1
|
Li C, Terluk MR, Kartha RV. Nervonic acid, a long chain monounsaturated fatty acid, improves mitochondrial function in adrenomyeloneuropathy fibroblasts. Br J Pharmacol 2025. [PMID: 40400408 DOI: 10.1111/bph.70044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/12/2025] [Accepted: 03/02/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND AND PURPOSE Nervonic acid plays a vital role in maintaining normal brain and neuronal function. Nervonic acid has gained increasing attention because of its potential neuroprotective and anti-inflammatory properties. Nonetheless, the beneficial effects of nervonic acid are yet to be fully investigated. Adrenomyeloneuropathy (AMN), a type of X-linked adrenoleukodystrophy (ALD), is a progressive inherited metabolic disease characterised by accumulation of saturated very long-chain fatty acids (VLCFAs) in plasma and tissues, leading to increasing oxidative stress, mitochondrial dysfunction, neuroinflammation, cognitive dysfunction and disability. We previously found that nervonic acid can biochemically reverse the accumulation of saturated VLCFAs and increase cellular ATP production in ALD. Here, we investigated nervonic acid as a potential therapy for ALD by assessing its impact on mitochondrial function. EXPERIMENTAL APPROACH We assessed the effect of nervonic acid on cellular bioenergetics and oxidative stress in AMN patient-derived fibroblasts. We employed Seahorse real-time cell metabolic analysis and imaging of cells treated with increasing concentrations of nervonic acid. Normal dermal fibroblasts served as the healthy control. KEY RESULTS AMN cells demonstrate significantly impaired basal respiration, ATP production, maximal respiration and spare respiratory capacity compared to healthy fibroblasts. These mitochondrial respiration parameters significantly improved on treatment with nervonic acid in a concentration-dependent manner. Nervonic acid treatment also significantly reduced mitochondria-derived and total cellular reactive oxygen species, indicating mitigation of total oxidative stress. CONCLUSION AND IMPLICATIONS Our findings indicate a new mechanism of action for nervonic acid in ALD and other mitochondrial dysfunction-associated diseases. This can also indirectly prevent downstream inflammation, thus altering disease progression.
Collapse
Affiliation(s)
- Chenxu Li
- Center for Orphan Drug Research, University of Minnesota, Minneapolis-Saint Paul, Minnesota, USA
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis-Saint Paul, Minnesota, USA
| | - Marcia R Terluk
- Center for Orphan Drug Research, University of Minnesota, Minneapolis-Saint Paul, Minnesota, USA
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis-Saint Paul, Minnesota, USA
| | - Reena V Kartha
- Center for Orphan Drug Research, University of Minnesota, Minneapolis-Saint Paul, Minnesota, USA
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis-Saint Paul, Minnesota, USA
| |
Collapse
|
2
|
Bougnères P, Le Stunff C. Revisiting the Pathogenesis of X-Linked Adrenoleukodystrophy. Genes (Basel) 2025; 16:590. [PMID: 40428412 PMCID: PMC12111468 DOI: 10.3390/genes16050590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2025] [Revised: 05/11/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND X-ALD is a white matter (WM) disease caused by mutations in the ABCD1 gene encoding the transporter of very-long-chain fatty acids (VLCFAs) into peroxisomes. Strikingly, the same ABCD1 mutation causes either devastating brain inflammatory demyelination during childhood or, more often, progressive spinal cord axonopathy starting in middle-aged adults. The accumulation of undegraded VLCFA in glial cell membranes and myelin has long been thought to be the central mechanism of X-ALD. METHODS This review discusses studies in mouse and drosophila models that have modified our views of X-ALD pathogenesis. RESULTS In the Abcd1 knockout (KO) mouse that mimics the spinal cord disease, the late manifestations of axonopathy are rapidly reversed by ABCD1 gene transfer into spinal cord oligodendrocytes (OLs). In a peroxin-5 KO mouse model, the selective impairment of peroxisomal biogenesis in OLs achieves an almost perfect phenocopy of cerebral ALD. A drosophila knockout model revealed that VLCFA accumulation in glial myelinating cells causes the production of a toxic lipid able to poison axons and activate inflammatory cells. Other mouse models showed the critical role of OLs in providing energy substrates to axons. In addition, studies on microglial changing substates have improved our understanding of neuroinflammation. CONCLUSIONS Animal models supporting a primary role of OLs and axonal pathology and a secondary role of microglia allow us to revisit of X-ALD mechanisms. Beyond ABCD1 mutations, pathogenesis depends on unidentified contributors, such as genetic background, cell-specific epigenomics, potential environmental triggers, and stochasticity of crosstalk between multiple cell types among billions of glial cells and neurons.
Collapse
Affiliation(s)
- Pierre Bougnères
- MIRCen Institute, Commissariat à l’Energie Atomique, Laboratoire des Maladies Neurodégénératives, 92260 Fontenay-aux-Roses, France
- NEURATRIS, 92260 Fontenay-aux-Roses, France
- Therapy Design Consulting, 94300 Vincennes, France
| | - Catherine Le Stunff
- MIRCen Institute, Commissariat à l’Energie Atomique, Laboratoire des Maladies Neurodégénératives, 92260 Fontenay-aux-Roses, France
- NEURATRIS, 92260 Fontenay-aux-Roses, France
- UMR1195 Inserm, University Paris Saclay, 94270 Le Kremlin-Bicêtre, France
| |
Collapse
|
3
|
Yu Y, Zhang L, Wen S, Li C, Tan L, Wu X, Zou Y, Liu T. Evaluation of visual function and morphological changes in patients with adrenoleukodystrophy using SS-OCT and PVEP. Doc Ophthalmol 2025:10.1007/s10633-025-10021-6. [PMID: 40347222 DOI: 10.1007/s10633-025-10021-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 04/11/2025] [Indexed: 05/12/2025]
Abstract
PURPOSE This study aims to evaluate visual function and morphological changes in patients with adrenoleukodystrophy (ALD) through sweep-source optical coherence tomography (SS-OCT) and patterned visual evoked potentials (PVEP), with the goal of providing more accurate and comprehensive data for the diagnosis, monitoring, and assessment of treatment efficacy in patients with ALD. METHODS This study included 46 ALD patients and 44 healthy controls. The thickness of the retinal nerve fiber layer (RNFL) around the optic disc, as well as the macular RNFL and ganglion cell-inner plexiform layer (GCIPL) thickness, was assessed using SS-OCT. The peak time and amplitude of the P100 wave were also measured using PVEP. Factors that were statistically significant in the univariate analysis were subjected to stepwise binary logistic regression analysis for further investigation. Predictive performance was evaluated by constructing receiver operating characteristic (ROC) curves and compared using DeLong's test. RESULTS Compared with the control patients, ALD patients presented a significant increase in the peak time of the P100 wave (P < 0.05); however, no notable difference in amplitude was observed (P > 0.05). Additionally, substantial decreases in the RNFL and GCIPL thicknesses were observed within the parafoveal ring, especially in the superior and nasal quadrants (P < 0.05). The area under the curve for the binary logistic stepwise regression model was 0.883, with a sensitivity of 0.95, which surpassed the performance of the individual parameters. CONCLUSION ALD patients present with abnormal retinal structures and a PVEP peak time delay. Combining these two parameters could increase the accuracy of an early ALD diagnosis.
Collapse
Affiliation(s)
- Yongqiu Yu
- Department of Ophthalmology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Lujie Zhang
- Department of Ophthalmology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Shuiqian Wen
- Department of Ophthalmology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Chongyi Li
- Department of Ophthalmology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Liang Tan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xian Wu
- Department of Ophthalmology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Yangcheng Zou
- Department of Ophthalmology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China
| | - Ting Liu
- Department of Ophthalmology, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042, China.
| |
Collapse
|
4
|
Weinhofer I, Rommer P, Berger J. Blood Biomarkers Reflecting Brain Pathology-From Common Grounds to Rare Frontiers. J Inherit Metab Dis 2025; 48:e70032. [PMID: 40325881 PMCID: PMC12053231 DOI: 10.1002/jimd.70032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 03/18/2025] [Accepted: 04/11/2025] [Indexed: 05/07/2025]
Abstract
Understanding pathological changes in the brain is essential for guiding treatment decisions in brain injuries and diseases. Despite significant advances in brain imaging techniques, clinical practice still faces challenges due to infrastructure reliance and high resource demands. This review explores the current knowledge on blood-based biomarkers that indicate brain pathology, which can assist in identifying at-risk patients, diagnosing patients, predicting disease progression, and treatment response. We focus on the inherited metabolic disorders X-linked adrenoleukodystrophy (X-ALD) and metachromatic leukodystrophy (MLD) which share remarkable phenotypic variability. Disease-specific increases in the lipid metabolites lyso-PC26:0 in X-ALD and sulfatides in MLD might contribute to predicting clinical manifestation. Disease-unspecific biomarkers for axonal damage (neurofilament light chain protein, NfL) and glial degeneration (glial fibrillary acidic protein, GFAP) are able to distinguish X-ALD and MLD phenotypes at the group level. The implementation of X-ALD into newborn screening programs in various countries, including several U.S. states, has increased the demand for predictive blood-based biomarkers capable of detecting the early conversion from the pre-symptomatic to the early neuroinflammatory cerebral form of X-ALD. Among different biomarkers, NfL has proven most effective in reflecting neuroinflammation and correlating with brain lesion volume and the magnetic resonance imaging (MRI)-based severity scores. We discuss how NfL has moved from initial proof-of-principle towards proof-of-concept studies in brain disorders such as multiple sclerosis and how this knowledge could be applied for the clinical implementation of NfL in rare inherited metabolic disorders such as X-ALD.
Collapse
Affiliation(s)
- Isabelle Weinhofer
- Department Pathobiology of the Nervous System, Center for Brain ResearchMedical University of ViennaViennaAustria
| | - Paulus Rommer
- Department of Neurology, Comprehensive Center for Clinical Neurosciences and Mental HealthMedical University of ViennaViennaAustria
| | - Johannes Berger
- Department Pathobiology of the Nervous System, Center for Brain ResearchMedical University of ViennaViennaAustria
| |
Collapse
|
5
|
Peacock DJSJ, Ferreira CR, Horvath G, Hoffmann GF, Blau N, Ebrahimi-Fakhari D. Clinical and biochemical footprints of inherited metabolic diseases: Ia. Movement disorders, updated. Mol Genet Metab 2025; 145:109084. [PMID: 40132382 DOI: 10.1016/j.ymgme.2025.109084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025]
Abstract
Movement disorders are a common manifestation of inherited metabolic diseases (IMDs), categorized into hyperkinetic movement disorders, hypokinetic-rigid syndromes, ataxia, and spasticity. We reviewed and updated the list of known metabolic disorders associated with movement disorders, identifying a total of 559 IMDs. We outlined the more common and treatable causes, sorted by the dominant movement disorder phenomenology, and provided a practical clinical approach for suspected IMDs presenting with movement disorders. This work represents an updated catalog in a series of articles aimed at creating and maintaining a comprehensive list of clinical and metabolic differential diagnoses based on system involvement.
Collapse
Affiliation(s)
- Dakota J S J Peacock
- Division of Neurology, Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, Canada
| | - Carlos R Ferreira
- Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| | - Gabriella Horvath
- Division of Biochemical Genetics, Department of Pediatrics, BC Children's Hospital, University of British Columbia, Vancouver, Canada
| | - Georg F Hoffmann
- University Children's Hospital, University Clinic Heidelberg, Germany
| | - Nenad Blau
- Division of Metabolism, University Children's Hospital, Zürich, Switzerland.
| | - Darius Ebrahimi-Fakhari
- Movement Disorders Program, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
6
|
Dong SW, Xiao LM, Sun YH, Li GH, Xie YX, Wang MW, Wang N, Chen WJ, Chen HZ. Novel ABCD1 Variants in X-Linked Adrenoleukodystrophy. Clin Genet 2025. [PMID: 40210590 DOI: 10.1111/cge.14752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/24/2025] [Accepted: 03/27/2025] [Indexed: 04/12/2025]
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a neurodegenerative disorder caused by mutations in the ABCD1 gene. We reported the clinical features and genetic findings of 17 X-ALD patients. Fifteen variants were identified, including five novel mutations: c.700dupC (p.Arg234Profs*67), c.743G>A (p.Gly248Asp), c.1469_1471delTGG (p.Val490del), c.1577C>A (p.Thr526Lys), and c.1658T>C (p.Leu553Pro), which were reported for the first time in X-ALD patients. Functional analysis confirmed the pathogenicity of novel variants at the protein and subcellular localization level. The p.Arg234Profs*67 mutant protein was undetectable, likely due to NMD-mediated mRNA degradation. This study expands the mutation spectrum and clinical profile of X-ALD, suggesting a potential correlation between the extent of protein dysfunction and disease severity.
