1
|
Manor J, Jangam SV, Chung HL, Bhagwat P, Andrews J, Chester H, Kondo S, Srivastav S, Botas J, Moser AB, Huguenin SM, Wangler MF. Genetic analysis of the X-linked Adrenoleukodystrophy ABCD1 gene in Drosophila uncovers a role in Peroxisomal dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614586. [PMID: 39386423 PMCID: PMC11463603 DOI: 10.1101/2024.09.23.614586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a progressive neurodegenerative disorder caused by a loss-of-function (LOF) mutation in the ATP-binding cassette subfamily D member 1 (ABCD1) gene, leading to the accumulation of very long-chain fatty acids (VLCFAs). This disorder exhibits striking heterogeneity; some male patients develop an early childhood neuroinflammatory demyelination disorder, while other patients, including adult males and most affected female carriers, experience a chronic progressive myelopathy. Adrenocortical failure is observed in almost all male patients, with age of onset varying sometimes being the first diagnostic finding. The gene underlying this spectrum of disease encodes an ATP-binding cassette (ABC) transporter that localizes to peroxisomes and facilitates VLCFA transport. X-ALD is considered a single peroxisomal component defect and does not play a direct role in peroxisome assembly. Drosophila models of other peroxisomal genes have provided mechanistic insight into some of the neurodegenerative mechanisms with reduced lifespan, retinal degeneration, and VLCFA accumulation. Here, we perform a genetic analysis of the fly ABCD1 ortholog Abcd1 (CG2316). Knockdown or deficiency of Abcd1 leads to VLCFA accumulation, salivary gland defects, locomotor impairment and retinal lipid abnormalities. Interestingly, there is also evidence of reduced peroxisomal numbers. Flies overexpressing the human cDNA for ABCD1 display a wing crumpling phenotype characteristic of the pex2 loss-of-function. Surprisingly, overexpression of human ABCD1 appears to inhibit or overwhelm peroxisomal biogenesis to levels similar to null mutations in fly pex2, pex16 and pex3. Drosophila Abcd1 is therefore implicated in peroxisomal number, and overexpression of the human ABCD1 gene acts a potent inhibitor of peroxisomal biogenesis in flies.
Collapse
Affiliation(s)
- Joshua Manor
- Metabolic Disease Unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Sharayu V Jangam
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Hyung-lok Chung
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, USA
- Department of Neurology, Weill Cornell Medical College, New York, NY, USA
| | - Pranjali Bhagwat
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Jonathan Andrews
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Hillary Chester
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Shu Kondo
- Tokyo University of Science, Faculty of Advanced Engineering, Department of Biological Science and Technology, Tokyo, Japan
| | - Saurabh Srivastav
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Juan Botas
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Ann B. Moser
- Hugo W Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Suzette M. Huguenin
- Hugo W Moser Research Institute, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Michael F Wangler
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| |
Collapse
|
2
|
Ali H, Yamanishi M, Sunagawa K, Kumon M, Hasi RY, Aihara M, Kawakami R, Tanaka T. Protective effect of oleic acid against very long-chain fatty acid-induced apoptosis in peroxisome-deficient CHO cells. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159452. [PMID: 38244676 DOI: 10.1016/j.bbalip.2024.159452] [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: 12/19/2023] [Revised: 01/14/2024] [Accepted: 01/15/2024] [Indexed: 01/22/2024]
Abstract
Very long-chain fatty acids (VLCFAs) are degraded exclusively in peroxisomes, as evidenced by the accumulation of VLCFAs in patients with certain peroxisomal disorders. Although accumulation of VLCFAs is considered to be associated with health issues, including neuronal degeneration, the mechanisms underlying VLCFAs-induced tissue degeneration remain unclear. Here, we report the toxic effect of VLCFA and protective effect of C18: 1 FA in peroxisome-deficient CHO cells. We examined the cytotoxicity of saturated and monounsaturated VLCFAs with chain-length at C20-C26, and found that longer and saturated VLCFA showed potent cytotoxicity at lower accumulation levels. Furthermore, the extent of VLCFA-induced toxicity was found to be associated with a decrease in cellular C18:1 FA levels. Notably, supplementation with C18:1 FA effectively rescued the cells from VLCFA-induced apoptosis without reducing the cellular VLCFAs levels, implying that peroxisome-deficient cells can survive in the presence of accumulated VLCFA, as long as the cells keep sufficient levels of cellular C18:1 FA. These results suggest a therapeutic potential of C18:1 FA in peroxisome disease and may provide new insights into the pharmacological effect of Lorenzo's oil, a 4:1 mixture of C18:1 and C22:1 FA.
Collapse
Affiliation(s)
- Hanif Ali
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima 770-8513, Japan
| | - Mone Yamanishi
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima 770-8513, Japan
| | - Keigo Sunagawa
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima 770-8513, Japan
| | - Mizuki Kumon
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima 770-8513, Japan
| | - Rumana Yesmin Hasi
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima 770-8513, Japan
| | - Mutsumi Aihara
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima 770-8513, Japan
| | - Ryushi Kawakami
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima 770-8513, Japan
| | - Tamotsu Tanaka
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima 770-8513, Japan.
| |
Collapse
|
3
|
Varma A, Weinstein J, Seabury J, Rosero S, Dilek N, Heatwole J, Engebrecht C, Khosa S, Chung K, Paker A, Woo A, Brooks G, Beals C, Gandhi R, Heatwole C. Patient-reported impact of symptoms in adrenoleukodystrophy (PRISM-ALD). Orphanet J Rare Dis 2024; 19:127. [PMID: 38504253 PMCID: PMC10953228 DOI: 10.1186/s13023-024-03129-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 03/03/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Adrenoleukodystrophy (ALD) is a multifaceted, X-linked, neurodegenerative disorder that comprises several clinical phenotypes. ALD affects patients through a variety of physical, emotional, social, and other disease-specific factors that collectively contribute to disease burden. To facilitate clinical care and research, it is important to identify which symptoms are most common and relevant to individuals with any subtype of ALD. METHODS We conducted semi-structured qualitative interviews and an international cross-sectional study to determine the most prevalent and important symptoms of ALD. Our study included adult participants with a diagnosis of ALD who were recruited from national and international patient registries. Responses were categorized by age, sex, disease phenotype, functional status, and other demographic and clinical features. RESULTS Seventeen individuals with ALD participated in qualitative interviews, providing 1709 direct quotes regarding their symptomatic burden. One hundred and nine individuals participated in the cross-sectional survey study, which inquired about 182 unique symptoms representing 24 distinct symptomatic themes. The symptomatic themes with the highest prevalence in the overall ALD sample cohort were problems with balance (90.9%), limitations with mobility or walking (87.3%), fatigue (86.4%), and leg weakness (86.4%). The symptomatic themes with the highest impact scores (on a 0-4 scale with 4 being the most severe) were trouble getting around (2.35), leg weakness (2.25), and problems with balance (2.21). A higher prevalence of symptomatic themes was associated with functional disability, employment disruption, and speech impairment. CONCLUSIONS There are many patient-relevant symptoms and themes that contribute to disease burden in individuals with ALD. These symptoms, identified by those having ALD, present key targets for further research and therapeutic development.
Collapse
Affiliation(s)
- Anika Varma
- Center for Health + Technology, University of Rochester, 265 Crittenden Blvd, CU 420694, Rochester, NY, 14642, USA.
| | - Jennifer Weinstein
- Center for Health + Technology, University of Rochester, 265 Crittenden Blvd, CU 420694, Rochester, NY, 14642, USA
| | - Jamison Seabury
- Center for Health + Technology, University of Rochester, 265 Crittenden Blvd, CU 420694, Rochester, NY, 14642, USA
| | - Spencer Rosero
- Center for Health + Technology, University of Rochester, 265 Crittenden Blvd, CU 420694, Rochester, NY, 14642, USA
| | - Nuran Dilek
- Department of Neurology, University of Rochester, 601 Elmwood Ave, Box 673, Rochester, NY, 14642, USA
| | | | - Charlotte Engebrecht
- Center for Health + Technology, University of Rochester, 265 Crittenden Blvd, CU 420694, Rochester, NY, 14642, USA
| | - Shaweta Khosa
- Center for Health + Technology, University of Rochester, 265 Crittenden Blvd, CU 420694, Rochester, NY, 14642, USA
| | - Kaitlin Chung
- Center for Health + Technology, University of Rochester, 265 Crittenden Blvd, CU 420694, Rochester, NY, 14642, USA
| | - Asif Paker
- SwanBio Therapeutics, 150 Monument Rd, Bala Cynwyd, PA, 19004, USA
| | - Amy Woo
- Autobahn Therapeutics, 9880 Campus Point Drive, San Diego, CA, 92121, USA
| | - Gregory Brooks
- Autobahn Therapeutics, 9880 Campus Point Drive, San Diego, CA, 92121, USA
| | - Chan Beals
- Autobahn Therapeutics, 9880 Campus Point Drive, San Diego, CA, 92121, USA
| | - Rohan Gandhi
- Autobahn Therapeutics, 9880 Campus Point Drive, San Diego, CA, 92121, USA
| | - Chad Heatwole
- Center for Health + Technology, University of Rochester, 265 Crittenden Blvd, CU 420694, Rochester, NY, 14642, USA
- Department of Neurology, University of Rochester, 601 Elmwood Ave, Box 673, Rochester, NY, 14642, USA
| |
Collapse
|
4
|
Sahasrabudhe SA, Terluk MR, Kartha RV. N-acetylcysteine Pharmacology and Applications in Rare Diseases-Repurposing an Old Antioxidant. Antioxidants (Basel) 2023; 12:1316. [PMID: 37507857 PMCID: PMC10376274 DOI: 10.3390/antiox12071316] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 07/30/2023] Open
Abstract
N-acetylcysteine (NAC), a precursor of cysteine and, thereby, glutathione (GSH), acts as an antioxidant through a variety of mechanisms, including oxidant scavenging, GSH replenishment, antioxidant signaling, etc. Owing to the variety of proposed targets, NAC has a long history of use as a prescription product and in wide-ranging applications that are off-label as an over-the-counter (OTC) product. Despite its discovery in the early 1960s and its development for various indications, systematic clinical pharmacology explorations of NAC pharmacokinetics (PK), pharmacodynamic targets, drug interactions, and dose-ranging are sorely limited. Although there are anecdotal instances of NAC benefits in a variety of diseases, a comprehensive review of the use of NAC in rare diseases does not exist. In this review, we attempt to summarize the existing literature focused on NAC explorations in rare diseases targeting mitochondrial dysfunction along with the history of NAC usage, approved indications, mechanisms of action, safety, and PK characterization. Further, we introduce the research currently underway on other structural derivatives of NAC and acknowledge the continuum of efforts through pre-clinical and clinical research to facilitate further therapeutic development of NAC or its derivatives for rare diseases.
Collapse
Affiliation(s)
- Siddhee A Sahasrabudhe
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, Rm 4-214, McGuire Translational Research Facility, 2001 6th St. SE, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Marcia R Terluk
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, Rm 4-214, McGuire Translational Research Facility, 2001 6th St. SE, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| | - Reena V Kartha
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, Rm 4-214, McGuire Translational Research Facility, 2001 6th St. SE, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
5
|
Ghasemi M, Rahgozar M, Kavousi K. Complex Disease Genes Identification Using a Heterogeneous Network Embedding Approach. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:875-882. [PMID: 35594221 DOI: 10.1109/tcbb.2022.3175598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Finding the causal relation between a gene and a disease using experimental approaches is a time-consuming and expensive task. However, computational approaches are cost-efficient methods for identifying candidate genes. This article proposes a new heterogeneous biological network embedding approach, named NetEM, to identify disease-associated genes. To evaluate NetEM, we examine six complex diseases, including peroxisomal disorders, sarcoma, grave's disease, lysosomal storage diseases, blood coagulation disorders, and cardiomyopathy hypertrophic. Our experiments indicate that NetEM outperforms three well-known state-of-the-art algorithms: Cardigan, DIAMOnD and GeneWanderer, in identifying disease genes. We examine TCGA data of Invasive Lobular Breast Cancer and CPTAC data of human glioblastoma as other case studies to evaluate NetEM using real data. This evaluation also indicates the validity of the method. The source codes of NetEM and data are available in the supplementary of this article.
Collapse
|
6
|
Ali H, Kobayashi M, Morito K, Hasi RY, Aihara M, Hayashi J, Kawakami R, Tsuchiya K, Sango K, Tanaka T. Peroxisomes attenuate cytotoxicity of very long-chain fatty acids. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159259. [PMID: 36460260 DOI: 10.1016/j.bbalip.2022.159259] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/13/2022] [Accepted: 11/10/2022] [Indexed: 12/02/2022]
Abstract
One of the major functions of peroxisomes in mammals is oxidation of very long-chain fatty acids (VLCFAs). Genetic defects in peroxisomal β-oxidation result in the accumulation of VLCFAs and lead to a variety of health problems, such as demyelination of nervous tissues. However, the mechanisms by which VLCFAs cause tissue degeneration have not been fully elucidated. Recently, we found that the addition of small amounts of isopropanol can enhance the solubility of saturated VLCFAs in an aqueous medium. In this study, we characterized the biological effect of extracellular VLCFAs in peroxisome-deficient Chinese hamster ovary (CHO) cells, neural crest-derived pheochromocytoma cells (PC12), and immortalized adult Fischer rat Schwann cells (IFRS1) using this solubilizing technique. C20:0 FA was the most toxic of the C16-C26 FAs tested in all cells. The basis of the toxicity of C20:0 FA was apoptosis and was observed at 5 μM and 30 μM in peroxisome-deficient and wild-type CHO cells, respectively. The sensitivity of wild-type CHO cells to cytotoxic C20:0 FA was enhanced in the presence of a peroxisomal β-oxidation inhibitor. Further, a positive correlation was evident between cell toxicity and the extent of intracellular accumulation of toxic FA. These results suggest that peroxisomes are pivotal in the detoxification of apoptotic VLCFAs by preventing their accumulation.
