1
|
Luetzen MA, Chakraborty R, Moreno-Ramos OA, Echeverri-Peña OY, Satta Y, Montaño AM. Purifying selection of the lysosomal enzymes arylsulfatase A and beta-galactocerebrosidase and their evolutionary impact on myelin integrity. J Lipid Res 2025; 66:100769. [PMID: 40054667 PMCID: PMC12008523 DOI: 10.1016/j.jlr.2025.100769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/02/2025] [Accepted: 03/03/2025] [Indexed: 04/10/2025] Open
Abstract
The myelin is responsible for providing stability to the axons of the nerve cells, but above all, to improve transmission speed of the nerve impulse in vertebrates. Over 70% of the myelin sheath is composed of lipids and the remaining portion by approximately 2,000 proteins. The myelin sheath has been constantly evolving, and it is known that unusually high concentrations of galactosylceramide (GalCer) and its sulfated form play a major role in the biophysical properties of the myelin. To gain insights of the evolutionary role of GalCer, we have studied two lysosomal enzymes involved in GalCer degradation, arylsulfatase A (ARSA) and galactocerebrosidase (GALC). Deficiency of ARSA or GALC causes demyelinating disorders. We conducted phylogenetic analyses of 105 ARSA and 110 GALC orthologs representing more than 600 million years ago of evolution. We examined i) low values of the ratio of nonsynonymous to synonymous nucleotide-substitution rates (dN/dS) indicating purifying selection and ii) negative selection of amino acids located in the active site preventing pathogenic mutations. Gene structure analyses showed evidence of rearrangement with gain and loss of exons while there were conserved regions mainly located around the active site. We also found a limited number of sites under positive selection pressure that do not cause alterations to the overall protein structure. Our results indicate that ARSA and GALC have been highly conserved during the evolutionary process to maintain the metabolism of GalCer, which is essential for the integrity of the white matter in vertebrate species.
Collapse
Affiliation(s)
- Matthew A Luetzen
- Department of Biochemistry and Molecular Biology, School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Richik Chakraborty
- School of Medicine, Saint Louis University, St. Louis, MO, USA; Clinical Trials Office, Georgetown Lombardi Comprehensive Cancer Center, Washington D.C., USA
| | - Oscar Andrés Moreno-Ramos
- Department of Pediatrics, School of Medicine, Saint Louis University, St Louis, MO, USA; Facultad de Ciencias, Departamento de Ciencias Biológicas, Centro de Investigaciones Genéticas en Enfermedades Humanas (CIGEN), Universidad de los Andes, Bogotá, Colombia
| | | | - Yoko Satta
- Department of Evolutionary Studies of Biosystems, SOKENDAI (The Graduate University for Advanced Studies), Hayama, Kanagawa, Japan
| | - Adriana M Montaño
- Department of Biochemistry and Molecular Biology, School of Medicine, Saint Louis University, St. Louis, MO, USA; Department of Pediatrics, School of Medicine, Saint Louis University, St Louis, MO, USA.
| |
Collapse
|
2
|
Renz-Polster H, Tremblay ME, Bienzle D, Fischer JE. The Pathobiology of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: The Case for Neuroglial Failure. Front Cell Neurosci 2022; 16:888232. [PMID: 35614970 PMCID: PMC9124899 DOI: 10.3389/fncel.2022.888232] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/13/2022] [Indexed: 12/20/2022] Open
Abstract
Although myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) has a specific and distinctive profile of clinical features, the disease remains an enigma because causal explanation of the pathobiological matrix is lacking. Several potential disease mechanisms have been identified, including immune abnormalities, inflammatory activation, mitochondrial alterations, endothelial and muscular disturbances, cardiovascular anomalies, and dysfunction of the peripheral and central nervous systems. Yet, it remains unclear whether and how these pathways may be related and orchestrated. Here we explore the hypothesis that a common denominator of the pathobiological processes in ME/CFS may be central nervous system dysfunction due to impaired or pathologically reactive neuroglia (astrocytes, microglia and oligodendrocytes). We will test this hypothesis by reviewing, in reference to the current literature, the two most salient and widely accepted features of ME/CFS, and by investigating how these might be linked to dysfunctional neuroglia. From this review we conclude that the multifaceted pathobiology of ME/CFS may be attributable in a unifying manner to neuroglial dysfunction. Because the two key features - post exertional malaise and decreased cerebral blood flow - are also recognized in a subset of patients with post-acute sequelae COVID, we suggest that our findings may also be pertinent to this entity.
Collapse
Affiliation(s)
- Herbert Renz-Polster
- Division of General Medicine, Center for Preventive Medicine and Digital Health Baden-Württemberg (CPD-BW), University Medicine Mannheim, Heidelberg University, Mannheim, Germany
| | - Marie-Eve Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec, Université Laval, Quebec, QC, Canada
- Département de Médecine Moléculaire, Université Laval, Quebec, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Center for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Dorothee Bienzle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Joachim E. Fischer
- Division of General Medicine, Center for Preventive Medicine and Digital Health Baden-Württemberg (CPD-BW), University Medicine Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
3
|
Montoro R, Heine VM, Kemp S, Engelen M. Evolution of adrenoleukodystrophy model systems. J Inherit Metab Dis 2021; 44:544-553. [PMID: 33373044 PMCID: PMC8248356 DOI: 10.1002/jimd.12357] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 01/09/2023]
Abstract
X-linked adrenoleukodystrophy (ALD) is a neurometabolic disorder affecting the adrenal glands, testes, spinal cord and brain. The disease is caused by mutations in the ABCD1 gene resulting in a defect in peroxisomal degradation of very long-chain fatty acids and their accumulation in plasma and tissues. Males with ALD have a near 100% life-time risk to develop myelopathy. The life-time prevalence to develop progressive cerebral white matter lesions (known as cerebral ALD) is about 60%. Adrenal insufficiency occurs in about 80% of male patients. In adulthood, 80% of women with ALD also develop myelopathy, but adrenal insufficiency or cerebral ALD are very rare. The complex clinical presentation and the absence of a genotype-phenotype correlation are complicating our understanding of the disease. In an attempt to understand the pathophysiology of ALD various model systems have been developed. While these model systems share the basic genetics and biochemistry of ALD they fail to fully recapitulate the complex neurodegenerative etiology of ALD. Each model system recapitulates certain aspects of the disorder. This exposes the complexity of ALD and therefore the challenge to create a comprehensive model system to fully understand ALD. In this review, we provide an overview of the different ALD modeling strategies from single-celled to multicellular organisms and from in vitro to in vivo approaches, and introduce how emerging iPSC-derived technologies could improve the understanding of this highly complex disorder.
Collapse
Affiliation(s)
- Roberto Montoro
- Department of Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Leukodystrophy Center, Amsterdam NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Vivi M. Heine
- Department of Child and Youth Psychiatry, Amsterdam UMC, Amsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdamThe Netherlands
- Department of Complex Trait Genetics, Centre for Neurogenomics and Cognitive Research, Amsterdam NeuroscienceVrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Stephan Kemp
- Department of Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Leukodystrophy Center, Amsterdam NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Amsterdam UMC, Amsterdam Gastroenterology & MetabolismUniversity of AmsterdamAmsterdamThe Netherlands
| | - Marc Engelen
- Department of Pediatric Neurology, Emma Children's Hospital, Amsterdam UMC, Amsterdam Leukodystrophy Center, Amsterdam NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
4
|
Murru E, Carta G, Manca C, Sogos V, Pistis M, Melis M, Banni S. Conjugated Linoleic Acid and Brain Metabolism: A Possible Anti-Neuroinflammatory Role Mediated by PPARα Activation. Front Pharmacol 2021; 11:587140. [PMID: 33505308 PMCID: PMC7832089 DOI: 10.3389/fphar.2020.587140] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/17/2020] [Indexed: 12/24/2022] Open
Abstract
Fatty acids play a crucial role in the brain as specific receptor ligands and as precursors of bioactive metabolites. Conjugated linoleic acid (CLA), a group of positional and geometric isomers of linoleic acid (LA, 18:2 n-6) present in meat and dairy products of ruminants and synthesized endogenously in non-ruminants and humans, has been shown to possess different nutritional properties associated with health benefits. Its ability to bind to peroxisome proliferator-activated receptor (PPAR) α, a nuclear receptor key regulator of fatty acid metabolism and inflammatory responses, partly mediates these beneficial effects. CLA is incorporated and metabolized into brain tissue where induces the biosynthesis of endogenous PPARα ligands palmitoylethanolamide (PEA) and oleoylethanolamide (OEA), likely through a positive feedback mechanism where PPARα activation sustains its own cellular effects through ligand biosynthesis. In addition to PPARα, PEA and OEA may as well bind to other receptors such as TRPV1, further extending CLA own anti-neuroinflammatory actions. Future studies are needed to investigate whether dietary CLA may exert anti-inflammatory activity, particularly in the setting of neurodegenerative diseases and neuropsychiatric disorders with a neuroinflammatory basis.
Collapse
Affiliation(s)
- Elisabetta Murru
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Gianfranca Carta
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Claudia Manca
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Valeria Sogos
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Marco Pistis
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy.,Neuroscience Institute, National Research Council of Italy (CNR), Cagliari, Italy
| | - Miriam Melis
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| | - Sebastiano Banni
- Department of Biomedical Sciences, University of Cagliari, Monserrato, Italy
| |
Collapse
|
5
|
Uzor NE, Scheihing DM, Kim GS, Moruno-Manchon JF, Zhu L, Reynolds CR, Stephenson JM, Holmes A, McCullough LD, Tsvetkov AS. Aging lowers PEX5 levels in cortical neurons in male and female mouse brains. Mol Cell Neurosci 2020; 107:103536. [PMID: 32777345 PMCID: PMC7484460 DOI: 10.1016/j.mcn.2020.103536] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/14/2020] [Accepted: 08/03/2020] [Indexed: 01/10/2023] Open
Abstract
Peroxisomes exist in nearly every cell, oxidizing fats, synthesizing lipids and maintaining redox balance. As the brain ages, multiple pathways are negatively affected, but it is currently unknown if peroxisomal proteins are affected by aging in the brain. While recent studies have investigated a PEX5 homolog in aging C. elegans models and found that it is reduced in aging, it is unclear if PEX5, a mammalian peroxisomal protein that plays a role in peroxisomal homeostasis and degradation, is affected in the aging brain. To answer this question, we first determined the amount of PEX5, in brain homogenates from young (3 months) and aged (26 through 32+ months of age) wild-type mice of both sexes. PEX5 protein was decreased in aged male brains, but this reduction was not significant in female brains. RNAScope and real-time qPCR analyses showed that Pex5 mRNA was also reduced in aged male brain cortices, but not in females. Immunohistochemistry assays of cortical neurons in young and aged male brains showed that the amount of neuronal PEX5 was reduced in aged male brains. Cortical neurons in aged female mice also had reduced PEX5 levels in comparison to younger female mice. In conclusion, total PEX5 levels and Pex5 gene expression both decrease with age in male brains, and neuronal PEX5 levels lower in an age-dependent manner in the cortices of animals of both sexes.