Collapse
Affiliation(s)
- Sen-Wei Dong
- Department of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Li-Mei Xiao
- Department of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yu-Hao Sun
- Department of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Gui-He Li
- Department of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Ying-Xuan Xie
- Department of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Meng-Wen Wang
- Department of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Ning Wang
- Department of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Wan-Jin Chen
- Department of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Hai-Zhu Chen
- Department of Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Neuroscience and Fujian Key Laboratory of Molecular Neurology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
7
|
Shang Q, Zhang X, Pu Y, Lin J, Ma P, Pan Y, Zhao M, Sun D, Cao L. LPCAT1, the Enzyme Responsible for Converting LPC to PC, Promotes OPC Differentiation In Vitro. J Cell Mol Med 2025; 29:e70387. [PMID: 39878319 PMCID: PMC11775935 DOI: 10.1111/jcmm.70387] [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: 04/25/2024] [Revised: 12/06/2024] [Accepted: 01/17/2025] [Indexed: 01/31/2025] Open
Abstract
Myelin is the key structure for high-speed information transmission and is formed by oligodendrocytes (OLs) which are differentiated from oligodendrocyte precursor cells (OPCs) in the central nervous system. Lipid is the main component of myelin and the role of lipid metabolism-related molecules in myelination attach increasing attention. Lysophosphatidylcholine acyltransferase 1 (LPCAT1) mediates the conversion of lysophosphatidylcholine (LPC) to phosphatidylcholine (PC), and its role in myelination draws our interest as LPC is a classical demyelination inducer and PC is a major component of myelin. In this work, LPCAT1 is found expressed in the oligodendrocyte lineage cells during myelination. In vitro experiments showed that the expression level of LPCAT1 gradually increased along with the differentiation process from OPCs to OLs, and over-expression and interference experiments showed that LPCAT1 promoted OPCs differentiation without affecting their proliferation or apoptosis. Mechanistically, the undertaker of LPCAT1's pro-differentiation role is not PC, but the phosphorylated mTOR which is a key regulator in OPCs differentiation. RNA sequencing analysis showed LPCAT1 promoted the expression of ZBTB20 which is an important transcription factor related to lipid metabolism and regulates mTOR phosphorylation. In vivo, complex myelin tomacula involving multiple axons was formed after conditionally knocking out LPCAT1 in oligodendrocyte lineage cells, but no obvious myelin thickness abnormalities were observed. Our results indicate that LPCAT1 is an important regulator of myelination, and lipid metabolism-related molecules may be new valuable targets for the treatment of diseases with myelin abnormalities.
Collapse
Affiliation(s)
- Qi Shang
- Department of Neurobiology, Key Laboratory of Molecular Neurobiology of the Ministry of EducationNaval Medical UniversityShanghaiChina
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Xin Zhang
- Department of Neurobiology, Key Laboratory of Molecular Neurobiology of the Ministry of EducationNaval Medical UniversityShanghaiChina
| | - Yingyan Pu
- Department of Neurobiology, Key Laboratory of Molecular Neurobiology of the Ministry of EducationNaval Medical UniversityShanghaiChina
| | - Junjian Lin
- Department of Neurobiology, Key Laboratory of Molecular Neurobiology of the Ministry of EducationNaval Medical UniversityShanghaiChina
| | - Peng Ma
- Department of Anesthesiology, Changzheng HospitalNaval Medical UniversityShanghaiChina
| | - Yuchen Pan
- Department of Neurobiology, Key Laboratory of Molecular Neurobiology of the Ministry of EducationNaval Medical UniversityShanghaiChina
| | - Ming Zhao
- Department of Neurology, Naval Specialty Medical CenterNaval Medical UniversityShanghaiChina
| | - Dingya Sun
- Department of Neurobiology, Key Laboratory of Molecular Neurobiology of the Ministry of EducationNaval Medical UniversityShanghaiChina
| | - Li Cao
- Department of Neurobiology, Key Laboratory of Molecular Neurobiology of the Ministry of EducationNaval Medical UniversityShanghaiChina
| |
Collapse
|
8
|
Vasireddy V, Maguire CA, Anderson DW, Ng C, Gong Y, Eichler F, Fourcade S, Guilera C, Onieva A, Sanchez A, Leal-Julià M, Verdés S, Dijkstra IM, Kemp S, Park H, Lutz T, Clark SW, Bosch A, Pujol A, Kozarsky K. An in vitro and in vivo efficacy evaluation of gene therapy candidate SBT101 in mouse models of adrenomyeloneuropathy and in NHPs. Mol Ther Methods Clin Dev 2024; 32:101354. [PMID: 39524975 PMCID: PMC11550353 DOI: 10.1016/j.omtm.2024.101354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024]
Abstract
Adrenomyeloneuropathy is a progressive neurodegenerative disease caused by pathogenic variants in the ABCD1 gene, resulting in very-long-chain fatty acid (VLCFA) accumulation that leads to dying-back axonopathy. Our candidate gene therapy, SBT101 (AAV9-human ABCD1 [hABCD1]), aims to ameliorate pathology by delivering functional copies of hABCD1 to the spinal cord. Transduced cells produce functional ABCD1 protein, thereby repairing the underlying biochemical defect. In vitro and in vivo mouse studies were conducted to assess the biochemical and functional efficacy of SBT101 and show effective delivery to target tissues involved in the disease pathology: spinal cord and dorsal root ganglia. Administration of SBT101 to mixed glial cell cultures from Abcd1-Null mice, and to male Abcd1 knockout (Abcd1 -/y ) and double-knockout (Abcd1 -/y /Abcd2 -/- ) mice led to increased hABCD1 production and reduced VLCFA. Double-knockout mice also exhibited improved grip strength. Furthermore, we conducted biodistribution and safety assessments in nonhuman primates. Six-hour intrathecal lumbar infusions demonstrated effective transduction throughout target tissues, supporting the clinical feasibility of the procedure. SBT101 was well tolerated, with no observed SBT101-related mortality or clinical signs. These findings not only provide preclinical efficacy data for SBT101 but also inform clinically relevant SBT101 dose selection for patients with adrenomyeloneuropathy.
Collapse
Affiliation(s)
| | - Casey A. Maguire
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Carrie Ng
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yi Gong
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Florian Eichler
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Stéphane Fourcade
- Neurometabolic Disease Laboratory, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospital Duran i Reynalds, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBERER), Institudo de Salud Carlos III (ISCIII), Madrid, Spain
| | - Cristina Guilera
- Neurometabolic Disease Laboratory, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospital Duran i Reynalds, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBERER), Institudo de Salud Carlos III (ISCIII), Madrid, Spain
| | - Andrea Onieva
- Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
- Unitat Mixta UAB-VHIR, Vall D’Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Angela Sanchez
- Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
- Unitat Mixta UAB-VHIR, Vall D’Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Marc Leal-Julià
- Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
- Unitat Mixta UAB-VHIR, Vall D’Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Sergi Verdés
- Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
- Unitat Mixta UAB-VHIR, Vall D’Hebron Institut de Recerca (VHIR), Barcelona, Spain
| | - Inge M.E. Dijkstra
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, the Netherlands
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Department of Laboratory Medicine, Amsterdam UMC location University of Amsterdam, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, the Netherlands
| | | | - Tiffany Lutz
- SwanBio Therapeutics, Inc., Philadelphia, PA, USA
| | | | - Assumpció Bosch
- Department of Biochemistry and Molecular Biology and Institute of Neurosciences, Universitat Autònoma de Barcelona, Barcelona, Spain
- Unitat Mixta UAB-VHIR, Vall D’Hebron Institut de Recerca (VHIR), Barcelona, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Institudo de Salud Carlos III (ISCIII), Madrid, Spain
| | - Aurora Pujol
- Neurometabolic Disease Laboratory, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Hospital Duran i Reynalds, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBERER), Institudo de Salud Carlos III (ISCIII), Madrid, Spain
- Catalan Institution of Research and Advanced Studies (ICREA), 08010 Barcelona, Catalonia, Spain
| | | |
Collapse
|
9
|
Zuo X, Chen Z. From gene to therapy: a review of deciphering the role of ABCD1 in combating X-Linked adrenoleukodystrophy. Lipids Health Dis 2024; 23:369. [PMID: 39529100 PMCID: PMC11552335 DOI: 10.1186/s12944-024-02361-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024] Open
Abstract
X-linked adrenoleukodystrophy (X-ALD) is a severe genetic disorder caused by ABCD1 mutations, resulting in the buildup of very-long-chain fatty acids, leading to significant neurological decline and adrenal insufficiency. Despite advancements in understanding the mechanisms of X-ALD, its pathophysiology remains incompletely understood, complicating the development of effective treatments. This review provides a comprehensive overview of X-ALD, with a focus on the genetic and biochemical roles of ABCD1 and the impacts of its mutations. Current therapeutic approaches are evaluated, discussing their limitations, and emphasizing the need to fully elucidate the pathogenesis of X-ALD. Additionally, this review highlights the importance of international collaboration to enhance systematic data collection and advance biomarker discovery, ultimately improving patient outcomes with X-ALD.
Collapse
Affiliation(s)
- Xinxin Zuo
- Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| | - Zeyu Chen
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| |
Collapse
|
10
|
Sarkar C, Lipinski MM. Role and Function of Peroxisomes in Neuroinflammation. Cells 2024; 13:1655. [PMID: 39404418 PMCID: PMC11476013 DOI: 10.3390/cells13191655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/25/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Peroxisomes are organelles involved in many cellular metabolic functions, including the degradation of very-long-chain fatty acids (VLCFAs; C ≥ 22), the initiation of ether-phospholipid synthesis, and the metabolism of reactive oxygen species. All of these processes are essential for the maintenance of cellular lipid and redox homeostasis, and their perturbation can trigger inflammatory response in immune cells, including in the central nervous system (CNS) resident microglia and astrocytes. Consistently, peroxisomal disorders, a group of congenital diseases caused by a block in peroxisomal biogenesis or the impairment of one of the peroxisomal enzymes, are associated with neuroinflammation. Peroxisomal function is also dysregulated in many neurodegenerative diseases and during brain aging, both of which are associated with neuroinflammation. This suggests that deciphering the role of peroxisomes in neuroinflammation may be important for understanding both congenital and age-related brain dysfunction. In this review, we discuss the current advances in understanding the role and function of peroxisomes in neuroinflammation.
Collapse
Affiliation(s)
- Chinmoy Sarkar
- Shock, Trauma and Anesthesiology Research (STAR) Center, Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Marta M. Lipinski
- Shock, Trauma and Anesthesiology Research (STAR) Center, Department of Anesthesiology and Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| |
Collapse
|
11
|
Chaudhary R, Rehman M, Agarwal V, Kumar A, Kaushik AS, Srivastava S, Srivastava S, Verma R, Rajinikanth PS, Mishra V. Terra incognita of glial cell dynamics in the etiology of leukodystrophies: Broadening disease and therapeutic perspectives. Life Sci 2024; 354:122953. [PMID: 39122110 DOI: 10.1016/j.lfs.2024.122953] [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: 02/05/2024] [Revised: 07/09/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Neuroglial cells, also known as glia, are primarily characterized as auxiliary cells within the central nervous system (CNS). The recent findings have shed light on their significance in numerous physiological processes and their involvement in various neurological disorders. Leukodystrophies encompass an array of rare and hereditary neurodegenerative conditions that were initially characterized by the deficiency, aberration, or degradation of myelin sheath within CNS. The primary cellular populations that experience significant alterations are astrocytes, oligodendrocytes and microglia. These glial cells are either structurally or metabolically impaired due to inherent cellular dysfunction. Alternatively, they may fall victim to the accumulation of harmful by-products resulting from metabolic disturbances. In either situation, the possible replacement of glial cells through the utilization of implanted tissue or stem cell-derived human neural or glial progenitor cells hold great promise as a therapeutic strategy for both the restoration of structural integrity through remyelination and the amelioration of metabolic deficiencies. Various emerging treatment strategies like stem cell therapy, ex-vivo gene therapy, infusion of adeno-associated virus vectors, emerging RNA-based therapies as well as long-term therapies have demonstrated success in pre-clinical studies and show promise for rapid clinical translation. Here, we addressed various leukodystrophies in a comprehensive and detailed manner as well as provide prospective therapeutic interventions that are being considered for clinical trials. Further, we aim to emphasize the crucial role of different glial cells in the pathogenesis of leukodystrophies. By doing so, we hope to advance our understanding of the disease, elucidate underlying mechanisms, and facilitate the development of potential treatment interventions.