Collapse
Affiliation(s)
- Hanif Ali
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima 770-8502, Japan; Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Miyu Kobayashi
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima 770-8502, Japan
| | - Katsuya Morito
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Rumana Yesmin Hasi
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima 770-8502, Japan
| | - Mutsumi Aihara
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima 770-8502, Japan
| | - Junji Hayashi
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima 770-8502, Japan
| | - Ryushi Kawakami
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima 770-8502, Japan
| | - Koichiro Tsuchiya
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Kazunori Sango
- Diabetic Neuropathy Project, Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| | - Tamotsu Tanaka
- Graduate School of Technology, Industrial and Social Sciences, Tokushima University, Tokushima 770-8502, Japan.
| |
Collapse
|
7
|
Fujiki Y, Okumoto K, Honsho M, Abe Y. Molecular insights into peroxisome homeostasis and peroxisome biogenesis disorders. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119330. [PMID: 35917894 DOI: 10.1016/j.bbamcr.2022.119330] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 06/15/2023]
Abstract
Peroxisomes are single-membrane organelles essential for cell metabolism including the β-oxidation of fatty acids, synthesis of etherlipid plasmalogens, and redox homeostasis. Investigations into peroxisome biogenesis and the human peroxisome biogenesis disorders (PBDs) have identified 14 PEX genes encoding peroxins involved in peroxisome biogenesis and the mutation of PEX genes is responsible for the PBDs. Many recent findings have further advanced our understanding of the biology, physiology, and consequences of a functional deficit of peroxisomes. In this Review, we discuss cell defense mechanisms that counteract oxidative stress by 1) a proapoptotic Bcl-2 factor BAK-mediated release to the cytosol of H2O2-degrading catalase from peroxisomes and 2) peroxisomal import suppression of catalase by Ser232-phosphorylation of Pex14, a docking protein for the Pex5-PTS1 complex. With respect to peroxisome division, the important issue of how the energy-rich GTP is produced and supplied for the division process was recently addressed by the discovery of a nucleoside diphosphate kinase-like protein, termed DYNAMO1 in a lower eukaryote, which has a mammalian homologue NME3. In regard to the mechanisms underlying the pathogenesis of PBDs, a new PBD model mouse defective in Pex14 manifests a dysregulated brain-derived neurotrophic factor (BDNF)-TrkB pathway, an important signaling pathway for cerebellar morphogenesis. Communications between peroxisomes and other organelles are also addressed.
Collapse
Affiliation(s)
- Yukio Fujiki
- Medical Institute of Bioregulation, Institute of Rheological Functions of Food, Collaboration Program, Kyushu University, 3-1-1 Maidashi, Fukuoka 812-8582, Japan.
| | - Kanji Okumoto
- Department of Biology and Graduate School of Systems Life Sciences, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| | - Masanori Honsho
- Medical Institute of Bioregulation, Institute of Rheological Functions of Food, Collaboration Program, Kyushu University, 3-1-1 Maidashi, Fukuoka 812-8582, Japan
| | - Yuichi Abe
- Faculty of Arts and Science, Kyushu University, 744 Motooka, Fukuoka 819-0395, Japan
| |
Collapse
|
8
|
Monternier PA, Parasar P, Theurey P, Gluais Dagorn P, Kaur N, Nagaraja TN, Fouqueray P, Bolze S, Moller DE, Singh J, Hallakou-Bozec S. Beneficial Effects of the Direct AMP-Kinase Activator PXL770 in In Vitro and In Vivo Models of X-Linked Adrenoleukodystrophy. J Pharmacol Exp Ther 2022; 382:208-222. [PMID: 35764327 PMCID: PMC11047065 DOI: 10.1124/jpet.122.001208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/09/2022] [Indexed: 11/22/2022] Open
Abstract
X-linked adrenoleukodystrophy (ALD) is a severe orphan disease caused by mutations in the peroxisomal ABCD1 transporter gene, leading to toxic accumulation of Very Long-Chain Fatty Acids (VLCFA - in particular C26:0) resulting in inflammation, mitochondrial dysfunction and demyelination. AMP-activated protein kinase (AMPK) is downregulated in ALD, and its activation is implicated as a therapeutic target. PXL770 is the first direct allosteric AMPK activator with established clinical efficacy and tolerability. Methods: We investigated its effects in ALD patient-derived fibroblasts/lymphocytes and Abcd1 KO mouse glial cells. Readouts included VLCFA levels, mitochondrial function and mRNA levels of proinflammatory genes and compensatory transporters (ABCD2-3). After PXL770 treatment in Abcd1 KO mice, we assessed VLCFA levels in tissues, sciatic nerve axonal morphology by electronic microscopy and locomotor function by open-field/balance-beam tests. Results: In patients' cells and Abcd1 KO glial cells, PXL770 substantially decreased C26:0 levels (by ∼90%), improved mitochondrial respiration, reduced expression of multiple inflammatory genes and induced expression of ABCD2-3 In Abcd1 KO mice, PXL770 treatment normalized VLCFA in plasma and significantly reduced elevated levels in brain (-25%) and spinal cord (-32%) versus untreated (P < 0.001). Abnormal sciatic nerve axonal morphology was also improved along with amelioration of locomotor function. Conclusion: Direct AMPK activation exerts beneficial effects on several hallmarks of pathology in multiple ALD models in vitro and in vivo, supporting clinical development of PXL770 for this disease. Further studies would be needed to overcome limitations including small sample size for some parameters, lack of additional in vivo biomarkers and incomplete pharmacokinetic characterization. SIGNIFICANCE STATEMENT: Adrenoleukodystrophy is a rare and debilitating condition with no approved therapies, caused by accumulation of very long-chain fatty acids. AMPK is downregulated in the disease and has been implicated as a potential therapeutic target. PXL770 is a novel clinical stage direct AMPK activator. In these studies, we used PXL770 to achieve preclinical validation of direct AMPK activation for this disease - based on correction of key biochemical and functional readouts in vitro and in vivo, thus supporting clinical development.
Collapse
Affiliation(s)
- Pierre-Axel Monternier
- Poxel SA, Lyon, France (P.-A.M., P.T., P.G.D., P.F., S.B., D.E.M., S.H.-B.) and Departments of Neurology (P.P., N.K., J.S.) and Neurosurgery (T.N.N.), Henry Ford Health System, Detroit, Michigan
| | - Parveen Parasar
- Poxel SA, Lyon, France (P.-A.M., P.T., P.G.D., P.F., S.B., D.E.M., S.H.-B.) and Departments of Neurology (P.P., N.K., J.S.) and Neurosurgery (T.N.N.), Henry Ford Health System, Detroit, Michigan
| | - Pierre Theurey
- Poxel SA, Lyon, France (P.-A.M., P.T., P.G.D., P.F., S.B., D.E.M., S.H.-B.) and Departments of Neurology (P.P., N.K., J.S.) and Neurosurgery (T.N.N.), Henry Ford Health System, Detroit, Michigan
| | - Pascale Gluais Dagorn
- Poxel SA, Lyon, France (P.-A.M., P.T., P.G.D., P.F., S.B., D.E.M., S.H.-B.) and Departments of Neurology (P.P., N.K., J.S.) and Neurosurgery (T.N.N.), Henry Ford Health System, Detroit, Michigan
| | - Navtej Kaur
- Poxel SA, Lyon, France (P.-A.M., P.T., P.G.D., P.F., S.B., D.E.M., S.H.-B.) and Departments of Neurology (P.P., N.K., J.S.) and Neurosurgery (T.N.N.), Henry Ford Health System, Detroit, Michigan
| | - Tavarekere N Nagaraja
- Poxel SA, Lyon, France (P.-A.M., P.T., P.G.D., P.F., S.B., D.E.M., S.H.-B.) and Departments of Neurology (P.P., N.K., J.S.) and Neurosurgery (T.N.N.), Henry Ford Health System, Detroit, Michigan
| | - Pascale Fouqueray
- Poxel SA, Lyon, France (P.-A.M., P.T., P.G.D., P.F., S.B., D.E.M., S.H.-B.) and Departments of Neurology (P.P., N.K., J.S.) and Neurosurgery (T.N.N.), Henry Ford Health System, Detroit, Michigan
| | - Sébastien Bolze
- Poxel SA, Lyon, France (P.-A.M., P.T., P.G.D., P.F., S.B., D.E.M., S.H.-B.) and Departments of Neurology (P.P., N.K., J.S.) and Neurosurgery (T.N.N.), Henry Ford Health System, Detroit, Michigan
| | - David E Moller
- Poxel SA, Lyon, France (P.-A.M., P.T., P.G.D., P.F., S.B., D.E.M., S.H.-B.) and Departments of Neurology (P.P., N.K., J.S.) and Neurosurgery (T.N.N.), Henry Ford Health System, Detroit, Michigan
| | - Jaspreet Singh
- Poxel SA, Lyon, France (P.-A.M., P.T., P.G.D., P.F., S.B., D.E.M., S.H.-B.) and Departments of Neurology (P.P., N.K., J.S.) and Neurosurgery (T.N.N.), Henry Ford Health System, Detroit, Michigan
| | - Sophie Hallakou-Bozec
- Poxel SA, Lyon, France (P.-A.M., P.T., P.G.D., P.F., S.B., D.E.M., S.H.-B.) and Departments of Neurology (P.P., N.K., J.S.) and Neurosurgery (T.N.N.), Henry Ford Health System, Detroit, Michigan
| |
Collapse
|
9
|
Monternier P, Singh J, Parasar P, Theurey P, DeWitt S, Jacques V, Klett E, Kaur N, Nagaraja TN, Moller DE, Hallakou‐Bozec S. Therapeutic potential of deuterium-stabilized (R)-pioglitazone-PXL065-for X-linked adrenoleukodystrophy. J Inherit Metab Dis 2022; 45:832-847. [PMID: 35510808 PMCID: PMC9545763 DOI: 10.1002/jimd.12510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 11/18/2022]
Abstract
X-linked adrenoleukodystrophy (ALD) results from ABCD1 gene mutations which impair Very Long Chain Fatty Acids (VLCFA; C26:0 and C24:0) peroxisomal import and β-oxidation, leading to accumulation in plasma and tissues. Excess VLCFA drives impaired cellular functions (e.g. disrupted mitochondrial function), inflammation, and neurodegeneration. Major disease phenotypes include: adrenomyeloneuropathy (AMN), progressive spinal cord axonal degeneration, and cerebral ALD (C-ALD), inflammatory white matter demyelination and degeneration. No pharmacological treatment is available to-date for ALD. Pioglitazone, an anti-diabetic thiazolidinedione, exerts potential benefits in ALD models. Its mechanisms are genomic (PPARγ agonism) and nongenomic (mitochondrial pyruvate carrier-MPC, long-chain acyl-CoA synthetase 4-ACSL4, inhibition). However, its use is limited by PPARγ-driven side effects (e.g. weight gain, edema). PXL065 is a clinical-stage deuterium-stabilized (R)-enantiomer of pioglitazone which lacks PPARγ agonism but retains MPC activity. Here, we show that incubation of ALD patient-derived cells (both AMN and C-ALD) and glial cells from Abcd1-null mice with PXL065 resulted in: normalization of elevated VLCFA, improved mitochondrial function, and attenuated indices of inflammation. Compensatory peroxisomal transporter gene expression was also induced. Additionally, chronic treatment of Abcd1-null mice lowered VLCFA in plasma, brain and spinal cord and improved both neural histology (sciatic nerve) and neurobehavioral test performance. Several in vivo effects of PXL065 exceeded those achieved with pioglitazone. PXL065 was confirmed to lack PPARγ agonism but retained ACSL4 activity of pioglitazone. PXL065 has novel actions and mechanisms and exhibits a range of potential benefits in ALD models; further testing of this molecule in ALD patients is warranted.
Collapse
Affiliation(s)
| | - Jaspreet Singh
- Department of NeurologyHenry Ford Health SystemDetroitMichiganUSA
| | - Parveen Parasar
- Department of NeurologyHenry Ford Health SystemDetroitMichiganUSA
| | | | | | | | - Eric Klett
- Department of Medicine, Division of EndocrinologyUniversity of North Carolina School of MedicineChapel HillNorth CarolinaUSA
| | - Navtej Kaur
- Department of NeurologyHenry Ford Health SystemDetroitMichiganUSA
| | | | | | | |
Collapse
|
10
|
Terluk MR, Tieu J, Sahasrabudhe SA, Moser A, Watkins PA, Raymond GV, Kartha RV. Nervonic Acid Attenuates Accumulation of Very Long-Chain Fatty Acids and is a Potential Therapy for Adrenoleukodystrophy. Neurotherapeutics 2022; 19:1007-1017. [PMID: 35378685 PMCID: PMC9294126 DOI: 10.1007/s13311-022-01226-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2022] [Indexed: 12/23/2022] Open
Abstract
Adrenoleukodystrophy (ALD) is an X-linked inherited peroxisomal disorder due to mutations in the ALD protein and characterized by accumulation of very long-chain fatty acids (VLCFA), specifically hexacosanoic acid (C26:0). This can trigger other pathological processes such as mitochondrial dysfunction, oxidative stress, and inflammation, which if involves the brain tissues can result in a lethal form of the disease called childhood cerebral ALD. With the recent addition of ALD to the Recommended Uniform Screening Panel, there is an increase in the number of individuals who are identified with ALD. However, currently, there is no approved treatment for pre-symptomatic individuals that can arrest or delay symptom development. Here, we report our observations investigating nervonic acid, a monounsaturated fatty acid as a potential therapy for ALD. Using ALD patient-derived fibroblasts, we examined whether nervonic acid can reverse VLCFA accumulation similar to erucic acid, the active ingredient in Lorenzo's oil, a dietary intervention believed to alter disease course. We have shown that nervonic acid can reverse total lipid C26:0 accumulation in a concentration-dependent manner in ALD cell lines. Further, we show that nervonic acid can protect ALD fibroblasts from oxidative insults, presumably by increasing intracellular ATP production. Thus, nervonic acid can be a potential therapeutic for individuals with ALD, which can alter cellular biochemistry and improve its function.