Collapse
Affiliation(s)
- Ndidi-Ese Uzor
- University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA; Department of Neurology, the University of Texas McGovern Medical School at Houston, Houston 77030, TX, USA
| | - Diego Morales Scheihing
- Department of Neurology, the University of Texas McGovern Medical School at Houston, Houston 77030, TX, USA
| | - Gab Seok Kim
- Department of Neurology, the University of Texas McGovern Medical School at Houston, Houston 77030, TX, USA
| | - Jose Felix Moruno-Manchon
- Department of Neurology, the University of Texas McGovern Medical School at Houston, Houston 77030, TX, USA
| | - Liang Zhu
- Biostatistics and Epidemiology Research Design, University of Texas Health Science Center at Houston, Houston 77030, TX, USA
| | - Caroline R Reynolds
- Department of Neurology, the University of Texas McGovern Medical School at Houston, Houston 77030, TX, USA
| | - Jessica M Stephenson
- Department of Neurology, the University of Texas McGovern Medical School at Houston, Houston 77030, TX, USA
| | - Aleah Holmes
- Department of Neurology, the University of Texas McGovern Medical School at Houston, Houston 77030, TX, USA
| | - Louise D McCullough
- University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA; Department of Neurology, the University of Texas McGovern Medical School at Houston, Houston 77030, TX, USA; UTHealth Consortium on Aging, the University of Texas McGovern Medical School, Houston 77030, TX, USA
| | - Andrey S Tsvetkov
- University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX 77030, USA; Department of Neurology, the University of Texas McGovern Medical School at Houston, Houston 77030, TX, USA; UTHealth Consortium on Aging, the University of Texas McGovern Medical School, Houston 77030, TX, USA.
| |
Collapse
|
6
|
Piao L, Dorotea D, Jiang S, Koh EH, Oh GT, Ha H. Impaired Peroxisomal Fitness in Obese Mice, a Vicious Cycle Exacerbating Adipocyte Dysfunction via Oxidative Stress. Antioxid Redox Signal 2019; 31:1339-1351. [PMID: 31530170 PMCID: PMC6859694 DOI: 10.1089/ars.2018.7614] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Aims: Peroxisome is a critical organelle for fatty acid oxidation (FAO) and metabolism of reactive oxygen species (ROS). Increased oxidative stress in adipose tissue contributes to the development of insulin resistance and metabolic syndrome in obesity. This study aimed to investigate the role of peroxisomal fitness in maintaining adipocyte function, which has been under-rated in the obesity research area. Results: Reduced peroxisomal gene expressions in white adipose tissue (WAT) of obese mice suggested a close correlation between peroxisomes and obesity. Peroxisomal biogenesis factor 5 siRNA increased cellular ROS and inflammatory mediators in 3T3-L1 adipocytes. On the contrary, hydrogen peroxide or tumor necrosis factor-α treatment significantly decreased biogenesis- and function-related peroxisomal proteins, suggesting a positive feedback loop of ROS/inflammation and peroxisomal dysfunction. Correspondingly, catalase (a major peroxisomal antioxidant)-knockout mice fed with high-fat diet (HFD) exhibited suppressed peroxisomal proteins along with increased oxidative stress and accelerated obesity. In response to fenofibrate (a peroxisomal proliferator) treatment, WAT of HFD-fed wild-type mice showed not only increases in peroxisomal biogenesis and FAO but also attenuated features of adipocyte dysfunction and obesity. However, these results were not observed in peroxisome proliferator-activated receptor-alpha null obese mice. Innovation: Impaired peroxisomal fitness enhanced oxidative stress and inflammation in adipocytes, which exacerbates obesity. Conclusion: Adipose tissue peroxisomal homeostasis plays an important role in attenuating the features of obesity, and it can be a potential therapeutic target of obesity.
Collapse
Affiliation(s)
- Lingjuan Piao
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea.,Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Debra Dorotea
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Songling Jiang
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Eun Hee Koh
- Asan Institute for Life Sciences, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Internal Medicine, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Goo Taeg Oh
- Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Hunjoo Ha
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
7
|
Nury T, Sghaier R, Zarrouk A, Ménétrier F, Uzun T, Leoni V, Caccia C, Meddeb W, Namsi A, Sassi K, Mihoubi W, Riedinger JM, Cherkaoui-Malki M, Moreau T, Vejux A, Lizard G. Induction of peroxisomal changes in oligodendrocytes treated with 7-ketocholesterol: Attenuation by α-tocopherol. Biochimie 2018; 153:181-202. [PMID: 30031877 DOI: 10.1016/j.biochi.2018.07.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 07/17/2018] [Indexed: 02/08/2023]
Abstract
The involvement of organelles in cell death is well established especially for endoplasmic reticulum, lysosomes and mitochondria. However, the role of the peroxisome is not well known, though peroxisomal dysfunction favors a rupture of redox equilibrium. To study the role of peroxisomes in cell death, 158 N murine oligodendrocytes were treated with 7-ketocholesterol (7 KC: 25-50 μM, 24 h). The highest concentration is known to induce oxiapoptophagy (OXIdative stress + APOPTOsis + autoPHAGY), whereas the lowest concentration does not induce cell death. In those conditions (with 7 KC: 50 μM) morphological, topographical and functional peroxisome alterations associated with modifications of the cytoplasmic distribution of mitochondria, with mitochondrial dysfunction (loss of transmembrane mitochondrial potential, decreased level of cardiolipins) and oxidative stress were observed: presence of peroxisomes with abnormal sizes and shapes similar to those observed in Zellweger fibroblasts, lower cellular level of ABCD3, used as a marker of peroxisomal mass, measured by flow cytometry, lower mRNA and protein levels (measured by RT-qPCR and western blotting) of ABCD1 and ABCD3 (two ATP-dependent peroxisomal transporters), and of ACOX1 and MFP2 enzymes, and lower mRNA level of DHAPAT, involved in peroxisomal β-oxidation and plasmalogen synthesis, respectively, and increased levels of very long chain fatty acids (VLCFA: C24:0, C24:1, C26:0 and C26:1, quantified by gas chromatography coupled with mass spectrometry) metabolized by peroxisomal β-oxidation. In the presence of 7 KC (25 μM), slight mitochondrial dysfunction and oxidative stress were found, and no induction of apoptosis was detected; however, modifications of the cytoplasmic distribution of mitochondria and clusters of mitochondria were detected. The peroxisomal alterations observed with 7 KC (25 μM) were similar to those with 7 KC (50 μM). In addition, data obtained by transmission electron microcopy and immunofluorescence microscopy by dual staining with antibodies raised against p62, involved in autophagy, and ABCD3, support that 7 KC (25-50 μM) induces pexophagy. 7 KC (25-50 μM)-induced side effects were attenuated by α-tocopherol but not by α-tocotrienol, whereas the anti-oxidant properties of these molecules determined with the FRAP assay were in the same range. These data provide evidences that 7 KC, at concentrations inducing or not cell death, triggers morphological, topographical and functional peroxisomal alterations associated with minor or major mitochondrial changes.
Collapse
Affiliation(s)
- Thomas Nury
- Univ. Bourgogne Franche-Comté, Lab. Bio-PeroxIL, 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' (EA7270) / Inserm, Dijon, France
| | - Randa Sghaier
- Univ. Bourgogne Franche-Comté, Lab. Bio-PeroxIL, 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' (EA7270) / Inserm, Dijon, France; Univ. Monastir, Lab. Biotechnology, Monastir, Tunisia
| | - Amira Zarrouk
- Univ. Monastir, Lab-NAFS 'Nutrition - Functional Food & Vascular Diseases' LR12-ES-05, Monastir, Tunisia; Faculty of Medicine, Sousse, Tunisia
| | | | - Tugba Uzun
- Univ. Bourgogne Franche-Comté, Lab. Bio-PeroxIL, 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' (EA7270) / Inserm, Dijon, France
| | - Valerio Leoni
- Lab. Clinical Chemistry, Hospital of Varese, ASST-Settelaghi, Varese, Italy
| | - Claudio Caccia
- Unit of Medical Genetics and Neurogenetics, IRCCS Carlo Besta, Milano, Italy
| | - Wiem Meddeb
- Univ. Carthage, LMMA, IPEST, Tunis, and Fac. of Science of Bizerte, Bizerte, Tunisia
| | - Amira Namsi
- Univ. Bourgogne Franche-Comté, Lab. Bio-PeroxIL, 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' (EA7270) / Inserm, Dijon, France; Univ. Tunis El Manar, Lab. Neurophysiologie Fonctionnelle et Pathologie-UR11ES/09, Tunis, Tunisia
| | - Khouloud Sassi
- Univ. Bourgogne Franche-Comté, Lab. Bio-PeroxIL, 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' (EA7270) / Inserm, Dijon, France; Univ. Tunis El Manar, Fac. of Medicine, Lab of Onco-Hematology, Tunis, Tunisia
| | - Wafa Mihoubi
- Centre de Biotechnologie de Sfax, Lab. Biotechnologie Moléculaire des Eucaryotes, Sfax, Tunisia
| | - Jean-Marc Riedinger
- Centre de Lutte Contre le Cancer GF Leclerc, Laboratoire de Biologie Médicale, Dijon, France
| | - Mustapha Cherkaoui-Malki
- Univ. Bourgogne Franche-Comté, Lab. Bio-PeroxIL, 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' (EA7270) / Inserm, Dijon, France
| | - Thibault Moreau
- Univ. Bourgogne Franche-Comté, Lab. Bio-PeroxIL, 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' (EA7270) / Inserm, Dijon, France; Dept. of Neurology, Univ. Hospital of Dijon, France
| | - Anne Vejux
- Univ. Bourgogne Franche-Comté, Lab. Bio-PeroxIL, 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' (EA7270) / Inserm, Dijon, France
| | - Gérard Lizard
- Univ. Bourgogne Franche-Comté, Lab. Bio-PeroxIL, 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' (EA7270) / Inserm, Dijon, France.
| |
Collapse
|
8
|
Cai M, Sun X, Wang W, Lian Z, Wu P, Han S, Chen H, Zhang P. Disruption of peroxisome function leads to metabolic stress, mTOR inhibition, and lethality in liver cancer cells. Cancer Lett 2018; 421:82-93. [PMID: 29458144 DOI: 10.1016/j.canlet.2018.02.021] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/07/2018] [Accepted: 02/12/2018] [Indexed: 12/30/2022]
Abstract
Peroxisome houses a large number of enzymes involved in lipid and phytochemical oxidation as well as synthesis of bile acid and other specialized lipids. Peroxisome resident enzymes are imported into the organelle via a conserved cargo transport system composed of many peroxins, protein factors essential for the biogenesis of peroxisome. Among the peroxins, PEX5 plays a transporter role, and PEX2, 10, and 12 are thought to form a complex that functions as an E3 ubiquitin ligase to help recycle PEX5 in an ubiquitin modification-dependent process. Previous studies have demonstrated the importance of peroxins in postnatal development especially the development of nerve systems. These studies also show that peroxins or the function of peroxisomes is dispensable for cellular viability. In contrast, however, we report here that PEX2 and other peroxins are essential for the viability of liver cancer cells, probably through altering metabolism and signaling pathways. Our results suggest that peroxins may be potential targets of therapeutics against liver cancer.
