNFα. To clarify the underlying mechanisms, we identify BPI ligands of Gram-positive origin, specifically bacterial lipopeptides and lipoteichoic acids, and determine essential structural motifs for this interaction. Importantly, the interaction of BPI with these newly defined ligands significantly enhances the immune response in peripheral blood mononuclear cells (PBMCs) mediated by Gram-positive bacteria, and thereby ensures their sensitive perception. In conclusion, we define BPI as an immune enhancing pattern recognition molecule in Gram-positive infections.
Collapse
Affiliation(s)
- Sigrid Bülow
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Lisa Zeller
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Maren Werner
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Martina Toelge
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | - Jonas Holzinger
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| | | | | | - Franziska Waldow
- Division of Bioanalytical Chemistry, Priority Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Nicolas Gisch
- Division of Bioanalytical Chemistry, Priority Area Infections, Research Center Borstel, Leibniz Lung Center, Borstel, Germany
| | - Sven Hammerschmidt
- Department of Molecular Genetics and Infection Biology, Center for Functional Genomcis of Microbes, Interfaculty Institute for Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - André Gessner
- Institute of Clinical Microbiology and Hygiene, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
42
|
Stefan CJ. Building ER-PM contacts: keeping calm and ready on alarm. Curr Opin Cell Biol 2018; 53:1-8. [DOI: 10.1016/j.ceb.2018.03.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2018] [Revised: 03/20/2018] [Accepted: 03/22/2018] [Indexed: 11/28/2022]
|
43
|
Wideman JG, Balacco DL, Fieblinger T, Richards TA. PDZD8 is not the 'functional ortholog' of Mmm1, it is a paralog. F1000Res 2018; 7:1088. [PMID: 30109028 PMCID: PMC6069729 DOI: 10.12688/f1000research.15523.1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/06/2018] [Indexed: 11/20/2022] Open
Abstract
Authors of a recent paper demonstrate that, like ERMES (ER-mitochondria encounter structure) in fungal cells, PDZD8 (PDZ domain containing 8) tethers mitochondria to the ER in mammalian cells. However, identifying PDZD8 as a "functional ortholog" of yeast Mmm1 (maintenance of mitochondrial morphology protein 1) is at odds with the phylogenetic data. PDZD8 and Mmm1 are paralogs, not orthologs, which affects the interpretation of the data with respect to the evolution of ER-mitochondria tethering. Our phylogenetic analyses show that PDZD8 co-occurs with ERMES components in lineages closely related to animals solidifying its identity as a paralog of Mmm1. Additionally, we identify two related paralogs, one specific to flagellated fungi, and one present only in unicellular relatives of animals. These results point to a complex evolutionary history of ER-mitochondria tethering involving multiple gene gains and losses in the lineage leading to animals and fungi.
Collapse
Affiliation(s)
- Jeremy G Wideman
- Department of Biosciences, University of Exeter, Exeter, EX4 4QD, UK.,Wissenschaftskolleg zu Berlin, Berlin, 14193, Germany
| | - Dario L Balacco
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Tim Fieblinger
- Wissenschaftskolleg zu Berlin, Berlin, 14193, Germany.,Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, 22184, Sweden
| | - Thomas A Richards
- Department of Biosciences, University of Exeter, Exeter, EX4 4QD, UK
| |
Collapse
|
44
|
Hirabayashi Y, Kwon SK, Paek H, Pernice WM, Paul MA, Lee J, Erfani P, Raczkowski A, Petrey DS, Pon LA, Polleux F. ER-mitochondria tethering by PDZD8 regulates Ca 2+ dynamics in mammalian neurons. Science 2018; 358:623-630. [PMID: 29097544 DOI: 10.1126/science.aan6009] [Citation(s) in RCA: 331] [Impact Index Per Article: 47.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Revised: 08/21/2017] [Accepted: 09/20/2017] [Indexed: 01/06/2023]
Abstract
Interfaces between organelles are emerging as critical platforms for many biological responses in eukaryotic cells. In yeast, the ERMES complex is an endoplasmic reticulum (ER)-mitochondria tether composed of four proteins, three of which contain a SMP (synaptotagmin-like mitochondrial-lipid binding protein) domain. No functional ortholog for any ERMES protein has been identified in metazoans. Here, we identified PDZD8 as an ER protein present at ER-mitochondria contacts. The SMP domain of PDZD8 is functionally orthologous to the SMP domain found in yeast Mmm1. PDZD8 was necessary for the formation of ER-mitochondria contacts in mammalian cells. In neurons, PDZD8 was required for calcium ion (Ca2+) uptake by mitochondria after synaptically induced Ca2+-release from ER and thereby regulated cytoplasmic Ca2+ dynamics. Thus, PDZD8 represents a critical ER-mitochondria tethering protein in metazoans. We suggest that ER-mitochondria coupling is involved in the regulation of dendritic Ca2+ dynamics in mammalian neurons.