Collapse
Affiliation(s)
- Marcia R Terluk
- Center for Orphan Drug Research, University of Minnesota, McGuire Translational Research Facility, 2001 6th St SE, Minneapolis, MN, 55455, USA
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Julianne Tieu
- Center for Orphan Drug Research, University of Minnesota, McGuire Translational Research Facility, 2001 6th St SE, Minneapolis, MN, 55455, USA
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Siddhee A Sahasrabudhe
- Center for Orphan Drug Research, University of Minnesota, McGuire Translational Research Facility, 2001 6th St SE, Minneapolis, MN, 55455, USA
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Ann Moser
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, 21205, USA
- Department of Neurology, John Hopkins University, Baltimore, MD, 21287, USA
| | - Paul A Watkins
- Hugo W. Moser Research Institute at Kennedy Krieger, Baltimore, MD, 21205, USA
- Department of Neurology, John Hopkins University, Baltimore, MD, 21287, USA
| | - Gerald V Raymond
- Department of Neurology, John Hopkins University, Baltimore, MD, 21287, USA
- Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Reena V Kartha
- Center for Orphan Drug Research, University of Minnesota, McGuire Translational Research Facility, 2001 6th St SE, Minneapolis, MN, 55455, USA.
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
11
|
Kim J, Bai H. Peroxisomal Stress Response and Inter-Organelle Communication in Cellular Homeostasis and Aging. Antioxidants (Basel) 2022; 11:192. [PMID: 35204075 PMCID: PMC8868334 DOI: 10.3390/antiox11020192] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/15/2022] [Accepted: 01/16/2022] [Indexed: 12/20/2022] Open
Abstract
Peroxisomes are key regulators of cellular and metabolic homeostasis. These organelles play important roles in redox metabolism, the oxidation of very-long-chain fatty acids (VLCFAs), and the biosynthesis of ether phospholipids. Given the essential role of peroxisomes in cellular homeostasis, peroxisomal dysfunction has been linked to various pathological conditions, tissue functional decline, and aging. In the past few decades, a variety of cellular signaling and metabolic changes have been reported to be associated with defective peroxisomes, suggesting that many cellular processes and functions depend on peroxisomes. Peroxisomes communicate with other subcellular organelles, such as the nucleus, mitochondria, endoplasmic reticulum (ER), and lysosomes. These inter-organelle communications are highly linked to the key mechanisms by which cells surveil defective peroxisomes and mount adaptive responses to protect them from damages. In this review, we highlight the major cellular changes that accompany peroxisomal dysfunction and peroxisomal inter-organelle communication through membrane contact sites, metabolic signaling, and retrograde signaling. We also discuss the age-related decline of peroxisomal protein import and its role in animal aging and age-related diseases. Unlike other organelle stress response pathways, such as the unfolded protein response (UPR) in the ER and mitochondria, the cellular signaling pathways that mediate stress responses to malfunctioning peroxisomes have not been systematically studied and investigated. Here, we coin these signaling pathways as "peroxisomal stress response pathways". Understanding peroxisomal stress response pathways and how peroxisomes communicate with other organelles are important and emerging areas of peroxisome research.
Collapse
Affiliation(s)
- Jinoh Kim
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA 50011, USA
| | - Hua Bai
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
12
|
Zhou J, Terluk MR, Orchard PJ, Cloyd JC, Kartha RV. N-Acetylcysteine Reverses the Mitochondrial Dysfunction Induced by Very Long-Chain Fatty Acids in Murine Oligodendrocyte Model of Adrenoleukodystrophy. Biomedicines 2021; 9:biomedicines9121826. [PMID: 34944641 PMCID: PMC8698433 DOI: 10.3390/biomedicines9121826] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/24/2021] [Accepted: 11/28/2021] [Indexed: 11/16/2022] Open
Abstract
The accumulation of saturated very long-chain fatty acids (VLCFA, ≥C22:0) due to peroxisomal impairment leads to oxidative stress and neurodegeneration in X-linked adrenoleukodystrophy (ALD). Among the neural supporting cells, myelin-producing oligodendrocytes are the most sensitive to the detrimental effect of VLCFA. Here, we characterized the mitochondrial dysfunction and cell death induced by VLFCA, and examined whether N-acetylcysteine (NAC), an antioxidant, prevents the cytotoxicity. We exposed murine oligodendrocytes (158 N) to hexacosanoic acid (C26:0, 1-100 µM) for 24 h and measured reactive oxygen species (ROS) and cell death. Low concentrations of C26:0 (≤25 µM) induced a mild effect on cell survival with no alterations in ROS or total glutathione (GSH) concentrations. However, analysis of the mitochondrial status of cells treated with C26:0 (25 µM) revealed depletion in mitochondrial GSH (mtGSH) and a decrease in the inner membrane potential. These results indicate that VLCFA disturbs the mitochondrial membrane potential causing ROS accumulation, oxidative stress, and cell death. We further tested whether NAC (500 µM) can prevent the mitochondria-specific effects of VLCFA in C26:0-treated oligodendrocytes. Our results demonstrate that NAC improves mtGSH levels and mitochondrial function in oligodendrocytes, indicating that it has potential use in the treatment of ALD and related disorders.
Collapse
Affiliation(s)
- Jie Zhou
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 2001 6th Street SE, Minneapolis, MN 55455, USA; (J.Z.); (M.R.T.); (J.C.C.)
| | - Marcia R. Terluk
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 2001 6th Street SE, Minneapolis, MN 55455, USA; (J.Z.); (M.R.T.); (J.C.C.)
| | - Paul J. Orchard
- Division of Pediatric Blood and Marrow Transplantation, Department of Pediatrics, Medical School, University of Minnesota, 425 East River Parkway, Minneapolis, MN 55455, USA;
| | - James C. Cloyd
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 2001 6th Street SE, Minneapolis, MN 55455, USA; (J.Z.); (M.R.T.); (J.C.C.)
| | - Reena V. Kartha
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, 2001 6th Street SE, Minneapolis, MN 55455, USA; (J.Z.); (M.R.T.); (J.C.C.)
- Correspondence: ; Tel.: +1-612-626-2436
| |
Collapse
|
13
|
Manor J, Chung H, Bhagwat PK, Wangler MF. ABCD1 and X-linked adrenoleukodystrophy: A disease with a markedly variable phenotype showing conserved neurobiology in animal models. J Neurosci Res 2021; 99:3170-3181. [PMID: 34716609 PMCID: PMC9665428 DOI: 10.1002/jnr.24953] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/30/2021] [Accepted: 08/15/2021] [Indexed: 12/12/2022]
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a phenotypically heterogeneous disorder involving defective peroxisomal β-oxidation of very long-chain fatty acids (VLCFAs), due to mutation in the ABCD1 gene. X-ALD is the most common peroxisomal inborn error of metabolism and confers a high degree of morbidity and mortality. Remarkably, a subset of patients exhibit a cerebral form with inflammatory invasion of the central nervous system and extensive demyelination, while in others only dying-back axonopathy or even isolated adrenal insufficiency is seen, without genotype-phenotype correlation. X-ALD's biochemical signature is marked elevation of VLCFAs in blood, a finding that has been utilized for massive newborn screening for early diagnosis. Investigational gene therapy approaches hold promises for improved outcomes. However, the pathophysiological mechanisms of the disease remain poorly understood, limiting investigation of targeted therapeutic options. Animal models for the disease recapitulate the biochemical signature of VLCFA accumulation and demonstrate mitochondrially generated reactive oxygen species, oxidative damage, increased glial death, and axonal damage. Most strikingly, however, cerebral invasion of leukocytes and demyelination were not observed in any animal model for X-ALD, reflecting upon pathological processes that are yet to be discovered. This review summarizes the current disease models in animals, the lessons learned from these models, and the gaps that remained to be filled in order to assist in therapeutic investigations for ALD.
Collapse
Affiliation(s)
- Joshua Manor
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
- Texas Children’s Hospital, Houston, Texas, USA
| | - Hyunglok Chung
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Pranjali K. Bhagwat
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| | - Michael F. Wangler
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, Texas, USA
| |
Collapse
|
14
|
Tieu JH, Sahasrabudhe SA, Orchard PJ, Cloyd JC, Kartha RV. Translational and clinical pharmacology considerations in drug repurposing for X-linked adrenoleukodystrophy-A rare peroxisomal disorder. Br J Clin Pharmacol 2021; 88:2552-2563. [PMID: 34558098 DOI: 10.1111/bcp.15090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/09/2021] [Accepted: 09/11/2021] [Indexed: 12/28/2022] Open
Abstract
X-linked adrenoleukodystrophy (X-ALD) is an inherited, neurodegenerative rare disease that can result in devastating symptoms of blindness, gait disturbances and spastic quadriparesis due to progressive demyelination. Typically, the disease progresses rapidly, causing death within the first decade of life. With limited treatments available, efforts to determine an effective therapy that can alter disease progression or mitigate symptoms have been undertaken for many years, particularly through drug repurposing. Repurposing has generally been guided through clinical experience and small trials. At this time, none of the drug candidates have been approved for use, which may be due, in part, to the lack of pharmacokinetic/pharmacodynamic information on the repurposed medications in the target patient population. Greater consideration for the disease pathophysiology, drug pharmacology and potential drug-target interactions, specifically at the site of action, would improve drug repurposing and facilitate drug development. Incorporating advanced translational and clinical pharmacological approaches in preclinical studies and early-stage clinical trials will improve the success of repurposed drugs for X-ALD as well as other rare diseases.
Collapse
Affiliation(s)
- Julianne H Tieu
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Siddhee A Sahasrabudhe
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Paul J Orchard
- Division of Pediatric Blood and Marrow Transplantation, Department of Pediatrics, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - James C Cloyd
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Reena V Kartha
- Center for Orphan Drug Research, Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
15
|
Peroxisomal ABC Transporters: An Update. Int J Mol Sci 2021; 22:ijms22116093. [PMID: 34198763 PMCID: PMC8201181 DOI: 10.3390/ijms22116093] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 12/12/2022] Open
Abstract
ATP-binding cassette (ABC) transporters constitute one of the largest superfamilies of conserved proteins from bacteria to mammals. In humans, three members of this family are expressed in the peroxisomal membrane and belong to the subfamily D: ABCD1 (ALDP), ABCD2 (ALDRP), and ABCD3 (PMP70). These half-transporters must dimerize to form a functional transporter, but they are thought to exist primarily as tetramers. They possess overlapping but specific substrate specificity, allowing the transport of various lipids into the peroxisomal matrix. The defects of ABCD1 and ABCD3 are responsible for two genetic disorders called X-linked adrenoleukodystrophy and congenital bile acid synthesis defect 5, respectively. In addition to their role in peroxisome metabolism, it has recently been proposed that peroxisomal ABC transporters participate in cell signaling and cell control, particularly in cancer. This review presents an overview of the knowledge on the structure, function, and mechanisms involving these proteins and their link to pathologies. We summarize the different in vitro and in vivo models existing across the species to study peroxisomal ABC transporters and the consequences of their defects. Finally, an overview of the known and possible interactome involving these proteins, which reveal putative and unexpected new functions, is shown and discussed.
Collapse
|
16
|
Ravi A, Palamiuc L, Loughran RM, Triscott J, Arora GK, Kumar A, Tieu V, Pauli C, Reist M, Lew RJ, Houlihan SL, Fellmann C, Metallo C, Rubin MA, Emerling BM. PI5P4Ks drive metabolic homeostasis through peroxisome-mitochondria interplay. Dev Cell 2021; 56:1661-1676.e10. [PMID: 33984270 DOI: 10.1016/j.devcel.2021.04.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/29/2021] [Accepted: 04/21/2021] [Indexed: 12/16/2022]
Abstract
PI5P4Ks are a class of phosphoinositide kinases that phosphorylate PI-5-P to PI-4,5-P2. Distinct localization of phosphoinositides is fundamental for a multitude of cellular functions. Here, we identify a role for peroxisomal PI-4,5-P2 generated by the PI5P4Ks in maintaining energy balance. We demonstrate that PI-4,5-P2 regulates peroxisomal fatty acid oxidation by mediating trafficking of lipid droplets to peroxisomes, which is essential for sustaining mitochondrial metabolism. Using fluorescent-tagged lipids and metabolite tracing, we show that loss of the PI5P4Ks significantly impairs lipid uptake and β-oxidation in the mitochondria. Further, loss of PI5P4Ks results in dramatic alterations in mitochondrial structural and functional integrity, which under nutrient deprivation is further exacerbated, causing cell death. Notably, inhibition of the PI5P4Ks in cancer cells and mouse tumor models leads to decreased cell viability and tumor growth, respectively. Together, these studies reveal an unexplored role for PI5P4Ks in preserving metabolic homeostasis, which is necessary for tumorigenesis.