Collapse
Affiliation(s)
- Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Mujeeba Rehman
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Vipul Agarwal
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Anand Kumar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Arjun Singh Kaushik
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Siddhi Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Sukriti Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Rajkumar Verma
- University of Connecticut School of Medicine, 200 Academic Way, Farmington, CT 06032, USA
| | - P S Rajinikanth
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Vikas Mishra
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India.
| |
Collapse
|
12
|
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
|
13
|
Haschka T, Lamari F, Mochel F, Zujovic V. Innovative tree-based method for sampling molecular conformations: exploring the ATP-binding cassette subfamily D member 1 (ABCD1) transporter as a case study. Front Mol Biosci 2024; 11:1440529. [PMID: 39148631 PMCID: PMC11325183 DOI: 10.3389/fmolb.2024.1440529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024] Open
Abstract
We introduce a novel tree-based method for visualizing molecular conformation sampling. Our method offers enhanced precision in highlighting conformational differences and facilitates the observation of local minimas within proteins fold space. The projection of empirical laboratory data on the tree allows us to create a link between protein conformations and disease relevant data. To demonstrate the efficacy of our approach, we applied it to the ATP-binding cassette subfamily D member 1 (ABCD1) transporter responsible for very long-chain fatty acids (VLCFAs) import into peroxisomes. The genetic disorder called X-linked adrenoleukodystrophy (XALD) is characterized by the accumulation of VLCFA due to pathogenic variants in the ABCD1 gene. Using in silico molecular simulation, we examined the behavior of 16 prevalent mutations alongside the wild-type protein, exploring both inward and outward open forms of the transporter through molecular simulations. We evaluated from resulting trajectories the energy potential related to the ABCD1 interactions with ATP molecules. We categorized XALD patients based on the severity and progression of their disease, providing a unique clinical perspective. By integrating this data into our numerical framework, our study aimed to uncover the molecular underpinnings of XALD, offering new insights into disease progression. As we explored molecular trajectories and conformations resulting from our study, the tree-based method not only contributes valuable insights into XALD but also lays a solid foundation for forthcoming drug design studies. We advocate for the broader adoption of our innovative approach, proposing it as a valuable tool for researchers engaged in molecular simulation studies.
Collapse
Affiliation(s)
- Thomas Haschka
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital Pitié la Salpétrière University Hospital, DMU Neuroscience 6, Paris, France
| | - Foudil Lamari
- UF Biochimie des Maladies Neuro-métaboliques, Service de Biochimie Métabolique, APHP, Hôpital Pitié la Salpétrière University Hospital, Paris, France
| | - Fanny Mochel
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital Pitié la Salpétrière University Hospital, DMU Neuroscience 6, Paris, France
| | - Violetta Zujovic
- Sorbonne Université, Institut du Cerveau - Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital Pitié la Salpétrière University Hospital, DMU Neuroscience 6, Paris, France
| |
Collapse
|
14
|
Vaia Y, Bruschi F, Tagi VM, Tosi M, Montanari C, Zuccotti G, Tonduti D, Verduci E. Microbiota gut-brain axis: implications for pediatric-onset leukodystrophies. Front Nutr 2024; 11:1417981. [PMID: 39070252 PMCID: PMC11272617 DOI: 10.3389/fnut.2024.1417981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024] Open
Abstract
Neurodegenerative disorders are a group of diseases characterized by progressive degeneration of the nervous system, leading to a gradual loss of previously acquired motor, sensory and/or cognitive functions. Leukodystrophies are amongst the most frequent childhood-onset neurodegenerative diseases and primarily affect the white matter of the brain, often resulting in neuro-motor disability. Notably, gastrointestinal (GI) symptoms and complications, such as gastroesophageal reflux disease (GERD) and dysphagia, significantly impact patients' quality of life, highlighting the need for comprehensive management strategies. Gut dysbiosis, characterized by microbial imbalance, has been implicated in various GI disorders and neurodegenerative diseases. This narrative review explores the intricate relationship between GI symptoms, Gut Microbiota (GM), and neurodegeneration. Emerging evidence underscores the profound influence of GM on neurological functions via the microbiota gut-brain axis. Animal models have demonstrated alterations in GM composition associated with neuroinflammation and neurodegeneration. Our single-centre experience reveals a high prevalence of GI symptoms in leukodystrophy population, emphasizing the importance of gastroenterological assessment and nutritional intervention in affected children. The bidirectional relationship between GI disorders and neurodegeneration suggests a potential role of gut dysbiosis in disease progression. Prospective studies investigating the GM in leukodystrophies are essential to understand the role of gut-brain axis dysfunction in disease progression and identify novel therapeutic targets. In conclusion, elucidating the interplay between GI disorders, GM, and neurodegeneration holds promise for precision treatments aimed at improving patient outcomes and quality of life.
Collapse
Affiliation(s)
- Ylenia Vaia
- C.O.A.L.A. (Center for Diagnosis and Treatment of Leukodystrophies), Unit of Pediatric Neurology, V. Buzzi Children’s Hospital, Milan, Italy
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Fabio Bruschi
- C.O.A.L.A. (Center for Diagnosis and Treatment of Leukodystrophies), Unit of Pediatric Neurology, V. Buzzi Children’s Hospital, Milan, Italy
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Veronica Maria Tagi
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
- Department of Pediatrics, V. Buzzi Children's Hospital, Milan, Italy
| | - Martina Tosi
- Department of Pediatrics, V. Buzzi Children's Hospital, Milan, Italy
- Department of Health Sciences, University of Milan, Milan, Italy
| | - Chiara Montanari
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
- Department of Pediatrics, V. Buzzi Children's Hospital, Milan, Italy
| | - Gianvincenzo Zuccotti
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
- Department of Pediatrics, V. Buzzi Children's Hospital, Milan, Italy
| | - Davide Tonduti
- C.O.A.L.A. (Center for Diagnosis and Treatment of Leukodystrophies), Unit of Pediatric Neurology, V. Buzzi Children’s Hospital, Milan, Italy
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
| | - Elvira Verduci
- Department of Health Sciences, University of Milan, Milan, Italy
- Metabolic Diseases Unit, Department of Pediatrics, V. Buzzi Children’s Hospital, University of Milan, Milan, Italy
| |
Collapse
|
15
|
Parasar P, Kaur N, Singh J. Pathophysiology of X-Linked Adrenoleukodystrophy: Updates on Molecular Mechanisms. JOURNAL OF BIOTECHNOLOGY AND BIOMEDICINE 2024; 7:277-288. [PMID: 39056013 PMCID: PMC11271253 DOI: 10.26502/jbb.2642-91280151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/28/2024]
Abstract
X-ALD, an inherited monogenic metabolic disorder affecting the CNS and adrenal white matter, is caused by mutations in ABCD1 gene leading to defective fatty acid oxidation in the peroxisomes. This results in accumulation of very long-chain fatty acids, VLCFA, into brain, spinal cord, and body fluids. A single ABCD1mutation does not clearly explain the severity and diverse clinical spectrum of X-ALD phenotypes which suggests that not only genetic but also other modifier genes, epigenetic factors, and environmental factors play a role and contribute to neuroinflammation, mitochondrial dysfunctions, oxidative stress, and metabolic defects seen in phenotypes of ALD. In this review we discuss genotype and phenotype correlation and clinical spectra of X-ALD, previous and recent modifier genetic factors of X-ALD, including novel role of microRNAs (miRNAs) in pathology and as biomarkers. We also discuss the mechanistic interplay of miRNAs and metabolic pathways and potential of targeting miRNAs for X-ALD.
Collapse
Affiliation(s)
- Parveen Parasar
- Department of Neurology, Henry Ford Health, Detroit, MI 48202, USA
| | - Navtej Kaur
- Department of Neurology, Henry Ford Health, Detroit, MI 48202, USA
| | - Jaspreet Singh
- Department of Neurology, Henry Ford Health, Detroit, MI 48202, USA
- Department of Physiology, Michigan State University, Lansing, MI 48824, USA
| |
Collapse
|
16
|
Granadeiro L, Zarralanga VE, Rosa R, Franquinho F, Lamas S, Brites P. Ataxia with giant axonopathy in Acbd5-deficient mice halted by adeno-associated virus gene therapy. Brain 2024; 147:1457-1473. [PMID: 38066620 DOI: 10.1093/brain/awad407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/27/2023] [Accepted: 11/07/2023] [Indexed: 04/06/2024] Open
Abstract
Acyl-CoA binding domain containing 5 (ACBD5) is a critical player in handling very long chain fatty acids (VLCFA) en route for peroxisomal β-oxidation. Mutations in ACBD5 lead to the accumulation of VLCFA and patients present retinal dystrophy, ataxia, psychomotor delay and a severe leukodystrophy. Using CRISPR/Cas9, we generated and characterized an Acbd5 Gly357* mutant allele. Gly357* mutant mice recapitulated key features of the human disorder, including reduced survival, impaired locomotion and reflexes, loss of photoreceptors, and demyelination. The ataxic presentation of Gly357* mice involved the loss of cerebellar Purkinje cells and a giant axonopathy throughout the CNS. Lipidomic studies provided evidence for the extensive lipid dysregulation caused by VLCFA accumulation. Following a proteomic survey, functional studies in neurons treated with VLCFA unravelled a deregulated cytoskeleton with reduced actin dynamics and increased neuronal filopodia. We also show that an adeno-associated virus-mediated gene delivery ameliorated the gait phenotypes and the giant axonopathy, also improving myelination and astrocyte reactivity. Collectively, we established a mouse model with significance for VLCFA-related disorders. The development of relevant neuropathological outcomes enabled the understanding of mechanisms modulated by VLCFA and the evaluation of the efficacy of preclinical therapeutic interventions.
Collapse
Affiliation(s)
- Luis Granadeiro
- Neurolipid Biology, Instituto de Investigação e Inovação em Saúde da Universidade do Porto - i3S and Instituto de Biologia Molecular e Celular - IBMC, 4200-135 Porto, Portugal
- Graduate Program in Molecular and Cell Biology, Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, 4050-313 Porto, Portugal
| | - Violeta Enríquez Zarralanga
- Neurolipid Biology, Instituto de Investigação e Inovação em Saúde da Universidade do Porto - i3S and Instituto de Biologia Molecular e Celular - IBMC, 4200-135 Porto, Portugal
| | - Ricardo Rosa
- Neurolipid Biology, Instituto de Investigação e Inovação em Saúde da Universidade do Porto - i3S and Instituto de Biologia Molecular e Celular - IBMC, 4200-135 Porto, Portugal
| | - Filipa Franquinho
- Animal Facility, Instituto de Investigação e Inovação em Saúde da Universidade do Porto - i3S, 4200-135 Porto, Portugal
| | - Sofia Lamas
- Animal Facility, Instituto de Investigação e Inovação em Saúde da Universidade do Porto - i3S, 4200-135 Porto, Portugal
| | - Pedro Brites
- Neurolipid Biology, Instituto de Investigação e Inovação em Saúde da Universidade do Porto - i3S and Instituto de Biologia Molecular e Celular - IBMC, 4200-135 Porto, Portugal
| |
Collapse
|
17
|
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
|
18
|
Yska HAF, Engelen M, Bugiani M. The pathology of X-linked adrenoleukodystrophy: tissue specific changes as a clue to pathophysiology. Orphanet J Rare Dis 2024; 19:138. [PMID: 38549180 PMCID: PMC10976706 DOI: 10.1186/s13023-024-03105-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 02/23/2024] [Indexed: 04/02/2024] Open
Abstract
Although the pathology of X-linked adrenoleukodystrophy (ALD) is well described, it represents the end-stage of neurodegeneration. It is still unclear what cell types are initially involved and what their role is in the disease process. Revisiting the seminal post-mortem studies from the 1970s can generate new hypotheses on pathophysiology. This review describes (histo)pathological changes of the brain and spinal cord in ALD. It aims at integrating older works with current insights and at providing an overarching theory on the pathophysiology of ALD. The data point to an important role for axons and glia in the pathology of both the myelopathy and leukodystrophy of ALD. In-depth pathological analyses with new techniques could help further unravel the sequence of events behind the pathology of ALD.
Collapse
Affiliation(s)
- Hemmo A F Yska
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam UMC location University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.
| | - Marc Engelen
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam UMC location University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Marianna Bugiani
- Department of Pediatrics/Child Neurology, VU University Medical Centre, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Pathology, VU University Medical Centre, Amsterdam Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
19
|
Rao Y, Peng B. Allogenic microglia replacement: A novel therapeutic strategy for neurological disorders. FUNDAMENTAL RESEARCH 2024; 4:237-245. [PMID: 38933508 PMCID: PMC11197774 DOI: 10.1016/j.fmre.2023.02.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/17/2022] [Accepted: 02/19/2023] [Indexed: 03/29/2023] Open
Abstract
Microglia are resident immune cells in the central nervous system (CNS) that play vital roles in CNS development, homeostasis and disease pathogenesis. Genetic defects in microglia lead to microglial dysfunction, which in turn leads to neurological disorders. The correction of the specific genetic defects in microglia in these disorders can lead to therapeutic effects. Traditional genetic defect correction approaches are dependent on viral vector-based genetic defect corrections. However, the viruses used in these approaches, including adeno-associated viruses, lentiviruses and retroviruses, do not primarily target microglia; therefore, viral vector-based genetic defect corrections are ineffective in microglia. Microglia replacement is a novel approach to correct microglial genetic defects via replacing microglia of genetic defects with allogenic healthy microglia. In this paper, we systematically review the history, rationale and therapeutic perspectives of microglia replacement, which would be a novel strategy for treating CNS disorders.
