1
|
Esmaeili M, Nasr-Esfahani MH, Shoaraye Nejati A, Safaeinejad Z, Atefi A, L. Megraw T, Ghaedi K. PPARgamma dependent PEX11beta counteracts the suppressive role of SIRT1 on neural differentiation of HESCs. PLoS One 2024; 19:e0298274. [PMID: 38753762 PMCID: PMC11098471 DOI: 10.1371/journal.pone.0298274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 01/18/2024] [Indexed: 05/18/2024] Open
Abstract
The membrane peroxisomal proteins PEX11, play a crucial role in peroxisome proliferation by regulating elongation, membrane constriction, and fission of pre-existing peroxisomes. In this study, we evaluated the function of PEX11B gene in neural differentiation of human embryonic stem cell (hESC) by inducing shRNAi-mediated knockdown of PEX11B expression. Our results demonstrate that loss of PEX11B expression led to a significant decrease in the expression of peroxisomal-related genes including ACOX1, PMP70, PEX1, and PEX7, as well as neural tube-like structures and neuronal markers. Inhibition of SIRT1 using pharmacological agents counteracted the effects of PEX11B knockdown, resulting in a relative increase in PEX11B expression and an increase in differentiated neural tube-like structures. However, the neuroprotective effects of SIRT1 were eliminated by PPAR inhibition, indicating that PPARɣ may mediate the interaction between PEX11B and SIRT1. Our findings suggest that both SIRT1 and PPARɣ have neuroprotective effects, and also this study provides the first indication for a potential interaction between PEX11B, SIRT1, and PPARɣ during hESC neural differentiation.
Collapse
Affiliation(s)
- Maryam Esmaeili
- Department of Cellular Biotechnology, Royan Institute for Biotechnology, Cell Science Research Center, ACECR, Isfahan, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Cellular Biotechnology, Royan Institute for Biotechnology, Cell Science Research Center, ACECR, Isfahan, Iran
| | - Alireza Shoaraye Nejati
- Department of Cellular Biotechnology, Royan Institute for Biotechnology, Cell Science Research Center, ACECR, Isfahan, Iran
| | - Zahra Safaeinejad
- Department of Cellular Biotechnology, Royan Institute for Biotechnology, Cell Science Research Center, ACECR, Isfahan, Iran
| | - Atefeh Atefi
- Department of Cellular Biotechnology, Royan Institute for Biotechnology, Cell Science Research Center, ACECR, Isfahan, Iran
| | - Timothy L. Megraw
- Department of Biomedical Sciences, Florida State University College of Medicine, West Call Street, Tallahassee, FL, United States of America
| | - Kamran Ghaedi
- Department of Cellular Biotechnology, Royan Institute for Biotechnology, Cell Science Research Center, ACECR, Isfahan, Iran
- Faculty of Biological Science and Technology, Department of Cell and Molecular Biology and Microbiology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
2
|
Colasante C, Bonilla-Martinez R, Berg T, Windhorst A, Baumgart-Vogt E. Peroxisomes during postnatal development of mouse endocrine and exocrine pancreas display cell-type- and stage-specific protein composition. Cell Tissue Res 2023:10.1007/s00441-023-03766-6. [PMID: 37126142 DOI: 10.1007/s00441-023-03766-6] [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: 12/20/2022] [Accepted: 03/15/2023] [Indexed: 05/02/2023]
Abstract
Peroxisomal dysfunction unhinges cellular metabolism by causing the accumulation of toxic metabolic intermediates (e.g. reactive oxygen species, very -chain fatty acids, phytanic acid or eicosanoids) and the depletion of important lipid products (e.g. plasmalogens, polyunsaturated fatty acids), leading to various proinflammatory and devastating pathophysiological conditions like metabolic syndrome and age-related diseases including diabetes. Because the peroxisomal antioxidative marker enzyme catalase is low abundant in Langerhans islet cells, peroxisomes were considered scarcely present in the endocrine pancreas. Recently, studies demonstrated that the peroxisomal metabolism is relevant for pancreatic cell functionality. During the postnatal period, significant changes occur in the cell structure and the metabolism to trigger the final maturation of the pancreas, including cell proliferation, regulation of energy metabolism, and activation of signalling pathways. Our aim in this study was to (i) morphometrically analyse the density of peroxisomes in mouse endocrine versus exocrine pancreas and (ii) investigate how the distribution and the abundance of peroxisomal proteins involved in biogenesis, antioxidative defence and fatty acid metabolism change during pancreatic maturation in the postnatal period. Our results prove that endocrine and exocrine pancreatic cells contain high amounts of peroxisomes with heterogeneous protein content indicating that distinct endocrine and exocrine cell types require a specific set of peroxisomal proteins depending on their individual physiological functions. We further show that significant postnatal changes occur in the peroxisomal compartment of different pancreatic cells that are most probably relevant for the metabolic maturation and differentiation of the pancreas during the development from birth to adulthood.
Collapse
Affiliation(s)
- Claudia Colasante
- Institute for Anatomy and Cell Biology, Medical Cell Biology, Justus Liebig -University, Aulweg 123, 35392, Giessen, Germany
| | - Rocio Bonilla-Martinez
- Institute for Anatomy and Cell Biology, Medical Cell Biology, Justus Liebig -University, Aulweg 123, 35392, Giessen, Germany
| | - Timm Berg
- Institute for Anatomy and Cell Biology, Medical Cell Biology, Justus Liebig -University, Aulweg 123, 35392, Giessen, Germany
| | - Anita Windhorst
- Institute for Medical Informatic, Justus Liebig University, Rudolf-Buchheim-Str. 6, 35392, Gießen, Germany
| | - Eveline Baumgart-Vogt
- Institute for Anatomy and Cell Biology, Medical Cell Biology, Justus Liebig -University, Aulweg 123, 35392, Giessen, Germany.
| |
Collapse
|
3
|
Abe Y, Wanders RJA, Waterham HR, Mandel H, Falik-Zaccai TC, Ishihara N, Fujiki Y. Genetic defects in peroxisome morphogenesis (Pex11β, dynamin-like protein 1, and nucleoside diphosphate kinase 3) affect docosahexaenoic acid-phospholipid metabolism. J Inherit Metab Dis 2023; 46:273-285. [PMID: 36522796 DOI: 10.1002/jimd.12582] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Peroxisomes are essential organelles involved in lipid metabolisms including plasmalogen biosynthesis and β-oxidation of very long-chain fatty acids. Peroxisomes proliferate by the growth and division of pre-existing peroxisomes. The peroxisomal membrane is elongated by Pex11β and then divided by the dynamin-like GTPase, DLP1 (also known as DRP1 encoded by DNM1L gene), which also functions as a fission factor for mitochondria. Nucleoside diphosphate kinase 3 (NME3) localized in both peroxisomes and mitochondria generates GTP for DLP1 activity. Deficiencies of either of these factors induce abnormal morphology of peroxisomes and/or mitochondria, and are associated with central nervous system dysfunction. To investigate whether the impaired division of peroxisomes affects lipid metabolisms, we assessed the phospholipid composition of cells lacking each of the different division factors. In fibroblasts from the patients deficient in DLP1, NME3, or Pex11β, docosahexaenoic acid (DHA, C22:6)-containing phospholipids were found to be decreased. Conversely, the levels of several fatty acids such as arachidonic acid (AA, C20:4) and oleic acid (C18:1) were elevated. Mouse embryonic fibroblasts from Drp1- and Pex11β-knockout mice also showed a decrease in the levels of phospholipids containing DHA and AA. Collectively, these results suggest that the dynamics of organelle morphology exert marked effects on the fatty acid composition of phospholipids.
Collapse
Affiliation(s)
- Yuichi Abe
- Faculty of Arts and Science, Kyushu University, Fukuoka, Japan
| | - Ronald J A Wanders
- Departments of Pediatrics, EMMA Children's Hospital & Laboratory Division, Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam, The Netherlands
| | - Hans R Waterham
- Departments of Pediatrics, EMMA Children's Hospital & Laboratory Division, Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centers, Location Academic Medical Center, Amsterdam, The Netherlands
| | - Hanna Mandel
- Galilee Medical Center, Institute of Human Genetics, Nahariya, Israel
| | - Tzipora C Falik-Zaccai
- Galilee Medical Center, Institute of Human Genetics, Nahariya, Israel
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Naotada Ishihara
- Department of Biological Sciences, Osaka University, Osaka, Japan
| | - Yukio Fujiki
- Medical Institute of Bioregulation, Institute of Rheological Functions of Food-Kyushu University Collaboration Program, Kyushu University, Fukuoka, Japan
- Graduate School of Science, University of Hyogo, Hyogo, Japan
| |
Collapse
|
4
|
Peroxisomes Are Highly Abundant and Heterogeneous in Human Parotid Glands. Int J Mol Sci 2023; 24:ijms24054783. [PMID: 36902220 PMCID: PMC10003153 DOI: 10.3390/ijms24054783] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/18/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
The parotid gland is one of the major salivary glands producing a serous secretion, and it plays an essential role in the digestive and immune systems. Knowledge of peroxisomes in the human parotid gland is minimal; furthermore, the peroxisomal compartment and its enzyme composition in the different cell types of the human parotid gland have never been subjected to a detailed investigation. Therefore, we performed a comprehensive analysis of peroxisomes in the human parotid gland's striated duct and acinar cells. We combined biochemical techniques with various light and electron microscopy techniques to determine the localization of parotid secretory proteins and different peroxisomal marker proteins in parotid gland tissue. Moreover, we analyzed the mRNA of numerous gene encoding proteins localized in peroxisomes using real-time quantitative PCR. The results confirm the presence of peroxisomes in all striated duct and acinar cells of the human parotid gland. Immunofluorescence analyses for various peroxisomal proteins showed a higher abundance and more intense staining in striated duct cells compared to acinar cells. Moreover, human parotid glands comprise high quantities of catalase and other antioxidative enzymes in discrete subcellular regions, suggesting their role in protection against oxidative stress. This study provides the first thorough description of parotid peroxisomes in different parotid cell types of healthy human tissue.
Collapse
|
5
|
Kocherlakota S, Swinkels D, Van Veldhoven PP, Baes M. Mouse Models to Study Peroxisomal Functions and Disorders: Overview, Caveats, and Recommendations. Methods Mol Biol 2023; 2643:469-500. [PMID: 36952207 DOI: 10.1007/978-1-0716-3048-8_34] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Abstract
During the last three decades many mouse lines were created or identified that are deficient in one or more peroxisomal functions. Different methodologies were applied to obtain global, hypomorph, cell type selective, inducible, and knockin mice. Whereas some models closely mimic pathologies in patients, others strongly deviate or no human counterpart has been reported. Often, mice, apparently endowed with a stronger transcriptional adaptation, have to be challenged with dietary additions or restrictions in order to trigger phenotypic changes. Depending on the inactivated peroxisomal protein, several approaches can be taken to validate the loss-of-function. Here, an overview is given of the available mouse models and their most important characteristics.
Collapse
Affiliation(s)
- Sai Kocherlakota
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Daniëlle Swinkels
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Paul P Van Veldhoven
- Laboratory of Peroxisome Biology and Intracellular Communication, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Myriam Baes
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
6
|
Wanders RJA, Baes M, Ribeiro D, Ferdinandusse S, Waterham HR. The physiological functions of human peroxisomes. Physiol Rev 2023; 103:957-1024. [PMID: 35951481 DOI: 10.1152/physrev.00051.2021] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Peroxisomes are subcellular organelles that play a central role in human physiology by catalyzing a range of unique metabolic functions. The importance of peroxisomes for human health is exemplified by the existence of a group of usually severe diseases caused by an impairment in one or more peroxisomal functions. Among others these include the Zellweger spectrum disorders, X-linked adrenoleukodystrophy, and Refsum disease. To fulfill their role in metabolism, peroxisomes require continued interaction with other subcellular organelles including lipid droplets, lysosomes, the endoplasmic reticulum, and mitochondria. In recent years it has become clear that the metabolic alliance between peroxisomes and other organelles requires the active participation of tethering proteins to bring the organelles physically closer together, thereby achieving efficient transfer of metabolites. This review intends to describe the current state of knowledge about the metabolic role of peroxisomes in humans, with particular emphasis on the metabolic partnership between peroxisomes and other organelles and the consequences of genetic defects in these processes. We also describe the biogenesis of peroxisomes and the consequences of the multiple genetic defects therein. In addition, we discuss the functional role of peroxisomes in different organs and tissues and include relevant information derived from model systems, notably peroxisomal mouse models. Finally, we pay particular attention to a hitherto underrated role of peroxisomes in viral infections.