Collapse
Affiliation(s)
- Archna Ravi
- Cell and Molecular Biology of Cancer Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Lavinia Palamiuc
- Cell and Molecular Biology of Cancer Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Ryan M Loughran
- Cell and Molecular Biology of Cancer Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Joanna Triscott
- Department of Biomedical Research and Bern Center for Precision Medicine, University of Bern and Inselspital Bern, Bern 3008, Switzerland
| | - Gurpreet K Arora
- Cell and Molecular Biology of Cancer Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Avi Kumar
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Vivian Tieu
- Cell and Molecular Biology of Cancer Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Chantal Pauli
- Institute of Pathology and Molecular Pathology, University Hospital Zürich and the University of Zurich (UZH), Zurich 8006, Switzerland
| | - Matthias Reist
- Department of Biomedical Research and Bern Center for Precision Medicine, University of Bern and Inselspital Bern, Bern 3008, Switzerland
| | - Rachel J Lew
- Gladstone Institutes, San Francisco, CA 94158, USA
| | - Shauna L Houlihan
- Cancer Biology and Genetics Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Christof Fellmann
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Cellular and Molecular Pharmacology, School of Medicine, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Christian Metallo
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Mark A Rubin
- Department of Biomedical Research and Bern Center for Precision Medicine, University of Bern and Inselspital Bern, Bern 3008, Switzerland
| | - Brooke M Emerling
- Cell and Molecular Biology of Cancer Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
17
|
Duncan GJ, Simkins TJ, Emery B. Neuron-Oligodendrocyte Interactions in the Structure and Integrity of Axons. Front Cell Dev Biol 2021; 9:653101. [PMID: 33763430 PMCID: PMC7982542 DOI: 10.3389/fcell.2021.653101] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/15/2021] [Indexed: 12/12/2022] Open
Abstract
The myelination of axons by oligodendrocytes is a highly complex cell-to-cell interaction. Oligodendrocytes and axons have a reciprocal signaling relationship in which oligodendrocytes receive cues from axons that direct their myelination, and oligodendrocytes subsequently shape axonal structure and conduction. Oligodendrocytes are necessary for the maturation of excitatory domains on the axon including nodes of Ranvier, help buffer potassium, and support neuronal energy metabolism. Disruption of the oligodendrocyte-axon unit in traumatic injuries, Alzheimer's disease and demyelinating diseases such as multiple sclerosis results in axonal dysfunction and can culminate in neurodegeneration. In this review, we discuss the mechanisms by which demyelination and loss of oligodendrocytes compromise axons. We highlight the intra-axonal cascades initiated by demyelination that can result in irreversible axonal damage. Both the restoration of oligodendrocyte myelination or neuroprotective therapies targeting these intra-axonal cascades are likely to have therapeutic potential in disorders in which oligodendrocyte support of axons is disrupted.
Collapse
Affiliation(s)
- Greg J. Duncan
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Tyrell J. Simkins
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- Vollum Institute, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, VA Portland Health Care System, Portland, OR, United States
| | - Ben Emery
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
18
|
X-linked adrenoleukodystrophy caused by a novel mutation presenting with various phenotypes in a Taiwanese family. Clin Chim Acta 2020; 514:100-106. [PMID: 33359056 DOI: 10.1016/j.cca.2020.12.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 11/13/2020] [Accepted: 12/14/2020] [Indexed: 11/23/2022]
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a peroxisomal disorder that primarily affects the white matter of central nervous system and the adrenal cortex. It is caused by mutations in the adenosine triphosphate-binding cassette, subfamily D, member 1 (ABCD1) gene that results in elevated plasma levels of very long chain fatty acids (VLCFAs). The disease is characterized by an unpredictable variation in phenotypic expressions, including childhood cerebral form (CCALD) and adrenomyeloneuropathy (AMN). Genetic analysis is a reliable method for the diagnosis of X-ALD. We reported a 46-year-old male admitted to Department of Neurology, Chang Gung Memorial Hospital with progressive paraparesis and Addison's disease, which was diagnosed when he was around 20-year-old. Plasma levels of VLCFA showed that his C26:0, C24:0/C22:0 and C26:0/C22:0 ratios were significantly elevated. A novel missense mutation (p.Arg163Cys) caused by the nucleotide change c.487C > T in exon 1 was identified in the ABCD1 gene of the proband and his subclinical family members. In this article, we reviewed the mutations that had been reported at the same position with different phenotypes. Given that the nerve conduction study (NCS) of the proband demonstrated a rare finding of demyelinating polyneuropathy with conduction blocks, we also reviewed the findings of NCS in patients with AMN in literature.
Collapse
|
19
|
Coppa A, Guha S, Fourcade S, Parameswaran J, Ruiz M, Moser AB, Schlüter A, Murphy MP, Lizcano JM, Miranda-Vizuete A, Dalfó E, Pujol A. The peroxisomal fatty acid transporter ABCD1/PMP-4 is required in the C. elegans hypodermis for axonal maintenance: A worm model for adrenoleukodystrophy. Free Radic Biol Med 2020; 152:797-809. [PMID: 32017990 PMCID: PMC7611262 DOI: 10.1016/j.freeradbiomed.2020.01.177] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/20/2020] [Accepted: 01/20/2020] [Indexed: 02/07/2023]
Abstract
Adrenoleukodystrophy is a neurometabolic disorder caused by a defective peroxisomal ABCD1 transporter of very long-chain fatty acids (VLCFAs). Its pathogenesis is incompletely understood. Here we characterize a nematode model of X-ALD with loss of the pmp-4 gene, the worm orthologue of ABCD1. These mutants recapitulate the hallmarks of X-ALD: i) VLCFAs accumulation and impaired mitochondrial redox homeostasis and ii) axonal damage coupled to locomotor dysfunction. Furthermore, we identify a novel role for PMP-4 in modulating lipid droplet dynamics. Importantly, we show that the mitochondria targeted antioxidant MitoQ normalizes lipid droplets size, and prevents axonal degeneration and locomotor disability, highlighting its therapeutic potential. Moreover, PMP-4 acting solely in the hypodermis rescues axonal and locomotion abnormalities, suggesting a myelin-like role for the hypodermis in providing essential peroxisomal functions for the nematode nervous system.
Collapse
Affiliation(s)
- Andrea Coppa
- Neurometabolic Diseases Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Spain
| | - Sanjib Guha
- Neurometabolic Diseases Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Spain
| | - Stéphane Fourcade
- Neurometabolic Diseases Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Spain; CIBERER U759, Center for Biomedical Research on Rare Diseases, Spain
| | - Janani Parameswaran
- Neurometabolic Diseases Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Spain; CIBERER U759, Center for Biomedical Research on Rare Diseases, Spain
| | - Montserrat Ruiz
- Neurometabolic Diseases Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Spain; CIBERER U759, Center for Biomedical Research on Rare Diseases, Spain
| | - Ann B Moser
- Peroxisomal Diseases Laboratory, Kennedy Krieger Institute, 707 N. Broadway, Baltimore, MD, 21205, USA
| | - Agatha Schlüter
- Neurometabolic Diseases Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Spain; CIBERER U759, Center for Biomedical Research on Rare Diseases, Spain
| | | | - Jose Miguel Lizcano
- Departament de Bioquímica i Biologia Molecular, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra (Barcelona), Spain
| | - Antonio Miranda-Vizuete
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío /CSIC/ Universidad de Sevilla, E-41013, Sevilla, Spain
| | - Esther Dalfó
- Departament de Bioquímica i Biologia Molecular, Institut de Neurociències, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra (Barcelona), Spain; Faculty of Medicine, University of Vic-Central University of Catalonia (UVic-UCC), 08500, Vic, Spain.
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Spain; CIBERER U759, Center for Biomedical Research on Rare Diseases, Spain; ICREA (Institució Catalana de Recerca i Estudis Avançats), Barcelona, Spain.
| |
Collapse
|
20
|
Abstract
Owing to their ability to efficiently generate ATP required to sustain normal cell function, mitochondria are often considered the 'powerhouses of the cell'. However, our understanding of the role of mitochondria in cell biology recently expanded when we recognized that they are key platforms for a plethora of cell signalling cascades. This functional versatility is tightly coupled to constant reshaping of the cellular mitochondrial network in a series of processes, collectively referred to as mitochondrial membrane dynamics and involving organelle fusion and fission (division) as well as ultrastructural remodelling of the membrane. Accordingly, mitochondrial dynamics influence and often orchestrate not only metabolism but also complex cell signalling events, such as those involved in regulating cell pluripotency, division, differentiation, senescence and death. Reciprocally, mitochondrial membrane dynamics are extensively regulated by post-translational modifications of its machinery and by the formation of membrane contact sites between mitochondria and other organelles, both of which have the capacity to integrate inputs from various pathways. Here, we discuss mitochondrial membrane dynamics and their regulation and describe how bioenergetics and cellular signalling are linked to these dynamic changes of mitochondrial morphology.
Collapse
|
21
|
Deori NM, Kale A, Maurya PK, Nagotu S. Peroxisomes: role in cellular ageing and age related disorders. Biogerontology 2018; 19:303-324. [PMID: 29968207 DOI: 10.1007/s10522-018-9761-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 06/27/2018] [Indexed: 12/12/2022]
Abstract
Peroxisomes are dynamic organelles essential for optimum functioning of a eukaryotic cell. Biogenesis of these organelles and the diverse functions performed by them have been extensively studied in the past decade. Their ability to perform functions depending on the cell type and growth conditions is unique and remarkable. Oxidation of fatty acids and reactive oxygen species metabolism are the two most important functions of these ubiquitous organelles. They are often referred to as both source and sink of reactive oxygen species in a cell. Recent research connects peroxisome dysfunction to fatal oxidative damage associated with ageing-related diseases/disorders. It is now widely accepted that mitochondria and peroxisomes are required to maintain oxidative balance in a cell. However, our understanding on the inter-dependence of these organelles to maintain cellular homeostasis of reactive oxygen species is still in its infancy. Herein, we summarize findings that highlight the role of peroxisomes in cellular reactive oxygen species metabolism, ageing and age-related disorders.
Collapse
Affiliation(s)
- Nayan M Deori
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Avinash Kale
- UM-DAE, Centre for Excellence in Basic Sciences, Health Centre, University of Mumbai, Mumbai, 400098, India
| | - Pawan K Maurya
- Interdisciplinary Laboratory for Clinical Neuroscience (LiNC), Universidade Federal de Sao Paulo-UNIFESP, Sao Paulo, Brazil
| | - Shirisha Nagotu
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
22
|
Dietary rescue of lipotoxicity-induced mitochondrial damage in Peroxin19 mutants. PLoS Biol 2018; 16:e2004893. [PMID: 29920513 PMCID: PMC6025876 DOI: 10.1371/journal.pbio.2004893] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 06/29/2018] [Accepted: 06/01/2018] [Indexed: 11/19/2022] Open
Abstract
Mutations in peroxin (PEX) genes lead to loss of peroxisomes, resulting in the formation of peroxisomal biogenesis disorders (PBDs) and early lethality. Studying PBDs and their animal models has greatly contributed to our current knowledge about peroxisomal functions. Very-long-chain fatty acid (VLCFA) accumulation has long been suggested as a major disease-mediating factor, although the exact pathological consequences are unclear. Here, we show that a Drosophila Pex19 mutant is lethal due to a deficit in medium-chain fatty acids (MCFAs). Increased lipolysis mediated by Lipase 3 (Lip3) leads to accumulation of free fatty acids and lipotoxicity. Administration of MCFAs prevents lipolysis and decreases the free fatty acid load. This drastically increases the survival rate of Pex19 mutants without reducing VLCFA accumulation. We identified a mediator of MCFA-induced lipolysis repression, the ceramide synthase Schlank, which reacts to MCFA supplementation by increasing its repressive action on lip3. This shifts our understanding of the key defects in peroxisome-deficient cells away from elevated VLCFA levels toward elevated lipolysis and shows that loss of this important organelle can be compensated by a dietary adjustment. Peroxisomes are organelles that contain several enzymes and fatty acids required for many metabolic tasks in the cell, and upon peroxisome loss, their educts accumulate. One example is the accumulation of very-long-chain fatty acids (VLCFAs) with a chain length of more than 20 carbons. These fatty acids cannot be oxidized in mitochondria but are exclusively degraded in peroxisomes. Lowering increased VLCFA levels is sometimes attempted as a treatment option for human disorders with peroxisomal dysfunction, although its effectiveness remains unclear. Here, we have analyzed this process in Drosophila melanogaster and found that peroxisomal loss results not only in VLCFA accumulation but also in a reduction of medium-chain fatty acids (MCFAs). We could show that this is due to a state of high lipolysis and increased mitochondrial activity. By supplementation with MCFAs from coconut oil, we were able to rescue mitochondrial damage and lethality observed in peroxisome-deficient flies. We found that this process is mediated by the ceramide synthase Schlank, which acts as a transcription factor and shuttles between nuclear membrane and endoplasmic reticulum (ER) in response to MCFA availability. We conclude that peroxisome loss triggers the accumulation of free fatty acids and mitochondrial damage in flies and that these effects can be rescued by a diet rich in MCFAs.