Collapse
Affiliation(s)
- Yanxia Rao
- Department of Laboratory Animal Science, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Bo Peng
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, MOE Innovative Center for New Drug Development of Immune Inflammatory Diseases, Fudan University, Shanghai 200000, China
- Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| |
Collapse
|
20
|
Gong Y, Laheji F, Berenson A, Li Y, Moser A, Qian A, Frosch M, Sadjadi R, Hahn R, Maguire CA, Eichler F. Role of Basal Forebrain Neurons in Adrenomyeloneuropathy in Mice and Humans. Ann Neurol 2024; 95:442-458. [PMID: 38062617 PMCID: PMC10949091 DOI: 10.1002/ana.26849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 11/28/2023] [Accepted: 12/05/2023] [Indexed: 12/27/2023]
Abstract
OBJECTIVE X-linked adrenoleukodystrophy is caused by mutations in the peroxisomal half-transporter ABCD1. The most common manifestation is adrenomyeloneuropathy, a hereditary spastic paraplegia of adulthood. The present study set out to understand the role of neuronal ABCD1 in mice and humans with adrenomyeloneuropathy. METHODS Neuronal expression of ABCD1 during development was assessed in mice and humans. ABCD1-deficient mice and human brain tissues were examined for corresponding pathology. Next, we silenced ABCD1 in cholinergic Sh-sy5y neurons to investigate its impact on neuronal function. Finally, we tested adeno-associated virus vector-mediated ABCD1 delivery to the brain in mice with adrenomyeloneuropathy. RESULTS ABCD1 is highly expressed in neurons located in the periaqueductal gray matter, basal forebrain and hypothalamus. In ABCD1-deficient mice (Abcd1-/y), these structures showed mild accumulations of α-synuclein. Similarly, healthy human controls had high expression of ABCD1 in deep gray nuclei, whereas X-ALD patients showed increased levels of phosphorylated tau, gliosis, and complement activation in those same regions, albeit not to the degree seen in neurodegenerative tauopathies. Silencing ABCD1 in Sh-sy5y neurons impaired expression of functional proteins and decreased acetylcholine levels, similar to observations in plasma of Abcd1-/y mice. Notably, hind limb clasping in Abcd1-/y mice was corrected through transduction of ABCD1 in basal forebrain neurons following intracerebroventricular gene delivery. INTERPRETATION Our study suggests that the basal forebrain-cortical cholinergic pathway may contribute to dysfunction in adrenomyeloneuropathy. Rescuing peroxisomal transport activity in basal forebrain neurons and supporting glial cells might represent a viable therapeutic strategy. ANN NEUROL 2024;95:442-458.
Collapse
Affiliation(s)
- Yi Gong
- Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston
| | - Fiza Laheji
- Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston
| | - Anna Berenson
- Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston
| | - Yedda Li
- Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston
| | - Ann Moser
- Peroxisome Disease Lab, Hugo W Moser Research Institute, Baltimore, MD, USA
| | - April Qian
- Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston
| | - Matthew Frosch
- Massachusetts General Hospital, Department of Neuropathology, Harvard Medical School, Boston
| | - Reza Sadjadi
- Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston
| | - Ryan Hahn
- Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston
| | - Casey A. Maguire
- Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston
| | - Florian Eichler
- Massachusetts General Hospital, Department of Neurology, Harvard Medical School, Boston
| |
Collapse
|
21
|
Siwek T, Zwiernik B, Jezierska-Woźniak K, Jezierska K, Mycko MP, Selmaj KW. Intrathecal administration of mesenchymal stem cells in patients with adrenomyeloneuropathy. Front Neurol 2024; 15:1345503. [PMID: 38370525 PMCID: PMC10869536 DOI: 10.3389/fneur.2024.1345503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/17/2024] [Indexed: 02/20/2024] Open
Abstract
Background and objectives X-linked adrenomyeloneuropathy (AMN) is an inherited neurodegenerative disorder associated with mutations in the ABCD1 gene and the accumulation of very long-chain fatty acids (VLFCAs) in plasma and tissues. Currently, there is no effective treatment for AMN. We have aimed to evaluate the therapeutic effects of mesenchymal stem cell (MSC) transplantation in patients with AMN. Methods This is a small cohort open-label study with patients with AMN diagnosed and treated at the University Hospital in Olsztyn, Poland. All patients met clinical, biochemical, MRI, and neuropsychological criteria for AMN. MSCs derived from Wharton jelly, 20 × 106 cells, were administered intrathecally three times every 2 months, and patients were followed up for an additional 3 months. The primary outcome measures included a blinded assessment of lower limb muscle strength with the Medical Research Council Manual Muscle Testing scale at baseline and on every month visits until the end of the study. Additional outcomes included measurements of the timed 25-feet walk (T25FW) and VLFCA serum ratio. Results Three male patients with AMN with an age range of 26-37 years participated in this study. All patients experienced increased muscle strength in the lower limbs at the end of the study versus baseline. The power grade increased by 25-43% at the baseline. In addition, all patients showed an improvement trend in walking speed measured with the T25FW test. Treatment with MSCs in patients with AMN appeared to be safe and well tolerated. Discussion The results of this study demonstrated that intrathecal administration of WJ-MSC improves motor symptoms in patients with AMN. The current findings lend support to the safety and feasibility of MSC therapy as a potentially viable treatment option for patients with AMN.
Collapse
Affiliation(s)
- Tomasz Siwek
- Department of Neurology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
- University Hospital, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Beata Zwiernik
- Department of Neurology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
- University Hospital, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Katarzyna Jezierska-Woźniak
- Laboratory for Regenerative Medicine, Department of Neurosurgery, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Kamila Jezierska
- University Hospital, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Marcin P. Mycko
- Department of Neurology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
- University Hospital, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
| | - Krzysztof W. Selmaj
- Department of Neurology, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland
- Center of Neurology, Lodz, Poland
| |
Collapse
|
22
|
Raas Q, Wood A, Stevenson TJ, Swartwood S, Liu S, Kannan RM, Kannan S, Bonkowsky JL. Generation and characterization of a zebrafish gain-of-function ACOX1 Mitchell disease model. Front Pediatr 2024; 12:1326886. [PMID: 38357503 PMCID: PMC10864527 DOI: 10.3389/fped.2024.1326886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/05/2024] [Indexed: 02/16/2024] Open
Abstract
Background Mitchell syndrome is a rare, neurodegenerative disease caused by an ACOX1 gain-of-function mutation (c.710A>G; p.N237S), with fewer than 20 reported cases. Affected patients present with leukodystrophy, seizures, and hearing loss. ACOX1 serves as the rate-limiting enzyme in peroxisomal beta-oxidation of very long-chain fatty acids. The N237S substitution has been shown to stabilize the active ACOX1 dimer, resulting in dysregulated enzymatic activity, increased oxidative stress, and glial damage. Mitchell syndrome lacks a vertebrate model, limiting insights into the pathophysiology of ACOX1-driven white matter damage and neuroinflammatory insults. Methods We report a patient presenting with rapidly progressive white matter damage and neurological decline, who was eventually diagnosed with an ACOX1 N237S mutation through whole genome sequencing. We developed a zebrafish model of Mitchell syndrome using transient ubiquitous overexpression of the human ACOX1 N237S variant tagged with GFP. We assayed zebrafish behavior, oligodendrocyte numbers, expression of white matter and inflammatory transcripts, and analysis of peroxisome counts. Results The patient experienced progressive leukodystrophy and died 2 years after presentation. The transgenic zebrafish showed a decreased swimming ability, which was restored with the reactive microglia-targeted antioxidant dendrimer-N-acetyl-cysteine conjugate. The mutants showed no effect on oligodendrocyte counts but did display activation of the integrated stress response (ISR). Using a novel SKL-targeted mCherry reporter, we found that mutants had reduced density of peroxisomes. Conclusions We developed a vertebrate (zebrafish) model of Mitchell syndrome using transient ubiquitous overexpression of the human ACOX1 N237S variant. The transgenic mutants exhibited motor impairment and showed signs of activated ISR, but interestingly, there were no changes in oligodendrocyte counts. However, the mutants exhibited a deficiency in the number of peroxisomes, suggesting a possible shared mechanism with the Zellweger spectrum disorders.
Collapse
Affiliation(s)
- Quentin Raas
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, United States
- Laboratory of Translational Research for Neurological Disorders, Imagine Institute, Université de Paris, INSERM UMR 1163, Paris, France
| | - Austin Wood
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Tamara J. Stevenson
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Shanna Swartwood
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Suzanne Liu
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Rangaramanujam M. Kannan
- Department of Ophthalmology, Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sujatha Kannan
- Department of Ophthalmology, Center for Nanomedicine, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Joshua L. Bonkowsky
- Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, United States
- Center for Personalized Medicine, Primary Children’s Hospital, Salt Lake City, UT, United States
| |
Collapse
|
23
|
Thakkar RN, Patel D, Kioutchoukova IP, Al-Bahou R, Reddy P, Foster DT, Lucke-Wold B. Leukodystrophy Imaging: Insights for Diagnostic Dilemmas. Med Sci (Basel) 2024; 12:7. [PMID: 38390857 PMCID: PMC10885080 DOI: 10.3390/medsci12010007] [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: 10/31/2023] [Revised: 12/09/2023] [Accepted: 12/13/2023] [Indexed: 02/24/2024] Open
Abstract
Leukodystrophies, a group of rare demyelinating disorders, mainly affect the CNS. Clinical presentation of different types of leukodystrophies can be nonspecific, and thus, imaging techniques like MRI can be used for a more definitive diagnosis. These diseases are characterized as cerebral lesions with characteristic demyelinating patterns which can be used as differentiating tools. In this review, we talk about these MRI study findings for each leukodystrophy, associated genetics, blood work that can help in differentiation, emerging diagnostics, and a follow-up imaging strategy. The leukodystrophies discussed in this paper include X-linked adrenoleukodystrophy, metachromatic leukodystrophy, Krabbe's disease, Pelizaeus-Merzbacher disease, Alexander's disease, Canavan disease, and Aicardi-Goutières Syndrome.
Collapse
Affiliation(s)
- Rajvi N. Thakkar
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Drashti Patel
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | | | - Raja Al-Bahou
- College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Pranith Reddy
- College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Devon T. Foster
- College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, 1600 SW Archer Rd., Gainesville, FL 32610, USA
| |
Collapse
|
24
|
Sevin C, Mochel F. Hematopoietic stem cell transplantation in leukodystrophies. HANDBOOK OF CLINICAL NEUROLOGY 2024; 204:355-366. [PMID: 39322389 DOI: 10.1016/b978-0-323-99209-1.00017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
More than 50 leukodystrophies have been described. This group of inherited disorders affects myelin development and/or maintenance and can manifest from birth to adulthood. Neuroinflammation is a hallmark of some leukodystrophies, explaining in part the therapeutic benefit of hematopoietic stem cell transplantation (HSCT). Indeed, in addition to supplying the CNS with myelomonocyte donor cells expressing the deficient protein or enzyme, HSCT allows the restoration of normal microglia function, which may act on neuroinflammation. In this chapter, we explore the rationale, indication, and outcome of HSCT in Cerebral Adrenoleukodystrophy (CALD), Metachromatic Leukodystrophy (MLD), Krabbe Disease (KD), and Adult-onset Leukoencephalopathy with Axonal Spheroids and Pigmented Glia (ALSP), which are among the most frequent leukodystrophies. For these leukodystrophies, HSCT may modify notably the natural history and improve CNS-related deficits, provided that the procedure is performed early into the disease course. In addition, we discuss the recent development of ex vivo gene therapy for CALD and MLD as a promising alternative to allograft.
Collapse
Affiliation(s)
- Caroline Sevin
- AP-HP, Kremlin-Bicêtre University Hospital, Department of Neuropediatrics, Reference Center for Pediatric Leukodystrophies, Paris, France; INSERM U 1127, CNRS UMR 7225, Sorbonne Université, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau, ICM, Paris, France
| | - Fanny Mochel
- INSERM U 1127, CNRS UMR 7225, Sorbonne Université, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau, ICM, Paris, France; AP-HP, Pitié-Salpêtrière University Hospital, Department of Medical Genetics, Reference Centers for Adult Neurometabolic Diseases and Adult Leukodystrophies, Paris, France.
| |
Collapse
|
25
|
Vigne S, Pot C. Implication of Oxysterols and Phytosterols in Aging and Human Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1440:231-260. [PMID: 38036883 DOI: 10.1007/978-3-031-43883-7_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Cholesterol is easily oxidized and can be transformed into numerous oxidation products, among which oxysterols. Phytosterols are plant sterols related to cholesterol. Both oxysterols and phytosterols can have an impact on human health and diseases.Cholesterol is a member of the sterol family that plays essential roles in biological processes, including cell membrane stability and myelin formation. Cholesterol can be metabolized into several molecules including bile acids, hormones, and oxysterols. On the other hand, phytosterols are plant-derived compounds structurally related to cholesterol, which can also have an impact on human health. Here, we review the current knowledge about the role of oxysterols and phytosterols on human health and focus on the impact of their pathways on diseases of the central nervous system (CNS), autoimmune diseases, including inflammatory bowel diseases (IBD), vascular diseases, and cancer in both experimental models and human studies. We will first discuss the implications of oxysterols and then of phytosterols in different human diseases.