Collapse
Affiliation(s)
- Mengjiao Cai
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University Medical College, Xi'an 710061, China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China; Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xiao Sun
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University Medical College, Xi'an 710061, China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Wenchao Wang
- Shanghai ProfLeader Biotech Co, Shanghai 200231, China
| | - Zhusheng Lian
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University Medical College, Xi'an 710061, China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Ping Wu
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China
| | - Suxia Han
- Department of Oncology, The First Affiliated Hospital, Xi'an Jiaotong University Medical College, Xi'an 710061, China.
| | - Huan Chen
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China.
| | - Pumin Zhang
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Proteome Research Center, Beijing Institute of Lifeomics, Beijing 102206, China; Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
9
|
Lin JP, Mironova YA, Shrager P, Giger RJ. LRP1 regulates peroxisome biogenesis and cholesterol homeostasis in oligodendrocytes and is required for proper CNS myelin development and repair. eLife 2017; 6:30498. [PMID: 29251594 PMCID: PMC5752207 DOI: 10.7554/elife.30498] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 12/15/2017] [Indexed: 01/01/2023] Open
Abstract
Low-density lipoprotein receptor-related protein-1 (LRP1) is a large endocytic and signaling molecule broadly expressed by neurons and glia. In adult mice, global inducible (Lrp1flox/flox;CAG-CreER) or oligodendrocyte (OL)-lineage specific ablation (Lrp1flox/flox;Pdgfra-CreER) of Lrp1 attenuates repair of damaged white matter. In oligodendrocyte progenitor cells (OPCs), Lrp1 is required for cholesterol homeostasis and differentiation into mature OLs. Lrp1-deficient OPC/OLs show a strong increase in the sterol-regulatory element-binding protein-2 yet are unable to maintain normal cholesterol levels, suggesting more global metabolic deficits. Mechanistic studies revealed a decrease in peroxisomal biogenesis factor-2 and fewer peroxisomes in OL processes. Treatment of Lrp1−/− OPCs with cholesterol or activation of peroxisome proliferator-activated receptor-γ with pioglitazone alone is not sufficient to promote differentiation; however, when combined, cholesterol and pioglitazone enhance OPC differentiation into mature OLs. Collectively, our studies reveal a novel role for Lrp1 in peroxisome biogenesis, lipid homeostasis, and OPC differentiation during white matter development and repair.
Collapse
Affiliation(s)
- Jing-Ping Lin
- Department of Cell and Developmental Biology, University of Michigan School of Medicine, Ann Arbor, MI, United States
| | - Yevgeniya A Mironova
- Cellular and Molecular Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Peter Shrager
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, United States
| | - Roman J Giger
- Department of Cell and Developmental Biology, University of Michigan School of Medicine, Ann Arbor, MI, United States.,Cellular and Molecular Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States.,Interdepartmental Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
10
|
Moruno-Manchon JF, Uzor NE, Kesler SR, Wefel JS, Townley DM, Nagaraja AS, Pradeep S, Mangala LS, Sood AK, Tsvetkov AS. Peroxisomes contribute to oxidative stress in neurons during doxorubicin-based chemotherapy. Mol Cell Neurosci 2017; 86:65-71. [PMID: 29180229 DOI: 10.1016/j.mcn.2017.11.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 11/21/2017] [Accepted: 11/23/2017] [Indexed: 12/14/2022] Open
Abstract
Doxorubicin, a commonly used anti-neoplastic agent, causes severe neurotoxicity. Doxorubicin promotes thinning of the brain cortex and accelerates brain aging, leading to cognitive impairment. Oxidative stress induced by doxorubicin contributes to cellular damage. In addition to mitochondria, peroxisomes also generate reactive oxygen species (ROS) and promote cell senescence. Here, we investigated if doxorubicin affects peroxisomal homeostasis in neurons. We demonstrate that the number of peroxisomes is increased in doxorubicin-treated neurons and in the brains of mice which underwent doxorubicin-based chemotherapy. Pexophagy, the specific autophagy of peroxisomes, is downregulated in neurons, and peroxisomes produce more ROS. 2-hydroxypropyl-β-cyclodextrin (HPβCD), an activator of the transcription factor TFEB, which regulates expression of genes involved in autophagy and lysosome function, mitigates damage of pexophagy and decreases ROS production induced by doxorubicin. We conclude that peroxisome-associated oxidative stress induced by doxorubicin may contribute to neurotoxicity, cognitive dysfunction, and accelerated brain aging in cancer patients and survivors. Peroxisomes might be a valuable new target for mitigating neuronal damage caused by chemotherapy drugs and for slowing down brain aging in general.
Collapse
Affiliation(s)
- Jose F Moruno-Manchon
- Department of Neurobiology and Anatomy, The University of Texas, Houston Medical School, Houston, TX, United States
| | - Ndidi-Ese Uzor
- Department of Neurobiology and Anatomy, The University of Texas, Houston Medical School, Houston, TX, United States; The University of Texas Graduate School of Biomedical Sciences, Houston, TX, United States
| | - Shelli R Kesler
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jeffrey S Wefel
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Debra M Townley
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States
| | - Archana Sidalaghatta Nagaraja
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sunila Pradeep
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lingegowda S Mangala
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, United States; Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Andrey S Tsvetkov
- Department of Neurobiology and Anatomy, The University of Texas, Houston Medical School, Houston, TX, United States; The University of Texas Graduate School of Biomedical Sciences, Houston, TX, United States.
| |
Collapse
|
11
|
Geric I, Tyurina YY, Krysko O, Krysko DV, De Schryver E, Kagan VE, Van Veldhoven PP, Baes M, Verheijden S. Lipid homeostasis and inflammatory activation are disturbed in classically activated macrophages with peroxisomal β-oxidation deficiency. Immunology 2017; 153:342-356. [PMID: 28940384 DOI: 10.1111/imm.12844] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 09/13/2017] [Accepted: 09/17/2017] [Indexed: 01/07/2023] Open
Abstract
Macrophage activation is characterized by pronounced metabolic adaptation. Classically activated macrophages show decreased rates of mitochondrial fatty acid oxidation and oxidative phosphorylation and acquire a glycolytic state together with their pro-inflammatory phenotype. In contrast, alternatively activated macrophages require oxidative phosphorylation and mitochondrial fatty acid oxidation for their anti-inflammatory function. Although it is evident that mitochondrial metabolism is regulated during macrophage polarization and essential for macrophage function, little is known on the regulation and role of peroxisomal β-oxidation during macrophage activation. In this study, we show that peroxisomal β-oxidation is strongly decreased in classically activated bone-marrow-derived macrophages (BMDM) and mildly induced in alternatively activated BMDM. To examine the role of peroxisomal β-oxidation in macrophages, we used Mfp2-/- BMDM lacking the key enzyme of this pathway. Impairment of peroxisomal β-oxidation in Mfp2-/- BMDM did not cause lipid accumulation but rather an altered distribution of lipid species with very-long-chain fatty acids accumulating in the triglyceride and phospholipid fraction. These lipid alterations in Mfp2-/- macrophages led to decreased inflammatory activation of Mfp2-/- BMDM and peritoneal macrophages evidenced by impaired production of several inflammatory cytokines and chemokines, but did not affect anti-inflammatory polarization. The disturbed inflammatory responses of Mfp2-/- macrophages did not affect immune cell infiltration, as mice with selective elimination of MFP2 from myeloid cells showed normal monocyte and neutrophil influx upon challenge with zymosan. Together, these data demonstrate that peroxisomal β-oxidation is involved in fine-tuning the phenotype of macrophages, probably by influencing the dynamic lipid profile during macrophage polarization.
Collapse
Affiliation(s)
- Ivana Geric
- Department of Pharmaceutical and Pharmacological Sciences, Cell Metabolism, KU Leuven - University of Leuven, Leuven, Belgium
| | - Yulia Y Tyurina
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Olga Krysko
- Department of Oto-Rhino-Laryngology, The Upper Airway Research Laboratory, Hospital, Ghent University Ghent, Ghent, Belgium
| | - Dmitri V Krysko
- Molecular Signalling and Cell Death Unit, VIB, Centre for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Evelyn De Schryver
- Department of Cellular and Molecular Medicine, LIPIT, KU Leuven - University of Leuven, Leuven, Belgium
| | - Valerian E Kagan
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Paul P Van Veldhoven
- Department of Cellular and Molecular Medicine, LIPIT, KU Leuven - University of Leuven, Leuven, Belgium
| | - Myriam Baes
- Department of Pharmaceutical and Pharmacological Sciences, Cell Metabolism, KU Leuven - University of Leuven, Leuven, Belgium
| | - Simon Verheijden
- Department of Clinical and Experimental Medicine, Translational Research Centre for Gastrointestinal Disorders (TARGID), KU Leuven - University of Leuven, Leuven, Belgium
| |
Collapse
|
12
|
Argan Oil-Mediated Attenuation of Organelle Dysfunction, Oxidative Stress and Cell Death Induced by 7-Ketocholesterol in Murine Oligodendrocytes 158N. Int J Mol Sci 2017; 18:ijms18102220. [PMID: 29065513 PMCID: PMC5666899 DOI: 10.3390/ijms18102220] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/18/2017] [Accepted: 10/20/2017] [Indexed: 02/07/2023] Open
Abstract
Argan oil is widely used in Morocco in traditional medicine. Its ability to treat cardiovascular diseases is well-established. However, nothing is known about its effects on neurodegenerative diseases, which are often associated with increased oxidative stress leading to lipid peroxidation and the formation of 7-ketocholesterol (7KC) resulting from cholesterol auto-oxidation. As 7KC induces oxidative stress, inflammation and cell death, it is important to identify compounds able to impair its harmful effects. These compounds may be either natural or synthetic molecules or mixtures of molecules such as oils. In this context: (i) the lipid profiles of dietary argan oils from Berkane and Agadir (Morocco) in fatty acids, phytosterols, tocopherols and polyphenols were determined by different chromatographic techniques; and (ii) their anti-oxidant and cytoprotective effects in 158N murine oligodendrocytes cultured with 7KC (25-50 µM; 24 h) without and with argan oil (0.1% v/v) or α-tocopherol (400 µM, positive control) were evaluated with complementary techniques of cellular and molecular biology. Among the unsaturated fatty acids present in argan oils, oleate (C18:1 n-9) and linoleate (C18:1 n-6) were the most abundant; the highest quantities of saturated fatty acids were palmitate (C16:0) and stearate (C18:0). Several phytosterols were found, mainly schottenol and spinasterol (specific to argan oil), cycloartenol, β-amyrin and citrostadienol. α- and γ-tocopherols were also present. Tyrosol and protocatechic acid were the only polyphenols detected. Argan and extra virgin olive oils have many compounds in common, principally oleate and linoleate, and tocopherols. Kit Radicaux Libres (KRL) and ferric reducing antioxidant power (FRAP) tests showed that argan and extra virgin olive oils have anti-oxidant properties. Argan oils were able to attenuate the cytotoxic effects of 7KC on 158N cells: loss of cell adhesion, cell growth inhibition, increased plasma membrane permeability, mitochondrial, peroxisomal and lysosomal dysfunction, and the induction of oxiapoptophagy (OXIdation + APOPTOsis + autoPHAGY). Altogether, our data obtained in 158N oligodendrocytes provide evidence that argan oil is able to counteract the toxic effects of 7KC on nerve cells, thus suggesting that some of its compounds could prevent or mitigate neurodegenerative diseases to the extent that they are able to cross the blood-brain barrier.