Collapse
|
23
|
Florea A, Varga AP, Matei HV. Ultrastructural variability of mitochondrial cristae induced in vitro by bee (Apis mellifera) venom and its derivatives, melittin and phospholipase A2, in isolated rat adrenocortical mitochondria. Micron 2018; 112:42-54. [PMID: 29908421 DOI: 10.1016/j.micron.2018.06.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/08/2018] [Accepted: 06/08/2018] [Indexed: 12/20/2022]
Abstract
We tested the ability of bee venom (BV), melittin (Mlt), and phospholipase A2 (PLA) - used in 5 concentrations each (5, 10, 15, 20 and 40 μg/100 μl) - to promote ultrastructural changes and reorganization of cristae in vitro in mitochondria isolated from rat adrenal cortex after a protocol optimized by us. Thus, apart from two control grups (CI and CS), in which the mitochondria were suspended into saline buffer and isolation medium respectively, 15 more groups of mitochondria were constituted, corresponding to the five different doses of the three substance tested (BV5 to M40; M5 to M40 and P5 to P40). The ultrastructural effects were quantified on transmission electron micrographs using a morphometry software. Values of 84.49 nm and 95.45 nm were calculated for median diameters of mitochondrial cristae in two control groups. Large and very large vesicular cristae, many with 2 or 3 membranes, were generated depending on dose among normal cristae in all treated groups. In the BV and Mlt treated groups, after an initial increase (up to 127.27 nm in V15 group and 151.2 nm in M10 group) due to stimulation of cristae fusion, the cristae diameter diminished as the doses increased, mainly by the collapse of the cristae. In the PLA treated groups, the cristae diameter increased continuously from 83.84 nm to 136.01 nm, by stimulated fusion of cristae, only the two largest doses promoting the collapse of cristae in some mitochondria. The highest percentage of abnormal cristae was found in the Mlt treated groups and next in BV treated groups. All substances tested produced pronounced ultrastructural variability of mitochondrial cristae in vitro: they also changed (depending on dose) mitochondrial shapes, generated matrix debris and the highest concentrations of BV and Mlt were responsible for mitochondrial breakdown. These ultrastructural alterations of mitochondrial criste in the presence of the BV molecules suggest a reduced capacity of adrenocortical mitochondria to synthetize steroid hormones consequently to BV envenomations and partially explain the toxic effects of the BV.
Collapse
Affiliation(s)
- Adrian Florea
- Department of Cell and Molecular Biology, Faculty of Medicine, "Iuliu Haţieganu" University of Medicine and Pharmacy, 6 L. Pasteur St., 400349 Cluj-Napoca, Romania.
| | - Andrei Patrick Varga
- Department of Cell and Molecular Biology, Faculty of Medicine, "Iuliu Haţieganu" University of Medicine and Pharmacy, 6 L. Pasteur St., 400349 Cluj-Napoca, Romania; Jibou County Hospital, 28 Libertatii St., 455200 Jibou, Romania.
| | - Horea Vladi Matei
- Department of Cell and Molecular Biology, Faculty of Medicine, "Iuliu Haţieganu" University of Medicine and Pharmacy, 6 L. Pasteur St., 400349 Cluj-Napoca, Romania.
| |
Collapse
|
24
|
Systematic mapping of contact sites reveals tethers and a function for the peroxisome-mitochondria contact. Nat Commun 2018; 9:1761. [PMID: 29720625 PMCID: PMC5932058 DOI: 10.1038/s41467-018-03957-8] [Citation(s) in RCA: 200] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 03/22/2018] [Indexed: 12/15/2022] Open
Abstract
The understanding that organelles are not floating in the cytosol, but rather held in an organized yet dynamic interplay through membrane contact sites, is altering the way we grasp cell biological phenomena. However, we still have not identified the entire repertoire of contact sites, their tethering molecules and functions. To systematically characterize contact sites and their tethering molecules here we employ a proximity detection method based on split fluorophores and discover four potential new yeast contact sites. We then focus on a little-studied yet highly disease-relevant contact, the Peroxisome-Mitochondria (PerMit) proximity, and uncover and characterize two tether proteins: Fzo1 and Pex34. We genetically expand the PerMit contact site and demonstrate a physiological function in β-oxidation of fatty acids. Our work showcases how systematic analysis of contact site machinery and functions can deepen our understanding of these structures in health and disease.
Collapse
|
25
|
Castro IG, Schuldiner M, Zalckvar E. Mind the Organelle Gap - Peroxisome Contact Sites in Disease. Trends Biochem Sci 2018; 43:199-210. [PMID: 29395653 PMCID: PMC6252078 DOI: 10.1016/j.tibs.2018.01.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 12/23/2017] [Accepted: 01/02/2018] [Indexed: 12/20/2022]
Abstract
The eukaryotic cell is organized as a complex grid system where membrane-bound cellular compartments, organelles, must be localized to the right place at the right time. One way to facilitate correct organelle localization and organelle cooperation is through membrane contact sites, areas of close proximity between two organelles that are bridged by protein/lipid complexes. It is now clear that all organelles physically contact each other. The main focus of this review is contact sites of peroxisomes, central metabolic hubs whose defects lead to a variety of diseases. New peroxisome contacts, their tethering complexes and functions have been recently discovered. However, if and how peroxisome contacts contribute to the development of peroxisome-related diseases is still a mystery.
Collapse
Affiliation(s)
- Inês Gomes Castro
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Maya Schuldiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel.
| | - Einat Zalckvar
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
26
|
Singh I, Samuvel DJ, Choi S, Saxena N, Singh AK, Won J. Combination therapy of lovastatin and AMP-activated protein kinase activator improves mitochondrial and peroxisomal functions and clinical disease in experimental autoimmune encephalomyelitis model. Immunology 2018; 154:434-451. [PMID: 29331024 DOI: 10.1111/imm.12893] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 12/29/2017] [Indexed: 01/04/2023] Open
Abstract
Recent studies report that loss and dysfunction of mitochondria and peroxisomes contribute to the myelin and axonal damage in multiple sclerosis (MS). In this study, we investigated the efficacy of a combination of lovastatin and AMP-activated protein kinase (AMPK) activator (AICAR) on the loss and dysfunction of mitochondria and peroxisomes and myelin and axonal damage in spinal cords, relative to the clinical disease symptoms, using a mouse model of experimental autoimmune encephalomyelitis (EAE, a model for MS). We observed that lovastatin and AICAR treatments individually provided partial protection of mitochondria/peroxisomes and myelin/axons, and therefore partial attenuation of clinical disease in EAE mice. However, treatment of EAE mice with the lovastatin and AICAR combination provided greater protection of mitochondria/peroxisomes and myelin/axons, and greater improvement in clinical disease compared with individual drug treatments. In spinal cords of EAE mice, lovastatin-mediated inhibition of RhoA and AICAR-mediated activation of AMPK cooperatively enhanced the expression of the transcription factors and regulators (e.g. PPARα/β, SIRT-1, NRF-1, and TFAM) required for biogenesis and the functions of mitochondria (e.g. OXPHOS, MnSOD) and peroxisomes (e.g. PMP70 and catalase). In summary, these studies document that oral medication with a combination of lovastatin and AICAR, which are individually known to have immunomodulatory effects, provides potent protection and repair of inflammation-induced loss and dysfunction of mitochondria and peroxisomes as well as myelin and axonal abnormalities in EAE. As statins are known to provide protection in progressive MS (Phase II study), these studies support that supplementation statin treatment with an AMPK activator may provide greater efficacy against MS.
Collapse
Affiliation(s)
- Inderjit Singh
- Charles P. Darby Children's Research Institute, Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA.,Research Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| | - Devadoss J Samuvel
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Seungho Choi
- Charles P. Darby Children's Research Institute, Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Nishant Saxena
- Charles P. Darby Children's Research Institute, Department of Pediatrics, Medical University of South Carolina, Charleston, SC, USA
| | - Avtar K Singh
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA.,Pathology and Laboratory Medicine Service, Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA
| | - Jeseong Won
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
27
|
Strachan LR, Stevenson TJ, Freshner B, Keefe MD, Miranda Bowles D, Bonkowsky JL. A zebrafish model of X-linked adrenoleukodystrophy recapitulates key disease features and demonstrates a developmental requirement for abcd1 in oligodendrocyte patterning and myelination. Hum Mol Genet 2018; 26:3600-3614. [PMID: 28911205 DOI: 10.1093/hmg/ddx249] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 06/26/2017] [Indexed: 01/02/2023] Open
Abstract
X-linked adrenoleukodystrophy (ALD) is a devastating inherited neurodegenerative disease caused by defects in the ABCD1 gene and affecting peripheral and central nervous system myelin. ABCD1 encodes a peroxisomal transmembrane protein required for very long chain fatty acid (VLCFA) metabolism. We show that zebrafish (Danio rerio) Abcd1 is highly conserved at the amino acid level with human ABCD1, and during development is expressed in homologous regions including the central nervous system and adrenal glands. We used TALENs to generate five zebrafish abcd1 mutant allele lines introducing premature stop codons in exon 1, as well as obtained an abcd1 allele from the Zebrafish Mutation Project carrying a point mutation in a splice donor site. Similar to patients with ALD, zebrafish abcd1 mutants have elevated VLCFA levels. Interestingly, we found that CNS development of the abcd1 mutants is disrupted, with hypomyelination in the spinal cord, abnormal patterning and decreased numbers of oligodendrocytes, and increased cell death. By day of life five abcd1 mutants demonstrate impaired motor function, and overall survival to adulthood of heterozygous and homozygous mutants is decreased. Expression of human ABCD1 in oligodendrocytes rescued apoptosis in the abcd1 mutant. In summary, we have established a zebrafish model of ALD that recapitulates key features of human disease pathology and which reveals novel features of underlying disease pathogenesis.
Collapse
Affiliation(s)
- Lauren R Strachan
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Tamara J Stevenson
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Briana Freshner
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Matthew D Keefe
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - D Miranda Bowles
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Joshua L Bonkowsky
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| |
Collapse
|
28
|
Zhang Y, Guo D, Tang Y. Autonomic dysfunction in a patient with X-linked adrenoleukodystrophy. Int J Neurosci 2017; 128:783-784. [PMID: 29284317 DOI: 10.1080/00207454.2017.1405953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
X-linked adrenoleukodystrophy is an inherited disease caused by abnormal accumulation of very long chain fatty acids. The diagnosis of X-linked adrenoleukodystrophy can be confirmed with the mutation of ABCD1 gene. The main symptom of the X-linked adrenoleukodystrophy is spastic paraparesis, and autonomic dysfunction is rare in X-linked adrenoleukodystrophy. Here, we presented an X-ALD case of a 46-year-old Asian male with severe autonomic dysfunction. Impairment of the autonomic nervous system may closely relate to mitochondrial defect.
Collapse
Affiliation(s)
- Yifan Zhang
- a Department of Neurology, Xuan Wu Hospital , Capital Medical University , Beijing , P. R. China
| | - Dongmei Guo
- a Department of Neurology, Xuan Wu Hospital , Capital Medical University , Beijing , P. R. China
| | - Yi Tang
- a Department of Neurology, Xuan Wu Hospital , Capital Medical University , Beijing , P. R. China
| |
Collapse
|
29
|
The Peroxisome-Mitochondria Connection: How and Why? Int J Mol Sci 2017; 18:ijms18061126. [PMID: 28538669 PMCID: PMC5485950 DOI: 10.3390/ijms18061126] [Citation(s) in RCA: 225] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 05/15/2017] [Accepted: 05/20/2017] [Indexed: 12/14/2022] Open
Abstract
Over the past decades, peroxisomes have emerged as key regulators in overall cellular lipid and reactive oxygen species metabolism. In mammals, these organelles have also been recognized as important hubs in redox-, lipid-, inflammatory-, and innate immune-signaling networks. To exert these activities, peroxisomes must interact both functionally and physically with other cell organelles. This review provides a comprehensive look of what is currently known about the interconnectivity between peroxisomes and mitochondria within mammalian cells. We first outline how peroxisomal and mitochondrial abundance are controlled by common sets of cis- and trans-acting factors. Next, we discuss how peroxisomes and mitochondria may communicate with each other at the molecular level. In addition, we reflect on how these organelles cooperate in various metabolic and signaling pathways. Finally, we address why peroxisomes and mitochondria have to maintain a healthy relationship and why defects in one organelle may cause dysfunction in the other. Gaining a better insight into these issues is pivotal to understanding how these organelles function in their environment, both in health and disease.
Collapse
|
30
|
Mitochondria-organelle contact sites: the plot thickens. Biochem Soc Trans 2017; 45:477-488. [PMID: 28408488 DOI: 10.1042/bst20160130] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 02/07/2017] [Accepted: 02/13/2017] [Indexed: 01/30/2023]
Abstract
Membrane contact sites (MCSs) are areas of close apposition between the membranes of two different organelles that enable non-vesicular transfer of ions and lipids. Recent studies reveal that mitochondria maintain contact sites with organelles other than the endoplasmic reticulum such as the vacuole, plasma membrane and peroxisomes. This review focuses on novel findings achieved mainly in yeast regarding tethers, function and regulation of mitochondria-organelle contact sites. The emerging network of MCSs linking virtually all cellular organelles is highly dynamic and integrated with cellular metabolism.
Collapse
|
31
|
Larsen PA, Lutz MW, Hunnicutt KE, Mihovilovic M, Saunders AM, Yoder AD, Roses AD. The Alu neurodegeneration hypothesis: A primate-specific mechanism for neuronal transcription noise, mitochondrial dysfunction, and manifestation of neurodegenerative disease. Alzheimers Dement 2017; 13:828-838. [PMID: 28242298 PMCID: PMC6647845 DOI: 10.1016/j.jalz.2017.01.017] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 01/12/2017] [Accepted: 01/24/2017] [Indexed: 01/13/2023]
Abstract
It is hypothesized that retrotransposons have played a fundamental role in primate evolution and that enhanced neurologic retrotransposon activity in humans may underlie the origin of higher cognitive function. As a potential consequence of this enhanced activity, it is likely that neurons are susceptible to deleterious retrotransposon pathways that can disrupt mitochondrial function. An example is observed in the TOMM40 gene, encoding a β-barrel protein critical for mitochondrial preprotein transport. Primate-specific Alu retrotransposons have repeatedly inserted into TOMM40 introns, and at least one variant associated with late-onset Alzheimer’s disease originated from an Alu insertion event. We provide evidence of enriched Alu content in mitochondrial genes and postulate that Alus can disrupt mitochondrial populations in neurons, thereby setting the stage for progressive neurologic dysfunction. This Alu neurodegeneration hypothesis is compatible with decades of research and offers a plausible mechanism for the disruption of neuronal mitochondrial homeostasis, ultimately cascading into neurodegenerative disease.