Collapse
Affiliation(s)
- Solenne Vigne
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Epalinges, Lausanne, Switzerland
| | - Caroline Pot
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Epalinges, Lausanne, Switzerland.
| |
Collapse
|
26
|
Villoria-González A, Zierfuss B, Parzer P, Heuböck E, Zujovic V, Waidhofer-Söllner P, Ponleitner M, Rommer P, Göpfert J, Forss-Petter S, Berger J, Weinhofer I. Efficacy of HDAC Inhibitors in Driving Peroxisomal β-Oxidation and Immune Responses in Human Macrophages: Implications for Neuroinflammatory Disorders. Biomolecules 2023; 13:1696. [PMID: 38136568 PMCID: PMC10741867 DOI: 10.3390/biom13121696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Elevated levels of saturated very long-chain fatty acids (VLCFAs) in cell membranes and secreted lipoparticles have been associated with neurotoxicity and, therefore, require tight regulation. Excessive VLCFAs are imported into peroxisomes for degradation by β-oxidation. Impaired VLCFA catabolism due to primary or secondary peroxisomal alterations is featured in neurodegenerative and neuroinflammatory disorders such as X-linked adrenoleukodystrophy and multiple sclerosis (MS). Here, we identified that healthy human macrophages upregulate the peroxisomal genes involved in β-oxidation during myelin phagocytosis and pro-inflammatory activation, and that this response is impaired in peripheral macrophages and phagocytes in brain white matter lesions in MS patients. The pharmacological targeting of VLCFA metabolism and peroxisomes in innate immune cells could be favorable in the context of neuroinflammation and neurodegeneration. We previously identified the epigenetic histone deacetylase (HDAC) inhibitors entinostat and vorinostat to enhance VLCFA degradation and pro-regenerative macrophage polarization. However, adverse side effects currently limit their use in chronic neuroinflammation. Here, we focused on tefinostat, a monocyte/macrophage-selective HDAC inhibitor that has shown reduced toxicity in clinical trials. By using a gene expression analysis, peroxisomal β-oxidation assay, and live imaging of primary human macrophages, we assessed the efficacy of tefinostat in modulating VLCFA metabolism, phagocytosis, chemotaxis, and immune function. Our results revealed the significant stimulation of VLCFA degradation with the upregulation of genes involved in peroxisomal β-oxidation and interference with immune cell recruitment; however, tefinostat was less potent than the class I HDAC-selective inhibitor entinostat in promoting a regenerative macrophage phenotype. Further research is needed to fully explore the potential of class I HDAC inhibition and downstream targets in the context of neuroinflammation.
Collapse
Affiliation(s)
- Andrea Villoria-González
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria; (A.V.-G.)
| | - Bettina Zierfuss
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria; (A.V.-G.)
- Department of Neuroscience, Centre de Recherche du CHUM, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Patricia Parzer
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria; (A.V.-G.)
| | - Elisabeth Heuböck
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria; (A.V.-G.)
| | - Violetta Zujovic
- Institut du Cerveau—Paris Brain Institute—ICM, Inserm, CNRS, APHP, Hôpital Pitié Salpétrière—University Hospital, Sorbonne University, DMU Neuroscience 6, 75013 Paris, France
| | - Petra Waidhofer-Söllner
- Division of Immune Receptors and T Cell Activation, Institute of Immunology Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Markus Ponleitner
- Department of Neurology, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, 1090 Vienna, Austria
| | - Paulus Rommer
- Department of Neurology, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, 1090 Vienna, Austria
| | - Jens Göpfert
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute, University of Tübingen, 72770 Reutlingen, Germany
| | - Sonja Forss-Petter
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria; (A.V.-G.)
| | - Johannes Berger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria; (A.V.-G.)
| | - Isabelle Weinhofer
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria; (A.V.-G.)
| |
Collapse
|
27
|
Poisson LM, Kaur N, Felicella MM, Singh J. System-based integrated metabolomics and microRNA analysis identifies potential molecular alterations in human X-linked cerebral adrenoleukodystrophy brain. Hum Mol Genet 2023; 32:3249-3262. [PMID: 37656183 PMCID: PMC10656705 DOI: 10.1093/hmg/ddad144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 08/24/2023] [Indexed: 09/02/2023] Open
Abstract
X-linked adrenoleukodystrophy is a severe demyelinating neurodegenerative disease mainly affecting males. The severe cerebral adrenoleukodystrophy (cALD) phenotype has a poor prognosis and underlying mechanism of onset and progression of neuropathology remains poorly understood. In this study we aim to integrate metabolomic and microRNA (miRNA) datasets to identify variances associated with cALD. Postmortem brain tissue samples from five healthy controls (CTL) and five cALD patients were utilized in this study. White matter from ALD patients was obtained from normal-appearing areas, away from lesions (NLA) and from the periphery of lesions- plaque shadow (PLS). Metabolomics was performed by gas chromatography coupled with time-of-flight mass spectrometry and miRNA expression analysis was performed by next generation sequencing (RNAseq). Principal component analysis revealed that among the three sample groups (CTL, NLA and PLS) there were 19 miRNA, including several novel miRNA, of which 17 were increased with disease severity and 2 were decreased. Untargeted metabolomics revealed 13 metabolites with disease severity-related patterns with 7 increased and 6 decreased with disease severity. Ingenuity pathway analysis of differentially altered metabolites and miRNA comparing CTL with NLA and NLA with PLS, identified several hubs of metabolite and signaling molecules and their upstream regulation by miRNA. The transomic approach to map the crosstalk between miRNA and metabolomics suggests involvement of specific molecular and metabolic pathways in cALD and offers opportunity to understand the complex underlying mechanism of disease severity in cALD.
Collapse
Affiliation(s)
- Laila M Poisson
- Department of Public Health Science, Henry Ford Hospital, 2799 West Grand Blvd, Detroit, Michigan 48202, United States
| | - Navtej Kaur
- Department of Neurology, Henry Ford Hospital, 2799 West Grand Blvd, Detroit, Michigan 48202, United States
| | - Michelle M Felicella
- Department of Pathology, Henry Ford Hospital, 2799 West Grand Blvd, Detroit, Michigan 48202, United States
| | - Jaspreet Singh
- Department of Neurology, Henry Ford Hospital, 2799 West Grand Blvd, Detroit, Michigan 48202, United States
| |
Collapse
|
28
|
Weinhofer I, Rommer P, Gleiss A, Ponleitner M, Zierfuss B, Waidhofer-Söllner P, Fourcade S, Grabmeier-Pfistershammer K, Reinert MC, Göpfert J, Heine A, Yska HAF, Casasnovas C, Cantarín V, Bergner CG, Mallack E, Forss-Petter S, Aubourg P, Bley A, Engelen M, Eichler F, Lund TC, Pujol A, Köhler W, Kühl JS, Berger J. Biomarker-based risk prediction for the onset of neuroinflammation in X-linked adrenoleukodystrophy. EBioMedicine 2023; 96:104781. [PMID: 37683329 PMCID: PMC10497986 DOI: 10.1016/j.ebiom.2023.104781] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/21/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
BACKGROUND X-linked adrenoleukodystrophy (X-ALD) is highly variable, ranging from slowly progressive adrenomyeloneuropathy to severe brain demyelination and inflammation (cerebral ALD, CALD) affecting males with childhood peak onset. Risk models integrating blood-based biomarkers to indicate CALD onset, enabling timely interventions, are lacking. Therefore, we evaluated the prognostic value of blood biomarkers in addition to current neuroimaging predictors for early detection of CALD. METHODS We measured blood biomarkers in a retrospective, male CALD risk-assessment cohort consisting of 134 X-ALD patients and 66 controls and in a phenotype-blinded validation set (25 X-ALD boys, 4-13 years) using Simoa®and Luminex® technologies. FINDINGS Among 25 biomarkers indicating axonal damage, astrocye/microglia activation, or immune-cell recruitment, neurofilament light chain (NfL) had the highest prognostic value for early indication of childhood/adolescent CALD. A plasma NfL cut-off level of 8.33 pg/mL, determined in the assessment cohort, correctly discriminated CALD with an accuracy of 96% [95% CI: 80-100] in the validation group. Multivariable logistic regression models revealed that combining NfL with GFAP or cytokines/chemokines (IL-15, IL-12p40, CXCL8, CCL11, CCL22, and IL-4) that were significantly elevated in CALD vs healthy controls had no additional benefit for detecting neuroinflammation. Some cytokines/chemokines were elevated only in childhood/adolescent CALD and already upregulated in asymptomatic X-ALD children (IL-15, IL-12p40, and CCL7). In adults, NfL levels distinguished CALD but were lower than in childhood/adolescent CALD patients with similar (MRI) lesion severity. Blood GFAP did not differentiate CALD from non-inflammatory X-ALD. INTERPRETATION Biomarker-based risk prediction with a plasma NfL cut-off value of 8.33 pg/mL, determined by ROC analysis, indicates CALD onset with high sensitivity and specificity in childhood X-ALD patients. A specific pro-inflammatory cytokine/chemokine profile in asymptomatic X-ALD boys may indicate a primed, immanent inflammatory state aligning with peak onset of CALD. Age-related differences in biomarker levels in adult vs childhood CALD patients warrants caution in predicting onset and progression of CALD in adults. Further evaluations are needed to assess clinical utility of the NfL cut-off for risk prognosis of CALD onset. FUNDING Austrian Science Fund, European Leukodystrophy Association.
Collapse
Affiliation(s)
- Isabelle Weinhofer
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria.
| | - Paulus Rommer
- Department of Neurology, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Andreas Gleiss
- Institute of Clinical Biometrics, Center for Medical Data Science, Medical University of Vienna, Vienna, Austria
| | - Markus Ponleitner
- Department of Neurology, Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Vienna, Austria
| | - Bettina Zierfuss
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria; Department of Neuroscience, Centre de Recherche du CHUM, Université de Montréal, Montréal, Canada
| | - Petra Waidhofer-Söllner
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Austria
| | - Stéphane Fourcade
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Catalonia, Spain; Biomedical Research Networking Center on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Katharina Grabmeier-Pfistershammer
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Austria
| | - Marie-Christine Reinert
- Division of Pediatric Neurology, Department of Pediatrics and Adolescent Medicine, University Medical Center Göttingen, Göttingen, Germany
| | - Jens Göpfert
- Applied Biomarkers and Immunoassays Working Group, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Anne Heine
- Applied Biomarkers and Immunoassays Working Group, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Hemmo A F Yska
- Department of Pediatric Neurology, Amsterdam Public Health, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Carlos Casasnovas
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Catalonia, Spain; Biomedical Research Networking Center on Rare Diseases (CIBERER), ISCIII, Madrid, Spain; Neuromuscular Unit, Neurology Department, Hospital Universitario Bellvitge, Bellvitge Biomedical Research Unit, Barcelona, Spain
| | - Verónica Cantarín
- Infant Jesus Children´s Hospital and Biomedical Research Networking Center on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Caroline G Bergner
- Department of Neurology, Leukodystrophy Clinic, University of Leipzig Medical Center, Leipzig, Germany
| | - Eric Mallack
- Leukodystrophy Center, Division of Child Neurology, Department of Pediatrics, Weill Cornell Medical College, NewYork-Presbyterian Hospital, New York, NY, USA
| | - Sonja Forss-Petter
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Patrick Aubourg
- Kremlin-Bicêtre-Hospital, University Paris-Saclay, Paris, France
| | - Annette Bley
- Department of Pediatrics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Marc Engelen
- Department of Pediatric Neurology, Amsterdam Public Health, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - Florian Eichler
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Troy C Lund
- Pediatric Blood and Marrow Transplant Program, Global Pediatrics, Division of Pediatric Blood and Marrow Transplantation, MCRB, University of Minnesota, Minneapolis, MN, USA
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Catalonia, Spain; Biomedical Research Networking Center on Rare Diseases (CIBERER), ISCIII, Madrid, Spain
| | - Wolfgang Köhler
- Department of Neurology, Leukodystrophy Clinic, University of Leipzig Medical Center, Leipzig, Germany
| | - Jörn-Sven Kühl
- Department of Pediatric Oncology, Hematology and Hemostaseology, University Hospital Leipzig, Leipzig, Germany
| | - Johannes Berger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
29
|
Buda A, Forss-Petter S, Hua R, Jaspers Y, Lassnig M, Waidhofer-Söllner P, Kemp S, Kim P, Weinhofer I, Berger J. ABCD1 Transporter Deficiency Results in Altered Cholesterol Homeostasis. Biomolecules 2023; 13:1333. [PMID: 37759733 PMCID: PMC10526550 DOI: 10.3390/biom13091333] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 08/25/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
X-linked adrenoleukodystrophy (X-ALD), the most common peroxisomal disorder, is caused by mutations in the peroxisomal transporter ABCD1, resulting in the accumulation of very long-chain fatty acids (VLCFA). Strongly affected cell types, such as oligodendrocytes, adrenocortical cells and macrophages, exhibit high cholesterol turnover. Here, we investigated how ABCD1 deficiency affects cholesterol metabolism in human X-ALD patient-derived fibroblasts and CNS tissues of Abcd1-deficient mice. Lipidome analyses revealed increased levels of cholesterol esters (CE), containing both saturated VLCFA and mono/polyunsaturated (V)LCFA. The elevated CE(26:0) and CE(26:1) levels remained unchanged in LXR agonist-treated Abcd1 KO mice despite reduced total C26:0. Under high-cholesterol loading, gene expression of SOAT1, converting cholesterol to CE and lipid droplet formation were increased in human X-ALD fibroblasts versus healthy control fibroblasts. However, the expression of NCEH1, catalysing CE hydrolysis and the cholesterol transporter ABCA1 and cholesterol efflux were also upregulated. Elevated Soat1 and Abca1 expression and lipid droplet content were confirmed in the spinal cord of X-ALD mice, where expression of the CNS cholesterol transporter Apoe was also elevated. The extent of peroxisome-lipid droplet co-localisation appeared low and was not impaired by ABCD1-deficiency in cholesterol-loaded primary fibroblasts. Finally, addressing steroidogenesis, progesterone-induced cortisol release was amplified in X-ALD fibroblasts. These results link VLCFA to cholesterol homeostasis and justify further consideration of therapeutic approaches towards reducing VLCFA and cholesterol levels in X-ALD.