Collapse
Affiliation(s)
- Ronald J A Wanders
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,United for Metabolic Diseases, Amsterdam, The Netherlands
| | - Myriam Baes
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Daniela Ribeiro
- Institute of Biomedicine (iBiMED) and Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Sacha Ferdinandusse
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,United for Metabolic Diseases, Amsterdam, The Netherlands
| | - Hans R Waterham
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,United for Metabolic Diseases, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Fujiki Y, Okumoto K, Honsho M, Abe Y. Molecular insights into peroxisome homeostasis and peroxisome biogenesis disorders. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119330. [PMID: 35917894 DOI: 10.1016/j.bbamcr.2022.119330] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 06/15/2023]
Abstract
Peroxisomes are single-membrane organelles essential for cell metabolism including the β-oxidation of fatty acids, synthesis of etherlipid plasmalogens, and redox homeostasis. Investigations into peroxisome biogenesis and the human peroxisome biogenesis disorders (PBDs) have identified 14 PEX genes encoding peroxins involved in peroxisome biogenesis and the mutation of PEX genes is responsible for the PBDs. Many recent findings have further advanced our understanding of the biology, physiology, and consequences of a functional deficit of peroxisomes. In this Review, we discuss cell defense mechanisms that counteract oxidative stress by 1) a proapoptotic Bcl-2 factor BAK-mediated release to the cytosol of H2O2-degrading catalase from peroxisomes and 2) peroxisomal import suppression of catalase by Ser232-phosphorylation of Pex14, a docking protein for the Pex5-PTS1 complex. With respect to peroxisome division, the important issue of how the energy-rich GTP is produced and supplied for the division process was recently addressed by the discovery of a nucleoside diphosphate kinase-like protein, termed DYNAMO1 in a lower eukaryote, which has a mammalian homologue NME3. In regard to the mechanisms underlying the pathogenesis of PBDs, a new PBD model mouse defective in Pex14 manifests a dysregulated brain-derived neurotrophic factor (BDNF)-TrkB pathway, an important signaling pathway for cerebellar morphogenesis. Communications between peroxisomes and other organelles are also addressed.
Collapse
Affiliation(s)
- Yukio Fujiki
- Medical Institute of Bioregulation, Institute of Rheological Functions of Food, Collaboration Program, Kyushu University, 3-1-1 Maidashi, Fukuoka 812-8582, Japan.
| | - Kanji Okumoto
- Department of Biology and Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Masanori Honsho
- Medical Institute of Bioregulation, Institute of Rheological Functions of Food, Collaboration Program, Kyushu University, 3-1-1 Maidashi, Fukuoka 812-8582, Japan
| | - Yuichi Abe
- Faculty of Arts and Science, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| |
Collapse
|
8
|
Jiang S, Uddin MJ, Yu X, Piao L, Dorotea D, Oh GT, Ha H. Peroxisomal Fitness: A Potential Protective Mechanism of Fenofibrate against High Fat Diet-Induced Non-Alcoholic Fatty Liver Disease in Mice. Diabetes Metab J 2022; 46:829-842. [PMID: 35746892 PMCID: PMC9723204 DOI: 10.4093/dmj.2021.0274] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 03/15/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) has been increasing in association with the epidemic of obesity and diabetes. Peroxisomes are single membrane-enclosed organelles that play a role in the metabolism of lipid and reactive oxygen species. The present study examined the role of peroxisomes in high-fat diet (HFD)-induced NAFLD using fenofibrate, a peroxisome proliferator-activated receptor α (PPARα) agonist. METHODS Eight-week-old male C57BL/6J mice were fed either a normal diet or HFD for 12 weeks, and fenofibrate (50 mg/kg/day) was orally administered along with the initiation of HFD. RESULTS HFD-induced liver injury as measured by increased alanine aminotransferase, inflammation, oxidative stress, and lipid accumulation was effectively prevented by fenofibrate. Fenofibrate significantly increased the expression of peroxisomal genes and proteins involved in peroxisomal biogenesis and function. HFD-induced attenuation of peroxisomal fatty acid oxidation was also significantly restored by fenofibrate, demonstrating the functional significance of peroxisomal fatty acid oxidation. In Ppara deficient mice, fenofibrate failed to maintain peroxisomal biogenesis and function in HFD-induced liver injury. CONCLUSION The present data highlight the importance of PPARα-mediated peroxisomal fitness in the protective effect of fenofibrate against NAFLD.
Collapse
Affiliation(s)
- Songling Jiang
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, College of Pharmacy, Seoul, Korea
| | - Md Jamal Uddin
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, College of Pharmacy, Seoul, Korea
| | - Xiaoying Yu
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, College of Pharmacy, Seoul, Korea
| | - Lingjuan Piao
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, College of Pharmacy, Seoul, Korea
| | - Debra Dorotea
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, College of Pharmacy, Seoul, Korea
| | - Goo Taeg Oh
- Department of Life Sciences, Ewha Womans University, Seoul, Korea
| | - Hunjoo Ha
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, College of Pharmacy, Seoul, Korea
| |
Collapse
|
9
|
Fission Impossible (?)-New Insights into Disorders of Peroxisome Dynamics. Cells 2022; 11:cells11121922. [PMID: 35741050 PMCID: PMC9221819 DOI: 10.3390/cells11121922] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/09/2022] [Accepted: 06/10/2022] [Indexed: 11/16/2022] Open
Abstract
Peroxisomes are highly dynamic and responsive organelles, which can adjust their morphology, number, intracellular position, and metabolic functions according to cellular needs. Peroxisome multiplication in mammalian cells involves the concerted action of the membrane-shaping protein PEX11β and division proteins, such as the membrane adaptors FIS1 and MFF, which recruit the fission GTPase DRP1 to the peroxisomal membrane. The latter proteins are also involved in mitochondrial division. Patients with loss of DRP1, MFF or PEX11β function have been identified, showing abnormalities in peroxisomal (and, for the shared proteins, mitochondrial) dynamics as well as developmental and neurological defects, whereas the metabolic functions of the organelles are often unaffected. Here, we provide a timely update on peroxisomal membrane dynamics with a particular focus on peroxisome formation by membrane growth and division. We address the function of PEX11β in these processes, as well as the role of peroxisome–ER contacts in lipid transfer for peroxisomal membrane expansion. Furthermore, we summarize the clinical phenotypes and pathophysiology of patients with defects in the key division proteins DRP1, MFF, and PEX11β as well as in the peroxisome–ER tether ACBD5. Potential therapeutic strategies for these rare disorders with limited treatment options are discussed.
Collapse
|
10
|
Stocks B, Gonzalez-Franquesa A, Borg ML, Björnholm M, Niu L, Zierath JR, Deshmukh AS. Integrated Liver and Plasma Proteomics in Obese Mice Reveals Complex Metabolic Regulation. Mol Cell Proteomics 2022; 21:100207. [PMID: 35093608 PMCID: PMC8928073 DOI: 10.1016/j.mcpro.2022.100207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/23/2022] [Indexed: 11/28/2022] Open
Abstract
Obesity leads to the development of nonalcoholic fatty liver disease (NAFLD) and associated alterations to the plasma proteome. To elucidate the underlying changes associated with obesity, we performed liquid chromatography-tandem mass spectrometry in the liver and plasma of obese leptin-deficient ob/ob mice and integrated these data with publicly available transcriptomic and proteomic datasets of obesity and metabolic diseases in preclinical and clinical cohorts. We quantified 7173 and 555 proteins in the liver and plasma proteomes, respectively. The abundance of proteins related to fatty acid metabolism were increased, alongside peroxisomal proliferation in ob/ob liver. Putatively secreted proteins and the secretory machinery were also dysregulated in the liver, which was mirrored by a substantial alteration of the plasma proteome. Greater than 50% of the plasma proteins were differentially regulated, including NAFLD biomarkers, lipoproteins, the 20S proteasome, and the complement and coagulation cascades of the immune system. Integration of the liver and plasma proteomes identified proteins that were concomitantly regulated in the liver and plasma in obesity, suggesting that the systemic abundance of these plasma proteins is regulated by secretion from the liver. Many of these proteins are systemically regulated during type 2 diabetes and/or NAFLD in humans, indicating the clinical importance of liver-plasma cross talk and the relevance of our investigations in ob/ob mice. Together, these analyses yield a comprehensive insight into obesity and provide an extensive resource for obesity research in a prevailing model organism.
Collapse
Affiliation(s)
- Ben Stocks
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Alba Gonzalez-Franquesa
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Melissa L Borg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Marie Björnholm
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Lili Niu
- Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Juleen R Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Atul S Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
11
|
Carmichael RE, Schrader M. Determinants of Peroxisome Membrane Dynamics. Front Physiol 2022; 13:834411. [PMID: 35185625 PMCID: PMC8853631 DOI: 10.3389/fphys.2022.834411] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 01/12/2022] [Indexed: 11/13/2022] Open
Abstract
Organelles within the cell are highly dynamic entities, requiring dramatic morphological changes to support their function and maintenance. As a result, organelle membranes are also highly dynamic, adapting to a range of topologies as the organelle changes shape. In particular, peroxisomes—small, ubiquitous organelles involved in lipid metabolism and reactive oxygen species homeostasis—display a striking plasticity, for example, during the growth and division process by which they proliferate. During this process, the membrane of an existing peroxisome elongates to form a tubule, which then constricts and ultimately undergoes scission to generate new peroxisomes. Dysfunction of this plasticity leads to diseases with developmental and neurological phenotypes, highlighting the importance of peroxisome dynamics for healthy cell function. What controls the dynamics of peroxisomal membranes, and how this influences the dynamics of the peroxisomes themselves, is just beginning to be understood. In this review, we consider how the composition, biophysical properties, and protein-lipid interactions of peroxisomal membranes impacts on their dynamics, and in turn on the biogenesis and function of peroxisomes. In particular, we focus on the effect of the peroxin PEX11 on the peroxisome membrane, and its function as a major regulator of growth and division. Understanding the roles and regulation of peroxisomal membrane dynamics necessitates a multidisciplinary approach, encompassing knowledge across a range of model species and a number of fields including lipid biochemistry, biophysics and computational biology. Here, we present an integrated overview of our current understanding of the determinants of peroxisome membrane dynamics, and reflect on the outstanding questions still remaining to be solved.