Collapse
|
13
|
Roth AD, Núñez MT. Oligodendrocytes: Functioning in a Delicate Balance Between High Metabolic Requirements and Oxidative Damage. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 949:167-181. [PMID: 27714689 DOI: 10.1007/978-3-319-40764-7_8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The study of the metabolic interactions between myelinating glia and the axons they ensheath has blossomed into an area of research much akin to the elucidation of the role of astrocytes in tripartite synapses (Tsacopoulos and Magistretti in J Neurosci 16:877-885, 1996). Still, unlike astrocytes, rich in cytochrome-P450 and other anti-oxidative defense mechanisms (Minn et al. in Brain Res Brain Res Rev 16:65-82, 1991; Wilson in Can J Physiol Pharmacol. 75:1149-1163, 1997), oligodendrocytes can be easily damaged and are particularly sensitive to both hypoxia and oxidative stress, especially during their terminal differentiation phase and while generating myelin sheaths. In the present review, we will focus in the metabolic complexity of oligodendrocytes, particularly during the processes of differentiation and myelin deposition, and with a specific emphasis in the context of oxidative stress and the intricacies of the iron metabolism of the most iron-loaded cells of the central nervous system (CNS).
Collapse
Affiliation(s)
- Alejandro D Roth
- Department of Biology, Faculty of Science, University of Chile, Santiago, Chile.
| | - Marco T Núñez
- Department of Biology, Faculty of Science, University of Chile, Santiago, Chile
| |
Collapse
|
14
|
Beckers L, Stroobants S, Verheijden S, West B, D'Hooge R, Baes M. Specific suppression of microgliosis cannot circumvent the severe neuropathology in peroxisomal β-oxidation-deficient mice. Mol Cell Neurosci 2017; 80:123-133. [PMID: 28286294 DOI: 10.1016/j.mcn.2017.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/21/2017] [Accepted: 03/05/2017] [Indexed: 12/22/2022] Open
Abstract
An important hallmark of various neurodegenerative disorders is the proliferation and activation of microglial cells, the resident immune cells of the central nervous system (CNS). Mice that lack multifunctional protein-2 (MFP2), the key enzyme in peroxisomal β-oxidation, develop excessive microgliosis that positively correlates with behavioral deficits whereas no neuronal loss occurs. However, the precise contribution of neuroinflammation to the fatal neuropathology of MFP2 deficiency remains largely unknown. Here, we first attempted to suppress the inflammatory response by administering various anti-inflammatory drugs but they failed to reduce microgliosis. Subsequently, Mfp2-/- mice were treated with the selective colony-stimulating factor 1 receptor (CSF1R) inhibitor PLX5622 as microglial proliferation and survival is dependent on CSF1R signaling. This resulted in the elimination of >95% of microglia from control mice but only 70% of the expanded microglial population from Mfp2-/- mice. Despite microglial diminution in Mfp2-/- brain, inflammatory markers remained unaltered and residual microglia persisted in a reactive state. CSF1R inhibition did not prevent neuronal dysfunction, cognitive decline and clinical deterioration of Mfp2-/- mice. Collectively, the unaltered inflammatory profile despite suppressed microgliosis concurrent with persevering clinical decline strengthens our hypothesis that neuroinflammation importantly contributes to the Mfp2-/- phenotype.
Collapse
Affiliation(s)
- L Beckers
- KU Leuven - University of Leuven, Department of Pharmaceutical and Pharmacological Sciences, Cell Metabolism, B-3000 Leuven, Belgium
| | - S Stroobants
- KU Leuven - University of Leuven, Faculty of Psychology and Educational Sciences, Biological Psychology, B-3000 Leuven, Belgium
| | - S Verheijden
- KU Leuven - University of Leuven, Department of Pharmaceutical and Pharmacological Sciences, Cell Metabolism, B-3000 Leuven, Belgium
| | - B West
- Plexxikon Inc., Berkeley, CA 94710, USA
| | - R D'Hooge
- KU Leuven - University of Leuven, Faculty of Psychology and Educational Sciences, Biological Psychology, B-3000 Leuven, Belgium
| | - M Baes
- KU Leuven - University of Leuven, Department of Pharmaceutical and Pharmacological Sciences, Cell Metabolism, B-3000 Leuven, Belgium.
| |
Collapse
|
15
|
Protective Effects of α-Tocopherol, γ-Tocopherol and Oleic Acid, Three Compounds of Olive Oils, and No Effect of Trolox, on 7-Ketocholesterol-Induced Mitochondrial and Peroxisomal Dysfunction in Microglial BV-2 Cells. Int J Mol Sci 2016; 17:ijms17121973. [PMID: 27897980 PMCID: PMC5187773 DOI: 10.3390/ijms17121973] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 11/08/2016] [Accepted: 11/17/2016] [Indexed: 01/18/2023] Open
Abstract
Lipid peroxidation products, such as 7-ketocholesterol (7KC), may be increased in the body fluids and tissues of patients with neurodegenerative diseases and trigger microglial dysfunction involved in neurodegeneration. It is therefore important to identify synthetic and natural molecules able to impair the toxic effects of 7KC. We determined the impact of 7KC on murine microglial BV-2 cells, especially its ability to trigger mitochondrial and peroxisomal dysfunction, and evaluated the protective effects of α- and γ-tocopherol, Trolox, and oleic acid (OA). Multiple complementary chemical assays, flow cytometric and biochemical methods were used to evaluate the antioxidant and cytoprotective properties of these molecules. According to various complementary assays to estimate antioxidant activity, only α-, and γ-tocopherol, and Trolox had antioxidant properties. However, only α-tocopherol, γ-tocopherol and OA were able to impair 7KC-induced loss of mitochondrial transmembrane potential, which is associated with increased permeability to propidium iodide, an indicator of cell death. In addition, α-and γ-tocopherol, and OA were able to prevent the decrease in Abcd3 protein levels, which allows the measurement of peroxisomal mass, and in mRNA levels of Abcd1 and Abcd2, which encode for two transporters involved in peroxisomal β-oxidation. Thus, 7KC-induced side effects are associated with mitochondrial and peroxisomal dysfunction which can be inversed by natural compounds, thus supporting the hypothesis that the composition of the diet can act on the function of organelles involved in neurodegenerative diseases.
Collapse
|
16
|
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
|
17
|
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: 129] [Impact Index Per Article: 12.9] [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
|
18
|
Jodeiri Farshbaf M, Ghaedi K, Megraw TL, Curtiss J, Shirani Faradonbeh M, Vaziri P, Nasr-Esfahani MH. Does PGC1α/FNDC5/BDNF Elicit the Beneficial Effects of Exercise on Neurodegenerative Disorders? Neuromolecular Med 2015; 18:1-15. [PMID: 26611102 DOI: 10.1007/s12017-015-8370-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 08/22/2015] [Indexed: 12/20/2022]
Abstract
Neurodegenerative disorders such as Alzheimer's, Parkinson's and Huntington's diseases have high prevalence among the elderly. Many strategies have been established to alleviate the symptoms experienced by affected individuals. Recent studies have shown that exercise helps patients with neurological disorders to regain lost physical abilities. PGC1α/FNDC5/BDNF has emerged recently as a critical pathway for neuroprotection. PGC1α is a highly conserved co-activator of transcription factors that preserves and protects neurons against destruction. PGC1α regulates FNDC5 and its processed and secreted peptide Irisin, which has been proposed to play a critical role in energy expenditure and to promote neural differentiation of mouse embryonic stem cells. FNDC5 may also increase the expression of the neurotrophic factor BDNF, a neuroprotective agent, in the hippocampus. BDNF is secreted from hippocampus, amygdala, cerebral cortex and hypothalamus neurons and initiates intracellular signaling pathways through TrkB receptors. These pathways have positive feedback on CREB activities and lead to enhancement in PGC1α expression in neurons. Therefore, FNDC5 could behave as a key regulator in neuronal survival and development. This review presents recent findings on the PGC1α/FNDC5/BDNF pathway and its role in neuroprotection, and discusses the controversial promise of irisin as a mediator of the positive benefits of exercise.
Collapse
Affiliation(s)
- Mohammad Jodeiri Farshbaf
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Royan Street, Salman Street, Khorasgan, Isfahan, 8165131378, Iran.,Department of Biology, School of Sciences, University of Isfahan, Hezarjerib Street, Azadi Square, Isfahan, 8174673441, Iran.,Department of Biology, New Mexico State University, Las Cruces, NM, USA
| | - Kamran Ghaedi
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Royan Street, Salman Street, Khorasgan, Isfahan, 8165131378, Iran. .,Department of Biology, School of Sciences, University of Isfahan, Hezarjerib Street, Azadi Square, Isfahan, 8174673441, Iran.
| | - Timothy L Megraw
- Department of Biomedical Sciences, Florida State University College of Medicine, West Call Street, Tallahassee, FL, 32306-4300, USA.
| | - Jennifer Curtiss
- Department of Biology, New Mexico State University, Las Cruces, NM, USA
| | - Mahsa Shirani Faradonbeh
- Department of Biology, School of Sciences, University of Isfahan, Hezarjerib Street, Azadi Square, Isfahan, 8174673441, Iran
| | - Pooneh Vaziri
- Department of Biology, New Mexico State University, Las Cruces, NM, USA
| | - Mohammad Hossein Nasr-Esfahani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Royan Street, Salman Street, Khorasgan, Isfahan, 8165131378, Iran.
| |
Collapse
|
19
|
Wang XM, Yik WY, Zhang P, Lu W, Huang N, Kim BR, Shibata D, Zitting M, Chow RH, Moser AB, Steinberg SJ, Hacia JG. Induced pluripotent stem cell models of Zellweger spectrum disorder show impaired peroxisome assembly and cell type-specific lipid abnormalities. Stem Cell Res Ther 2015; 6:158. [PMID: 26319495 PMCID: PMC4553005 DOI: 10.1186/s13287-015-0149-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 05/26/2015] [Accepted: 08/07/2015] [Indexed: 01/08/2023] Open
Abstract
Introduction Zellweger spectrum disorder (PBD-ZSD) is a disease continuum caused by mutations in a subset of PEX genes required for normal peroxisome assembly and function. They highlight the importance of peroxisomes in the development and functions of the central nervous system, liver, and other organs. To date, the underlying bases for the cell-type specificity of disease are not fully elucidated. Methods Primary skin fibroblasts from seven PBD-ZSD patients with biallelic PEX1, PEX10, PEX12, or PEX26 mutations and three healthy donors were transduced with retroviral vectors expressing Yamanaka reprogramming factors. Candidate induced pluripotent stem cells (iPSCs) were subject to global gene expression, DNA methylation, copy number variation, genotyping, in vitro differentiation and teratoma formation assays. Confirmed iPSCs were differentiated into neural progenitor cells (NPCs), neurons, oligodendrocyte precursor cells (OPCs), and hepatocyte-like cell cultures with peroxisome assembly evaluated by microscopy. Saturated very long chain fatty acid (sVLCFA) and plasmalogen levels were determined in primary fibroblasts and their derivatives. Results iPSCs were derived from seven PBD-ZSD patient-derived fibroblasts with mild to severe peroxisome assembly defects. Although patient and control skin fibroblasts had similar gene expression profiles, genes related to mitochondrial functions and organelle cross-talk were differentially expressed among corresponding iPSCs. Mitochondrial DNA levels were consistent among patient and control fibroblasts, but varied among all iPSCs. Relative to matching controls, sVLCFA levels were elevated in patient-derived fibroblasts, reduced in patient-derived iPSCs, and not significantly different in patient-derived NPCs. All cell types derived from donors with biallelic null mutations in a PEX gene showed plasmalogen deficiencies. Reporter gene assays compatible with high content screening (HCS) indicated patient-derived OPC and hepatocyte-like cell cultures had impaired peroxisome assembly. Conclusions Normal peroxisome activity levels are not required for cellular reprogramming of skin fibroblasts. Patient iPSC gene expression profiles were consistent with hypotheses highlighting the role of altered mitochondrial activities and organelle cross-talk in PBD-ZSD pathogenesis. sVLCFA abnormalities dramatically differed among patient cell types, similar to observations made in iPSC models of X-linked adrenoleukodystrophy. We propose that iPSCs could assist investigations into the cell type-specificity of peroxisomal activities, toxicology studies, and in HCS for targeted therapies for peroxisome-related disorders. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0149-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiao-Ming Wang
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California, USA.