Collapse
Affiliation(s)
- Agnieszka Buda
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Sonja Forss-Petter
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Rong Hua
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5G 1A8, Canada
| | - Yorrick Jaspers
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Centers, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Mark Lassnig
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Petra Waidhofer-Söllner
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Centers, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Peter Kim
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5G 1A8, Canada
| | - Isabelle Weinhofer
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Johannes Berger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
30
|
Martinović K, Bauer J, Kunze M, Berger J, Forss-Petter S. Abcd1 deficiency accelerates cuprizone-induced oligodendrocyte loss and axonopathy in a demyelinating mouse model of X-linked adrenoleukodystrophy. Acta Neuropathol Commun 2023; 11:98. [PMID: 37331971 PMCID: PMC10276915 DOI: 10.1186/s40478-023-01595-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 05/30/2023] [Indexed: 06/20/2023] Open
Abstract
X-linked adrenoleukodystrophy (X-ALD), the most frequent, inherited peroxisomal disease, is caused by mutations in the ABCD1 gene encoding a peroxisomal lipid transporter importing very long-chain fatty acids (VLCFAs) from the cytosol into peroxisomes for degradation via β-oxidation. ABCD1 deficiency results in accumulation of VLCFAs in tissues and body fluids of X-ALD patients with a wide range of phenotypic manifestations. The most severe variant, cerebral X-ALD (CALD) is characterized by progressive inflammation, loss of the myelin-producing oligodendrocytes and demyelination of the cerebral white matter. Whether the oligodendrocyte loss and demyelination in CALD are caused by a primary cell autonomous defect or injury to oligodendrocytes or by a secondary effect of the inflammatory reaction remains unresolved. To address the role of X-ALD oligodendrocytes in demyelinating pathophysiology, we combined the Abcd1 deficient X-ALD mouse model, in which VLCFAs accumulate without spontaneous demyelination, with the cuprizone model of toxic demyelination. In mice, the copper chelator cuprizone induces reproducible demyelination in the corpus callosum, followed by remyelination upon cuprizone removal. By immunohistochemical analyses of oligodendrocytes, myelin, axonal damage and microglia activation during de-and remyelination, we found that the mature oligodendrocytes of Abcd1 KO mice are more susceptible to cuprizone-induced cell death compared to WT mice in the early demyelinating phase. Furthermore, this effect was mirrored by a greater extent of acute axonal damage during demyelination in the KO mice. Abcd1 deficiency did not affect the function of microglia in either phase of the treatment. Also, the proliferation and differentiation of oligodendrocyte precursor cells and remyelination progressed at similar rates in both genotypes. Taken together, our findings point to an effect of Abcd1 deficiency on mature oligodendrocytes and the oligodendrocyte-axon unit, leading to increased vulnerability in the context of a demyelinating insult.
Collapse
Affiliation(s)
- Ksenija Martinović
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| | - Jan Bauer
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| | - Markus Kunze
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| | - Johannes Berger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| | - Sonja Forss-Petter
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| |
Collapse
|
31
|
Ishikawa R, Nakamori M, Takenaka M, Aoki S, Yamazaki Y, Hashiguchi A, Takashima H, Maruyama H. Case report: Mitochondrial trifunctional protein deficiency caused by HADHB gene mutation (c.1175C>T) characterized by higher brain dysfunction followed by neuropathy, presented gadolinium enhancement on brain imaging in an adult patient. Front Neurol 2023; 14:1187822. [PMID: 37388542 PMCID: PMC10299898 DOI: 10.3389/fneur.2023.1187822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 05/23/2023] [Indexed: 07/01/2023] Open
Abstract
Mitochondrial trifunctional protein (MTP) deficiency is an autosomal recessive disorder caused by impaired metabolism of long-chain fatty acids (LCFAs). Childhood and late-onset MTP deficiency is characterized by myopathy/rhabdomyolysis and peripheral neuropathy; however, the features are unclear. A 44-year-old woman was clinically diagnosed with Charcot-Marie-Tooth disease at 3 years of age due to gait disturbance. Her activity and voluntary speech gradually decreased in her 40s. Cognitive function was evaluated and brain imaging tests were performed. The Mini-Mental State Examination and frontal assessment battery scores were 25/30 and 10/18, respectively, suggesting higher brain dysfunction. Peripheral nerve conduction studies revealed axonal impairments. Brain computed tomography showed significant calcification. Magnetic resonance imaging revealed an increased gadolinium contrast-enhanced signal in the white matter, suggesting demyelination of the central nervous system (CNS) due to LCFAs. The diagnosis of MTP deficiency was confirmed through genetic examination. Administration of L-carnitine and a medium-chain fatty triglyceride diet was initiated, and the progression of higher brain dysfunction was retarded within 1 year. This patient's presentation was suggestive of CNS demyelination. The presence of brain calcification, higher brain dysfunction, or gadolinium enhancement in the white matter in patients with peripheral neuropathy may be suggestive of MTP deficiency.
Collapse
Affiliation(s)
- Ruoyi Ishikawa
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Masahiro Nakamori
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Megumi Takenaka
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Shiro Aoki
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Yu Yamazaki
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| | - Akihiro Hashiguchi
- Department of Neurology and Geriatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Hiroshi Takashima
- Department of Neurology and Geriatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Hirofumi Maruyama
- Department of Clinical Neuroscience and Therapeutics, Hiroshima University Graduate School of Biomedical and Health Sciences, Hiroshima, Japan
| |
Collapse
|
32
|
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
|
33
|
Affiliation(s)
- Vivian Szymczuk
- Metabolic Bone Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA; Pediatric Endocrinology Inter-Institute Training Program, National Institute of Child Health and Development, National Institutes of Health, Bethesda, MD, USA
| | - Nadia Merchant
- Division of Endocrinology, Children's National Hospital, Washington, DC, USA; Department of Pediatrics, George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
34
|
Wanders RJA, Baes M, Ribeiro D, Ferdinandusse S, Waterham HR. The physiological functions of human peroxisomes. Physiol Rev 2023; 103:957-1024. [PMID: 35951481 DOI: 10.1152/physrev.00051.2021] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Peroxisomes are subcellular organelles that play a central role in human physiology by catalyzing a range of unique metabolic functions. The importance of peroxisomes for human health is exemplified by the existence of a group of usually severe diseases caused by an impairment in one or more peroxisomal functions. Among others these include the Zellweger spectrum disorders, X-linked adrenoleukodystrophy, and Refsum disease. To fulfill their role in metabolism, peroxisomes require continued interaction with other subcellular organelles including lipid droplets, lysosomes, the endoplasmic reticulum, and mitochondria. In recent years it has become clear that the metabolic alliance between peroxisomes and other organelles requires the active participation of tethering proteins to bring the organelles physically closer together, thereby achieving efficient transfer of metabolites. This review intends to describe the current state of knowledge about the metabolic role of peroxisomes in humans, with particular emphasis on the metabolic partnership between peroxisomes and other organelles and the consequences of genetic defects in these processes. We also describe the biogenesis of peroxisomes and the consequences of the multiple genetic defects therein. In addition, we discuss the functional role of peroxisomes in different organs and tissues and include relevant information derived from model systems, notably peroxisomal mouse models. Finally, we pay particular attention to a hitherto underrated role of peroxisomes in viral infections.
Collapse
Affiliation(s)
- Ronald J A Wanders
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,United for Metabolic Diseases, Amsterdam, The Netherlands
| | - Myriam Baes
- Laboratory of Cell Metabolism, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Daniela Ribeiro
- Institute of Biomedicine (iBiMED) and Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Sacha Ferdinandusse
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,United for Metabolic Diseases, Amsterdam, The Netherlands
| | - Hans R Waterham
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands.,United for Metabolic Diseases, Amsterdam, The Netherlands
| |
Collapse
|
35
|
Zierfuss B, Buda A, Villoria-González A, Logist M, Fabjan J, Parzer P, Battin C, Vandersteene S, Dijkstra IME, Waidhofer-Söllner P, Grabmeier-Pfistershammer K, Steinberger P, Kemp S, Forss-Petter S, Berger J, Weinhofer I. Saturated very long-chain fatty acids regulate macrophage plasticity and invasiveness. J Neuroinflammation 2022; 19:305. [PMID: 36528616 PMCID: PMC9759912 DOI: 10.1186/s12974-022-02664-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/04/2022] [Indexed: 12/23/2022] Open
Abstract
Saturated very long-chain fatty acids (VLCFA, ≥ C22), enriched in brain myelin and innate immune cells, accumulate in X-linked adrenoleukodystrophy (X-ALD) due to inherited dysfunction of the peroxisomal VLCFA transporter ABCD1. In its severest form, X-ALD causes cerebral myelin destruction with infiltration of pro-inflammatory skewed monocytes/macrophages. How VLCFA levels relate to macrophage activation is unclear. Here, whole transcriptome sequencing of X-ALD macrophages indicated that VLCFAs prime human macrophage membranes for inflammation and increased expression of factors involved in chemotaxis and invasion. When added externally to mimic lipid release in demyelinating X-ALD lesions, VLCFAs did not activate toll-like receptors in primary macrophages. In contrast, VLCFAs provoked pro-inflammatory responses through scavenger receptor CD36-mediated uptake, cumulating in JNK signalling and expression of matrix-degrading enzymes and chemokine release. Following pro-inflammatory LPS activation, VLCFA levels increased also in healthy macrophages. With the onset of the resolution, VLCFAs were rapidly cleared in control macrophages by increased peroxisomal VLCFA degradation through liver-X-receptor mediated upregulation of ABCD1. ABCD1 deficiency impaired VLCFA homeostasis and prolonged pro-inflammatory gene expression upon LPS treatment. Our study uncovers a pivotal role for ABCD1, a protein linked to neuroinflammation, and associated peroxisomal VLCFA degradation in regulating macrophage plasticity.
Collapse
Affiliation(s)
- Bettina Zierfuss
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
- Department of Neuroscience, Centre de Recherche du CHUM, Université de Montréal, Montréal, H2X 0A9, Canada
| | - Agnieszka Buda
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Andrea Villoria-González
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Maxime Logist
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
- Department of Chronic Diseases and Metabolism, Translational Research in GastroIntestinal Disorders, KU Leuven, 3000, Leuven, Belgium
| | - Jure Fabjan
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Patricia Parzer
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Claire Battin
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Streggi Vandersteene
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Inge M E Dijkstra
- Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Petra Waidhofer-Söllner
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Katharina Grabmeier-Pfistershammer
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Peter Steinberger
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090, Vienna, Austria
| | - Stephan Kemp
- Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Sonja Forss-Petter
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Johannes Berger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria
| | - Isabelle Weinhofer
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090, Vienna, Austria.
| |
Collapse
|
36
|
Chegodaev D, Gusev V, Lvova O, Pavlova P. Possible role of ketone bodies in the generation of burst suppression electroencephalographic pattern. Front Neurosci 2022; 16:1021035. [PMID: 36590288 PMCID: PMC9800049 DOI: 10.3389/fnins.2022.1021035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
|
37
|
Ebrahimi-Fakhari D, Saffari A, Pearl PL. Childhood-onset hereditary spastic paraplegia and its treatable mimics. Mol Genet Metab 2022; 137:436-444. [PMID: 34183250 PMCID: PMC8843241 DOI: 10.1016/j.ymgme.2021.06.006] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 06/18/2021] [Accepted: 06/19/2021] [Indexed: 12/24/2022]
Abstract
Early-onset forms of hereditary spastic paraplegia and inborn errors of metabolism that present with spastic diplegia are among the most common "mimics" of cerebral palsy. Early detection of these heterogenous genetic disorders can inform genetic counseling, anticipatory guidance, and improve outcomes, particularly where specific treatments exist. The diagnosis relies on clinical pattern recognition, biochemical testing, neuroimaging, and increasingly next-generation sequencing-based molecular testing. In this short review, we summarize the clinical and molecular understanding of: 1) childhood-onset and complex forms of hereditary spastic paraplegia (SPG5, SPG7, SPG11, SPG15, SPG35, SPG47, SPG48, SPG50, SPG51, SPG52) and, 2) the most common inborn errors of metabolism that present with phenotypes that resemble hereditary spastic paraplegia.