Collapse
Affiliation(s)
- Ruth E Carmichael
- College of Life and Environmental Sciences, Biosciences, University of Exeter, Exeter, United Kingdom
| | - Michael Schrader
- College of Life and Environmental Sciences, Biosciences, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
12
|
Garikapati V, Colasante C, Baumgart-Vogt E, Spengler B. Sequential lipidomic, metabolomic, and proteomic analyses of serum, liver, and heart tissue specimens from peroxisomal biogenesis factor 11α knockout mice. Anal Bioanal Chem 2022; 414:2235-2250. [PMID: 35083512 PMCID: PMC8821073 DOI: 10.1007/s00216-021-03860-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 11/25/2021] [Accepted: 12/20/2021] [Indexed: 12/16/2022]
Abstract
Peroxisomes are versatile single membrane-enclosed cytoplasmic organelles, involved in reactive oxygen species (ROS) and lipid metabolism and diverse other metabolic processes. Peroxisomal disorders result from mutations in Pex genes-encoded proteins named peroxins (PEX proteins) and single peroxisomal enzyme deficiencies. The PEX11 protein family (α, β, and γ isoforms) plays an important role in peroxisomal proliferation and fission. However, their specific functions and the metabolic impact caused by their deficiencies have not been precisely characterized. To understand the systemic molecular alterations caused by peroxisomal defects, here we utilized untreated peroxisomal biogenesis factor 11α knockout (Pex11α KO) mouse model and performed serial relative-quantitative lipidomic, metabolomic, and proteomic analyses of serum, liver, and heart tissue homogenates. We demonstrated significant specific changes in the abundances of multiple lipid species, polar metabolites, and proteins and dysregulated metabolic pathways in distinct biological specimens of the Pex11α KO adult mice in comparison to the wild type (WT) controls. Overall, the present study reports comprehensive semi-quantitative molecular omics information of the Pex11α KO mice, which might serve in the future as a reference for a better understanding of the roles of Pex11α and underlying pathophysiological mechanisms of peroxisomal biogenesis disorders.
Collapse
Affiliation(s)
- Vannuruswamy Garikapati
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, 35392, Giessen, Germany.,Institute for Anatomy and Cell Biology II, Division of Medical Cell Biology, Justus Liebig University Giessen, 35392, Giessen, Germany
| | - Claudia Colasante
- Institute for Anatomy and Cell Biology II, Division of Medical Cell Biology, Justus Liebig University Giessen, 35392, Giessen, Germany
| | - Eveline Baumgart-Vogt
- Institute for Anatomy and Cell Biology II, Division of Medical Cell Biology, Justus Liebig University Giessen, 35392, Giessen, Germany.
| | - Bernhard Spengler
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, 35392, Giessen, Germany.
| |
Collapse
|
13
|
Zientara-Rytter KM, Mahalingam SS, Farré JC, Carolino K, Subramani S. Recognition and Chaperoning by Pex19, Followed by Trafficking and Membrane Insertion of the Peroxisome Proliferation Protein, Pex11. Cells 2022; 11:cells11010157. [PMID: 35011719 PMCID: PMC8750153 DOI: 10.3390/cells11010157] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 01/27/2023] Open
Abstract
Pex11, an abundant peroxisomal membrane protein (PMP), is required for division of peroxisomes and is robustly imported to peroxisomal membranes. We present a comprehensive analysis of how the Pichia pastoris Pex11 is recognized and chaperoned by Pex19, targeted to peroxisome membranes and inserted therein. We demonstrate that Pex11 contains one Pex19-binding site (Pex19-BS) that is required for Pex11 insertion into peroxisomal membranes by Pex19, but is non-essential for peroxisomal trafficking. We provide extensive mutational analyses regarding the recognition of Pex19-BS in Pex11 by Pex19. Pex11 also has a second, Pex19-independent membrane peroxisome-targeting signal (mPTS) that is preserved among Pex11-family proteins and anchors the human HsPex11γ to the outer leaflet of the peroxisomal membrane. Thus, unlike most PMPs, Pex11 can use two mechanisms of transport to peroxisomes, where only one of them depends on its direct interaction with Pex19, but the other does not. However, Pex19 is necessary for membrane insertion of Pex11. We show that Pex11 can self-interact, using both homo- and/or heterotypic interactions involving its N-terminal helical domains. We demonstrate that Pex19 acts as a chaperone by interacting with the Pex19-BS in Pex11, thereby protecting Pex11 from spontaneous oligomerization that would otherwise cause its aggregation and subsequent degradation.
Collapse
|
14
|
Takashima S, Takemoto S, Toyoshi K, Ohba A, Shimozawa N. Zebrafish model of human Zellweger syndrome reveals organ-specific accumulation of distinct fatty acid species and widespread gene expression changes. Mol Genet Metab 2021; 133:307-323. [PMID: 34016526 DOI: 10.1016/j.ymgme.2021.05.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 05/04/2021] [Accepted: 05/04/2021] [Indexed: 11/24/2022]
Abstract
In Zellweger syndrome (ZS), lack of peroxisome function causes physiological and developmental abnormalities in many organs such as the brain, liver, muscles, and kidneys, but little is known about the exact pathogenic mechanism. By disrupting the zebrafish pex2 gene, we established a disease model for ZS and found that it exhibits pathological features and metabolic changes similar to those observed in human patients. By comprehensive analysis of the fatty acid profile, we found organ-specific accumulation and reduction of distinct fatty acid species, such as an accumulation of ultra-very-long-chain polyunsaturated fatty acids (ultra-VLC-PUFAs) in the brains of pex2 mutant fish. Transcriptome analysis using microarray also revealed mutant-specific gene expression changes that might lead to the symptoms, including reduction of crystallin, troponin, parvalbumin, and fatty acid metabolic genes. Our data indicated that the loss of peroxisomes results in widespread metabolic and gene expression changes beyond the causative peroxisomal function. These results suggest the genetic and metabolic basis of the pathology of this devastating human disease.
Collapse
Affiliation(s)
- Shigeo Takashima
- Division of Genomics Research, Life Science Research Center, Gifu University, Gifu, Japan; Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan; United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan.
| | - Shoko Takemoto
- Division of Genomics Research, Life Science Research Center, Gifu University, Gifu, Japan
| | - Kayoko Toyoshi
- Division of Genomics Research, Life Science Research Center, Gifu University, Gifu, Japan
| | - Akiko Ohba
- Division of Genomics Research, Life Science Research Center, Gifu University, Gifu, Japan
| | - Nobuyuki Shimozawa
- Division of Genomics Research, Life Science Research Center, Gifu University, Gifu, Japan; Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, Japan; United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Gifu, Japan
| |
Collapse
|
15
|
Watermann C, Meyer MT, Valerius KP, Kleefeldt F, Wagner S, Wittekindt C, Klussmann JP, Ergün S, Baumgart-Vogt E, Karnati S. Peroxisomes in the mouse parotid glands: An in-depth morphological and molecular analysis. Ann Anat 2021; 238:151778. [PMID: 34091056 DOI: 10.1016/j.aanat.2021.151778] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/16/2021] [Accepted: 05/10/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND The parotid gland is a major salivary gland that has important roles in the digestive and immune system. Peroxisomes are ubiquitous, single-membrane-bound organelles that are present in all eukaryotic cells. Peroxisomes help mediate lipid and reactive oxygen species metabolism, as well as polyunsaturated fatty acid, cholesterol and plasmalogen synthesis. Much of the knowledge on peroxisomes has derived from metabolic organs, however no detailed knowledge is available on peroxisomes in the parotid glands. We thus aimed to comprehensively delineate the localization and characterization of peroxisomal proteins in the murine parotid gland. METHODS We characterized peroxisomes in the acinar and striated duct cells of the murine parotid gland by fluorescence and electron microscopy, as well as protein and mRNA expression analyses for important peroxisomal genes and proteins. RESULTS We found that peroxisomes are present in all cell types of the mouse parotid gland, however, exhibit notable cell-specific differences in their abundance and enzyme content. We also observed that mouse parotid glands contain high levels of peroxisomal β-oxidation enzymes (including Acox1, Mfp2 and Acaa1), catalase and other peroxisomal anti-oxidative enzymes. CONCLUSIONS This data suggests that peroxisomes are highly abundant in the murine parotid gland and might help to protect against oxidative stress. This comprehensive description of peroxisomes in the parotid gland lays the groundwork for further research concerning their role in the pathogenesis of parotid gland diseases and tumors.
Collapse
Affiliation(s)
- Christoph Watermann
- Institute for Anatomy and Cell Biology II, Medical Cell Biology, Justus Liebig University, Giessen D-35385, Germany; Department of Otorhinolaryngology, Head and Neck Surgery, Justus Liebig University, Giessen D-35392, Germany
| | - Malin T Meyer
- Institute for Anatomy and Cell Biology II, Medical Cell Biology, Justus Liebig University, Giessen D-35385, Germany; Department of Otorhinolaryngology, Head and Neck Surgery, Justus Liebig University, Giessen D-35392, Germany
| | - Klaus P Valerius
- Institute for Anatomy and Cell Biology II, Medical Cell Biology, Justus Liebig University, Giessen D-35385, Germany
| | - Florian Kleefeldt
- Institute for Anatomy and Cell Biology, Julius Maximilians University, Würzburg D-97070, Germany
| | - Steffen Wagner
- Department of Otorhinolaryngology, Head and Neck Surgery, Justus Liebig University, Giessen D-35392, Germany
| | - Claus Wittekindt
- Department of Otorhinolaryngology, Head and Neck Surgery, Justus Liebig University, Giessen D-35392, Germany
| | - Jens P Klussmann
- Department of Otorhinolaryngology, Head and Neck Surgery, Justus Liebig University, Giessen D-35392, Germany; Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, Cologne D-50931, Germany
| | - Süleyman Ergün
- Institute for Anatomy and Cell Biology, Julius Maximilians University, Würzburg D-97070, Germany
| | - Eveline Baumgart-Vogt
- Institute for Anatomy and Cell Biology II, Medical Cell Biology, Justus Liebig University, Giessen D-35385, Germany
| | - Srikanth Karnati
- Institute for Anatomy and Cell Biology II, Medical Cell Biology, Justus Liebig University, Giessen D-35385, Germany; Institute for Anatomy and Cell Biology, Julius Maximilians University, Würzburg D-97070, Germany.
| |
Collapse
|
16
|
Blair HR, Tomas C, Miwa S, Heath A, Russell A, Ginkel MV, Gunn D, Walker M. Peroxisomes and pancreatic beta-cell lipo-dysfunction. J Diabetes Complications 2021; 35:107843. [PMID: 33419633 DOI: 10.1016/j.jdiacomp.2020.107843] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/20/2020] [Indexed: 10/22/2022]
Abstract
AIMS Pancreatic beta-cell lipo-dysfunction decreases insulin secretion and predisposes to the development of type 2 diabetes. Through targeted Pex11β knockdown and peroxisome depletion, our aim was to investigate the specific contribution of peroxisomes to palmitate mediated pancreatic beta-cell dysfunction. METHODS MIN6 cells were transfected with probes targeted against Pex11β, a regulator of peroxisome abundance, or with scrambled control probes. Peroxisome abundance was measured by PMP-70 protein expression. 48 h post transfection, cells were incubated with 250 μM palmitate or BSA control for a further 48 h before measurement of glucose stimulated insulin secretion and of reactive oxygen species. RESULTS Pex11β knockdown decreased target gene expression by >80% compared with the scrambled control (P<0.001). This led to decreased PMP-70 expression (p<0.01) and a 22% decrease in peroxisome number (p<0.05). At 25 mM glucose, palmitate treatment decreased insulin secretion by 64% in the scrambled control cells (2.54±0.25 vs 7.07±0.83 [mean±SEM] ng/h/μg protein; Palmitate vs BSA P<0.001), but by just 37% in the Pex11β knockdown cells. Comparing responses in the presence of palmitate, insulin secretion at 25 mM glucose was significantly greater in the Pex11β knockdown cells compared with the scrambled controls (4.04±0.46 vs 2.54±0.25 ng/h/μg protein; p<0.05). Reactive oxygen species generation with palmitate was lower in the Pex11β knockdown cells compared with the scrambled controls (P<0.001). CONCLUSION Pex11β knockdown decreased peroxisome abundance, decreased palmitate mediated reactive oxygen species generation, and reversed the inhibitory effect of palmitate on insulin secretion. These findings reveal a distinct role of peroxisomes in palmitate mediated beta-cell dysfunction.