Collapse
Affiliation(s)
- Peter A Larsen
- Department of Biology, Duke University, Durham, NC, USA.
| | - Michael W Lutz
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA
| | | | - Mirta Mihovilovic
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA
| | - Ann M Saunders
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA
| | - Anne D Yoder
- Department of Biology, Duke University, Durham, NC, USA; Duke Lemur Center, Duke University, Durham, NC, USA
| | - Allen D Roses
- Department of Neurology, Duke University School of Medicine, Durham, NC, USA; Zinfandel Pharmaceuticals, Inc, Durham, NC, USA
| |
Collapse
|
32
|
Schönfeld P, Reiser G. Brain Lipotoxicity of Phytanic Acid and Very Long-chain Fatty Acids. Harmful Cellular/Mitochondrial Activities in Refsum Disease and X-Linked Adrenoleukodystrophy. Aging Dis 2016; 7:136-49. [PMID: 27114847 PMCID: PMC4809606 DOI: 10.14336/ad.2015.0823] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Accepted: 08/23/2015] [Indexed: 02/02/2023] Open
Abstract
It is increasingly understood that in the aging brain, especially in the case of patients suffering from neurodegenerative diseases, some fatty acids at pathologically high concentrations exert detrimental activities. To study such activities, we here analyze genetic diseases, which are due to compromised metabolism of specific fatty acids, either the branched-chain phytanic acid or very long-chain fatty acids (VLCFAs). Micromolar concentrations of phytanic acid or of VLCFAs disturb the integrity of neural cells by impairing Ca2+ homeostasis, enhancing oxidative stress or de-energizing mitochondria. Finally, these combined harmful activities accelerate cell death. Mitochondria are more severely targeted by phytanic acid than by VLCFAs. The insertion of VLCFAs into the inner membrane distorts the arrangement of membrane constituents and their functional interactions. Phytanic acid exerts specific protonophoric activity, induces reactive oxygen species (ROS) generation, and reduces ATP generation. A clear inhibition of the Na+, K+-ATPase activity by phytanic acid has also been reported. In addition to the instantaneous effects, a chronic exposure of brain cells to low micromolar concentrations of phytanic acid may produce neuronal damage in Refsum disease by altering epigenetic transcriptional regulation. Myelin-producing oligodendrocytes respond with particular sensitivity to VLCFAs. Deleterious activity of VLCFAs on energy-dependent mitochondrial functions declines with increasing the hydrocarbon chain length (C22:0 > C24:0 > C26:0). In contrast, the reverse sequence holds true for cell death induction by VLCFAs (C22:0 < C24:0 < C26:0). In adrenoleukodystrophy, the uptake of VLCFAs by peroxisomes is impaired by defects of the ABCD1 transporter. Studying mitochondria from ABCD1-deficient and wild-type mice proves that the energy-dependent functions are not altered in the disease model. Thus, a defective ABCD1 apparently exerts no obvious adaptive pressure on mitochondria. Further research has to elucidate the detailed mechanistic basis for the failures causing fatty acid-mediated neurodegeneration and should help to provide possible therapeutic interventions.
Collapse
Affiliation(s)
| | - Georg Reiser
- Institut für Neurobiochemie (Institut für Inflammation und Neurodegeneration), Medizinische Fakultät der Otto-von-Guericke-Universität Magdeburg, Magdeburg, Germany
| |
Collapse
|
33
|
Fourcade S, Ferrer I, Pujol A. Oxidative stress, mitochondrial and proteostasis malfunction in adrenoleukodystrophy: A paradigm for axonal degeneration. Free Radic Biol Med 2015; 88:18-29. [PMID: 26073123 DOI: 10.1016/j.freeradbiomed.2015.05.041] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/07/2015] [Accepted: 05/26/2015] [Indexed: 12/15/2022]
Abstract
Peroxisomal and mitochondrial malfunction, which are highly intertwined through redox regulation, in combination with defective proteostasis, are hallmarks of the most prevalent multifactorial neurodegenerative diseases-including Alzheimer's (AD) and Parkinson's disease (PD)-and of the aging process, and are also found in inherited conditions. Here we review the interplay between oxidative stress and axonal degeneration, taking as groundwork recent findings on pathomechanisms of the peroxisomal neurometabolic disease adrenoleukodystrophy (X-ALD). We explore the impact of chronic redox imbalance caused by the excess of very long-chain fatty acids (VLCFA) on mitochondrial respiration and biogenesis, and discuss how this impairs protein quality control mechanisms essential for neural cell survival, such as the proteasome and autophagy systems. As consequence, prime molecular targets in the pathogenetic cascade emerge, such as the SIRT1/PGC-1α axis of mitochondrial biogenesis, and the inhibitor of autophagy mTOR. Thus, we propose that mitochondria-targeted antioxidants; mitochondrial biogenesis boosters such as the antidiabetic pioglitazone and the SIRT1 ligand resveratrol; and the autophagy activator temsirolimus, a derivative of the mTOR inhibitor rapamycin, hold promise as disease-modifying therapies for X-ALD.
Collapse
Affiliation(s)
- Stéphane Fourcade
- Neurometabolic Diseases Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, 08908 Barcelona, Spain; Institut of Neuropathology, Pathologic Anatomy Service, Bellvitge Biomedical Research Institute, IDIBELL-Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, 08908 Barcelona, Spain; Center for Biomedical Research on Rare Diseases (CIBERER), U759, ISCIII, Spain.
| | - Isidre Ferrer
- Institut of Neuropathology, Pathologic Anatomy Service, Bellvitge Biomedical Research Institute, IDIBELL-Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, 08908 Barcelona, Spain; Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Spain
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospitalet de Llobregat, 08908 Barcelona, Spain; Institut of Neuropathology, Pathologic Anatomy Service, Bellvitge Biomedical Research Institute, IDIBELL-Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, 08908 Barcelona, Spain; Center for Biomedical Research on Rare Diseases (CIBERER), U759, ISCIII, Spain; Catalan Institution of Research and Advanced Studies (ICREA), Barcelona 08010, Catalonia, Spain.
| |
Collapse
|
34
|
No peroxisome is an island - Peroxisome contact sites. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:1061-9. [PMID: 26384874 DOI: 10.1016/j.bbamcr.2015.09.016] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/10/2015] [Accepted: 09/11/2015] [Indexed: 10/23/2022]
Abstract
In order to optimize their multiple cellular functions, peroxisomes must collaborate and communicate with the surrounding organelles. A common way of communication between organelles is through physical membrane contact sites where membranes of two organelles are tethered, facilitating exchange of small molecules and intracellular signaling. In addition contact sites are important for controlling processes such as metabolism, organelle trafficking, inheritance and division. How peroxisomes rely on contact sites for their various cellular activities is only recently starting to be appreciated and explored and the extent of peroxisomal communication, their contact sites and their functions are less characterized. In this review we summarize the identified peroxisomal contact sites, their tethering complexes and their potential physiological roles. Additionally, we highlight some of the preliminary evidence that exists in the field for unexplored peroxisomal contact sites.
Collapse
|
35
|
Schrader M, Costello J, Godinho LF, Islinger M. Peroxisome-mitochondria interplay and disease. J Inherit Metab Dis 2015; 38:681-702. [PMID: 25687155 DOI: 10.1007/s10545-015-9819-7] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 01/21/2015] [Accepted: 01/26/2015] [Indexed: 12/16/2022]
Abstract
Peroxisomes and mitochondria are ubiquitous, highly dynamic organelles with an oxidative type of metabolism in eukaryotic cells. Over the years, substantial evidence has been provided that peroxisomes and mitochondria exhibit a close functional interplay which impacts on human health and development. The so-called "peroxisome-mitochondria connection" includes metabolic cooperation in the degradation of fatty acids, a redox-sensitive relationship, an overlap in key components of the membrane fission machineries and cooperation in anti-viral signalling and defence. Furthermore, combined peroxisome-mitochondria disorders with defects in organelle division have been revealed. In this review, we present the latest progress in the emerging field of peroxisomal and mitochondrial interplay in mammals with a particular emphasis on cooperative fatty acid β-oxidation, redox interplay, organelle dynamics, cooperation in anti-viral signalling and the resulting implications for disease.
Collapse
Affiliation(s)
- Michael Schrader
- College of Life and Environmental Sciences, Biosciences, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter, EX4 4QD, UK,
| | | | | | | |
Collapse
|
36
|
Lismont C, Nordgren M, Van Veldhoven PP, Fransen M. Redox interplay between mitochondria and peroxisomes. Front Cell Dev Biol 2015; 3:35. [PMID: 26075204 PMCID: PMC4444963 DOI: 10.3389/fcell.2015.00035] [Citation(s) in RCA: 132] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/09/2015] [Indexed: 12/14/2022] Open
Abstract
Reduction-oxidation or “redox” reactions are an integral part of a broad range of cellular processes such as gene expression, energy metabolism, protein import and folding, and autophagy. As many of these processes are intimately linked with cell fate decisions, transient or chronic changes in cellular redox equilibrium are likely to contribute to the initiation and progression of a plethora of human diseases. Since a long time, it is known that mitochondria are major players in redox regulation and signaling. More recently, it has become clear that also peroxisomes have the capacity to impact redox-linked physiological processes. To serve this function, peroxisomes cooperate with other organelles, including mitochondria. This review provides a comprehensive picture of what is currently known about the redox interplay between mitochondria and peroxisomes in mammals. We first outline the pro- and antioxidant systems of both organelles and how they may function as redox signaling nodes. Next, we critically review and discuss emerging evidence that peroxisomes and mitochondria share an intricate redox-sensitive relationship and cooperate in cell fate decisions. Key issues include possible physiological roles, messengers, and mechanisms. We also provide examples of how data mining of publicly-available datasets from “omics” technologies can be a powerful means to gain additional insights into potential redox signaling pathways between peroxisomes and mitochondria. Finally, we highlight the need for more studies that seek to clarify the mechanisms of how mitochondria may act as dynamic receivers, integrators, and transmitters of peroxisome-derived mediators of oxidative stress. The outcome of such studies may open up exciting new avenues for the community of researchers working on cellular responses to organelle-derived oxidative stress, a research field in which the role of peroxisomes is currently highly underestimated and an issue of discussion.
Collapse
Affiliation(s)
- Celien Lismont
- Laboratory of Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, KU Leuven - University of Leuven Leuven, Belgium
| | - Marcus Nordgren
- Laboratory of Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, KU Leuven - University of Leuven Leuven, Belgium
| | - Paul P Van Veldhoven
- Laboratory of Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, KU Leuven - University of Leuven Leuven, Belgium
| | - Marc Fransen
- Laboratory of Lipid Biochemistry and Protein Interactions, Department of Cellular and Molecular Medicine, KU Leuven - University of Leuven Leuven, Belgium
| |
Collapse
|
37
|
Kruska N, Schönfeld P, Pujol A, Reiser G. Astrocytes and mitochondria from adrenoleukodystrophy protein (ABCD1)-deficient mice reveal that the adrenoleukodystrophy-associated very long-chain fatty acids target several cellular energy-dependent functions. Biochim Biophys Acta Mol Basis Dis 2015; 1852:925-36. [PMID: 25583114 DOI: 10.1016/j.bbadis.2015.01.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 12/03/2014] [Accepted: 01/05/2015] [Indexed: 12/21/2022]
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a severe neurodegenerative disorder resulting from defective ABCD1 transport protein. ABCD1 mediates peroxisomal uptake of free very-long-chain fatty acids (VLCFA) as well as their CoA-esters. Consequently, VLCFA accumulate in patients' plasma and tissues, which is considered as pathogenic X-ALD triggering factor. Clinical symptoms are mostly manifested in neural tissues and adrenal gland. Here, we investigate astrocytes from wild-type control and a genetic X-ALD mouse model (Abcd1-knockout), exposed to supraphysiological VLCFA (C22:0, C24:0 and C26:0) concentrations. They exhibit multiple impairments of energy metabolism. Furthermore, brain mitochondria from Abcd1(-/-) mice and wild-type control respond similarly to VLCFA with increased ROS generation, impaired oxidative ATP synthesis and diminished Ca(2+) uptake capacity, suggesting that a defective ABCD1 exerts no adaptive pressure on mitochondria. In contrast, astrocytes from Abcd1(-/-) mice respond more sensitively to VLCFA than wild-type control astrocytes. Moreover, long-term application of VLCFA induces high ROS generation, and strong in situ depolarization of mitochondria, and, in Abcd1(-/-) astrocytes, severely diminishes the capability to revert oxidized pyridine nucleotides to NAD(P)H. In addition, observed differences in responses of mitochondria and astrocytes to the hydrocarbon chain length of VLCFA suggest that detrimental VLCFA activities in astrocytes involve defective cellular functions other than mitochondria. In summary, we clearly demonstrate that VLCFA increase the vulnerability of Abcd1(-/-) astrocytes.