| | - Wing Yan Yik
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California, USA.
| | - Peilin Zhang
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California, USA.
| | - Wange Lu
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California, USA.
| | - Ning Huang
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California, USA.
| | - Bo Ram Kim
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California, USA.
| | - Darryl Shibata
- Department of Pathology, University of Southern California, Los Angeles, California, USA.
| | - Madison Zitting
- Department of Physiology and Biophysics, University of Southern California, Los Angeles, California, USA.
| | - Robert H Chow
- Department of Physiology and Biophysics, University of Southern California, Los Angeles, California, USA.
| | - Ann B Moser
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA.
| | - Steven J Steinberg
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, Maryland, USA.
| | - Joseph G Hacia
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
20
|
Walbrecq G, Wang B, Becker S, Hannotiau A, Fransen M, Knoops B. Antioxidant cytoprotection by peroxisomal peroxiredoxin-5. Free Radic Biol Med 2015; 84:215-226. [PMID: 25772011 DOI: 10.1016/j.freeradbiomed.2015.02.032] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 02/19/2015] [Accepted: 02/27/2015] [Indexed: 10/23/2022]
Abstract
Peroxiredoxin-5 (PRDX5) is a thioredoxin peroxidase that reduces hydrogen peroxide, alkyl hydroperoxides, and peroxynitrite. This enzyme is present in the cytosol, mitochondria, peroxisomes, and nucleus in human cells. Antioxidant cytoprotective functions have been previously documented for cytosolic, mitochondrial, and nuclear mammalian PRDX5. However, the exact function of PRDX5 in peroxisomes is still not clear. The aim of this work was to determine the function of peroxisomal PRDX5 in mammalian cells and, more specifically, in glial cells. To study the role of PRDX5 in peroxisomes, the endogenous expression of PRDX5 in murine oligodendrocyte 158N cells was silenced by RNA interference. In addition, human PRDX5 was also overexpressed in peroxisomes using a vector coding for human PRDX5, whose unconventional peroxisomal targeting sequence 1 (PTS1; SQL) was replaced by the prototypical PTS1 SKL. Stable 158N clones were obtained. The antioxidant cytoprotective function of peroxisomal PRDX5 against peroxisomal and mitochondrial KillerRed-mediated reactive oxygen species production as well as H2O2 was examined using MTT viability assays, roGFP2, and C11-BOBIPY probes. Altogether our results show that peroxisomal PRDX5 protects 158N oligodendrocytes against peroxisomal and mitochondrial KillerRed- and H2O2-induced oxidative stress.
Collapse
Affiliation(s)
- Geoffroy Walbrecq
- Group of Animal Molecular and Cellular Biology, Institut des Sciences de la Vie, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium
| | - Bo Wang
- Laboratory of Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Sarah Becker
- Group of Animal Molecular and Cellular Biology, Institut des Sciences de la Vie, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium
| | - Amandine Hannotiau
- Group of Animal Molecular and Cellular Biology, Institut des Sciences de la Vie, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium
| | - Marc Fransen
- Laboratory of Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Bernard Knoops
- Group of Animal Molecular and Cellular Biology, Institut des Sciences de la Vie, Université Catholique de Louvain, 1348 Louvain-la-Neuve, Belgium.
| |
Collapse
|
21
|
Young JM, Nelson JW, Cheng J, Zhang W, Mader S, Davis CM, Morrison RS, Alkayed NJ. Peroxisomal biogenesis in ischemic brain. Antioxid Redox Signal 2015; 22:109-20. [PMID: 25226217 PMCID: PMC4281844 DOI: 10.1089/ars.2014.5833] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
AIMS Peroxisomes are highly adaptable and dynamic organelles, adjusting their size, number, and enzyme composition to changing environmental and metabolic demands. We determined whether peroxisomes respond to ischemia, and whether peroxisomal biogenesis is an adaptive response to cerebral ischemia. RESULTS Focal cerebral ischemia induced peroxisomal biogenesis in peri-infarct neurons, which was associated with a corresponding increase in peroxisomal antioxidant enzyme catalase. Peroxisomal biogenesis was also observed in primary cultured cortical neurons subjected to ischemic insult induced by oxygen-glucose deprivation (OGD). A catalase inhibitor increased OGD-induced neuronal death. Moreover, preventing peroxisomal proliferation by knocking down dynamin-related protein 1 (Drp1) exacerbated neuronal death induced by OGD, whereas enhancing peroxisomal biogenesis pharmacologically using a peroxisome proliferator-activated receptor-alpha agonist protected against neuronal death induced by OGD. INNOVATION This is the first documentation of ischemia-induced peroxisomal biogenesis in mammalian brain using a combined in vivo and in vitro approach, electron microscopy, high-resolution laser-scanning confocal microscopy, and super-resolution structured illumination microscopy. CONCLUSION Our findings suggest that neurons respond to ischemic injury by increasing peroxisome biogenesis, which serves a protective function, likely mediated by enhanced antioxidant capacity of neurons.
Collapse
Affiliation(s)
- Jennifer M Young
- 1 Department of Anesthesiology & Perioperative Medicine, The Knight Cardiovascular Institute, Oregon Health & Science University , Portland, Oregon
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Peripheral nervous system defects in a mouse model for peroxisomal biogenesis disorders. Dev Biol 2014; 395:84-95. [PMID: 25176044 DOI: 10.1016/j.ydbio.2014.08.026] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 08/19/2014] [Accepted: 08/20/2014] [Indexed: 12/19/2022]
Abstract
Peroxisome biogenesis disorders (PBD) are autosomal recessive disorders in humans characterized by skeletal, eye and brain abnormalities. Despite the fact that neurological deficits, including peripheral nervous system (PNS) defects, can be observed at birth in some PBD patients including those with PEX10 mutations, the embryological basis of the PNS defects is unclear. Using a forward genetic screen, we identified a mouse model for Pex10 deficiency that exhibits neurological abnormalities during fetal development. Homozygous Pex10 mutant mouse embryos display biochemical abnormalities related to a PBD deficiency. During late embryogenesis, Pex10 homozygous mutant mice experience progressive loss of movement and at birth they become cyanotic and die shortly thereafter. Homozygous Pex10 mutant fetuses display decreased integrity of axons and synapses, over-extension of axons in the diaphragm and decreased Schwann cell numbers. Our neuropathological, molecular and electrophysiological studies provide new insights into the embryological basis of the PNS deficits in a PBD model. Our findings identify PEX10 function, and likely other PEX proteins, as an essential component of the spinal locomotor circuit.
Collapse
|
23
|
Di Cesare Mannelli L, Zanardelli M, Micheli L, Ghelardini C. PPAR- γ impairment alters peroxisome functionality in primary astrocyte cell cultures. BIOMED RESEARCH INTERNATIONAL 2014; 2014:546453. [PMID: 24729976 PMCID: PMC3960521 DOI: 10.1155/2014/546453] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 01/04/2014] [Accepted: 01/07/2014] [Indexed: 12/18/2022]
Abstract
Peroxisomes provide glial cells with protective functions against the harmful effects of H2O2 on neurons and peroxisome impairment results in nervous lesions. Agonists of the γ -subtype of the Peroxisome-Proliferator-Activated-Receptors (PPAR) have been proposed as neuroprotective agents in neurodegenerative disorders. Nevertheless, the role of PPAR- γ alterations in pathophysiological mechanisms and the relevance of peroxisome functions in the PPAR- γ effects are not yet clear. In a primary cell culture of rat astrocytes, the irreversible PPAR- γ antagonist GW9662 concentration-dependently decreased the activity of catalase, the most important antioxidant defense enzyme in peroxisomes. Catalase functionality recovered in a few days and the PPAR- γ agonist rosiglitazone promoted reversal of enzymatic damage. The reversible antagonist G3335 reduced both the activity and expression of catalase in a rosiglitazone-prevented manner. G3335 reduced also the glutathione reductase expression, indicating that enzyme involved in glutathione regeneration was compromised. Neither the PPAR- α target gene Acyl-Coenzyme-A-oxidase-1 nor the mitochondrial detoxifying enzyme NADH:ubiquinone-oxidoreductase (NDFUS3) was altered by PPAR- γ inhibition. In conclusion, PPAR- γ inhibition induced impairment of catalase in astrocytes. A general decrease of the antioxidant defenses of the cell suggests that a PPAR- γ hypofunction could participate in neurodegenerative mechanisms through peroxisomal damage. This series of experiments could be a useful model for studying compounds able to restore peroxisome functionality.