Collapse
Affiliation(s)
- Helen R Blair
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Cara Tomas
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Satomi Miwa
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Alan Heath
- Unilever Discover, Colworth Science Park, Sharnbrook, Bedford, UK
| | - Alison Russell
- Unilever Discover, Colworth Science Park, Sharnbrook, Bedford, UK
| | | | - David Gunn
- Unilever Discover, Colworth Science Park, Sharnbrook, Bedford, UK
| | - Mark Walker
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
17
|
Azadi AS, Carmichael RE, Kovacs WJ, Koster J, Kors S, Waterham HR, Schrader M. A Functional SMAD2/3 Binding Site in the PEX11β Promoter Identifies a Role for TGFβ in Peroxisome Proliferation in Humans. Front Cell Dev Biol 2020; 8:577637. [PMID: 33195217 PMCID: PMC7644849 DOI: 10.3389/fcell.2020.577637] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/01/2020] [Indexed: 01/10/2023] Open
Abstract
In mammals, peroxisomes perform crucial functions in cellular metabolism, signaling and viral defense which are essential to the viability of the organism. Molecular cues triggered by changes in the cellular environment induce a dynamic response in peroxisomes, which manifests itself as a change in peroxisome number, altered enzyme levels and adaptations to the peroxisomal morphology. How the regulation of this process is integrated into the cell's response to different stimuli, including the signaling pathways and factors involved, remains unclear. Here, a cell-based peroxisome proliferation assay has been applied to investigate the ability of different stimuli to induce peroxisome proliferation. We determined that serum stimulation, long-chain fatty acid supplementation and TGFβ application all increase peroxisome elongation, a prerequisite for proliferation. Time-resolved mRNA expression during the peroxisome proliferation cycle revealed a number of peroxins whose expression correlated with peroxisome elongation, including the β isoform of PEX11, but not the α or γ isoforms. An initial map of putative regulatory motif sites in the respective promoters showed a difference between binding sites in PEX11α and PEX11β, suggesting that these genes may be regulated by distinct pathways. A functional SMAD2/3 binding site in PEX11β points to the involvement of the TGFβ signaling pathway in expression of this gene and thus peroxisome proliferation/dynamics in humans.
Collapse
Affiliation(s)
- Afsoon S Azadi
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Ruth E Carmichael
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Werner J Kovacs
- Institute of Molecular Health Sciences, Swiss Federal Institute of Technology in Zürich (ETH Zürich), Zurich, Switzerland
| | - Janet Koster
- Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, Netherlands
| | - Suzan Kors
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| | - Hans R Waterham
- Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, Netherlands
| | - Michael Schrader
- Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom
| |
Collapse
|
18
|
Mammalian Homologue NME3 of DYNAMO1 Regulates Peroxisome Division. Int J Mol Sci 2020; 21:ijms21218040. [PMID: 33126676 PMCID: PMC7662248 DOI: 10.3390/ijms21218040] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/22/2020] [Accepted: 10/25/2020] [Indexed: 12/21/2022] Open
Abstract
Peroxisomes proliferate by sequential processes comprising elongation, constriction, and scission of peroxisomal membrane. It is known that the constriction step is mediated by a GTPase named dynamin-like protein 1 (DLP1) upon efficient loading of GTP. However, mechanism of fuelling GTP to DLP1 remains unknown in mammals. We earlier show that nucleoside diphosphate (NDP) kinase-like protein, termed dynamin-based ring motive-force organizer 1 (DYNAMO1), generates GTP for DLP1 in a red alga, Cyanidioschyzon merolae. In the present study, we identified that nucleoside diphosphate kinase 3 (NME3), a mammalian homologue of DYNAMO1, localizes to peroxisomes. Elongated peroxisomes were observed in cells with suppressed expression of NME3 and fibroblasts from a patient lacking NME3 due to the homozygous mutation at the initiation codon of NME3. Peroxisomes proliferated by elevation of NME3 upon silencing the expression of ATPase family AAA domain containing 1, ATAD1. In the wild-type cells expressing catalytically-inactive NME3, peroxisomes were elongated. These results suggest that NME3 plays an important role in peroxisome division in a manner dependent on its NDP kinase activity. Moreover, the impairment of peroxisome division reduces the level of ether-linked glycerophospholipids, ethanolamine plasmalogens, implying the physiological importance of regulation of peroxisome morphology.
Collapse
|
19
|
Analysis of the Level of Plasmid-Derived mRNA in the Presence of Residual Plasmid DNA by Two-Step Quantitative RT-PCR. Methods Protoc 2020; 3:mps3020040. [PMID: 32456168 PMCID: PMC7359704 DOI: 10.3390/mps3020040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/19/2020] [Accepted: 05/21/2020] [Indexed: 11/17/2022] Open
Abstract
In transfection experiments with mammalian cells aiming to overexpress a specific protein, it is often necessary to correctly quantify the level of the recombinant and the corresponding endogenous mRNA. In our case, mouse calvarial osteoblasts were transfected with a vector containing the complete Pex11β cDNA (plasmid DNA). The Pex11β mRNA level, as calculated using the RT-qPCR product, was unrealistically higher (>1000-fold) in transfected compared to non-transfected cells, and we assumed that there were large amounts of contaminating plasmid DNA in the RNA sample. Thus, we searched for a simple way to distinguish between plasmid-derived mRNA, endogenous genome-derived mRNA and plasmid DNA, with minimal changes to standard RT-PCR techniques. We succeeded by performing a plasmid mRNA-specific reverse transcription, and the plasmid cDNA was additionally tagged with a nonsense tail. A subsequent standard qPCR was conducted using appropriate PCR primers annealing to the plasmid cDNA and to the nonsense tail. Using this method, we were able to determine the specific amount of mRNA derived from the transfected plasmid DNA in comparison to the endogenous genome-derived mRNA, and thus the transfection and transcription efficiency.
Collapse
|
20
|
Fujiki Y, Abe Y, Imoto Y, Tanaka AJ, Okumoto K, Honsho M, Tamura S, Miyata N, Yamashita T, Chung WK, Kuroiwa T. Recent insights into peroxisome biogenesis and associated diseases. J Cell Sci 2020; 133:133/9/jcs236943. [PMID: 32393673 DOI: 10.1242/jcs.236943] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Peroxisomes are single-membrane organelles present in eukaryotes. The functional importance of peroxisomes in humans is represented by peroxisome-deficient peroxisome biogenesis disorders (PBDs), including Zellweger syndrome. Defects in the genes that encode the 14 peroxins that are required for peroxisomal membrane assembly, matrix protein import and division have been identified in PBDs. A number of recent findings have advanced our understanding of the biology, physiology and consequences of functional defects in peroxisomes. In this Review, we discuss a cooperative cell defense mechanisms against oxidative stress that involves the localization of BAK (also known as BAK1) to peroxisomes, which alters peroxisomal membrane permeability, resulting in the export of catalase, a peroxisomal enzyme. Another important recent finding is the discovery of a nucleoside diphosphate kinase-like protein that has been shown to be essential for how the energy GTP is generated and provided for the fission of peroxisomes. With regard to PBDs, we newly identified a mild mutation, Pex26-F51L that causes only hearing loss. We will also discuss findings from a new PBD model mouse defective in Pex14, which manifested dysregulation of the BDNF-TrkB pathway, an essential signaling pathway in cerebellar morphogenesis. Here, we thus aim to provide a current view of peroxisome biogenesis and the molecular pathogenesis of PBDs.
Collapse
Affiliation(s)
- Yukio Fujiki
- Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yuichi Abe
- Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yuuta Imoto
- Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Akemi J Tanaka
- Department of Pediatrics, Columbia University Medical Center, New York, New York 10019, USA
| | - Kanji Okumoto
- Department of Biology, Kyushu University, 744 Motooka Nishi-ku, Fukuoka 819-0395, Japan
| | - Masanori Honsho
- Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Shigehiko Tamura
- Faculty of Arts and Science, Kyushu University, 744 Motooka Nishi-ku, Fukuoka 819-0395, Japan
| | - Non Miyata
- Chemistry, Faculty of Sciences, Kyushu University, 744 Motooka Nishi-ku, Fukuoka 819-0395, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871
| | - Wendy K Chung
- Department of Pediatrics, Columbia University Medical Center, New York, New York 10019, USA
| | - Tsuneyoshi Kuroiwa
- Department of Chemical and Biological Science, Faculty of Science, Japan Women's University, 2-8-1 Mejirodai, Bunkyo-ku, Tokyo 112-8681, Japan
| |
Collapse
|
21
|
Okumoto K, Tamura S, Honsho M, Fujiki Y. Peroxisome: Metabolic Functions and Biogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1299:3-17. [PMID: 33417203 DOI: 10.1007/978-3-030-60204-8_1] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Peroxisome is an organelle conserved in almost all eukaryotic cells with a variety of functions in cellular metabolism, including fatty acid β-oxidation, synthesis of ether glycerolipid plasmalogens, and redox homeostasis. Such metabolic functions and the exclusive importance of peroxisomes have been highlighted in fatal human genetic disease called peroxisomal biogenesis disorders (PBDs). Recent advances in this field have identified over 30 PEX genes encoding peroxins as essential factors for peroxisome biogenesis in various species from yeast to humans. Functional delineation of the peroxins has revealed that peroxisome biogenesis comprises the processes, involving peroxisomal membrane assembly, matrix protein import, division, and proliferation. Catalase, the most abundant peroxisomal enzyme, catalyzes decomposition of hydrogen peroxide. Peroxisome plays pivotal roles in the cellular redox homeostasis and the response to oxidative stresses, depending on intracellular localization of catalase.
Collapse
Affiliation(s)
- Kanji Okumoto
- Department of Biology, Faculty of Sciences, Kyushu University, Fukuoka, Japan
| | | | | | - Yukio Fujiki
- Institute of Rheological Functions of Food, Fukuoka, Japan. .,Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
22
|
Abe Y, Tamura S, Honsho M, Fujiki Y. A Mouse Model System to Study Peroxisomal Roles in Neurodegeneration of Peroxisome Biogenesis Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1299:119-143. [PMID: 33417212 DOI: 10.1007/978-3-030-60204-8_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Fourteen PEX genes are currently identified as genes responsible for peroxisome biogenesis disorders (PBDs). Patients with PBDs manifest as neurodegenerative symptoms such as neuronal migration defect and malformation of the cerebellum. To address molecular mechanisms underlying the pathogenesis of PBDs, mouse models for the PBDs have been generated by targeted disruption of Pex genes. Pathological phenotypes and metabolic abnormalities in Pex-knockout mice well resemble those of the patients with PBDs. The mice with tissue- or cell type-specific inactivation of Pex genes have also been established by using a Cre-loxP system. The genetically modified mice reveal that pathological phenotypes of PBDs are mediated by interorgan and intercellular communications. Despite the illustrations of detailed pathological phenotypes in the mutant mice, mechanistic insights into pathogenesis of PBDs are still underway. In this chapter, we overview the phenotypes of Pex-inactivated mice and the current understanding of the pathogenesis underlying PBDs.
Collapse
Affiliation(s)
- Yuichi Abe
- Faculty of Arts and Science, Kyushu University, Fukuoka, Japan
| | | | | | - Yukio Fujiki
- Institute of Rheological Functions of Food, Fukuoka, Japan. .,Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
23
|
Honsho M, Okumoto K, Tamura S, Fujiki Y. Peroxisome Biogenesis Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1299:45-54. [PMID: 33417206 DOI: 10.1007/978-3-030-60204-8_4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Peroxisomes are presented in all eukaryotic cells and play essential roles in many of lipid metabolic pathways, including β-oxidation of fatty acids and synthesis of ether-linked glycerophospholipids, such as plasmalogens. Impaired peroxisome biogenesis, including defects of membrane assembly, import of peroxisomal matrix proteins, and division of peroxisome, causes peroxisome biogenesis disorders (PBDs). Fourteen complementation groups of PBDs are found, and their complementing genes termed PEXs are isolated. Several new mutations in peroxins from patients with mild PBD phenotype or patients with phenotypes unrelated to the commonly observed impairments of PBD patients are found by next-generation sequencing. Exploring a dysfunctional step(s) caused by the mutation is important for unveiling the pathogenesis of novel mutation by means of cellular and biochemical analyses.