Collapse
Affiliation(s)
- Yusuke Hirabayashi
- Department of Neuroscience, Columbia University Medical Center, Columbia University, New York, NY 10027, USA.,Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA.,Kavli Institute for Brain Science, Columbia University, New York, NY 10027, USA.,Japan Science and Technology Agency (JST), Precursory Research for Embryonic Science and Technology (PRESTO), Tokyo, Japan
| | - Seok-Kyu Kwon
- Department of Neuroscience, Columbia University Medical Center, Columbia University, New York, NY 10027, USA.,Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA.,Kavli Institute for Brain Science, Columbia University, New York, NY 10027, USA
| | - Hunki Paek
- Department of Neuroscience, Columbia University Medical Center, Columbia University, New York, NY 10027, USA.,Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA.,Kavli Institute for Brain Science, Columbia University, New York, NY 10027, USA
| | - Wolfgang M Pernice
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Maëla A Paul
- Department of Neuroscience, Columbia University Medical Center, Columbia University, New York, NY 10027, USA.,Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA.,Kavli Institute for Brain Science, Columbia University, New York, NY 10027, USA
| | - Jinoh Lee
- Department of Neuroscience, Columbia University Medical Center, Columbia University, New York, NY 10027, USA.,Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA.,Kavli Institute for Brain Science, Columbia University, New York, NY 10027, USA
| | - Parsa Erfani
- Department of Neuroscience, Columbia University Medical Center, Columbia University, New York, NY 10027, USA.,Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA.,Kavli Institute for Brain Science, Columbia University, New York, NY 10027, USA
| | - Ashleigh Raczkowski
- Simons Electron Microscopy Center, New York Structural Biology Center (NYSBC), New York, NY 10027, USA
| | - Donald S Petrey
- Center for Computational Biology and Bioinformatics, Department of Systems Biology, Columbia University, New York, NY 10032, USA.,Howard Hughes Medical Institute, Columbia University, New York, NY 10032, USA
| | - Liza A Pon
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, USA.,Institute of Human Nutrition, Columbia University, New York, NY 10032, USA
| | - Franck Polleux
- Department of Neuroscience, Columbia University Medical Center, Columbia University, New York, NY 10027, USA. .,Mortimer B. Zuckerman Mind Brain Behavior Institute, Columbia University, New York, NY 10027, USA.,Kavli Institute for Brain Science, Columbia University, New York, NY 10027, USA
| |
Collapse
|
45
|
Pridans C, Sauter KA, Irvine KM, Davis GM, Lefevre L, Raper A, Rojo R, Nirmal AJ, Beard P, Cheeseman M, Hume DA. Macrophage colony-stimulating factor increases hepatic macrophage content, liver growth, and lipid accumulation in neonatal rats. Am J Physiol Gastrointest Liver Physiol 2018; 314:G388-G398. [PMID: 29351395 PMCID: PMC5899243 DOI: 10.1152/ajpgi.00343.2017] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Signaling via the colony-stimulating factor 1 receptor (CSF1R) controls the survival, differentiation, and proliferation of macrophages. Mutations in CSF1 or CSF1R in mice and rats have pleiotropic effects on postnatal somatic growth. We tested the possible application of pig CSF1-Fc fusion protein as a therapy for low birth weight (LBW) at term, using a model based on maternal dexamethasone treatment in rats. Neonatal CSF1-Fc treatment did not alter somatic growth and did not increase the blood monocyte count. Instead, there was a substantial increase in the size of liver in both control and LBW rats, and the treatment greatly exacerbated lipid droplet accumulation seen in the dexamethasone LBW model. These effects were reversed upon cessation of treatment. Transcriptional profiling of the livers supported histochemical evidence of a large increase in