Collapse
Affiliation(s)
- Lorenzo Di Cesare Mannelli
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino-(Neurofarba)-Sezione di Farmacologia e Tossicologia, Università di Firenze, Viale Pieraccini 6, 50139 Florence, Italy
| | - Matteo Zanardelli
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino-(Neurofarba)-Sezione di Farmacologia e Tossicologia, Università di Firenze, Viale Pieraccini 6, 50139 Florence, Italy
| | - Laura Micheli
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino-(Neurofarba)-Sezione di Farmacologia e Tossicologia, Università di Firenze, Viale Pieraccini 6, 50139 Florence, Italy
| | - Carla Ghelardini
- Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino-(Neurofarba)-Sezione di Farmacologia e Tossicologia, Università di Firenze, Viale Pieraccini 6, 50139 Florence, Italy
| |
Collapse
|
24
|
Trompier D, Vejux A, Zarrouk A, Gondcaille C, Geillon F, Nury T, Savary S, Lizard G. Brain peroxisomes. Biochimie 2014; 98:102-10. [DOI: 10.1016/j.biochi.2013.09.009] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 09/12/2013] [Indexed: 02/06/2023]
|
25
|
Van Veldhoven PP, Baes M. Peroxisome deficient invertebrate and vertebrate animal models. Front Physiol 2013; 4:335. [PMID: 24319432 PMCID: PMC3837297 DOI: 10.3389/fphys.2013.00335] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 11/01/2013] [Indexed: 11/29/2022] Open
Abstract
Although peroxisomes are ubiquitous organelles in all animal species, their importance for the functioning of tissues and organs remains largely unresolved. Because peroxins are essential for the biogenesis of peroxisomes, an obvious approach to investigate their physiological role is to inactivate a Pex gene or to suppress its translation. This has been performed in mice but also in more primitive organisms including D. melanogaster, C. elegans, and D. rerio, and the major findings and abnormalities in these models will be highlighted. Although peroxisomes are generally not essential for embryonic development and organogenesis, a generalized inactivity of peroxisomes affects lifespan and posthatching/postnatal growth, proving that peroxisomal metabolism is necessary for the normal maturation of these organisms. Strikingly, despite the wide variety of model organisms, corresponding tissues are affected including the central nervous system and the testis. By inactivating peroxisomes in a cell type selective way in the brain of mice, it was also demonstrated that peroxisomes are necessary to prevent neurodegeneration. As these peroxisome deficient model organisms recapitulate pathologies of patients affected with peroxisomal diseases, their further analysis will contribute to the elucidation of still elusive pathogenic mechanisms.
Collapse
Affiliation(s)
| | - Myriam Baes
- Laboratory of Cellular Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU LeuvenLeuven, Belgium
| |
Collapse
|
26
|
Braverman NE, D'Agostino MD, MacLean GE. Peroxisome biogenesis disorders: Biological, clinical and pathophysiological perspectives. ACTA ACUST UNITED AC 2013; 17:187-96. [DOI: 10.1002/ddrr.1113] [Citation(s) in RCA: 101] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 05/17/2012] [Indexed: 01/08/2023]
|
27
|
Verheijden S, Bottelbergs A, Krysko O, Krysko DV, Beckers L, De Munter S, Van Veldhoven PP, Wyns S, Kulik W, Nave KA, Ramer MS, Carmeliet P, Kassmann CM, Baes M. Peroxisomal multifunctional protein-2 deficiency causes neuroinflammation and degeneration of Purkinje cells independent of very long chain fatty acid accumulation. Neurobiol Dis 2013; 58:258-69. [PMID: 23777740 DOI: 10.1016/j.nbd.2013.06.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 05/27/2013] [Accepted: 06/07/2013] [Indexed: 01/03/2023] Open
Abstract
Although peroxisome biogenesis and β-oxidation disorders are well known for their neurodevelopmental defects, patients with these disorders are increasingly diagnosed with neurodegenerative pathologies. In order to investigate the cellular mechanisms of neurodegeneration in these patients, we developed a mouse model lacking multifunctional protein 2 (MFP2, also called D-bifunctional protein), a central enzyme of peroxisomal β-oxidation, in all neural cells (Nestin-Mfp2(-/-)) or in oligodendrocytes (Cnp-Mfp2(-/-)) and compared these models with an already established general Mfp2 knockout. Nestin-Mfp2 but not Cnp-Mfp2 knockout mice develop motor disabilities and ataxia, similar to the general mutant. Deterioration of motor performance correlates with the demise of Purkinje cell axons in the cerebellum, which precedes loss of Purkinje cells and cerebellar atrophy. This closely mimics spinocerebellar ataxias of patients affected with mild peroxisome β-oxidation disorders. However, general knockouts have a much shorter life span than Nestin-Mfp2 knockouts which is paralleled by a disparity in activation of the innate immune system. Whereas in general mutants a strong and chronic proinflammatory reaction proceeds throughout the brain, elimination of MFP2 from neural cells results in minor neuroinflammation. Neither the extent of the inflammatory reaction nor the cerebellar degeneration could be correlated with levels of very long chain fatty acids, substrates of peroxisomal β-oxidation. In conclusion, MFP2 has multiple tasks in the adult brain, including the maintenance of Purkinje cells and the prevention of neuroinflammation but this is not mediated by its activity in oligodendrocytes nor by its role in very long chain fatty acid degradation.
Collapse
Affiliation(s)
- Simon Verheijden
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Camacho A, Huang JK, Delint-Ramirez I, Yew Tan C, Fuller M, Lelliott CJ, Vidal-Puig A, Franklin RJM. Peroxisome proliferator-activated receptor gamma-coactivator-1 alpha coordinates sphingolipid metabolism, lipid raft composition and myelin protein synthesis. Eur J Neurosci 2013; 38:2672-83. [DOI: 10.1111/ejn.12281] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 05/14/2013] [Accepted: 05/16/2013] [Indexed: 01/19/2023]
Affiliation(s)
- Alberto Camacho
- Metabolic Research Laboratories; Institute of Metabolic Science; Addenbrooke's Treatment Centre; Addenbrooke's Hospital; University of Cambridge; Cambridge; UK
| | - Jeffrey K. Huang
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine; Cambridge; UK
| | - Ilse Delint-Ramirez
- Department of Pharmacology; Faculty of Medicine; Autonomous University of Nuevo León; Monterrey; Mexico
| | - Chong Yew Tan
- Metabolic Research Laboratories; Institute of Metabolic Science; Addenbrooke's Treatment Centre; Addenbrooke's Hospital; University of Cambridge; Cambridge; UK
| | - Maria Fuller
- Department of Genetics and Molecular Pathology; SA Pathology; Adelaide; SA; Australia
| | | | - Antonio Vidal-Puig
- Metabolic Research Laboratories; Institute of Metabolic Science; Addenbrooke's Treatment Centre; Addenbrooke's Hospital; University of Cambridge; Cambridge; UK
| | - Robin J. M. Franklin
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine; Cambridge; UK
| |
Collapse
|
29
|
Morita M, Shinbo S, Asahi A, Imanaka T. Very long chain fatty acid β-oxidation in astrocytes: contribution of the ABCD1-dependent and -independent pathways. Biol Pharm Bull 2013; 35:1972-9. [PMID: 23123468 DOI: 10.1248/bpb.b12-00411] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Very long chain fatty acid (VLCFA) metabolism in astrocytes is important for the maintenance of myelin structure in central nervous system. To analyze the contribution of the ABCD1-dependent and -independent pathways to VLCFA metabolism in astrocytes, we prepared human glioblastoma U87 cells with a silencing of ABCD1 and primary astrocytes from abcd1-deficient mice, and measured fatty acid β-oxidation in the presence or absence of a potent inhibitor of carnitine palmitoyltransferase I, 2-[5-(4-chlorophenyl)pentyl]oxirane-2-carboxylate (POCA). In U87 cells, C24:0 β-oxidation was decreased to ca. 70% of the control in the presence of POCA, and the activity was further decreased to ca. 20% by the silencing of ABCD1. In mouse primary astrocytes, C24:0 β-oxidation was also decreased to ca. 70% of the control in the presence of POCA. The C24:0 β-oxidation in Abcd1-deficient primary astrocytes was ca. 60% of the wild-type cells and the activity was further decreased to ca. 25% in the presence of POCA. Compared to human skin fibroblasts, in which VLCFA β-oxidation is not significantly inhibited by POCA, approximately one-third of the overall VLCFA β-oxidation was inhibited in both types of astrocytic cells. These results suggest that VLCFA is indeed β-oxidized in ABCD1-dependent pathway, but the ABCD1-independent peroxisomal and mitochondrial β-oxidation pathways significantly contribute to VLCFA β-oxidation in astrocytic cells.
Collapse
Affiliation(s)
- Masashi Morita
- Department of Biological Chemistry, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930–0194, Japan.
| | | | | | | |
Collapse
|
30
|
Palmitoylethanolamide is a disease-modifying agent in peripheral neuropathy: pain relief and neuroprotection share a PPAR-alpha-mediated mechanism. Mediators Inflamm 2013; 2013:328797. [PMID: 23533304 PMCID: PMC3596927 DOI: 10.1155/2013/328797] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 12/27/2012] [Accepted: 01/02/2013] [Indexed: 12/23/2022] Open
Abstract
Neuropathic syndromes which are evoked by lesions to the peripheral or central nervous system are extremely difficult to treat, and available drugs rarely joint an antihyperalgesic with a neurorestorative effect. N-Palmitoylethanolamine (PEA) exerts antinociceptive effects in several animal models and inhibits peripheral inflammation in rodents. Aimed to evaluate the antineuropathic properties of PEA, a damage of the sciatic nerve was induced in mice by chronic constriction injury (CCI) and a subcutaneous daily treatment with 30 mg kg(-1) PEA was performed. On the day 14, PEA prevented pain threshold alterations. Histological studies highlighted that CCI induced oedema and an important infiltrate of CD86 positive cells in the sciatic nerve. Moreover, osmicated preparations revealed a decrease in axon diameter and myelin thickness. Repeated treatments with PEA reduced the presence of oedema and macrophage infiltrate, and a significant higher myelin sheath, axonal diameter, and a number of fibers were observable. In PPAR- α null mice PEA treatment failed to induce pain relief as well as to rescue the peripheral nerve from inflammation and structural derangement. These results strongly suggest that PEA, via a PPAR- α -mediated mechanism, can directly intervene in the nervous tissue alterations responsible for pain, starting to prevent macrophage infiltration.
Collapse
|
31
|
Martens K, Bottelbergs A, Peeters A, Jacobs F, Espeel M, Carmeliet P, Van Veldhoven PP, Baes M. Peroxisome deficient aP2-Pex5 knockout mice display impaired white adipocyte and muscle function concomitant with reduced adrenergic tone. Mol Genet Metab 2012; 107:735-47. [PMID: 23141464 DOI: 10.1016/j.ymgme.2012.10.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 10/16/2012] [Accepted: 10/16/2012] [Indexed: 12/21/2022]
Abstract
Peroxisomes are essential for intermediary lipid metabolism, but the role of these organelles has been primarily studied in the liver. We recently generated aP2-Pex5 conditional knockout mice that due to the nonselectivity of the aP2 promoter, not only had dysfunctional peroxisomes in the adipose tissue but also in the central and peripheral nervous system, besides some other tissues. Peroxisomes were however intact in the liver, heart, pancreas and muscle. Surprisingly, these mice not only showed dysfunctional white adipose tissue with increased fat mass and reduced lipolysis but also the skeletal muscle was affected including impaired shivering thermogenesis, reduced motor performance and increased insulin resistance. Non-shivering thermogenesis by brown adipose tissue was not altered. Strongly reduced levels of plasma adrenaline and to a lesser extent noradrenaline, impaired expression of catecholamine synthesizing enzymes in the adrenal medulla and reversal of all pathologies after administration of the β-agonist isoproterenol indicated that β-adrenergic signaling was reduced. Based on normal white adipose and muscle function in Nestin-Pex5 and Wnt-Pex5 knockout mice respectively, it is unlikely that peroxisome absence from the central and peripheral nervous system caused the phenotype. We conclude that peroxisomal metabolism is necessary to maintain the adrenergic tone in mice, which in turn determines metabolic homeostasis.