Collapse
Affiliation(s)
| | - Kanji Okumoto
- Department of Biology, Faculty of Sciences, Kyushu University Graduate School, Fukuoka, Japan
| | | | - Yukio Fujiki
- Institute of Rheological Functions of Food, Fukuoka, Japan. .,Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
24
|
Tian Y, Zhang L, Li Y, Gao J, Yu H, Guo Y, Jia L. Variant analysis of PEX11B gene from a family with peroxisome biogenesis disorder 14B by whole exome sequencing. Mol Genet Genomic Med 2019; 8. [PMID: 31724321 PMCID: PMC6978261 DOI: 10.1002/mgg3.1042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 08/30/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Peroxisome biogenesis disorder 14B (PBD14B) is an autosomal recessive peroxisome biogenesis disorder characterized clinically by mild intellectual disability, congenital cataracts, progressive hearing loss, and polyneuropathy peroxisome biogenesis disorders are genetically heterogeneous group of disorders caused by biallelic mutations in peroxin (PEX) genes. METHODOLOGY/LABORATORY EXAMINATION DNA of the family was extracted and sequenced by whole exome sequencing. The results were validated with Sanger sequencing analyzed with Bioinformatics software. RESULTS Sequencing result showed that the patient has carried a homozygous variant of c.277C>T of the PEX11B gene. The patient's brother has carried a homozygous variant of c.277C>T of the PEX11B gene and their variants of c.277C>T of the PEX11B gene were inherited, respectively, from his mother and father. DISCUSSION AND CONCLUSION The homozygous variant of c.277C>T of the PEX11B gene probably underlie the disease in this child and her brother.
Collapse
Affiliation(s)
- Yuan Tian
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Linlin Zhang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ying Li
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinshuang Gao
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Haiyang Yu
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yaqing Guo
- Department of Clinical Laboratory, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liting Jia
- Screening Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
25
|
Lismont C, Koster J, Provost S, Baes M, Van Veldhoven PP, Waterham HR, Fransen M. Deciphering the potential involvement of PXMP2 and PEX11B in hydrogen peroxide permeation across the peroxisomal membrane reveals a role for PEX11B in protein sorting. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2019; 1861:182991. [DOI: 10.1016/j.bbamem.2019.05.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/09/2019] [Accepted: 05/21/2019] [Indexed: 02/07/2023]
|
26
|
Garikapati V, Karnati S, Bhandari DR, Baumgart-Vogt E, Spengler B. High-resolution atmospheric-pressure MALDI mass spectrometry imaging workflow for lipidomic analysis of late fetal mouse lungs. Sci Rep 2019; 9:3192. [PMID: 30816198 PMCID: PMC6395778 DOI: 10.1038/s41598-019-39452-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 01/17/2019] [Indexed: 12/19/2022] Open
Abstract
Mass spectrometry imaging (MSI) provides label-free, non-targeted molecular and spatial information of the biomolecules within tissue. Lipids play important roles in lung biology, e.g. as surfactant, preventing alveolar collapse during normal and forced respiration. Lipidomic characterization of late fetal mouse lungs at day 19 of gestation (E19) has not been performed yet. In this study we employed high-resolution atmospheric pressure scanning microprobe matrix-assisted laser desorption/ionization MSI for the lipidomic analysis of E19 mouse lungs. Molecular species of different lipid classes were imaged in E19 lung sections at high spatial and mass resolution in positive- and negative-ion mode. Lipid species were characterized based on accurate mass and on-tissue tandem mass spectrometry. In addition, a dedicated sample preparation protocol, homogenous deposition of matrices on tissue surfaces and data processing parameters were optimized for the comparison of signal intensities of lipids between different tissue sections of E19 lungs of wild type and Pex11β-knockout mice. Our study provides lipid information of E19 mouse lungs, optimized experimental and data processing strategies for the direct comparison of signal intensities of metabolites (lipids) among the tissue sections from MSI experiments. To best of our knowledge, this is the first MSI and lipidomic study of E19 mouse lungs.
Collapse
Affiliation(s)
- Vannuruswamy Garikapati
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Giessen, Germany.,Institute for Anatomy and Cell Biology II, Division of Medical Cell Biology, Justus Liebig University Giessen, Giessen, Germany
| | - Srikanth Karnati
- Institute for Anatomy and Cell Biology II, Division of Medical Cell Biology, Justus Liebig University Giessen, Giessen, Germany.,Institute for Anatomy and Cell Biology, Julius Maximilians University Würzburg, Würzburg, Germany
| | - Dhaka Ram Bhandari
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Giessen, Germany
| | - Eveline Baumgart-Vogt
- Institute for Anatomy and Cell Biology II, Division of Medical Cell Biology, Justus Liebig University Giessen, Giessen, Germany
| | - Bernhard Spengler
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, Giessen, Germany.
| |
Collapse
|
27
|
Chen C, Wang H, Chen B, Chen D, Lu C, Li H, Qian Y, Tan Y, Weng H, Cai L. Pex11a deficiency causes dyslipidaemia and obesity in mice. J Cell Mol Med 2018; 23:2020-2031. [PMID: 30585412 PMCID: PMC6378206 DOI: 10.1111/jcmm.14108] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 11/20/2018] [Accepted: 12/03/2018] [Indexed: 11/29/2022] Open
Abstract
Peroxisomes play a central role in lipid metabolism. We previously demonstrated that Pex11a deficiency impairs peroxisome abundance and fatty acid β‐oxidation and results in hepatic triglyceride accumulation. The role of Pex11a in dyslipidaemia and obesity is investigated here with Pex11a knockout mice (Pex11a−/−). Metabolic phenotypes including tissue weight, glucose tolerance, insulin sensitivity, cholesterol levels, fatty acid profile, oxygen consumption, physical activity were assessed in wild‐type (WT) and Pex11a−/− fed with a high‐fat diet. Molecular changes and peroxisome abundance in adipose tissue were evaluated through qRT‐PCR, Western blotting, and Immunofluorescence. Pex11a−/− showed increased fat mass, decreased skeletal muscle, higher cholesterol levels, and more severely impaired glucose and insulin tolerance. Pex11a−/− consumed less oxygen, indicating a decrease in fatty acid oxidation, which is consistent with the accumulation of very long‐ and long‐chain fatty acids. Adipose palmitic acid (C16:0) levels were elevated in Pex11a−/−, which may be because of dramatically increased fatty acid synthase mRNA and protein levels. Furthermore, Pex11a deficiency increased ventricle size and macrophage infiltration, which are related to the reduced physical activity. These data demonstrate that Pex11a deficiency impairs physical activity and energy expenditure, decreases fatty acid β‐oxidation, increases de novo lipogenesis and results in dyslipidaemia and obesity.
Collapse
Affiliation(s)
- Congcong Chen
- Chinese-American Research Institute for Pediatrics & Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Chashan University-Town, Wenzhou, China.,Department of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,Department of Pharmacy, Jinhua Central Hospital, Jinhua, China
| | - Hongwei Wang
- Hepatobiliary and Pancreatic Surgery Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bicheng Chen
- Hepatobiliary and Pancreatic Surgery Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Deyuan Chen
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chaosheng Lu
- Chinese-American Research Institute for Pediatrics & Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Chashan University-Town, Wenzhou, China
| | - Haiyan Li
- Chinese-American Research Institute for Pediatrics & Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Chashan University-Town, Wenzhou, China
| | - Yan Qian
- Chinese-American Research Institute for Pediatrics & Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Chashan University-Town, Wenzhou, China
| | - Yi Tan
- Chinese-American Research Institute for Pediatrics & Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Chashan University-Town, Wenzhou, China.,Department of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,Pediatric Research Institute, Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Huachun Weng
- Chinese-American Research Institute for Pediatrics & Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Chashan University-Town, Wenzhou, China
| | - Lu Cai
- Chinese-American Research Institute for Pediatrics & Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Chashan University-Town, Wenzhou, China.,Department of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.,Pediatric Research Institute, Departments of Pediatrics, Radiation Oncology, Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| |
Collapse
|
28
|
Jean Beltran PM, Cook KC, Hashimoto Y, Galitzine C, Murray LA, Vitek O, Cristea IM. Infection-Induced Peroxisome Biogenesis Is a Metabolic Strategy for Herpesvirus Replication. Cell Host Microbe 2018; 24:526-541.e7. [PMID: 30269970 PMCID: PMC6195127 DOI: 10.1016/j.chom.2018.09.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/12/2018] [Accepted: 08/20/2018] [Indexed: 12/28/2022]
Abstract
Viral proteins have evolved to target cellular organelles and usurp their functions for virus replication. Despite the knowledge of these critical functions for several organelles, little is known about peroxisomes during infection. Peroxisomes are primarily metabolic organelles with important functions in lipid metabolism. Here, we discovered that the enveloped viruses human cytomegalovirus (HCMV) and herpes simplex virus type 1 (HSV-1) induce the biogenesis of and unique morphological changes to peroxisomes to support their replication. Targeted proteomic quantification revealed a global virus-induced upregulation of peroxisomal proteins. Mathematical modeling and microscopy structural analysis show that infection triggers peroxisome growth and fission, leading to increased peroxisome numbers and irregular disc-like structures. HCMV-induced peroxisome biogenesis increased the phospholipid plasmalogen, thereby enhancing virus production. Peroxisome regulation and dependence were not observed for the non-enveloped adenovirus. Our findings uncover a role of peroxisomes in viral pathogenesis, with likely implications for multiple enveloped viruses.
Collapse
Affiliation(s)
- Pierre M Jean Beltran
- Department of Molecular Biology, Princeton University, Lewis Thomas Laboratory, Washington Road, Princeton, NJ 08544, USA
| | - Katelyn C Cook
- Department of Molecular Biology, Princeton University, Lewis Thomas Laboratory, Washington Road, Princeton, NJ 08544, USA
| | - Yutaka Hashimoto
- Department of Molecular Biology, Princeton University, Lewis Thomas Laboratory, Washington Road, Princeton, NJ 08544, USA
| | - Cyril Galitzine
- College of Science, College of Computer and Information Science, Northeastern University, Boston, MA 02115, USA
| | - Laura A Murray
- Department of Molecular Biology, Princeton University, Lewis Thomas Laboratory, Washington Road, Princeton, NJ 08544, USA
| | - Olga Vitek
- College of Science, College of Computer and Information Science, Northeastern University, Boston, MA 02115, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Lewis Thomas Laboratory, Washington Road, Princeton, NJ 08544, USA.
| |
Collapse
|
29
|
Deori NM, Kale A, Maurya PK, Nagotu S. Peroxisomes: role in cellular ageing and age related disorders. Biogerontology 2018; 19:303-324. [PMID: 29968207 DOI: 10.1007/s10522-018-9761-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 06/27/2018] [Indexed: 12/12/2022]
Abstract
Peroxisomes are dynamic organelles essential for optimum functioning of a eukaryotic cell. Biogenesis of these organelles and the diverse functions performed by them have been extensively studied in the past decade. Their ability to perform functions depending on the cell type and growth conditions is unique and remarkable. Oxidation of fatty acids and reactive oxygen species metabolism are the two most important functions of these ubiquitous organelles. They are often referred to as both source and sink of reactive oxygen species in a cell. Recent research connects peroxisome dysfunction to fatal oxidative damage associated with ageing-related diseases/disorders. It is now widely accepted that mitochondria and peroxisomes are required to maintain oxidative balance in a cell. However, our understanding on the inter-dependence of these organelles to maintain cellular homeostasis of reactive oxygen species is still in its infancy. Herein, we summarize findings that highlight the role of peroxisomes in cellular reactive oxygen species metabolism, ageing and age-related disorders.