macrophages with a resident Kupffer cell phenotype and revealed increased expression of many genes implicated in lipid droplet formation. There was no further increase in hepatocyte proliferation over the already high rates in neonatal liver. In conclusion, treatment of neonatal rats with CSF1-Fc caused an increase in liver size and hepatic lipid accumulation, due to Kupffer cell expansion and/or activation rather than hepatocyte proliferation. Increased liver macrophage numbers and expression of endocytic receptors could mitigate defective clearance functions in neonates. NEW & NOTEWORTHY This study is based on extensive studies in mice and pigs of the role of CSF1/CSF1R in macrophage development and postnatal growth. We extended the study to neonatal rats as a possible therapy for low birth weight. Unlike our previous studies in mice and pigs, there was no increase in hepatocyte proliferation and no increase in monocyte numbers. Instead, neonatal rats treated with CSF1 displayed reversible hepatic steatosis and Kupffer cell expansion.
Collapse
Affiliation(s)
- Clare Pridans
- 1The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom,2Medical Research Council Centre for Inflammation Research, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, United Kingdom
| | - Kristin A. Sauter
- 1The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Katharine M. Irvine
- 3Mater Research-University of Queensland, Translational Research Institute, Woolloongabba, Australia
| | - Gemma M. Davis
- 1The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Lucas Lefevre
- 1The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Anna Raper
- 1The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Rocio Rojo
- 1The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Ajit J. Nirmal
- 1The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Philippa Beard
- 1The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom,4The Pirbright Institute, Surrey, United Kingdom
| | - Michael Cheeseman
- 1The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - David A. Hume
- 1The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom,2Medical Research Council Centre for Inflammation Research, University of Edinburgh, The Queen's Medical Research Institute, Edinburgh, United Kingdom,3Mater Research-University of Queensland, Translational Research Institute, Woolloongabba, Australia
| |
Collapse
|
46
|
Bian X, Saheki Y, De Camilli P. Ca 2+ releases E-Syt1 autoinhibition to couple ER-plasma membrane tethering with lipid transport. EMBO J 2018; 37:219-234. [PMID: 29222176 PMCID: PMC5770786 DOI: 10.15252/embj.201797359] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 11/08/2017] [Accepted: 11/10/2017] [Indexed: 11/09/2022] Open
Abstract
The extended synaptotagmins (E-Syts) are endoplasmic reticulum (ER) proteins that bind the plasma membrane (PM) via C2 domains and transport lipids between them via SMP domains. E-Syt1 tethers and transports lipids in a Ca2+-dependent manner, but the role of Ca2+ in this regulation is unclear. Of the five C2 domains of E-Syt1, only C2A and C2C contain Ca2+-binding sites. Using liposome-based assays, we show that Ca2+ binding to C2C promotes E-Syt1-mediated membrane tethering by releasing an inhibition that prevents C2E from interacting with PI(4,5)P2-rich membranes, as previously suggested by studies in semi-permeabilized cells. Importantly, Ca2+ binding to C2A enables lipid transport by releasing a charge-based autoinhibitory interaction between this domain and the SMP domain. Supporting these results, E-Syt1 constructs defective in Ca2+ binding in either C2A or C2C failed to rescue two defects in PM lipid homeostasis observed in E-Syts KO cells, delayed diacylglycerol clearance from the PM and impaired Ca2+-triggered phosphatidylserine scrambling. Thus, a main effect of Ca2+ on E-Syt1 is to reverse an autoinhibited state and to couple membrane tethering with lipid transport.