Collapse
Affiliation(s)
- Nicol Kruska
- Institut für Neurobiochemie, Medizinische Fakultät, Otto-von-Guericke-Universität Magdeburg, Leipziger Straße 44, D-39120 Magdeburg, Germany
| | - Peter Schönfeld
- Institut für Biochemie und Zellbiologie, Medizinische Fakultät, Otto-von-Guericke-Universität Magdeburg, Leipziger Straße 44, D-39120 Magdeburg, Germany
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory, ICREA and Institut d'Investigació Biomedica de Bellvitge (IDIBELL), Hospitalet de Llobregat 08908 Barcelona, Spain; Center for Biomedical Research in Rare Diseases (CIBERER), ISCIII Madrid, Spain
| | - Georg Reiser
- Institut für Neurobiochemie, Medizinische Fakultät, Otto-von-Guericke-Universität Magdeburg, Leipziger Straße 44, D-39120 Magdeburg, Germany.
| |
Collapse
|
38
|
Abcd2 is a strong modifier of the metabolic impairments in peritoneal macrophages of ABCD1-deficient mice. PLoS One 2014; 9:e108655. [PMID: 25255441 PMCID: PMC4177892 DOI: 10.1371/journal.pone.0108655] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 08/25/2014] [Indexed: 02/08/2023] Open
Abstract
The inherited peroxisomal disorder X-linked adrenoleukodystrophy (X-ALD), associated with neurodegeneration and inflammatory cerebral demyelination, is caused by mutations in the ABCD1 gene encoding the peroxisomal ATP-binding cassette (ABC) transporter ABCD1 (ALDP). ABCD1 transports CoA-esters of very long-chain fatty acids (VLCFA) into peroxisomes for degradation by β-oxidation; thus, ABCD1 deficiency results in VLCFA accumulation. The closest homologue, ABCD2 (ALDRP), when overexpressed, compensates for ABCD1 deficiency in X-ALD fibroblasts and in Abcd1-deficient mice. Microglia/macrophages have emerged as important players in the progression of neuroinflammation. Human monocytes, lacking significant expression of ABCD2, display severely impaired VLCFA metabolism in X-ALD. Here, we used thioglycollate-elicited primary mouse peritoneal macrophages (MPMΦ) from Abcd1 and Abcd2 single- and double-deficient mice to establish how these mutations affect VLCFA metabolism. By quantitative RT-PCR, Abcd2 mRNA was about half as abundant as Abcd1 mRNA in wild-type and similarly abundant in Abcd1-deficient MPMΦ. VLCFA (C26∶0) accumulated about twofold in Abcd1-deficient MPMΦ compared with wild-type controls, as measured by gas chromatography-mass spectrometry. In Abcd2-deficient macrophages VLCFA levels were normal. However, upon Abcd1/Abcd2 double-deficiency, VLCFA accumulation was markedly increased (sixfold) compared with Abcd1-deficient MPMΦ. Elovl1 mRNA, encoding the rate-limiting enzyme for elongation of VLCFA, was equally abundant across all genotypes. Peroxisomal β-oxidation of C26∶0 amounted to 62% of wild-type activity in Abcd1-deficient MPMΦ and was significantly more impaired (29% residual activity) upon Abcd1/Abcd2 double-deficiency. Single Abcd2 deficiency did not significantly compromise β-oxidation of C26∶0. Thus, the striking accumulation of VLCFA in double-deficient MPMΦ compared with single Abcd1 deficiency was due to the loss of ABCD2-mediated, compensatory transport of VLCFA into peroxisomes. We propose that moderate endogenous expression of Abcd2 in Abcd1-deficient murine macrophages prevents the severe metabolic phenotype observed in human X-ALD monocytes, which lack appreciable expression of ABCD2. This supports upregulation of ABCD2 as a therapeutic concept in X-ALD.
Collapse
|
39
|
Mohanty A, McBride HM. Emerging roles of mitochondria in the evolution, biogenesis, and function of peroxisomes. Front Physiol 2013; 4:268. [PMID: 24133452 PMCID: PMC3783979 DOI: 10.3389/fphys.2013.00268] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 09/10/2013] [Indexed: 12/19/2022] Open
Abstract
In the last century peroxisomes were thought to have an endosymbiotic origin. Along with mitochondria and chloroplasts, peroxisomes primarily regulate their numbers through the growth and division of pre-existing organelles, and they house specific machinery for protein import. These features were considered unique to endosymbiotic organelles, prompting the idea that peroxisomes were key cellular elements that helped facilitate the evolution of multicellular organisms. The functional similarities to mitochondria within mammalian systems expanded these ideas, as both organelles scavenge peroxide and reactive oxygen species, both organelles oxidize fatty acids, and at least in higher eukaryotes, the biogenesis of both organelles is controlled by common nuclear transcription factors of the PPAR family. Over the last decade it has been demonstrated that the fission machinery of both organelles is also shared, and that both organelles act as critical signaling platforms for innate immunity and other pathways. Taken together it is clear that the mitochondria and peroxisomes are functionally coupled, regulating cellular metabolism and signaling through a number of common mechanisms. However, recent work has focused primarily on the role of the ER in the biogenesis of peroxisomes, potentially overshadowing the critical importance of the mitochondria as a functional partner. In this review, we explore the mechanisms of functional coupling of the peroxisomes to the mitochondria/ER networks, providing some new perspectives on the potential contribution of the mitochondria to peroxisomal biogenesis.
Collapse
Affiliation(s)
- Abhishek Mohanty
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University Montreal, QC, Canada
| | | |
Collapse
|
40
|
Mitochondrial dysfunction and oxidative damage cooperatively fuel axonal degeneration in X-linked adrenoleukodystrophy. Biochimie 2013; 98:143-9. [PMID: 24076127 DOI: 10.1016/j.biochi.2013.09.012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Accepted: 09/14/2013] [Indexed: 12/18/2022]
Abstract
X-linked adrenoleukodystrophy (X-ALD) is the most frequent inherited monogenic demyelinating disease (minimal incidence 1:17,000). It is often lethal and currently lacks a satisfactory therapy. The disease is caused by loss of function of the ABCD1 gene, a peroxisomal ATP-binding cassette transporter, resulting in the accumulation of VLCFA (very long-chain fatty acids) in organs and plasma. Understanding of the aetiopathogenesis is a prerequisite for the development of novel therapeutic strategies. Functional genomics analysis of an ABCD1 null mouse, a mouse model for adrenomyeloneuropathy, has revealed presymptomatic alterations in several metabolic pathways converging on redox and bioenergetic homeostasis, with failure of mitochondrial OXPHOS disruption and mitochondrial depletion. These defects could be major contributors to the neurodegenerative cascade, as has been reported in several neurodegenerative disorders. Drugs targeting the redox imbalance/mitochondria dysfunction interplay have shown efficacy at halting axonal degeneration and associated disability in the mouse, and thus offer therapeutic hope.
Collapse
|
41
|
Wiesinger C, Kunze M, Regelsberger G, Forss-Petter S, Berger J. Impaired very long-chain acyl-CoA β-oxidation in human X-linked adrenoleukodystrophy fibroblasts is a direct consequence of ABCD1 transporter dysfunction. J Biol Chem 2013; 288:19269-79. [PMID: 23671276 DOI: 10.1074/jbc.m112.445445] [Citation(s) in RCA: 110] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
X-linked adrenoleukodystrophy (X-ALD), an inherited peroxisomal disorder, is caused by mutations in the ABCD1 gene encoding the peroxisomal ATP-binding cassette (ABC) transporter ABCD1 (adrenoleukodystrophy protein, ALDP). Biochemically, X-ALD is characterized by an accumulation of very long-chain fatty acids and partially impaired peroxisomal β-oxidation. In this study, we used primary human fibroblasts from X-ALD and Zellweger syndrome patients to investigate the peroxisomal β-oxidation defect. Our results show that the degradation of C26:0-CoA esters is as severely impaired as degradation of unesterified very long-chain fatty acids in X-ALD and is abolished in Zellweger syndrome. Interestingly, the β-oxidation rates for both C26:0-CoA and C22:0-CoA were similarly affected, although C22:0 does not accumulate in patient fibroblasts. Furthermore, we show that the β-oxidation defect in X-ALD is directly caused by ABCD1 dysfunction as blocking ABCD1 function with a specific antibody reduced β-oxidation to levels observed in X-ALD fibroblasts. By quantification of mRNA and protein levels of the peroxisomal ABC transporters and by blocking with specific antibodies, we found that residual β-oxidation activity toward C26:0-CoA in X-ALD fibroblasts is mediated by ABCD3, although the efficacy of ABCD3 appeared to be much lower than that of ABCD1. Finally, using isolated peroxisomes, we show that β-oxidation of C26:0-CoA is independent of additional CoA but requires a cytosolic factor of >10-kDa molecular mass that is resistant to N-ethylmaleimide and heat inactivation. In conclusion, our findings in human cells suggest that, in contrast to yeast cells, very long-chain acyl-CoA esters are transported into peroxisomes by ABCD1 independently of additional synthetase activity.
Collapse
Affiliation(s)
- Christoph Wiesinger
- Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| | | | | | | | | |
Collapse
|
42
|
López-Erauskin J, Galino J, Ruiz M, Cuezva JM, Fabregat I, Cacabelos D, Boada J, Martínez J, Ferrer I, Pamplona R, Villarroya F, Portero-Otín M, Fourcade S, Pujol A. Impaired mitochondrial oxidative phosphorylation in the peroxisomal disease X-linked adrenoleukodystrophy. Hum Mol Genet 2013; 22:3296-305. [PMID: 23604518 DOI: 10.1093/hmg/ddt186] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
X-linked adrenoleukodystrophy (X-ALD) is an inherited metabolic disorder of the nervous system characterized by axonopathy in spinal cords and/or cerebral demyelination, adrenal insufficiency and accumulation of very long-chain fatty acids (VLCFAs) in plasma and tissues. The disease is caused by malfunction of the ABCD1 gene, which encodes a peroxisomal transporter of VLCFAs or VLCFA-CoA. In the mouse, Abcd1 loss causes late onset axonal degeneration in the spinal cord, associated with locomotor disability resembling the most common phenotype in patients, adrenomyeloneuropathy. We have formerly shown that an excess of the VLCFA C26:0 induces oxidative damage, which underlies the axonal degeneration exhibited by the Abcd1(-) mice. In the present study, we sought to investigate the noxious effects of C26:0 on mitochondria function. Our data indicate that in X-ALD patients' fibroblasts, excess of C26:0 generates mtDNA oxidation and specifically impairs oxidative phosphorylation (OXPHOS) triggering mitochondrial ROS production from electron transport chain complexes. This correlates with impaired complex V phosphorylative activity, as visualized by high-resolution respirometry on spinal cord slices of Abcd1(-) mice. Further, we identified a marked oxidation of key OXPHOS system subunits in Abcd1(-) mouse spinal cords at presymptomatic stages. Altogether, our results illustrate some of the mechanistic intricacies by which the excess of a fatty acid targeted to peroxisomes activates a deleterious process of oxidative damage to mitochondria, leading to a multifaceted dysfunction of this organelle. These findings may be of relevance for patient management while unveiling novel therapeutic targets for X-ALD.
Collapse
Affiliation(s)
- J López-Erauskin
- Neurometabolic Diseases Laboratory, Institut d’Investigació Biomèdica de Bellvitge IDIBELL, L’Hospitalet de Llobregat, Barcelona, Catalonia, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Brose RD, Shin G, McGuinness MC, Schneidereith T, Purvis S, Dong GX, Keefer J, Spencer F, Smith KD. Activation of the stress proteome as a mechanism for small molecule therapeutics. Hum Mol Genet 2012; 21:4237-52. [PMID: 22752410 DOI: 10.1093/hmg/dds247] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Various small molecule pharmacologic agents with different known functions produce similar outcomes in diverse Mendelian and complex disorders, suggesting that they may induce common cellular effects. These molecules include histone deacetylase inhibitors, 4-phenylbutyrate (4PBA) and trichostatin A, and two small molecules without direct histone deacetylase inhibitor activity, hydroxyurea (HU) and sulforaphane. In some cases, the therapeutic effects of histone deacetylase inhibitors have been attributed to an increase in expression of genes related to the disease-causing gene. However, here we show that the pharmacological induction of mitochondrial biogenesis was necessary for the potentially therapeutic effects of 4PBA or HU in two distinct disease models, X-linked adrenoleukodystrophy and sickle cell disease. We hypothesized that a common cellular response to these four molecules is induction of mitochondrial biogenesis and peroxisome proliferation and activation of the stress proteome, or adaptive cell survival response. Treatment of human fibroblasts with these four agents induced mitochondrial and peroxisomal biogenesis as monitored by flow cytometry, immunofluorescence and/or western analyses. In treated normal human fibroblasts, all four agents induced the adaptive cell survival response: heat shock, unfolded protein, autophagic and antioxidant responses and the c-jun N-terminal kinase pathway, at the transcriptional and translational levels. Thus, activation of the evolutionarily conserved stress proteome and mitochondrial biogenesis may be a common cellular response to such small molecule therapy and a common basis of therapeutic action in various diseases. Modulation of this novel therapeutic target could broaden the range of treatable diseases without directly targeting the causative genetic abnormalities.