Collapse
Affiliation(s)
- Nayan M Deori
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Avinash Kale
- UM-DAE, Centre for Excellence in Basic Sciences, Health Centre, University of Mumbai, Mumbai, 400098, India
| | - Pawan K Maurya
- Interdisciplinary Laboratory for Clinical Neuroscience (LiNC), Universidade Federal de Sao Paulo-UNIFESP, Sao Paulo, Brazil
| | - Shirisha Nagotu
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
30
|
Mast FD, Herricks T, Strehler KM, Miller LR, Saleem RA, Rachubinski RA, Aitchison JD. ESCRT-III is required for scissioning new peroxisomes from the endoplasmic reticulum. J Cell Biol 2018; 217:2087-2102. [PMID: 29588378 PMCID: PMC5987711 DOI: 10.1083/jcb.201706044] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 01/23/2018] [Accepted: 03/05/2018] [Indexed: 12/11/2022] Open
Abstract
Dynamic control of peroxisome proliferation is integral to the peroxisome's many functions. The endoplasmic reticulum (ER) serves as a source of preperoxisomal vesicles (PPVs) that mature into peroxisomes during de novo peroxisome biogenesis and support growth and division of existing peroxisomes. However, the mechanism of PPV formation and release from the ER remains poorly understood. In this study, we show that endosomal sorting complexes required for transport (ESCRT)-III are required to release PPVs budding from the ER into the cytosol. Absence of ESCRT-III proteins impedes de novo peroxisome formation and results in an aberrant peroxisome population in vivo. Using a cell-free PPV budding assay, we show that ESCRT-III proteins Vps20 and Snf7 are necessary to release PPVs from the ER. ESCRT-III is therefore a positive effector of membrane scission for vesicles budding both away from and toward the cytosol. These findings have important implications for the evolutionary timing of emergence of peroxisomes and the rest of the internal membrane architecture of the eukaryotic cell.
Collapse
Affiliation(s)
- Fred D. Mast
- Center for Infectious Disease Research, Seattle, WA
- Institute for Systems Biology, Seattle, WA
| | - Thurston Herricks
- Center for Infectious Disease Research, Seattle, WA
- Institute for Systems Biology, Seattle, WA
| | - Kathleen M. Strehler
- Center for Infectious Disease Research, Seattle, WA
- Institute for Systems Biology, Seattle, WA
| | - Leslie R. Miller
- Center for Infectious Disease Research, Seattle, WA
- Institute for Systems Biology, Seattle, WA
| | - Ramsey A. Saleem
- Center for Infectious Disease Research, Seattle, WA
- Institute for Systems Biology, Seattle, WA
| | | | - John D. Aitchison
- Center for Infectious Disease Research, Seattle, WA
- Institute for Systems Biology, Seattle, WA
- Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
31
|
Colasante C, Chen J, Ahlemeyer B, Baumgart-Vogt E. Peroxisomes in cardiomyocytes and the peroxisome / peroxisome proliferator-activated receptor-loop. Thromb Haemost 2017; 113:452-63. [DOI: 10.1160/th14-06-0497] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 12/18/2014] [Indexed: 01/29/2023]
Abstract
SummaryIt is well established that the heart is strongly dependent on fatty acid metabolism. In cardiomyocytes there are two distinct sites for the β-oxidisation of fatty acids: the mitochondrion and the peroxisome. Although the metabolism of these two organelles is believed to be tightly coupled, the nature of this relationship has not been fully investigated. Recent research has established the significant contribution of mitochondrial function to cardiac ATP production under normal and pathological conditions. In contrast, limited information is available on peroxisomal function in the heart. This is despite these organelles harbouring metabolic pathways that are potentially cardioprotective, and findings that patients with peroxisomal diseases, such as adult Refsum’s disease, can develop heart failure. In this article, we provide a comprehensive overview on the current knowledge of peroxisomes and the regulation of lipid metabolism by PPARs in cardiomyocytes. We also present new experimental evidence on the differential expression of peroxisome-related genes in the heart chambers and demonstrate that even a mild peroxisomal biogenesis defect (Pex11α-/- ) can induce profound alterations in the cardiomyocyte’s peroxisomal compartment and related gene expression, including the concomitant deregulation of specific PPARs. The possible impact of peroxisomal dysfunction in the heart is discussed and a model for the modulation of myocardial metabolism via a peroxisome/PPAR-loop is proposed.
Collapse
|
32
|
Asare A, Levorse J, Fuchs E. Coupling organelle inheritance with mitosis to balance growth and differentiation. Science 2017; 355:355/6324/eaah4701. [PMID: 28154022 DOI: 10.1126/science.aah4701] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 09/26/2016] [Accepted: 11/23/2016] [Indexed: 01/03/2023]
Abstract
Balancing growth and differentiation is essential to tissue morphogenesis and homeostasis. How imbalances arise in disease states is poorly understood. To address this issue, we identified transcripts differentially expressed in mouse basal epidermal progenitors versus their differentiating progeny and those altered in cancers. We used an in vivo RNA interference screen to unveil candidates that altered the equilibrium between the basal proliferative layer and suprabasal differentiating layers forming the skin barrier. We found that epidermal progenitors deficient in the peroxisome-associated protein Pex11b failed to segregate peroxisomes properly and entered a mitotic delay that perturbed polarized divisions and skewed daughter fates. Together, our findings unveil a role for organelle inheritance in mitosis, spindle alignment, and the choice of daughter progenitors to differentiate or remain stem-like.
Collapse
Affiliation(s)
- Amma Asare
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY 10065, USA
| | - John Levorse
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY 10065, USA
| | - Elaine Fuchs
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
33
|
Abstract
Peroxisomes contribute to cell division and differentiation decisions in the mammalian epidermis
Collapse
Affiliation(s)
- Ulrike Gruneberg
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| | - Francis Barr
- Department of Biochemistry, University of Oxford, Oxford, UK.
| |
Collapse
|
34
|
Kim D, Song J, Kang Y, Park S, Kim YI, Kwak S, Lim D, Park R, Chun CH, Choe SK, Jin EJ. Fis1 depletion in osteoarthritis impairs chondrocyte survival and peroxisomal and lysosomal function. J Mol Med (Berl) 2016; 94:1373-1384. [PMID: 27497958 DOI: 10.1007/s00109-016-1445-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 06/16/2016] [Accepted: 06/29/2016] [Indexed: 11/26/2022]
Abstract
Cumulative evidence suggests the importance of organelle homeostasis in regulating metabolic functions in response to various cellular stresses. Particularly, the dynamism and health of the mitochondria-peroxisome network through fission and fusion are essential for cellular function; dysfunctional dynamism underlies the pathogenesis of several degenerative diseases including Parkinson's disease. Here, we investigated the role of Fis1 in cartilage homeostasis and its relevance to osteoarthritis (OA). We found that Fis1 is significantly suppressed in human OA chondrocytes compared to that in normal chondrocytes. Fis1 depletion through siRNA induced peroxisomal dysfunction. Moreover, Fis1 suppression altered miRNA profiles, especially those implicated in lysosomal regulation. Lysosomal destruction using LAMP-1-specific targeted nanorods or lysosomal dysfunction through chloroquine treatment resulted in enhanced chondrocyte apoptosis and/or suppression of autophagy. Accordingly, lysosomal activity and autophagy were severely decreased in OA chondrocytes despite abundant LAMP-1-positive organelles. Moreover, Fis1 morpholino-injected zebrafish embryos displayed lysosome accumulation, mitochondrial dysfunction, and peroxisome reduction. Collectively, these data suggest interconnected links among Fis1-modulated miRNA, lysosomes, and autophagy, which contributes to chondrocyte survival/apoptosis. This study represents the first functional study of Fis1 with its pathological relevance to OA. Our data suggest a new target for controlling cartilage-degenerative diseases, such as OA. KEY MESSAGE Fis1 suppression in OA chondrocytes induces accumulation and inhibition of lysosomes. Fis1 suppression alters miRNAs, especially those implicated in lysosomal regulation. Lysosomal destruction results in chondrocyte apoptosis and suppression of autophagy. Fis1 depletion in zebrafish causes lysosome accumulation, mitochondrial dysfunction, and peroxisome reduction. This is the first functional study of Fis1 and its pathological relevance to OA.
Collapse
Affiliation(s)
- Dongkyun Kim
- Department of Biological Sciences, College of Natural Sciences, Wonkwang University, Iksan, Chunbuk, 570-749, South Korea
| | - Jinsoo Song
- Department of Biological Sciences, College of Natural Sciences, Wonkwang University, Iksan, Chunbuk, 570-749, South Korea
| | - Yeonho Kang
- Department of Biological Sciences, College of Natural Sciences, Wonkwang University, Iksan, Chunbuk, 570-749, South Korea
| | - Sujung Park
- Department of Biological Sciences, College of Natural Sciences, Wonkwang University, Iksan, Chunbuk, 570-749, South Korea
| | - Yong-Il Kim
- Department of Microbiology, Wonkwang University School of Medicine, Iksan, Chunbuk, 570-749, South Korea
| | - Seongae Kwak
- Department of Microbiology, Wonkwang University School of Medicine, Iksan, Chunbuk, 570-749, South Korea
| | - Dongkwon Lim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 136-701, South Korea
| | - Raekil Park
- Department of Microbiology, Wonkwang University School of Medicine, Iksan, Chunbuk, 570-749, South Korea
| | - Churl-Hong Chun
- Department of Orthopedic Surgery, Wonkwang University School of Medicine, Iksan, Chunbuk, 570-749, South Korea
| | - Seong-Kyu Choe
- Department of Microbiology, Wonkwang University School of Medicine, Iksan, Chunbuk, 570-749, South Korea.
- Integrated Omics Institute, Wonkwang University, Iksan, Chunbuk, 570-749, South Korea.
| | - Eun-Jung Jin
- Department of Biological Sciences, College of Natural Sciences, Wonkwang University, Iksan, Chunbuk, 570-749, South Korea.
- Integrated Omics Institute, Wonkwang University, Iksan, Chunbuk, 570-749, South Korea.
| |
Collapse
|
35
|
Affiliation(s)
| | - Maria Daniela D'Agostino
- McGill University Department of Human Genetics and McGill University Health Center, Department of Medical Genetics, Montreal, QC, Canada
| | - Nancy Braverman
- McGill University Department of Human Genetics and Pediatrics, and The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
36
|
Peroxisome homeostasis: Mechanisms of division and selective degradation of peroxisomes in mammals. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:984-91. [DOI: 10.1016/j.bbamcr.2015.09.032] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 09/25/2015] [Accepted: 09/28/2015] [Indexed: 12/25/2022]
|
37
|
Human disorders of peroxisome metabolism and biogenesis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:922-33. [DOI: 10.1016/j.bbamcr.2015.11.015] [Citation(s) in RCA: 230] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 11/16/2015] [Accepted: 11/17/2015] [Indexed: 12/22/2022]
|
38
|
Zientara-Rytter K, Subramani S. Autophagic degradation of peroxisomes in mammals. Biochem Soc Trans 2016; 44:431-40. [PMID: 27068951 PMCID: PMC4958620 DOI: 10.1042/bst20150268] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Indexed: 12/21/2022]
Abstract
Peroxisomes are essential organelles required for proper cell function in all eukaryotic organisms. They participate in a wide range of cellular processes including the metabolism of lipids and generation, as well as detoxification, of hydrogen peroxide (H2O2). Therefore, peroxisome homoeostasis, manifested by the precise and efficient control of peroxisome number and functionality, must be tightly regulated in response to environmental changes. Due to the existence of many physiological disorders and diseases associated with peroxisome homoeostasis imbalance, the dynamics of peroxisomes have been widely examined. The increasing volume of reports demonstrating significant involvement of the autophagy machinery in peroxisome removal leads us to summarize current knowledge of peroxisome degradation in mammalian cells. In this review we present current models of peroxisome degradation. We particularly focus on pexophagy-the selective clearance of peroxisomes through autophagy. We also critically discuss concepts of peroxisome recognition for pexophagy, including signalling and selectivity factors. Finally, we present examples of the pathological effects of pexophagy dysfunction and suggest promising future directions.