Collapse
Affiliation(s)
- Xin Bian
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT, USA
| | - Yasunori Saheki
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT, USA
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Pietro De Camilli
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
- Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, New Haven, CT, USA
- Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
47
|
Masana L, Girona J, Ibarretxe D, Rodríguez-Calvo R, Rosales R, Vallvé JC, Rodríguez-Borjabad C, Guardiola M, Rodríguez M, Guaita-Esteruelas S, Oliva I, Martínez-Micaelo N, Heras M, Ferré R, Ribalta J, Plana N. Clinical and pathophysiological evidence supporting the safety of extremely low LDL levels-The zero-LDL hypothesis. J Clin Lipidol 2018; 12:292-299.e3. [PMID: 29398429 DOI: 10.1016/j.jacl.2017.12.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/18/2017] [Accepted: 12/20/2017] [Indexed: 11/30/2022]
Abstract
While the impact of very low concentrations of low-density lipoprotein cholesterol (LDL-C) on cardiovascular prevention is very reassuring, it is intriguing to know what effect these extremely low LDL-C concentrations have on lipid homoeostasis. The evidence supporting the safety of extremely low LDL levels comes from genetic studies and clinical drug trials. Individuals with lifelong low LDL levels due to mutations in genes associated with increased LDL-LDL receptor (LDLR) activity reveal no safety issues. Patients achieving extremely low LDL levels in the IMPROVE-IT and FOURIER, and the PROFICIO and ODYSSEY programs seem not to have an increased prevalence of adverse effects. The main concern regarding extremely low LDL-C plasma concentrations is the adequacy of the supply of cholesterol, and other molecules, to peripheral tissues. However, LDL proteomic and kinetic studies reaffirm that LDL is the final product of endogenous lipoprotein metabolism. Four of 5 LDL particles are cleared through the LDL-LDLR pathway in the liver. Given that mammalian cells have no enzymatic systems to degrade cholesterol, the LDL-LDLR pathway is the main mechanism for removal of cholesterol from the body. Our focus, therefore, is to review, from a physiological perspective, why such extremely low LDL-C concentrations do not appear to be detrimental. We suggest that extremely low LDL-C levels due to increased LDLR activity may be a surrogate of adequate LDL-LDLR pathway function.
Collapse
Affiliation(s)
- Luis Masana
- Unitat de Medicina Vascular i Metabolisme, Unitat de Recerca en Lipids i Arteriosclerosis, Sant Joan University Hospital, IISPV, CIBERDEM, Universitat Rovira I Virgili, Reus, Spain.
| | - Josefa Girona
- Unitat de Medicina Vascular i Metabolisme, Unitat de Recerca en Lipids i Arteriosclerosis, Sant Joan University Hospital, IISPV, CIBERDEM, Universitat Rovira I Virgili, Reus, Spain
| | - Daiana Ibarretxe
- Unitat de Medicina Vascular i Metabolisme, Unitat de Recerca en Lipids i Arteriosclerosis, Sant Joan University Hospital, IISPV, CIBERDEM, Universitat Rovira I Virgili, Reus, Spain
| | - Ricardo Rodríguez-Calvo
- Unitat de Medicina Vascular i Metabolisme, Unitat de Recerca en Lipids i Arteriosclerosis, Sant Joan University Hospital, IISPV, CIBERDEM, Universitat Rovira I Virgili, Reus, Spain
| | - Roser Rosales
- Unitat de Medicina Vascular i Metabolisme, Unitat de Recerca en Lipids i Arteriosclerosis, Sant Joan University Hospital, IISPV, CIBERDEM, Universitat Rovira I Virgili, Reus, Spain
| | - Joan-Carles Vallvé
- Unitat de Medicina Vascular i Metabolisme, Unitat de Recerca en Lipids i Arteriosclerosis, Sant Joan University Hospital, IISPV, CIBERDEM, Universitat Rovira I Virgili, Reus, Spain