Collapse
Affiliation(s)
- Katrin Martens
- Laboratory of Cell Metabolism, Department of Pharmaceutical Sciences, KU Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Peroxisomes are remarkably versatile cell organelles whose size, shape, number, and protein content can vary greatly depending on the organism, the developmental stage of the organism’s life cycle, and the environment in which the organism lives. The main functions usually associated with peroxisomes include the metabolism of lipids and reactive oxygen species. However, in recent years, it has become clear that these organelles may also act as intracellular signaling platforms that mediate developmental decisions by modulating extraperoxisomal concentrations of several second messengers. To fulfill their functions, peroxisomes physically and functionally interact with other cell organelles, including mitochondria and the endoplasmic reticulum. Defects in peroxisome dynamics can lead to organelle dysfunction and have been associated with various human disorders. The purpose of this paper is to thoroughly summarize and discuss the current concepts underlying peroxisome formation, multiplication, and degradation. In addition, this paper will briefly highlight what is known about the interplay between peroxisomes and other cell organelles and explore the physiological and pathological implications of this interorganellar crosstalk.
Collapse
Affiliation(s)
- Marc Fransen
- Department of Cellular and Molecular Medicine, KU Leuven, Herestraat 49, P.O. Box 601, 3000 Leuven, Belgium
| |
Collapse
|
33
|
Ro M, Park J, Nam M, Bang HJ, Yang J, Choi KS, Kim SK, Chung JH, Kwack K. Association between peroxisomal biogenesis factor 7 and autism spectrum disorders in a Korean population. J Child Neurol 2012; 27:1270-5. [PMID: 22378669 DOI: 10.1177/0883073811435507] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Autism spectrum disorder is a neurodevelopmental disorder characterized by deficits in social communication, impaired reciprocal social interaction, and repetitive patterns of behaviors or interests. Although the cause of autism spectrum disorder remains elusive, the present study identified peroxisomal biogenesis factor 7 (PEX7) as a gene associated with autism spectrum disorder, and this association was examined in a Korean population. PEX7 encodes a cytosolic receptor for peroxisome targeting signal 2 of peroxisomal matrix enzymes that are targeted to and translocated into the peroxisome. PEX7 defects are associated with rhizomelic chondrodysplasia punctata type 1 and Refsum disease. Mutations in PEX7 are related to a variety of mild to severe clinical symptoms, including mental retardation. The analysis of 9 intronic single nucleotide polymorphisms in 214 patients with autism spectrum disorder and 258 controls revealed the association of 2 single nucleotide polymorphisms and 1 haplotype with autism spectrum disorder (P < .05).
Collapse
Affiliation(s)
- MyungJa Ro
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Role of peroxisomes in ROS/RNS-metabolism: Implications for human disease. Biochim Biophys Acta Mol Basis Dis 2012; 1822:1363-73. [DOI: 10.1016/j.bbadis.2011.12.001] [Citation(s) in RCA: 412] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 11/25/2011] [Accepted: 12/02/2011] [Indexed: 12/27/2022]
|
35
|
Baes M, Van Veldhoven PP. Mouse models for peroxisome biogenesis defects and β-oxidation enzyme deficiencies. Biochim Biophys Acta Mol Basis Dis 2012; 1822:1489-500. [DOI: 10.1016/j.bbadis.2012.03.003] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Revised: 02/22/2012] [Accepted: 03/06/2012] [Indexed: 12/26/2022]
|
36
|
Cappa M, Bizzarri C, Petroni A, Carta G, Cordeddu L, Valeriani M, Vollono C, De Pasquale L, Blasevich M, Banni S. A mixture of oleic, erucic and conjugated linoleic acids modulates cerebrospinal fluid inflammatory markers and improve somatosensorial evoked potential in X-linked adrenoleukodystrophy female carriers. J Inherit Metab Dis 2012; 35:899-907. [PMID: 22189598 PMCID: PMC3432215 DOI: 10.1007/s10545-011-9432-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 11/23/2011] [Accepted: 11/28/2011] [Indexed: 12/03/2022]
Abstract
X-linked adrenoleukodystrophy is a rare inherited demyelinating disorder characterized by an abnormal accumulation of very long chain fatty acids, mainly hexacosanoic acid (26:0), due to a mutation of the gene encoding for a peroxisomal membrane protein. The only available, and partially effective, therapeutic treatment consists of dietary intake of a 4:1 mixture of triolein and trierucin, called Lorenzo's oil (LO), targeted to inhibit the elongation of docosanoic acid (22:0) to 26:0. In this study we tested whether, besides inhibiting elongation, an enhancement of peroxisomal beta oxidation induced by conjugated linoleic acid (CLA), will improve somatosensory evoked potentials and modify inflammatory markers in adrenoleukodystrophy females carriers. We enrolled five heterozygous women. They received a mixture of LO (40 g/day) with CLA (5 g/day) for 2 months. The therapeutic efficacy was evaluated by the means of plasma levels of 26:0, 26:0/22:0 ratio, modification of cerebrospinal fluid (CSF) inflammatory markers and somatosensory evoked potentials. Changes of fatty acid profile, and in particular CLA incorporation, were also evaluated in CSF and plasma. The results showed that CLA promptly passes the blood brain barrier and the mixture was able to lower both 26:0 and 26:0/22:0 ratio in plasma. The mixture improved somatosensory evoked potentials, which were previously found unchanged or worsened with dietary LO alone, and reduced IL-6 levels in CSF in three out of five patients. Our data suggest that the synergic activity of CLA and LO, by enhancing peroxisomal beta-oxidation and preventing 26:0 formation, improves the somatosensory evoked potentials and reduces neuroinflammation.
Collapse
Affiliation(s)
- Marco Cappa
- Unità Operativa di Endocrinologia, Ospedale Pediatrico Bambino Gesù, Roma, Italy
| | - Carla Bizzarri
- Unità Operativa di Endocrinologia, Ospedale Pediatrico Bambino Gesù, Roma, Italy
| | - Anna Petroni
- Dipartimento di Scienze Farmacologiche, Università di Milano, Milano, Italy
| | - Gianfranca Carta
- Dipartimento di Biologia Sperimentale, Università di Cagliari, Cittadella Universitaria, 09042 Monserrato, CA Italy
| | - Lina Cordeddu
- Dipartimento di Biologia Sperimentale, Università di Cagliari, Cittadella Universitaria, 09042 Monserrato, CA Italy
| | | | - Catello Vollono
- Unità Operativa di Neurologia, Ospedale Pediatrico Bambino Gesù, Roma, Italy
| | | | - Milena Blasevich
- Dipartimento di Scienze Farmacologiche, Università di Milano, Milano, Italy
| | - Sebastiano Banni
- Dipartimento di Biologia Sperimentale, Università di Cagliari, Cittadella Universitaria, 09042 Monserrato, CA Italy
- Nutrisearch srl, Pula, Cagliari Italy
| |
Collapse
|
37
|
Baarine M, Andréoletti P, Athias A, Nury T, Zarrouk A, Ragot K, Vejux A, Riedinger JM, Kattan Z, Bessede G, Trompier D, Savary S, Cherkaoui-Malki M, Lizard G. Evidence of oxidative stress in very long chain fatty acid – Treated oligodendrocytes and potentialization of ROS production using RNA interference-directed knockdown of ABCD1 and ACOX1 peroxisomal proteins. Neuroscience 2012; 213:1-18. [DOI: 10.1016/j.neuroscience.2012.03.058] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 03/11/2012] [Accepted: 03/29/2012] [Indexed: 10/28/2022]
|
38
|
Bottelbergs A, Verheijden S, Van Veldhoven PP, Just W, Devos R, Baes M. Peroxisome deficiency but not the defect in ether lipid synthesis causes activation of the innate immune system and axonal loss in the central nervous system. J Neuroinflammation 2012; 9:61. [PMID: 22458306 PMCID: PMC3419640 DOI: 10.1186/1742-2094-9-61] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 03/29/2012] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Mice with peroxisome deficiency in neural cells (Nestin-Pex5-/-) develop a neurodegenerative phenotype leading to motor and cognitive disabilities and early death. Major pathologies at the end stage of disease include severe demyelination, axonal degeneration and neuroinflammation. We now investigated the onset and progression of these pathological processes, and their potential interrelationship. In addition, the putative role of oxidative stress, the impact of plasmalogen depletion on the neurodegenerative phenotype, and the consequences of peroxisome elimination in the postnatal period were studied. METHODS Immunohistochemistry in association with gene expression analysis was performed on Nestin-Pex5-/- mice to document demyelination, axonal damage and neuroinflammation. Also Gnpat-/- mice, with selective plasmalogen deficiency and CMV-Tx-Pex5-/- mice, with tamoxifen induced generalized loss of peroxisomes were analysed. RESULTS Activation of the innate immune system is a very early event in the pathological process in Nestin-Pex5-/- mice which evolves in chronic neuroinflammation. The complement factor C1q, one of the earliest up regulated transcripts, was expressed on neurons and oligodendrocytes but not on microglia. Transcripts of other pro- and anti-inflammatory genes and markers of phagocytotic activity were already significantly induced before detecting pathologies with immunofluorescent staining. Demyelination, macrophage activity and axonal loss co-occurred throughout the brain. As in patients with mild peroxisome biogenesis disorders who develop regressive changes, demyelination in cerebellum and brain stem preceded major myelin loss in corpus callosum of both Nestin-Pex5-/- and CMV-Tx-Pex5-/- mice. These lesions were not accompanied by generalized oxidative stress throughout the brain. Although Gnpat-/- mice displayed dysmyelination and Purkinje cell axon damage in cerebellum, confirming previous observations, no signs of inflammation or demyelination aggravating with age were observed. CONCLUSIONS Peroxisome inactivity triggers a fast neuroinflammatory reaction, which is not solely due to the depletion of plasmalogens. In association with myelin abnormalities this causes axon damage and loss.
Collapse
Affiliation(s)
- Astrid Bottelbergs
- Laboratory of Cell Metabolism, Department of Pharmaceutical Sciences, K.U. Leuven, Leuven, Belgium
| | | | | | | | | | | |
Collapse
|
39
|
Pexophagy: the selective degradation of peroxisomes. Int J Cell Biol 2012; 2012:512721. [PMID: 22536249 PMCID: PMC3320016 DOI: 10.1155/2012/512721] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2011] [Accepted: 11/23/2011] [Indexed: 12/18/2022] Open
Abstract
Peroxisomes are single-membrane-bounded organelles present in the majority of eukaryotic cells. Despite the existence of great diversity among different species, cell types, and under different environmental conditions, peroxisomes contain enzymes involved in β-oxidation of fatty acids and the generation, as well as detoxification, of hydrogen peroxide. The exigency of all eukaryotic cells to quickly adapt to different environmental factors requires the ability to precisely and efficiently control peroxisome number and functionality. Peroxisome homeostasis is achieved by the counterbalance between organelle biogenesis and degradation. The selective degradation of superfluous or damaged peroxisomes is facilitated by several tightly regulated pathways. The most prominent peroxisome degradation system uses components of the general autophagy core machinery and is therefore referred to as “pexophagy.” In this paper we focus on recent developments in pexophagy and provide an overview of current knowledge and future challenges in the field. We compare different modes of pexophagy and mention shared and distinct features of pexophagy in yeast model systems, mammalian cells, and other organisms.