Collapse
Affiliation(s)
- Katarzyna Zientara-Rytter
- Section of Molecular Biology, Division of Biological Sciences, University California, San Diego, CA 92093-0322, U.S.A
| | - Suresh Subramani
- Section of Molecular Biology, Division of Biological Sciences, University California, San Diego, CA 92093-0322, U.S.A.
| |
Collapse
|
39
|
Kamisugi Y, Mitsuya S, El‐Shami M, Knight CD, Cuming AC, Baker A. Giant peroxisomes in a moss (Physcomitrella patens) peroxisomal biogenesis factor 11 mutant. THE NEW PHYTOLOGIST 2016; 209:576-89. [PMID: 26542980 PMCID: PMC4738463 DOI: 10.1111/nph.13739] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 10/01/2015] [Indexed: 05/22/2023]
Abstract
Peroxisomal biogenesis factor 11 (PEX11) proteins are found in yeasts, mammals and plants, and play a role in peroxisome morphology and regulation of peroxisome division. The moss Physcomitrella patens has six PEX11 isoforms which fall into two subfamilies, similar to those found in monocots and dicots. We carried out targeted gene disruption of the Phypa_PEX11-1 gene and compared the morphological and cellular phenotypes of the wild-type and mutant strains. The mutant grew more slowly and the development of gametophores was retarded. Mutant chloronemal filaments contained large cellular structures which excluded all other cellular organelles. Expression of fluorescent reporter proteins revealed that the mutant strain had greatly enlarged peroxisomes up to 10 μm in diameter. Expression of a vacuolar membrane marker confirmed that the enlarged structures were not vacuoles, or peroxisomes sequestered within vacuoles as a result of pexophagy. Phypa_PEX11 targeted to peroxisome membranes could rescue the knock out phenotype and interacted with Fission1 on the peroxisome membrane. Moss PEX11 functions in peroxisome division similar to PEX11 in other organisms but the mutant phenotype is more extreme and environmentally determined, making P. patens a powerful system in which to address mechanisms of peroxisome proliferation and division.
Collapse
Affiliation(s)
- Yasuko Kamisugi
- Centre for Plant SciencesFaculty of Biological SciencesUniversity of LeedsLeedsLS2 9JTUK
| | - Shiro Mitsuya
- Centre for Plant SciencesFaculty of Biological SciencesUniversity of LeedsLeedsLS2 9JTUK
| | - Mahmoud El‐Shami
- Centre for Plant SciencesFaculty of Biological SciencesUniversity of LeedsLeedsLS2 9JTUK
| | - Celia D. Knight
- Centre for Plant SciencesFaculty of Biological SciencesUniversity of LeedsLeedsLS2 9JTUK
| | - Andrew C. Cuming
- Centre for Plant SciencesFaculty of Biological SciencesUniversity of LeedsLeedsLS2 9JTUK
| | - Alison Baker
- Centre for Plant SciencesFaculty of Biological SciencesUniversity of LeedsLeedsLS2 9JTUK
| |
Collapse
|
40
|
FUJIKI Y. Peroxisome biogenesis and human peroxisome-deficiency disorders. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2016; 92:463-477. [PMID: 27941306 PMCID: PMC5328784 DOI: 10.2183/pjab.92.463] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Peroxisome is a single-membrane-bounded ubiquitous organelle containing a hundred different enzymes that catalyze various metabolic pathways such as β-oxidation of very long-chain fatty acids and synthesis of plasmalogens. To investigate peroxisome biogenesis and human peroxisome biogenesis disorders (PBDs) including Zellweger syndrome, more than a dozen different complementation groups of Chinese hamster ovary (CHO) cell mutants impaired in peroxisome biogenesis are isolated as a model experimental system. By taking advantage of rapid functional complementation assay of the CHO cell mutants, successful cloning of PEX genes encoding peroxins required for peroxisome assembly invaluably contributed to the accomplishment of cloning of pathogenic genes responsible for PBDs. Peroxins are divided into three groups: 1) peroxins including Pex3p, Pex16p and Pex19p, are responsible for peroxisome membrane biogenesis via Pex19p- and Pex3p-dependent class I and Pex19p- and Pex16p-dependent class II pathways; 2) peroxins that function in matrix protein import; 3) those such as Pex11pβ are involved in peroxisome division where DLP1, Mff, and Fis1 coordinately function.
Collapse
Affiliation(s)
- Yukio FUJIKI
- Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
- Correspondence should be addressed: Y. Fujiki, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan (e-mail: )
| |
Collapse
|
41
|
C22-bronchial and T7-alveolar epithelial cell lines of the immortomouse are excellent murine cell culture model systems to study pulmonary peroxisome biology and metabolism. Histochem Cell Biol 2015; 145:287-304. [DOI: 10.1007/s00418-015-1385-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2015] [Indexed: 10/22/2022]
|
42
|
Kandimalla R, Reddy PH. Multiple faces of dynamin-related protein 1 and its role in Alzheimer's disease pathogenesis. Biochim Biophys Acta Mol Basis Dis 2015; 1862:814-828. [PMID: 26708942 DOI: 10.1016/j.bbadis.2015.12.018] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/08/2015] [Accepted: 12/15/2015] [Indexed: 01/01/2023]
Abstract
Mitochondria play a large role in neuronal function by constantly providing energy, particularly at synapses. Recent studies suggest that amyloid beta (Aβ) and phosphorylated tau interact with the mitochondrial fission protein, dynamin-related protein 1 (Drp1), causing excessive fragmentation of mitochondria and leading to abnormal mitochondrial dynamics and synaptic degeneration in Alzheimer's disease (AD) neurons. Recent research also revealed Aβ-induced and phosphorylated tau-induced changes in mitochondria, particularly affecting mitochondrial shape, size, distribution and axonal transport in AD neurons. These changes affect mitochondrial health and, in turn, could affect synaptic function and neuronal damage and ultimately leading to memory loss and cognitive impairment in patients with AD. This article highlights recent findings in the role of Drp1 in AD pathogenesis. This article also highlights Drp1 and its relationships to glycogen synthase kinase 3, cyclin-dependent kinase 5, p53, and microRNAs in AD pathogenesis.
Collapse
Affiliation(s)
- Ramesh Kandimalla
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States
| | - P Hemachandra Reddy
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States; Cell Biology & Biochemistry, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States; Department of Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States; Department of Neurology, Texas Tech University Health Sciences Center, 3601 4(th) Street, MS 9424, Lubbock, TX 79430, United States; Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, 6630 S. Quaker Ste. E, MS 7495, Lubbock, TX 79413, United States.
| |
Collapse
|
43
|
Berger J, Dorninger F, Forss-Petter S, Kunze M. Peroxisomes in brain development and function. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:934-55. [PMID: 26686055 PMCID: PMC4880039 DOI: 10.1016/j.bbamcr.2015.12.005] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/04/2015] [Accepted: 12/09/2015] [Indexed: 12/26/2022]
Abstract
Peroxisomes contain numerous enzymatic activities that are important for mammalian physiology. Patients lacking either all peroxisomal functions or a single enzyme or transporter function typically develop severe neurological deficits, which originate from aberrant development of the brain, demyelination and loss of axonal integrity, neuroinflammation or other neurodegenerative processes. Whilst correlating peroxisomal properties with a compilation of pathologies observed in human patients and mouse models lacking all or individual peroxisomal functions, we discuss the importance of peroxisomal metabolites and tissue- and cell type-specific contributions to the observed brain pathologies. This enables us to deconstruct the local and systemic contribution of individual metabolic pathways to specific brain functions. We also review the recently discovered variability of pathological symptoms in cases with unexpectedly mild presentation of peroxisome biogenesis disorders. Finally, we explore the emerging evidence linking peroxisomes to more common neurological disorders such as Alzheimer’s disease, autism and amyotrophic lateral sclerosis. This article is part of a Special Issue entitled: Peroxisomes edited by Ralf Erdmann.
Collapse
Affiliation(s)
- Johannes Berger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria.
| | - Fabian Dorninger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria.
| | - Sonja Forss-Petter
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria.
| | - Markus Kunze
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria.
| |
Collapse
|
44
|
Qian G, Fan W, Ahlemeyer B, Karnati S, Baumgart-Vogt E. Peroxisomes in Different Skeletal Cell Types during Intramembranous and Endochondral Ossification and Their Regulation during Osteoblast Differentiation by Distinct Peroxisome Proliferator-Activated Receptors. PLoS One 2015; 10:e0143439. [PMID: 26630504 PMCID: PMC4668026 DOI: 10.1371/journal.pone.0143439] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2015] [Accepted: 11/04/2015] [Indexed: 01/10/2023] Open
Abstract
Ossification defects leading to craniofacial dysmorphism or rhizomelia are typical phenotypes in patients and corresponding knockout mouse models with distinct peroxisomal disorders. Despite these obvious skeletal pathologies, to date no careful analysis exists on the distribution and function of peroxisomes in skeletal tissues and their alterations during ossification. Therefore, we analyzed the peroxisomal compartment in different cell types of mouse cartilage and bone as well as in primary cultures of calvarial osteoblasts. The peroxisome number and metabolism strongly increased in chondrocytes during endochondral ossification from the reserve to the hypertrophic zone, whereas in bone, metabolically active osteoblasts contained a higher numerical abundance of this organelle than osteocytes. The high abundance of peroxisomes in these skeletal cell types is reflected by high levels of Pex11β gene expression. During culture, calvarial pre-osteoblasts differentiated into secretory osteoblasts accompanied by peroxisome proliferation and increased levels of peroxisomal genes and proteins. Since many peroxisomal genes contain a PPAR-responsive element, we analyzed the gene expression of PPARɑ/ß/ɣ in calvarial osteoblasts and MC3T3-E1 cells, revealing higher levels for PPARß than for PPARɑ and PPARɣ. Treatment with different PPAR agonists and antagonists not only changed the peroxisomal compartment and associated gene expression, but also induced complex alterations of the gene expression patterns of the other PPAR family members. Studies in M3CT3-E1 cells showed that the PPARß agonist GW0742 activated the PPRE-mediated luciferase expression and up-regulated peroxisomal gene transcription (Pex11, Pex13, Pex14, Acox1 and Cat), whereas the PPARß antagonist GSK0660 led to repression of the PPRE and a decrease of the corresponding mRNA levels. In the same way, treatment of calvarial osteoblasts with GW0742 increased in peroxisome number and related gene expression and accelerated osteoblast differentiation. Taken together, our results suggest that PPARß regulates the numerical abundance and metabolic function of peroxisomes via Pex11ß in parallel to osteoblast differentiation.