Collapse
Affiliation(s)
- Darius Ebrahimi-Fakhari
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; The Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA.
| | - Afshin Saffari
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Division of Child Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Phillip L Pearl
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
38
|
Jia Y, Zhang Y, Wang W, Lei J, Ying Z, Yang G. Structural and functional insights of the human peroxisomal ABC transporter ALDP. eLife 2022; 11:e75039. [PMID: 36374178 PMCID: PMC9683791 DOI: 10.7554/elife.75039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
Adrenoleukodystrophy protein (ALDP) is responsible for the transport of very-long-chain fatty acids (VLCFAs) and corresponding CoA-esters across the peroxisomal membrane. Dysfunction of ALDP leads to peroxisomal metabolic disorder exemplified by X-linked adrenoleukodystrophy (ALD). Hundreds of ALD-causing mutations have been identified on ALDP. However, the pathogenic mechanisms of these mutations are restricted to clinical description due to limited structural and biochemical characterization. Here we report the cryo-electron microscopy structure of human ALDP with nominal resolution at 3.4 Å. ALDP exhibits a cytosolic-facing conformation. Compared to other lipid ATP-binding cassette transporters, ALDP has two substrate binding cavities formed by the transmembrane domains. Such structural organization may be suitable for the coordination of VLCFAs. Based on the structure, we performed integrative analysis of the cellular trafficking, protein thermostability, ATP hydrolysis, and the transport activity of representative mutations. These results provide a framework for understanding the working mechanism of ALDP and pathogenic roles of disease-associated mutations.
Collapse
Affiliation(s)
- Yutian Jia
- State Key Laboratory for Agrobiotechnology, Department of Nutrition and Health, College of Biological Sciences, China Agricultural UniversityBeijingChina
| | - Yanming Zhang
- State Key Laboratory for Agrobiotechnology, Department of Nutrition and Health, College of Biological Sciences, China Agricultural UniversityBeijingChina
| | - Wenhao Wang
- State Key Laboratory for Agrobiotechnology, Department of Nutrition and Health, College of Biological Sciences, China Agricultural UniversityBeijingChina
| | - Jianlin Lei
- Technology Center for Protein Sciences, Ministry of Education Key Laboratory of Protein Sciences, School of Life Sciences, Tsinghua UniversityBeijingChina
| | - Zhengxin Ying
- State Key Laboratory for Agrobiotechnology, Department of Nutrition and Health, College of Biological Sciences, China Agricultural UniversityBeijingChina
| | - Guanghui Yang
- State Key Laboratory for Agrobiotechnology, Department of Nutrition and Health, College of Biological Sciences, China Agricultural UniversityBeijingChina
| |
Collapse
|
39
|
Shim YJ, Shin MK, Jung J, Koo B, Jang W. An in-silico approach to studying a very rare neurodegenerative disease using a disease with higher prevalence with shared pathways and genes: Cerebral adrenoleukodystrophy and Alzheimer’s disease. Front Mol Neurosci 2022; 15:996698. [PMID: 36245924 PMCID: PMC9553843 DOI: 10.3389/fnmol.2022.996698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 08/22/2022] [Indexed: 11/20/2022] Open
Abstract
Cerebral adrenoleukodystrophy (cALD) is a rare neurodegenerative disease characterized by inflammatory demyelination in the central nervous system. Another neurodegenerative disease with a high prevalence, Alzheimer’s disease (AD), shares many common features with cALD such as cognitive impairment and the alleviation of symptoms by erucic acid. We investigated cALD and AD in parallel to study the shared pathological pathways between a rare disease and a more common disease. The approach may expand the biological understandings and reveal novel therapeutic targets. Gene set enrichment analysis (GSEA) and weighted gene correlation network analysis (WGCNA) were conducted to identify both the resemblance in gene expression patterns and genes that are pathologically relevant in the two diseases. Within differentially expressed genes (DEGs), GSEA identified 266 common genes with similar up- or down-regulation patterns in cALD and AD. Among the interconnected genes in AD data, two gene sets containing 1,486 genes preserved in cALD data were selected by WGCNA that may significantly affect the development and progression of cALD. WGCNA results filtered by functional correlation via protein–protein interaction analysis overlapping with GSEA revealed four genes (annexin A5, beta-2-microglobulin, CD44 molecule, and fibroblast growth factor 2) that showed robust associations with the pathogeneses of cALD and AD, where they were highly involved in inflammation, apoptosis, and the mitogen-activated protein kinase pathway. This study provided an integrated strategy to provide new insights into a rare disease with scant publicly available data (cALD) using a more prevalent disorder with some pathological association (AD), which suggests novel druggable targets and drug candidates.
Collapse
Affiliation(s)
- Yu Jeong Shim
- Department of Life Science, Dongguk University, Goyang-si, South Korea
| | - Min Kyoung Shin
- Department of Life Science, Dongguk University, Goyang-si, South Korea
| | - Junghyun Jung
- Department of Life Science, Dongguk University, Goyang-si, South Korea
| | | | - Wonhee Jang
- Department of Life Science, Dongguk University, Goyang-si, South Korea
- *Correspondence: Wonhee Jang,
| |
Collapse
|
40
|
Weinhofer I, Buda A, Kunze M, Palfi Z, Traunfellner M, Hesse S, Villoria-Gonzalez A, Hofmann J, Hametner S, Regelsberger G, Moser AB, Eichler F, Kemp S, Bauer J, Kühl JS, Forss-Petter S, Berger J. Peroxisomal very long-chain fatty acid transport is targeted by herpesviruses and the antiviral host response. Commun Biol 2022; 5:944. [PMID: 36085307 PMCID: PMC9462615 DOI: 10.1038/s42003-022-03867-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 08/22/2022] [Indexed: 11/25/2022] Open
Abstract
Very long-chain fatty acids (VLCFA) are critical for human cytomegalovirus replication and accumulate upon infection. Here, we used Epstein-Barr virus (EBV) infection of human B cells to elucidate how herpesviruses target VLCFA metabolism. Gene expression profiling revealed that, despite a general induction of peroxisome-related genes, EBV early infection decreased expression of the peroxisomal VLCFA transporters ABCD1 and ABCD2, thus impairing VLCFA degradation. The mechanism underlying ABCD1 and ABCD2 repression involved RNA interference by the EBV-induced microRNAs miR-9-5p and miR-155, respectively, causing significantly increased VLCFA levels. Treatment with 25-hydroxycholesterol, an antiviral innate immune modulator produced by macrophages, restored ABCD1 expression and reduced VLCFA accumulation in EBV-infected B-lymphocytes, and, upon lytic reactivation, reduced virus production in control but not ABCD1-deficient cells. Finally, also other herpesviruses and coronaviruses target ABCD1 expression. Because viral infection might trigger neuroinflammation in X-linked adrenoleukodystrophy (X-ALD, inherited ABCD1 deficiency), we explored a possible link between EBV infection and cerebral X-ALD. However, neither immunohistochemistry of post-mortem brains nor analysis of EBV seropositivity in 35 X-ALD children supported involvement of EBV in the onset of neuroinflammation. Collectively, our findings indicate a previously unrecognized, pivotal role of ABCD1 in viral infection and host defence, prompting consideration of other viral triggers in cerebral X-ALD.
Collapse
Affiliation(s)
- Isabelle Weinhofer
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria.
| | - Agnieszka Buda
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Markus Kunze
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Zsofia Palfi
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Matthäus Traunfellner
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Sarah Hesse
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
- Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow, UK
| | - Andrea Villoria-Gonzalez
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Jörg Hofmann
- Institute of Virology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Simon Hametner
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Günther Regelsberger
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Vienna, Austria
| | - Ann B Moser
- Department of Neurogenetics, Hugo W. Moser Research Institute at Kennedy Krieger, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Florian Eichler
- Department of Neurology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Stephan Kemp
- Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam Gastroenterology Endocrinology Metabolism, University of Amsterdam, Amsterdam, The Netherlands
| | - Jan Bauer
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Jörn-Sven Kühl
- Department of Pediatric Oncology, Hematology, and Hemostaseology, University Hospital Leipzig, Leipzig, Germany
| | - Sonja Forss-Petter
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Johannes Berger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
41
|
Nowacki JC, Fields AM, Fu MM. Emerging cellular themes in leukodystrophies. Front Cell Dev Biol 2022; 10:902261. [PMID: 36003149 PMCID: PMC9393611 DOI: 10.3389/fcell.2022.902261] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 06/30/2022] [Indexed: 11/18/2022] Open
Abstract
Leukodystrophies are a broad spectrum of neurological disorders that are characterized primarily by deficiencies in myelin formation. Clinical manifestations of leukodystrophies usually appear during childhood and common symptoms include lack of motor coordination, difficulty with or loss of ambulation, issues with vision and/or hearing, cognitive decline, regression in speech skills, and even seizures. Many cases of leukodystrophy can be attributed to genetic mutations, but they have diverse inheritance patterns (e.g., autosomal recessive, autosomal dominant, or X-linked) and some arise from de novo mutations. In this review, we provide an updated overview of 35 types of leukodystrophies and focus on cellular mechanisms that may underlie these disorders. We find common themes in specialized functions in oligodendrocytes, which are specialized producers of membranes and myelin lipids. These mechanisms include myelin protein defects, lipid processing and peroxisome dysfunction, transcriptional and translational dysregulation, disruptions in cytoskeletal organization, and cell junction defects. In addition, non-cell-autonomous factors in astrocytes and microglia, such as autoimmune reactivity, and intercellular communication, may also play a role in leukodystrophy onset. We hope that highlighting these themes in cellular dysfunction in leukodystrophies may yield conceptual insights on future therapeutic approaches.
Collapse
|
42
|
Volmrich AM, Cuénant LM, Forghani I, Hsieh SL, Shapiro LT. ABCD1 Gene Mutations: Mechanisms and Management of Adrenomyeloneuropathy. Appl Clin Genet 2022; 15:111-123. [PMID: 35983253 PMCID: PMC9381027 DOI: 10.2147/tacg.s359479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 08/06/2022] [Indexed: 01/05/2023] Open
Abstract
Pathogenic variants in the ABCD1 gene on the X chromosome may result in widely heterogenous phenotypes, including adrenomyeloneuropathy (AMN). Affected males typically present in their third or fourth decade of life with progressive lower limb weakness and spasticity, and may develop signs and symptoms of adrenal insufficiency and/or cerebral demyelination. Heterozygous females may be asymptomatic, but may develop a later-onset and more slowly progressive spastic paraparesis. In this review, we describe the clinical presentation of AMN, as well as its diagnosis and management. The role of rehabilitative therapies and options for management of spasticity are highlighted.
Collapse
Affiliation(s)
- Alyssa M Volmrich
- Department of Physical Medicine & Rehabilitation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lauren M Cuénant
- Department of Physical Medicine & Rehabilitation, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Irman Forghani
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sharon L Hsieh
- MD/MPH Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lauren T Shapiro
- Department of Physical Medicine & Rehabilitation, University of Miami Miller School of Medicine, Miami, FL, USA
- Correspondence: Lauren T Shapiro, Department of Physical Medicine & Rehabilitation; University of Miami Miller School of Medicine, P.O. Box 016960 (C-206), Miami, FL, 33101, USA, Tel +1 305 243-6605, Fax +1 305 243-4650, Email
| |
Collapse
|
43
|
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
|
44
|
Gong Y, Laheji F, Berenson A, Qian A, Park SO, Kok R, Selig M, Hahn R, Sadjadi R, Kemp S, Eichler F. Peroxisome Metabolism Contributes to PIEZO2-Mediated Mechanical Allodynia. Cells 2022; 11:1842. [PMID: 35681537 PMCID: PMC9180358 DOI: 10.3390/cells11111842] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 05/21/2022] [Accepted: 06/01/2022] [Indexed: 11/17/2022] Open
Abstract
Mutations in the peroxisomal half-transporter ABCD1 cause X-linked adrenoleukodystrophy, resulting in elevated very long-chain fatty acids (VLCFA), progressive neurodegeneration and an associated pain syndrome that is poorly understood. In the nervous system of mice, we found ABCD1 expression to be highest in dorsal root ganglia (DRG), with satellite glial cells (SGCs) displaying higher expression than neurons. We subsequently examined sensory behavior and DRG pathophysiology in mice deficient in ABCD1 compared to wild-type mice. Beginning at 8 months of age, Abcd1-/y mice developed persistent mechanical allodynia. DRG had a greater number of IB4-positive nociceptive neurons expressing PIEZO2, the mechanosensitive ion channel. Blocking PIEZO2 partially rescued the mechanical allodynia. Beyond affecting neurons, ABCD1 deficiency impacted SGCs, as demonstrated by high levels of VLCFA, increased glial fibrillary acidic protein (GFAP), as well as genes disrupting neuron-SGC connectivity. These findings suggest that lack of the peroxisomal half-transporter ABCD1 leads to PIEZO2-mediated mechanical allodynia as well as SGC dysfunction. Given the known supportive role of SGCs to neurons, this elucidates a novel mechanism underlying pain in X-linked adrenoleukodystrophy.