| | - Cèlia Rodríguez-Borjabad
- Unitat de Medicina Vascular i Metabolisme, Unitat de Recerca en Lipids i Arteriosclerosis, Sant Joan University Hospital, IISPV, CIBERDEM, Universitat Rovira I Virgili, Reus, Spain
| | - Montserrat Guardiola
- Unitat de Medicina Vascular i Metabolisme, Unitat de Recerca en Lipids i Arteriosclerosis, Sant Joan University Hospital, IISPV, CIBERDEM, Universitat Rovira I Virgili, Reus, Spain
| | - Marina Rodríguez
- Unitat de Medicina Vascular i Metabolisme, Unitat de Recerca en Lipids i Arteriosclerosis, Sant Joan University Hospital, IISPV, CIBERDEM, Universitat Rovira I Virgili, Reus, Spain
| | - Sandra Guaita-Esteruelas
- Unitat de Medicina Vascular i Metabolisme, Unitat de Recerca en Lipids i Arteriosclerosis, Sant Joan University Hospital, IISPV, CIBERDEM, Universitat Rovira I Virgili, Reus, Spain
| | - Iris Oliva
- Unitat de Medicina Vascular i Metabolisme, Unitat de Recerca en Lipids i Arteriosclerosis, Sant Joan University Hospital, IISPV, CIBERDEM, Universitat Rovira I Virgili, Reus, Spain
| | - Neus Martínez-Micaelo
- Unitat de Medicina Vascular i Metabolisme, Unitat de Recerca en Lipids i Arteriosclerosis, Sant Joan University Hospital, IISPV, CIBERDEM, Universitat Rovira I Virgili, Reus, Spain
| | - Mercedes Heras
- Unitat de Medicina Vascular i Metabolisme, Unitat de Recerca en Lipids i Arteriosclerosis, Sant Joan University Hospital, IISPV, CIBERDEM, Universitat Rovira I Virgili, Reus, Spain
| | - Raimon Ferré
- Unitat de Medicina Vascular i Metabolisme, Unitat de Recerca en Lipids i Arteriosclerosis, Sant Joan University Hospital, IISPV, CIBERDEM, Universitat Rovira I Virgili, Reus, Spain
| | - Josep Ribalta
- Unitat de Medicina Vascular i Metabolisme, Unitat de Recerca en Lipids i Arteriosclerosis, Sant Joan University Hospital, IISPV, CIBERDEM, Universitat Rovira I Virgili, Reus, Spain
| | - Núria Plana
- Unitat de Medicina Vascular i Metabolisme, Unitat de Recerca en Lipids i Arteriosclerosis, Sant Joan University Hospital, IISPV, CIBERDEM, Universitat Rovira I Virgili, Reus, Spain
| |
Collapse
|
48
|
An innate defense peptide BPIFA1/SPLUNC1 restricts influenza A virus infection. Mucosal Immunol 2018; 11:71-81. [PMID: 28513596 DOI: 10.1038/mi.2017.45] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 04/17/2017] [Indexed: 02/04/2023]
Abstract
The airway epithelium secretes proteins that function in innate defense against infection. Bactericidal/permeability-increasing fold-containing family member A1 (BPIFA1) is secreted into airways and has a protective role during bacterial infections, but it is not known whether it also has an antiviral role. To determine a role in host defense against influenza A virus (IAV) infection and to find the underlying defense mechanism, we developed transgenic mouse models that are deficient in BPIFA1 and used these, in combination with in vitro three-dimensional mouse tracheal epithelial cell (mTEC) cultures, to investigate its antiviral properties. We show that BPIFA1 has a significant role in mucosal defense against IAV infection. BPIFA1 secretion was highly modulated after IAV infection. Mice deficient in BPIFA1 lost more weight after infection, supported a higher viral load and virus reached the peripheral lung earlier, indicative of a defect in the control of infection. Further analysis using mTEC cultures showed that BPIFA1-deficient cells bound more virus particles, displayed increased nuclear import of IAV ribonucleoprotein complexes, and supported higher levels of viral replication. Our results identify a critical role of BPIFA1 in the initial phase of infection by inhibiting the binding and entry of IAV into airway epithelial cells.