Collapse
|
40
|
Corthals AP. Multiple sclerosis is not a disease of the immune system. QUARTERLY REVIEW OF BIOLOGY 2012; 86:287-321. [PMID: 22384749 DOI: 10.1086/662453] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Multiple sclerosis is a complex neurodegenerative disease, thought to arise through autoimmunity against antigens of the central nervous system. The autoimmunity hypothesis fails to explain why genetic and environmental risk factors linked to the disease in one population tend to be unimportant in other populations. Despite great advances in documenting the cell and molecular mechanisms underlying MS pathophysiology, the autoimmunity framework has also been unable to develop a comprehensive explanation of the etiology of the disease. I propose a new framework for understanding MS as a dysfunction of the metabolism of lipids. Specifically, the homeostasis of lipid metabolism collapses during acute-phase inflammatory response triggered by a pathogen, trauma, or stress, starting a feedback loop of increased oxidative stress, inflammatory response, and proliferation of cytoxic foam cells that cross the blood brain barrier and both catabolize myelin and prevent remyelination. Understanding MS as a chronic metabolic disorder illuminates four aspects of disease onset and progression: 1) its pathophysiology; 2) genetic susceptibility; 3) environmental and pathogen triggers; and 4) the skewed sex ratio of patients. It also suggests new avenues for treatment.
Collapse
Affiliation(s)
- Angelique P Corthals
- Department of Sciences, John Jay College of Criminal Justice, City University of New York New York, New York 10019, USA.
| |
Collapse
|
41
|
Baarine M, Ragot K, Athias A, Nury T, Kattan Z, Genin EC, Andreoletti P, Ménétrier F, Riedinger JM, Bardou M, Lizard G. Incidence of Abcd1 level on the induction of cell death and organelle dysfunctions triggered by very long chain fatty acids and TNF-α on oligodendrocytes and astrocytes. Neurotoxicology 2012; 33:212-28. [DOI: 10.1016/j.neuro.2011.10.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2011] [Revised: 10/21/2011] [Accepted: 10/21/2011] [Indexed: 01/13/2023]
|
42
|
Morita M, Imanaka T. Peroxisomal ABC transporters: structure, function and role in disease. Biochim Biophys Acta Mol Basis Dis 2012; 1822:1387-96. [PMID: 22366764 DOI: 10.1016/j.bbadis.2012.02.009] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2011] [Revised: 01/07/2012] [Accepted: 02/08/2012] [Indexed: 12/20/2022]
Abstract
ATP-binding cassette (ABC) transporters belong to one of the largest families of membrane proteins, and are present in almost all living organisms from eubacteria to mammals. They exist on plasma membranes and intracellular compartments such as the mitochondria, peroxisomes, endoplasmic reticulum, Golgi apparatus and lysosomes, and mediate the active transport of a wide variety of substrates in a variety of different cellular processes. These include the transport of amino acids, polysaccharides, peptides, lipids and xenobiotics, including drugs and toxins. Three ABC transporters belonging to subfamily D have been identified in mammalian peroxisomes. The ABC transporters are half-size and assemble mostly as a homodimer after posttranslational transport to peroxisomal membranes. ABCD1/ALDP and ABCD2/ALDRP are suggested to be involved in the transport of very long chain acyl-CoA with differences in substrate specificity, and ABCD3/PMP70 is involved in the transport of long and branched chain acyl-CoA. ABCD1 is known to be responsible for X-linked adrenoleukodystrophy (X-ALD), an inborn error of peroxisomal β-oxidation of very long chain fatty acids. Here, we summarize recent advances and important points in our advancing understanding of how these ABC transporters target and assemble to peroxisomal membranes and perform their functions in physiological and pathological processes, including the neurodegenerative disease, X-ALD.
Collapse
Affiliation(s)
- Masashi Morita
- Department of Biological Chemistry, University of Toyama, Toyama, Japan
| | | |
Collapse
|
43
|
Fission and proliferation of peroxisomes. Biochim Biophys Acta Mol Basis Dis 2011; 1822:1343-57. [PMID: 22240198 DOI: 10.1016/j.bbadis.2011.12.014] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 12/22/2011] [Accepted: 12/23/2011] [Indexed: 01/12/2023]
Abstract
Peroxisomes are remarkably dynamic, multifunctional organelles, which react to physiological changes in their cellular environment and adopt their morphology, number, enzyme content and metabolic functions accordingly. At the organelle level, the key molecular machinery controlling peroxisomal membrane elongation and remodeling as well as membrane fission is becoming increasingly established and defined. Key players in peroxisome division are conserved in animals, plants and fungi, and key fission components are shared with mitochondria. However, the physiological stimuli and corresponding signal transduction pathways regulating and modulating peroxisome maintenance and proliferation are, despite a few exceptions, largely unexplored. There is emerging evidence that peroxisomal dynamics and proper regulation of peroxisome number and morphology are crucial for the physiology of the cell, as well as for the pathology of the organism. Here, we discuss several key aspects of peroxisomal fission and proliferation and highlight their association with certain diseases. We address signaling and transcriptional events resulting in peroxisome proliferation, and focus on novel findings concerning the key division components and their interplay. Finally, we present an updated model of peroxisomal growth and division. This article is part of a Special Issue entitled: Metabolic Functions and Biogenesis of Peroxisomes in Health and Disease.
Collapse
|
44
|
Kahn E, Baarine M, Dauphin A, Ragot K, Tissot N, Seguin A, Ménétrier F, Kattan Z, Bachelet CM, Frouin F, Lizard G. Impact of 7-ketocholesterol and very long chain fatty acids on oligodendrocyte lipid membrane organization: Evaluation via LAURDAN and FAMIS spectral image analysis. Cytometry A 2011; 79:293-305. [DOI: 10.1002/cyto.a.21017] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Revised: 12/02/2010] [Accepted: 12/05/2010] [Indexed: 02/04/2023]
|
45
|
Faust PL, Kaye EM, Powers JM. Myelin lesions associated with lysosomal and peroxisomal disorders. Expert Rev Neurother 2010; 10:1449-66. [PMID: 20819015 DOI: 10.1586/ern.10.127] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Abnormalities of myelin are common in lysosomal and peroxisomal disorders. Most display a primary loss of myelin in which the myelin sheath and/or oligodendrocytes are selectively targeted by diverse pathogenetic processes. The most severe and, hence, clinically relevant are heritable diseases predominantly of infants and children, the leukodystrophies: metachromatic, globoid cell (Krabbe disease) and adreno-leukodystrophy. Our still limited understanding of these diseases has derived from multiple sources: originally, neurological-neuropathologic-neurochemical correlative studies of the natural disease in humans or other mammals, which has been enhanced by more sophisticated and contemporary techniques of cell and molecular biology. Transgenic mouse models seem to be the most promising methodology, allowing the examination of the cellular role of lysosomes and peroxisomes for formation and maintenance of both myelin and axons, and providing initial platforms to evaluate therapies. Treatment options are woefully inadequate and in their nascent stages, but still inspire some hope for the future.
Collapse
Affiliation(s)
- Phyllis L Faust
- Department of Pathology and Cell Biology, Columbia University, 630 West 168th Street, New York, NY 10032, USA.
| | | | | |
Collapse
|
46
|
Abstract
The myelination of axons by glial cells was the last major step in the evolution of cells in the vertebrate nervous system, and white-matter tracts are key to the architecture of the mammalian brain. Cell biology and mouse genetics have provided insight into axon-glia signalling and the molecular architecture of the myelin sheath. Glial cells that myelinate axons were found to have a dual role by also supporting the long-term integrity of those axons. This function may be independent of myelin itself. Myelin abnormalities cause a number of neurological diseases, and may also contribute to complex neuropsychiatric disorders.
Collapse
Affiliation(s)
- Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Hermann-Rein-Strasse 3, 37075 Göttingen, Germany.
| |
Collapse
|
47
|
Chrast R, Saher G, Nave KA, Verheijen MHG. Lipid metabolism in myelinating glial cells: lessons from human inherited disorders and mouse models. J Lipid Res 2010; 52:419-34. [PMID: 21062955 DOI: 10.1194/jlr.r009761] [Citation(s) in RCA: 224] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The integrity of central and peripheral nervous system myelin is affected in numerous lipid metabolism disorders. This vulnerability was so far mostly attributed to the extraordinarily high level of lipid synthesis that is required for the formation of myelin, and to the relative autonomy in lipid synthesis of myelinating glial cells because of blood barriers shielding the nervous system from circulating lipids. Recent insights from analysis of inherited lipid disorders, especially those with prevailing lipid depletion and from mouse models with glia-specific disruption of lipid metabolism, shed new light on this issue. The particular lipid composition of myelin, the transport of lipid-associated myelin proteins, and the necessity for timely assembly of the myelin sheath all contribute to the observed vulnerability of myelin to perturbed lipid metabolism. Furthermore, the uptake of external lipids may also play a role in the formation of myelin membranes. In addition to an improved understanding of basic myelin biology, these data provide a foundation for future therapeutic interventions aiming at preserving glial cell integrity in metabolic disorders.
Collapse
Affiliation(s)
- Roman Chrast
- Department of Medical Genetics, University of Lausanne, Switzerland.
| | | | | | | |
Collapse
|
48
|
Islinger M, Cardoso MJR, Schrader M. Be different--the diversity of peroxisomes in the animal kingdom. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1803:881-97. [PMID: 20347886 DOI: 10.1016/j.bbamcr.2010.03.013] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2010] [Revised: 03/15/2010] [Accepted: 03/18/2010] [Indexed: 10/19/2022]
Abstract
Peroxisomes represent so-called "multipurpose organelles" as they contribute to various anabolic as well as catabolic pathways. Thus, with respect to the physiological specialization of an individual organ or animal species, peroxisomes exhibit a functional diversity, which is documented by significant variations in their proteome. These differences are usually regarded as an adaptational response to the nutritional and environmental life conditions of a specific organism. Thus, human peroxisomes can be regarded as an in part physiologically unique organellar entity fulfilling metabolic functions that differ from our animal model systems. In line with this, a profound understanding on how peroxisomes acquired functional heterogeneity in terms of an evolutionary and mechanistic background is required. This review summarizes our current knowledge on the heterogeneity of peroxisomal physiology, providing insights into the genetic and cell biological mechanisms, which lead to the differential localization or expression of peroxisomal proteins and further gives an overview on peroxisomal biochemical pathways, which are specialized in different animal species and organs. Moreover, it addresses the impact of proteome studies on our understanding of differential peroxisome function describing the utility of mass spectrometry and computer-assisted algorithms to identify peroxisomal target sequences for the detection of new organ- or species-specific peroxisomal proteins.
Collapse
Affiliation(s)
- M Islinger
- Department of Anatomy and Cell Biology, Ruprecht-Karls University, 69120 Heidelberg, Germany
| | | | | |
Collapse
|
49
|
Peroxisomes, lipid metabolism and lipotoxicity. Biochim Biophys Acta Mol Cell Biol Lipids 2010; 1801:272-80. [DOI: 10.1016/j.bbalip.2010.01.001] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Revised: 01/07/2010] [Accepted: 01/07/2010] [Indexed: 12/26/2022]
|