Collapse
Affiliation(s)
- Rebecca Deering Brose
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Brose RD, Avramopoulos D, Smith KD. SOD2 as a potential modifier of X-linked adrenoleukodystrophy clinical phenotypes. J Neurol 2012; 259:1440-7. [PMID: 22218650 DOI: 10.1007/s00415-011-6371-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 11/14/2011] [Accepted: 12/10/2011] [Indexed: 01/07/2023]
Abstract
X-linked adrenoleukodystrophy (XALD), a neurological disorder caused by mutations in the peroxisomal membrane protein gene ABCD1, presents as a rapidly progressing, inflammatory cerebral demyelination (cerebral cases) or a slowly progressing, distal axonopathy (non-cerebral cases). Specific ABCD1 defects do not explain this significant phenotypic variation. Patients have increased plasma and tissue very long chain fatty acid levels and increased cellular oxidative stress and oxidative damage. Superoxide dismutase 2 (SOD2), at candidate modifier locus 6q25.3, detoxifies superoxide radicals protecting against oxidative stress and damage. We tested an SOD2 variant C47T (Ala16Val) associated with reduced enzymatic activity as a potential modifier gene of cerebral demyelinating disease by comparing 117 cerebral XALD cases with 105 non-cerebral XALD cases. The hypoactive valine allele of the variant was associated with cerebral disease under a dominant model in the full data set (p = 0.04; ORT* = 1.90, 95% CI 1.01-3.56) and the non-childhood cerebral disease subset (p = 0.03; ORT* = 2.47, 95% CI 1.08-5.61). Three tag SNPs were genotyped to test for additional SNP or haplotype associations. A common haplotype, GTAC, which included the SOD2 valine allele, was associated with cerebral disease in the full data set (p = 0.03; OR = 1.75, 95% CI 1.11-2.75) and the non-childhood cerebral disease subset (p = 0.008; OR = 2.20, 95% CI 1.27-3.83). There was no association between childhood cerebral XALD and the C47T variant or the GTAC haplotype. Thus, reduced SOD2 activity may contribute to the development of cerebral demyelination in adolescent and adult XALD patients.
Collapse
Affiliation(s)
- Rebecca Deering Brose
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD, USA
| | | | | |
Collapse
|
45
|
Ahlemeyer B, Gottwald M, Baumgart-Vogt E. Deletion of a single allele of the Pex11β gene is sufficient to cause oxidative stress, delayed differentiation and neuronal death in mouse brain. Dis Model Mech 2011; 5:125-40. [PMID: 21954064 PMCID: PMC3255551 DOI: 10.1242/dmm.007708] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Impaired neuronal migration and cell death are commonly observed in patients with peroxisomal biogenesis disorders (PBDs), and in mouse models of this diseases. In Pex11β-deficient mice, we observed that the deletion of a single allele of the Pex11β gene (Pex11β+/− heterozygous mice) caused cell death in primary neuronal cultures prepared from the neocortex and cerebellum, although to a lesser extent as compared with the homozygous-null animals (Pex11β−/− mice). In corresponding brain sections, cell death was rare, but differences between the genotypes were similar to those found in vitro. Because PEX11β has been implicated in peroxisomal proliferation, we searched for alterations in peroxisomal abundance in the brain of heterozygous and homozygous Pex11β-null mice compared with wild-type animals. Deletion of one allele of the Pex11β gene slightly increased the abundance of peroxisomes, whereas the deletion of both alleles caused a 30% reduction in peroxisome number. The size of the peroxisomal compartment did not correlate with neuronal death. Similar to cell death, neuronal development was delayed in Pex11β+/− mice, and to a further extent in Pex11β−/− mice, as measured by a reduced mRNA and protein level of synaptophysin and a reduced protein level of the mature isoform of MAP2. Moreover, a gradual increase in oxidative stress was found in brain sections and primary neuronal cultures from wild-type to heterozygous to homozygous Pex11β-deficient mice. SOD2 was upregulated in neurons from Pex11β+/− mice, but not from Pex11β−/− animals, whereas the level of catalase remained unchanged in neurons from Pex11β+/− mice and was reduced in those from Pex11β−/− mice, suggesting a partial compensation of oxidative stress in the heterozygotes, but a failure thereof in the homozygous Pex11β−/− brain. In conclusion, we report the alterations in the brain caused by the deletion of a single allele of the Pex11β gene. Our data might lead to the reconsideration of the clinical treatment of PBDs and the common way of using knockout mouse models for studying autosomal recessive diseases.
Collapse
Affiliation(s)
- Barbara Ahlemeyer
- Institute for Anatomy and Cell Biology II, Division of Medical Cell Biology, University of Giessen, 35385 Giessen, Germany.
| | | | | |
Collapse
|
46
|
Miller WL, Auchus RJ. The molecular biology, biochemistry, and physiology of human steroidogenesis and its disorders. Endocr Rev 2011; 32:81-151. [PMID: 21051590 PMCID: PMC3365799 DOI: 10.1210/er.2010-0013] [Citation(s) in RCA: 1531] [Impact Index Per Article: 109.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Accepted: 08/20/2010] [Indexed: 02/08/2023]
Abstract
Steroidogenesis entails processes by which cholesterol is converted to biologically active steroid hormones. Whereas most endocrine texts discuss adrenal, ovarian, testicular, placental, and other steroidogenic processes in a gland-specific fashion, steroidogenesis is better understood as a single process that is repeated in each gland with cell-type-specific variations on a single theme. Thus, understanding steroidogenesis is rooted in an understanding of the biochemistry of the various steroidogenic enzymes and cofactors and the genes that encode them. The first and rate-limiting step in steroidogenesis is the conversion of cholesterol to pregnenolone by a single enzyme, P450scc (CYP11A1), but this enzymatically complex step is subject to multiple regulatory mechanisms, yielding finely tuned quantitative regulation. Qualitative regulation determining the type of steroid to be produced is mediated by many enzymes and cofactors. Steroidogenic enzymes fall into two groups: cytochrome P450 enzymes and hydroxysteroid dehydrogenases. A cytochrome P450 may be either type 1 (in mitochondria) or type 2 (in endoplasmic reticulum), and a hydroxysteroid dehydrogenase may belong to either the aldo-keto reductase or short-chain dehydrogenase/reductase families. The activities of these enzymes are modulated by posttranslational modifications and by cofactors, especially electron-donating redox partners. The elucidation of the precise roles of these various enzymes and cofactors has been greatly facilitated by identifying the genetic bases of rare disorders of steroidogenesis. Some enzymes not principally involved in steroidogenesis may also catalyze extraglandular steroidogenesis, modulating the phenotype expected to result from some mutations. Understanding steroidogenesis is of fundamental importance to understanding disorders of sexual differentiation, reproduction, fertility, hypertension, obesity, and physiological homeostasis.
Collapse
Affiliation(s)
- Walter L Miller
- Distinguished Professor of Pediatrics, University of California San Francisco, San Francisco, California 94143-0978, USA.
| | | |
Collapse
|
47
|
Florea A, Crăciun C. Abnormal mitochondrial cristae were experimentally generated by high doses of Apis mellifera venom in the rat adrenal cortex. Micron 2010; 42:434-42. [PMID: 21247771 DOI: 10.1016/j.micron.2010.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2010] [Revised: 12/17/2010] [Accepted: 12/21/2010] [Indexed: 11/29/2022]
Abstract
In the present study, Apis mellifera venom (AmV) was tested for its ability to cause ultrastructural changes in mitochondria of rat adrenal cortex in vivo. In order to achieve this goal, different AmV treatments were performed and the effects were quantified on transmission electron micrographs. In a first experimental group, AmV injected for 30 days in low daily doses (700 μg/kg) generated important ultrastructural changes in zona fasciculata. The mitochondrial ultrastructure was not affected, but the diameters of mitochondrial cristae (MC) were reduced (57.066 ± 7.795 nm) as compared to the MC diameters in the corresponding control groups (58.596 ± 6.603 nm, and 58.503 ± 5.708 nm). In adrenal glands collected from rats injected with a single, high dose of AmV (62 mg/kg), many ultrastructural changes were described. Mitochondria with normal, tubular MC (with diameter of 58.711 ± 5.907 nm) were observed in many cells, very close to the values calculated for the corresponding control group (58.639 ± 6.117 nm). However, the striking data reported herein concerned the ability of AmV high doses to promote dramatic alterations in the ultrastructure of these particular mitochondria, similar to those described in certain severe diseases. Thus, several types of abnormal mitochondria were observed, including mitochondria displaying lamellar and/or circular, concentric cristae and mitochondria devoid of cristae. The abnormal circular, concentric MC, with large inner (281.904 ± 158.588 nm) and outer (432.076 ± 230.372 nm) diameters, appeared to be the most stable form of MC in the adrenal cortex after the acute treatment with AmV. Among other ultrastructural aspects, these important changes indicated a high level of cytotoxicity of AmV in adrenocortical cells following in vivo experimental poisoning.
Collapse
Affiliation(s)
- Adrian Florea
- Laboratory of Electron Microscopy, Department of Cell and Molecular Biology, Iuliu Haţieganu University of Medicine and Pharmacy, 6 Pasteur St., 400349 Cluj-Napoca, Romania.
| | | |
Collapse
|
48
|
Ferrer I, Aubourg P, Pujol A. General aspects and neuropathology of X-linked adrenoleukodystrophy. Brain Pathol 2010; 20:817-30. [PMID: 20626743 DOI: 10.1111/j.1750-3639.2010.00390.x] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
X-adrenoleukodystrophy (X-ALD) is a metabolic, peroxisomal disease affecting the nervous system, adrenal cortex and testis resulting from inactivating mutations in ABCD1 gene which encodes a peroxisomal membrane half-adenosine triphosphate (ATP)-binding cassette transporter, ABCD1 (or ALDP), whose defect is associated with impaired peroxisomal beta-oxidation and accumulation of saturated very long-chain fatty acids (VLCFA) in tissues and body fluids. Several phenotypes are recognized in male patients including cerebral ALD in childhood, adolescence or adulthood, adrenomyeloneuropathy (AMN), Addison's disease and, eventually, gonadal insufficiency. Female carriers might present with mild to severe myeloneuropathy that resembles AMN. There is a lack of phenotype-genotype correlations, as the same ABCD1 gene mutation may be associated with different phenotypes in the same family, suggesting that genetic, epigenetic, environmental and stochastic factors are probably contributory to the development and course of the disease. Degenerative changes, like those seen in pure AMN without cerebral demyelination, are characterized by loss of axons and secondary myelin in the long tracts of the spinal cord, possibly related to the impaired lipid metabolism of VLCFAs and the associated alterations (ie, oxidative damage). Similar lesions are encountered following inactivation of ABCD1 in mice (ABCD1(-)). A different and more aggressive phenotype is secondary to cerebral demyelination, very often accompanied by inflammatory changes in the white matter of the brain and associated with activation of T lymphocytes, CD1 presentation and increased levels of cytokines, gamma-interferon, interleukin (IL)-1alpha, IL-2 and IL-6, Granulocyte macrophage colony-stimulating factor (GM-CSF), tumor necrosis factor-alpha, chemokines and chemokine receptors.
Collapse
Affiliation(s)
- Isidro Ferrer
- Institut Neuropatologia, Servei Anatomia Patològica, Institut d'Investigació Biomèdica de Bellvitge IDIBELL-Hospital Universitari de Bellvitge, Hospitalet de Llobregat, CIBERNED, Spain.
| | | | | |
Collapse
|
49
|
Berger J, Pujol A, Aubourg P, Forss-Petter S. Current and future pharmacological treatment strategies in X-linked adrenoleukodystrophy. Brain Pathol 2010; 20:845-56. [PMID: 20626746 DOI: 10.1111/j.1750-3639.2010.00393.x] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Mutations in the ABCD1 gene cause the clinical spectrum of the neurometabolic disorder X-linked adrenoleukodystrophy/adrenomyeloneuropathy (X-ALD/AMN). Currently, the most efficient therapeutic opportunity for patients with the cerebral form of X-ALD is hematopoietic stem cell transplantation and possibly gene therapy of autologous hematopoietic stem cells. Both treatments, however, are only accessible to a subset of X-ALD patients, mainly because of the lack of markers that can predict the onset of cerebral demyelination. Moreover, for female or male X-ALD patients with AMN, currently only unsatisfying therapeutic opportunities are available. Thus, this review focuses on current and urgently needed future pharmacological therapies. The treatment of adrenal and gonadal insufficiency is well established, whereas applications of immunomodulatory and immunosuppressive drugs have failed to prevent progression of cerebral neuroinflammation. The use of Lorenzo's oil and the inefficacy of lovastatin to normalize very-long-chain fatty acids in clinical trials as well as currently experimental and therefore possible future therapeutic strategies are reviewed. The latter include pharmacological gene therapy mediated by targeted upregulation of ABCD2, the closest homolog of ABCD1, antioxidative drug treatment, small molecule histone deacetylase inhibitors such as butyrates and valproic acid, and other neuroprotective attempts.
Collapse
Affiliation(s)
- Johannes Berger
- Center for Brain Research, Medical University of Vienna, Vienna, Austria.
| | | | | | | |
Collapse
|
50
|
A novel fluorescent sensor protein for visualization of redox states in the cytoplasm and in peroxisomes. Mol Cell Biol 2010; 30:3758-66. [PMID: 20498274 DOI: 10.1128/mcb.00121-10] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Reactive oxygen species are generated within peroxisomes during peroxisomal metabolism. However, due to technological difficulties, the intraperoxisomal redox state remain elusive, and the effect of peroxisome deficiency on the intracellular redox state is controversial. A newly developed, genetically encoded fluorescence resonance energy transfer (FRET) probe, Redoxfluor, senses the physiological redox state via its internal disulfide bonds, resulting in a change in the conformation of the protein leading to a FRET response. We made use of Redoxfluor to measure the redox states at the subcellular level in yeast and Chinese hamster ovary (CHO) cells. In wild-type peroxisomes harboring an intact fatty acid beta-oxidation system, the redox state within the peroxisomes was more reductive than that in the cytosol, despite the fact that reactive oxygen species were generated within the peroxisomes. Interestingly, we observed that the redox state of the cytosol of cell mutants for peroxisome assembly, regarded as models for a neurological metabolic disorder, was more reductive than that of the wild-type cells in yeast and CHO cells. Furthermore, Redoxfluor was utilized to develop an efficient system for the screening of drugs that moderate the abnormal cytosolic redox state in the mutant CHO cell lines for peroxisome assembly without affecting the redox state of normal cells.
Collapse
|