Collapse
Affiliation(s)
- Guofeng Qian
- Institute for Anatomy and Cell Biology, Medical Cell Biology, Justus-Liebig-University, Aulweg 123, 35385 Giessen, Germany
| | - Wei Fan
- Institute for Anatomy and Cell Biology, Medical Cell Biology, Justus-Liebig-University, Aulweg 123, 35385 Giessen, Germany
| | - Barbara Ahlemeyer
- Institute for Anatomy and Cell Biology, Medical Cell Biology, Justus-Liebig-University, Aulweg 123, 35385 Giessen, Germany
| | - Srikanth Karnati
- Institute for Anatomy and Cell Biology, Medical Cell Biology, Justus-Liebig-University, Aulweg 123, 35385 Giessen, Germany
| | - Eveline Baumgart-Vogt
- Institute for Anatomy and Cell Biology, Medical Cell Biology, Justus-Liebig-University, Aulweg 123, 35385 Giessen, Germany
- * E-mail:
| |
Collapse
|
45
|
Mindthoff S, Grunau S, Steinfort LL, Girzalsky W, Hiltunen JK, Erdmann R, Antonenkov VD. Peroxisomal Pex11 is a pore-forming protein homologous to TRPM channels. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:271-83. [PMID: 26597702 DOI: 10.1016/j.bbamcr.2015.11.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 10/16/2015] [Accepted: 11/16/2015] [Indexed: 01/16/2023]
Abstract
More than 30 proteins (Pex proteins) are known to participate in the biogenesis of peroxisomes-ubiquitous oxidative organelles involved in lipid and ROS metabolism. The Pex11 family of homologous proteins is responsible for division and proliferation of peroxisomes. We show that yeast Pex11 is a pore-forming protein sharing sequence similarity with TRPM cation-selective channels. The Pex11 channel with a conductance of Λ=4.1 nS in 1.0M KCl is moderately cation-selective (PK(+)/PCl(-)=1.85) and resistant to voltage-dependent closing. The estimated size of the channel's pore (r~0.6 nm) supports the notion that Pex11 conducts solutes with molecular mass below 300-400 Da. We localized the channel's selectivity determining sequence. Overexpression of Pex11 resulted in acceleration of fatty acids β-oxidation in intact cells but not in the corresponding lysates. The β-oxidation was affected in cells by expression of the Pex11 protein carrying point mutations in the selectivity determining sequence. These data suggest that the Pex11-dependent transmembrane traffic of metabolites may be a rate-limiting step in the β-oxidation of fatty acids. This conclusion was corroborated by analysis of the rate of β-oxidation in yeast strains expressing Pex11 with mutations mimicking constitutively phosphorylated (S165D, S167D) or unphosphorylated (S165A, S167A) protein. The results suggest that phosphorylation of Pex11 is a mechanism that can control the peroxisomal β-oxidation rate. Our results disclose an unexpected function of Pex11 as a non-selective channel responsible for transfer of metabolites across peroxisomal membrane. The data indicate that peroxins may be involved in peroxisomal metabolic processes in addition to their role in peroxisome biogenesis.
Collapse
Affiliation(s)
- Sabrina Mindthoff
- Institut für Biochemie und Pathobiochemie, Abt. Systembiochemie, Ruhr-Universität, Bochum, Germany
| | - Silke Grunau
- Faculty of Biochemistry and Molecular Medicine and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Laura L Steinfort
- Institut für Biochemie und Pathobiochemie, Abt. Systembiochemie, Ruhr-Universität, Bochum, Germany
| | - Wolfgang Girzalsky
- Institut für Biochemie und Pathobiochemie, Abt. Systembiochemie, Ruhr-Universität, Bochum, Germany
| | - J Kalervo Hiltunen
- Faculty of Biochemistry and Molecular Medicine and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Ralf Erdmann
- Institut für Biochemie und Pathobiochemie, Abt. Systembiochemie, Ruhr-Universität, Bochum, Germany.
| | - Vasily D Antonenkov
- Faculty of Biochemistry and Molecular Medicine and Biocenter Oulu, University of Oulu, Oulu, Finland.
| |
Collapse
|
46
|
Esmaeili M, Ghaedi K, Shoaraye Nejati A, Nematollahi M, Shiralyian H, Nasr-Esfahani MH. Pioglitazone significantly prevented decreased rate of neural differentiation of mouse embryonic stem cells which was reduced by Pex11β knock-down. Neuroscience 2015; 312:35-47. [PMID: 26562432 DOI: 10.1016/j.neuroscience.2015.11.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 11/03/2015] [Accepted: 11/03/2015] [Indexed: 12/12/2022]
Abstract
Peroxisomes constitute special cellular organelles which display a variety of metabolic functions including fatty acid oxidation and free radical elimination. Abundance of these flexible organelles varies in response to different environmental stimuli. It has been demonstrated that PEX11β, a peroxisomal membrane elongation factor, is involved in the regulation of size, shape and number of peroxisomes. To investigate the role of PEX11β in neural differentiation of mouse embryonic stem cells (mESCs), we generated a stably transduced mESCs line that derives the expression of a short hairpin RNA against Pex11β gene following doxycycline (Dox) induction. Knock-down of Pex11β, during neural differentiation, significantly reduced the expression of neural progenitor cells and mature neuronal markers (p<0.05) indicating that decreased expression of PEX11β suppresses neuronal maturation. Additionally, mRNA levels of other peroxisome-related genes such as PMP70, Pex11α, Catalase, Pex19 and Pex5 were also significantly reduced by Pex11β knock-down (p<0.05). Interestingly, pretreatment of transduced mESCs with peroxisome proliferator-activated receptor γ agonist (pioglitazone (Pio)) ameliorated the inhibitory effects of Pex11β knock down on neural differentiation. Pio also significantly (p<0.05) increased the expression of neural progenitor and mature neuronal markers besides the expression of peroxisomal genes in transduced mESC. Results elucidated the importance of Pex11β expression in neural differentiation of mESCs, thereby highlighting the essential role of peroxisomes in mammalian neural differentiation. The observation that Pio recovered peroxisomal function and improved neural differentiation of Pex11β knocked-down mESCs, proposes a potential new pharmacological implication of Pio for neurogenesis in patients with peroxisomal defects.
Collapse
Affiliation(s)
- M Esmaeili
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - K Ghaedi
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran; Division of Cellular and Molecular Biology, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran.
| | - A Shoaraye Nejati
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - M Nematollahi
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - H Shiralyian
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - M H Nasr-Esfahani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| |
Collapse
|
47
|
Gallego-Llamas J, Timms AE, Geister KA, Lindsay A, Beier DR. Variant mapping and mutation discovery in inbred mice using next-generation sequencing. BMC Genomics 2015; 16:913. [PMID: 26552429 PMCID: PMC4640199 DOI: 10.1186/s12864-015-2173-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 10/31/2015] [Indexed: 12/04/2022] Open
Abstract
Background The development of powerful new methods for DNA sequencing enable the discovery of sequence variants, their utilization for the mapping of mutant loci, and the identification of causal variants in a single step. We have applied this approach for the analysis of ENU-mutagenized mice maintained on an inbred background. Results We ascertained ENU-induced variants in four different phenotypically mutant lines. These were then used as informative markers for positional cloning of the mutated genes. We tested both whole genome (WGS) and whole exome (WES) datasets. Conclusion Both approaches were successful as a means to localize a region of homozygosity, as well as identifying mutations of candidate genes, which could be individually assessed. As expected, the WGS strategy was more reliable, since many more ENU-induced variants were ascertained. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2173-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jabier Gallego-Llamas
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA. .,Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Ave., Seattle, WA, 98101, USA.
| | - Andrew E Timms
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Ave., Seattle, WA, 98101, USA.
| | - Krista A Geister
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Ave., Seattle, WA, 98101, USA.
| | - Anna Lindsay
- Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Ave., Seattle, WA, 98101, USA.
| | - David R Beier
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA. .,Center for Developmental Biology and Regenerative Medicine, Seattle Children's Research Institute, 1900 Ninth Ave., Seattle, WA, 98101, USA.
| |
Collapse
|
48
|
Baes M, Van Veldhoven PP. Hepatic dysfunction in peroxisomal disorders. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:956-70. [PMID: 26453805 DOI: 10.1016/j.bbamcr.2015.09.035] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/25/2015] [Accepted: 09/28/2015] [Indexed: 12/18/2022]
Abstract
The peroxisomal compartment in hepatocytes hosts several essential metabolic conversions. These are defective in peroxisomal disorders that are either caused by failure to import the enzymes in the organelle or by mutations in the enzymes or in transporters needed to transfer the substrates across the peroxisomal membrane. Hepatic pathology is one of the cardinal features in disorders of peroxisome biogenesis and peroxisomal β-oxidation although it only rarely determines the clinical fate. In mouse models of these diseases liver pathologies also occur, although these are not always concordant with the human phenotype which might be due to differences in diet, expression of enzymes and backup mechanisms. Besides the morphological changes, we overview the impact of peroxisome malfunction on other cellular compartments including mitochondria and the ER. We further focus on the metabolic pathways that are affected such as bile acid formation, and dicarboxylic acid and branched chain fatty acid degradation. It appears that the association between deregulated metabolites and pathological events remains unclear.
Collapse
Affiliation(s)
- Myriam Baes
- Laboratory for Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, B-3000 Leuven, Belgium.
| | - Paul P Van Veldhoven
- Laboratory for Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, KU Leuven, B-3000 Leuven, Belgium.
| |
Collapse
|
49
|
Wang J, Li L, Zhang Z, Qiu H, Li D, Fang Y, Jiang H, Chai RY, Mao X, Wang Y, Sun G. One of Three Pex11 Family Members Is Required for Peroxisomal Proliferation and Full Virulence of the Rice Blast Fungus Magnaporthe oryzae. PLoS One 2015. [PMID: 26218097 PMCID: PMC4517885 DOI: 10.1371/journal.pone.0134249] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Peroxisomes play important roles in metabolisms of eukaryotes and infection of plant fungal pathogens. These organelles proliferate by de novo formation or division in response to environmental stimulation. Although the assembly of peroxisomes was documented in fungal pathogens, their division and its relationship to pathogenicity remain obscure. In present work, we analyzed the roles of three Pex11 family members in peroxisomal division and pathogenicity of the rice blast fungus Magnaporthe oryzae. Deletion of MoPEX11A led to fewer but enlarged peroxisomes, and impaired the separation of Woronin bodies from peroxisomes, while deletion of MoPEX11B or MoPEX11C put no evident impacts to peroxisomal profiles. MoPEX11A mutant exhibited typical peroxisome related defects, delayed conidial germination and appressoria formation, and decreased appressorial turgor and host penetration. As a result, the virulence of MoPEX11A mutant was greatly reduced. Deletion of MoPEX11B and MoPEX11C did not alter the virulence of the fungus. Further, double or triple deletions of the three genes were unable to enhance the virulence decrease in MoPEX11A mutant. Our data indicated that MoPEX11A is the main factor modulating peroxisomal division and is required for full virulence of the fungus.
Collapse
Affiliation(s)
- Jiaoyu Wang
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Plant Protection Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Ling Li
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Plant Protection Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
- School of Agricultural and Food Sciences, Zhejiang Agriculture and Forest University, Hangzhou, China
| | - Zhen Zhang
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Plant Protection Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Haiping Qiu
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Plant Protection Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Dongmei Li
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Plant Protection Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Yuan Fang
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Plant Protection Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
- College of Chemistry and Life Sciences, Zhejiang Normal University, Jinhua, China
| | - Hua Jiang
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Plant Protection Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Rong Yao Chai
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Plant Protection Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Xueqin Mao
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Plant Protection Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Yanli Wang
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Plant Protection Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Guochang Sun
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Plant Protection Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
- * E-mail:
| |
Collapse
|
50
|
Schrader M, Costello J, Godinho LF, Islinger M. Peroxisome-mitochondria interplay and disease. J Inherit Metab Dis 2015; 38:681-702. [PMID: 25687155 DOI: 10.1007/s10545-015-9819-7] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 01/21/2015] [Accepted: 01/26/2015] [Indexed: 12/16/2022]
Abstract
Peroxisomes and mitochondria are ubiquitous, highly dynamic organelles with an oxidative type of metabolism in eukaryotic cells. Over the years, substantial evidence has been provided that peroxisomes and mitochondria exhibit a close functional interplay which impacts on human health and development. The so-called "peroxisome-mitochondria connection" includes metabolic cooperation in the degradation of fatty acids, a redox-sensitive relationship, an overlap in key components of the membrane fission machineries and cooperation in anti-viral signalling and defence. Furthermore, combined peroxisome-mitochondria disorders with defects in organelle division have been revealed. In this review, we present the latest progress in the emerging field of peroxisomal and mitochondrial interplay in mammals with a particular emphasis on cooperative fatty acid β-oxidation, redox interplay, organelle dynamics, cooperation in anti-viral signalling and the resulting implications for disease.
Collapse
Affiliation(s)
- Michael Schrader
- College of Life and Environmental Sciences, Biosciences, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, EX4 4QD, UK,
| | | | | | | |
Collapse
|