Collapse
|
49
|
Hayes M, Choudhary V, Ojha N, Shin JJ, Han GS, Carman GM, Loewen CJ, Prinz WA, Levine T. Fat storage-inducing transmembrane (FIT or FITM) proteins are related to lipid phosphatase/phosphotransferase enzymes. ACTA ACUST UNITED AC 2017; 5:88-103. [PMID: 29417057 PMCID: PMC5798408 DOI: 10.15698/mic2018.02.614] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Fat storage-inducing transmembrane (FIT or FITM) proteins have been implicated in the partitioning of triacylglycerol to lipid droplets and the budding of lipid droplets from the ER. At the molecular level, the sole relevant interaction is that FITMs directly bind to triacyglycerol and diacylglycerol, but how they function at the molecular level is not known. Saccharomyces cerevisiae has two FITM homologues: Scs3p and Yft2p. Scs3p was initially identified because deletion leads to inositol auxotrophy, with an unusual sensitivity to addition of choline. This strongly suggests a role for Scs3p in phospholipid biosynthesis. Looking at the FITM family as widely as possible, we found that FITMs are widespread throughout eukaryotes, indicating presence in the last eukaryotic common ancestor. Protein alignments also showed that FITM sequences contain the active site of lipid phosphatase/phosphotransferase (LPT) enzymes. This large family transfers phosphate-containing headgroups either between lipids or in exchange for water. We confirmed the prediction that FITMs are related to LPTs by showing that single amino-acid substitutions in the presumptive catalytic site prevented their ability to rescue growth of the mutants on low inositol/high choline media when over-expressed. The substitutions also prevented rescue of other phenotypes associated with loss of FITM in yeast, including mistargeting of Opi1p, defective ER morphology, and aberrant lipid droplet budding. These results suggest that Scs3p, Yft2p and FITMs in general are LPT enzymes involved in an as yet unknown critical step in phospholipid metabolism.
Collapse
Affiliation(s)
- Matthew Hayes
- University College London Institute of Ophthalmology. 11-43 Bath Street, London, EC1V 9EL, UK
| | - Vineet Choudhary
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Namrata Ojha
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - John Jh Shin
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Gil-Soo Han
- Department of Food Science and Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey 08901, USA
| | - George M Carman
- Department of Food Science and Rutgers Center for Lipid Research, Rutgers University, New Brunswick, New Jersey 08901, USA
| | - Christopher Jr Loewen
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - William A Prinz
- Laboratory of Cell and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Timothy Levine
- University College London Institute of Ophthalmology. 11-43 Bath Street, London, EC1V 9EL, UK
| |
Collapse
|
50
|
Harmon M, Larkman P, Hardingham G, Jackson M, Skehel P. A Bi-fluorescence complementation system to detect associations between the Endoplasmic reticulum and mitochondria. Sci Rep 2017; 7:17467. [PMID: 29234100 PMCID: PMC5727038 DOI: 10.1038/s41598-017-17278-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/22/2017] [Indexed: 12/15/2022] Open
Abstract
Close contacts between the endoplasmic reticulum membrane and the mitochondrial outer membrane facilitate efficient transfer of lipids between the organelles and coordinate Ca2+ signalling and stress responses. Changes to this coupling is associated with a number of metabolic disorders and neurodegenerative diseases including Alzheimer’s, Parkinson’s and motor neuron disease. The distance between the two membranes at regions of close apposition is below the resolution of conventional light microscopy, which makes analysis of these interactions challenging. Here we describe a new bifluorescence complementation (BiFC) method that labels a subset of ER-mitochondrial associations in fixed and living cells. The total number of ER-mitochondria associations detected by this approach increases in response to tunicamycin-induced ER stress, serum deprivation or reduced levels of mitofusin 2 (MFN2). This method will facilitate the analysis of dynamic interactions between the ER and mitochondrial membranes.
Collapse
Affiliation(s)
- Mark Harmon
- Centre for Integrative Physiology, Euan MacDonald Centre for Motor Neurone Disease Research, The University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Philip Larkman
- Centre for Integrative Physiology, Euan MacDonald Centre for Motor Neurone Disease Research, The University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Giles Hardingham
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh Medical School, Edinburgh EH8 9XD, UK
| | - Mandy Jackson
- Centre for Integrative Physiology, Euan MacDonald Centre for Motor Neurone Disease Research, The University of Edinburgh, Edinburgh EH8 9XD, UK
| | - Paul Skehel
- Centre for Integrative Physiology, Euan MacDonald Centre for Motor Neurone Disease Research, The University of Edinburgh, Edinburgh EH8 9XD, UK.
| |
Collapse
|