Collapse
Affiliation(s)
- Yi Gong
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.G.); (F.L.); (A.B.); (A.Q.); (S.-O.P.); (M.S.); (R.H.); (R.S.)
| | - Fiza Laheji
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.G.); (F.L.); (A.B.); (A.Q.); (S.-O.P.); (M.S.); (R.H.); (R.S.)
| | - Anna Berenson
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.G.); (F.L.); (A.B.); (A.Q.); (S.-O.P.); (M.S.); (R.H.); (R.S.)
| | - April Qian
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.G.); (F.L.); (A.B.); (A.Q.); (S.-O.P.); (M.S.); (R.H.); (R.S.)
| | - Sang-O Park
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.G.); (F.L.); (A.B.); (A.Q.); (S.-O.P.); (M.S.); (R.H.); (R.S.)
| | - Rene Kok
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, Amsterdam Gastroenterology Endocrinology Metabolism, University of Amsterdam, 1105 Amsterdam, The Netherlands; (R.K.); (S.K.)
| | - Martin Selig
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.G.); (F.L.); (A.B.); (A.Q.); (S.-O.P.); (M.S.); (R.H.); (R.S.)
| | - Ryan Hahn
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.G.); (F.L.); (A.B.); (A.Q.); (S.-O.P.); (M.S.); (R.H.); (R.S.)
| | - Reza Sadjadi
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.G.); (F.L.); (A.B.); (A.Q.); (S.-O.P.); (M.S.); (R.H.); (R.S.)
| | - Stephan Kemp
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Center, Amsterdam Gastroenterology Endocrinology Metabolism, University of Amsterdam, 1105 Amsterdam, The Netherlands; (R.K.); (S.K.)
- Department of Pediatric Neurology, Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam Neuroscience, University of Amsterdam, 1105 Amsterdam, The Netherlands
| | - Florian Eichler
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; (Y.G.); (F.L.); (A.B.); (A.Q.); (S.-O.P.); (M.S.); (R.H.); (R.S.)
| |
Collapse
|
45
|
Umeda K. Unresolved issues in allogeneic hematopoietic cell transplantation for non-malignant diseases. Int J Hematol 2022; 116:41-47. [PMID: 35568772 DOI: 10.1007/s12185-022-03361-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 11/27/2022]
Abstract
Allogeneic hematopoietic cell transplantation (HCT) can be curative for a variety of non-malignant diseases (NMDs) as well as hematological malignancies. However, there are several fundamental differences between HCT for NMDs and hematological malignancies, which may necessitate the use of alternative HCT strategies. For example, these diseases differ in the intensity of conditioning regimen sufficient to improve disease. In addition, patients with NMDs are at higher risk of graft failure or mixed chimerism following HCT, and gain no or little survival benefit from graft-versus-host disease. Because more than 80% of patients with NMDs become long-term survivors, greater attention has been paid to late adverse effects and decreased of quality of life after HCT. This review addresses several unresolved issues in allogeneic HCT for patients with NMDs, such as (1) stem cell source, (2) conditioning regimen, (3) use of serotherapy or low-dose irradiation, and (4) therapeutic intervention for mixed chimerism. Resolving these issues may improve transplant outcomes in patients with NMDs.
Collapse
Affiliation(s)
- Katsutsugu Umeda
- Department of Pediatrics, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
46
|
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: 18] [Impact Index Per Article: 6.0] [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
|
47
|
Practical Genetics for the Neuroradiologist: Adding Value in Neurogenetic Disease. Acad Radiol 2022; 29 Suppl 3:S1-S27. [PMID: 33495073 DOI: 10.1016/j.acra.2020.12.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/19/2020] [Accepted: 12/27/2020] [Indexed: 11/23/2022]
Abstract
Genetic discoveries have transformed our understanding of many neurologic diseases. Identification of specific causal pathogenic variants has improved understanding of pathophysiology and enabled replacement of many confusing eponyms and acronyms with more meaningful and clinically relevant genetics-based terminology. In this era of rapid scientific advancement, multidisciplinary collaboration among pediatricians, neurologists, geneticists, radiologists, and other members of the health care team is increasingly important in the care of patients with genetic neurologic diseases. Radiologists familiar with neurogenetic disease add value by (1) recognizing constellations of characteristic imaging findings that are associated with a genetic disease before one is clinically suspected; (2) predicting the most likely genotypes for a given imaging phenotype in clinically suspected genetic disease; and (3) providing detailed and accurate descriptions of the imaging phenotype in challenging cases with unknown or uncertain genotypes. This review aims to increase awareness and understanding of pathogenic variants relating to neurologic disease by (1) briefly reviewing foundational knowledge of chromosomes, inheritance patterns, and mutagenesis; (2) providing concrete examples of and detailed information about specific neurologic diseases resulting from pathogenic variants; and (3) highlighting clinical and imaging features that are of greatest relevance for the radiologist.
Collapse
|
48
|
Mekhaeil M, Dev KK, Conroy MJ. Existing Evidence for the Repurposing of PARP-1 Inhibitors in Rare Demyelinating Diseases. Cancers (Basel) 2022; 14:cancers14030687. [PMID: 35158955 PMCID: PMC8833351 DOI: 10.3390/cancers14030687] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/23/2022] [Accepted: 01/27/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Poly (ADP-ribose) polymerase-1 (PARP-1) inhibitors are successful cancer therapeutics that impair DNA repair machinery, leading to an accumulation of DNA damage and consequently cell death. The shared underlying mechanisms driving malignancy and demyelinating disease, together with the success of anticancer drugs as repurposed therapeutics, makes the repurposing of PARP-1 inhibitors for demyelinating diseases a worthy concept to consider. In addition, PARP-1 inhibitors demonstrate notable neuroprotective effects in demyelinating disorders, including multiple sclerosis which is considered the archetypical demyelinating disease. Abstract Over the past decade, Poly (ADP-ribose) polymerase-1 (PARP-1) inhibitors have arisen as a novel and promising targeted therapy for breast cancer gene (BRCA)-mutated ovarian and breast cancer patients. Therapies targeting the enzyme, PARP-1, have since established their place as maintenance drugs for cancer. Here, we present existing evidence that implicates PARP-1 as a player in the development and progression of both malignancy and demyelinating disease. These findings, together with the proven clinical efficacy and marketed success of PARP-1 inhibitors in cancer, present the repurposing of these drugs for demyelinating diseases as a desirable therapeutic concept. Indeed, PARP-1 inhibitors are noted to demonstrate neuroprotective effects in demyelinating disorders such as multiple sclerosis and Parkinson’s disease, further supporting the use of these drugs in demyelinating, neuroinflammatory, and neurodegenerative diseases. In this review, we discuss the potential for repurposing PARP-1 inhibitors, with a focus on rare demyelinating diseases. In particular, we address the possible use of PARP-1 inhibitors in examples of rare leukodystrophies, for which there are a paucity of treatment options and an urgent need for novel therapeutic approaches.
Collapse
Affiliation(s)
- Marianna Mekhaeil
- Drug Development Research Group, Department of Physiology, School of Medicine, Trinity College Dublin, D18 DH50 Dublin, Ireland; (M.M.); (K.K.D.)
- Cancer Immunology Research Group, Department of Physiology, School of Medicine, Trinity College Dublin, D18 DH50 Dublin, Ireland
| | - Kumlesh Kumar Dev
- Drug Development Research Group, Department of Physiology, School of Medicine, Trinity College Dublin, D18 DH50 Dublin, Ireland; (M.M.); (K.K.D.)
| | - Melissa Jane Conroy
- Cancer Immunology Research Group, Department of Physiology, School of Medicine, Trinity College Dublin, D18 DH50 Dublin, Ireland
- Correspondence:
| |
Collapse
|
49
|
Structures of the human peroxisomal fatty acid transporter ABCD1 in a lipid environment. Commun Biol 2022; 5:7. [PMID: 35013584 PMCID: PMC8748874 DOI: 10.1038/s42003-021-02970-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 12/13/2021] [Indexed: 01/13/2023] Open
Abstract
The peroxisomal very long chain fatty acid (VLCFA) transporter ABCD1 is central to fatty acid catabolism and lipid biosynthesis. Its dysfunction underlies toxic cytosolic accumulation of VLCFAs, progressive demyelination, and neurological impairments including X-linked adrenoleukodystrophy (X-ALD). We present cryo-EM structures of ABCD1 in phospholipid nanodiscs in a nucleotide bound conformation open to the peroxisomal lumen and an inward facing conformation open to the cytosol at up to 3.5 Å resolution, revealing details of its transmembrane cavity and ATP dependent conformational spectrum. We identify features distinguishing ABCD1 from its closest homologs and show that coenzyme A (CoA) esters of VLCFAs modulate ABCD1 activity in a species dependent manner. Our data suggest a transport mechanism where the CoA moieties of VLCFA-CoAs enter the hydrophilic transmembrane domain while the acyl chains extend out into the surrounding membrane bilayer. The structures help rationalize disease causing mutations and may aid ABCD1 targeted structure-based drug design. Le et al. present cryo-EM structures of the peroxisomal very long chain fatty acid (VLCFA) transporter ABCD1 in phospholipid nanodiscs in a nucleotide-bound conformation open to the peroximsomal lumen and a conformation open to the cytosol. These structures provide the basis for structure-function studies to investigate VLCFA transport properties, disease-causing mutations, and drug design for disorders, such as X-linked adrenoleukodystrophy, associated with ABCD1 dysfunction.
Collapse
|
50
|
Dong B, Lv W, Xu L, Zhao Y, Sun X, Wang Z, Cheng B, Fu Z, Wang Y. Identification of Two Novel Mutations of ABCD1 Gene in Pedigrees with X-Linked Adrenoleukodystrophy and Review of the Literature. Int J Endocrinol 2022; 2022:5479781. [PMID: 35479665 PMCID: PMC9038410 DOI: 10.1155/2022/5479781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/18/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND X-linked adrenoleukodystrophy (ALD) is an inherited peroxisomal metabolism disorder, resulting from the loss-of-function mutation of ATP-binding cassette protein subfamily D1 (ABCD1) gene. The dysfunction of ALD protein, a peroxisomal ATP-binding cassette transporter, results in the excessive saturated very long-chain fatty acids (VLCFAs) accumulation in organs including the brain, spine, and adrenal cortex. X-ALD is characterized as the childhood, adolescent, adult cerebral ALD, adrenomyeloneuropathy (AMN), adrenal insufficiency, and asymptomatic phenotypes, exhibiting a high variety of clinical neurological manifestations with or without adrenocortical insufficiency. RESULTS In this study, we reported two cases of X-ALD, which were first diagnosed as adrenal insufficiency (Addison's disease) and treated with adrenocortical supplement. However, both of the cases progressed as neurological symptoms and signs after decades. Elevated VLCFAs level, brain MRI scan, and genetic analysis confirmed final diagnosis. In addition, we identified two novel mutations of ABCD1 gene, NM_000033.3 (ABCD1): c.874_876delGAG (p.Glu292del) and NM_000033.3 (ABCD1): c.96_97delCT (p.Tyr33Profs∗161), in exon 1 of ABCD1 gene. Sanger sequencing confirmed that the proband's mother of the first case was heterozygous carrying the same variant. Adrenal insufficiency-only type is very rare; however, it may be the starting performance of X-ALD. In addition, we summarized reported mutation sites and clinical manifestations to investigate the correlationship of phenotype-genotype of X-ALD. CONCLUSIONS The early warning manifestations should be noticed, and the probability of X-ALD should be considered. This report could be beneficial for the early diagnosis and genetic counseling for patients with X-ALD.
Collapse
Affiliation(s)
- Bingzi Dong
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Wenshan Lv
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Lili Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Yuhang Zhao
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Xiaofang Sun
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Zhongchao Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Bingfei Cheng
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Zhengju Fu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Yangang Wang
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| |
Collapse
|