1
|
Oudejans E, Witkamp D, Schonewille P, Adelman MR, Hu-A-Ng GV, Hoogterp L, van Rooijen-van Leeuwen G, van der Laan R, Kerindongo RP, Witvliet JJ, Weber NC, Preckel B, Abbink TEM, van der Knaap MS. Effect of Propofol and Sevoflurane on Vanishing White Matter Models. Pediatr Neurol 2025; 167:66-76. [PMID: 40215832 DOI: 10.1016/j.pediatrneurol.2025.03.009] [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: 08/21/2024] [Revised: 01/18/2025] [Accepted: 03/15/2025] [Indexed: 05/13/2025]
Abstract
BACKGROUND The leukodystrophy vanishing white matter (VWM) most often has its onset in childhood and is characterized by chronic decline and additionally stress-provoked acute neurological deterioration. Anesthesia is a stressor that can trigger this acute decline. The commonly used inhalational anesthetic sevoflurane has been reported to activate the integrated stress response (ISR), whereas the intravenous anesthetic propofol has not. We aimed to assess the differential effects of these anesthetics in models of VWM. METHODS We investigated the molecular effects of sevoflurane and propofol on the ISR in a murine neural cell line and in cultured astrocytes from patients with VWM and control subjects with immunostainings for prototypic ISR marker activating transcription factor 4 (ATF4). We determined the effects of these anesthetics on clinical signs and expression of ISR mRNAs in VWM and wild-type mice. RESULTS In the murine cell line, sevoflurane increased ATF4 accumulation compared with its vehicle (+157%); in contrast, propofol's vehicle without (-22%) and with (-23%) propofol decreased tunicamycin-increased levels of ATF4 to similar degree. Sevoflurane activated the ISR in astrocytes from control subjects (+17-33%) but not from patients with VWM (+1-2%). Propofol and its vehicle did not impact the ISR in astrocytes. The anesthetics did not have prolonged effects on motor skills in VWM mice but had small differential effects on ISR mRNA levels, consistently higher with sevoflurane than with propofol. CONCLUSIONS We observed differential effects of the anesthetics in cultured neural cells and on expression levels of ISR markers in VWM mice, but not on clinical signs in VWM mice.
Collapse
Affiliation(s)
- Ellen Oudejans
- Child Neurology, Emma Children's Hospital, Amsterdam Leukodystrophy Center, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands; Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands
| | - Diede Witkamp
- Child Neurology, Emma Children's Hospital, Amsterdam Leukodystrophy Center, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands; Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands
| | - Pleun Schonewille
- Child Neurology, Emma Children's Hospital, Amsterdam Leukodystrophy Center, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands; Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands
| | - Marcos Ross Adelman
- Child Neurology, Emma Children's Hospital, Amsterdam Leukodystrophy Center, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands; Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands
| | - Gino V Hu-A-Ng
- Child Neurology, Emma Children's Hospital, Amsterdam Leukodystrophy Center, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands; Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands
| | - Leoni Hoogterp
- Child Neurology, Emma Children's Hospital, Amsterdam Leukodystrophy Center, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands; Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands
| | - Gemma van Rooijen-van Leeuwen
- Child Neurology, Emma Children's Hospital, Amsterdam Leukodystrophy Center, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands; Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands
| | - Rika van der Laan
- Child Neurology, Emma Children's Hospital, Amsterdam Leukodystrophy Center, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands; Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands
| | - Raphaela P Kerindongo
- Laboratory of Experimental Intensive Care and Anesthesiology L.E.I.C.A, Department of Anesthesiology, Amsterdam University Medical Center, Amsterdam Cardiovascular Science (ACS), Amsterdam, The Netherlands
| | - Janneke J Witvliet
- Child Neurology, Emma Children's Hospital, Amsterdam Leukodystrophy Center, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands; Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands
| | - Nina C Weber
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands
| | - Benedikt Preckel
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands
| | - Truus E M Abbink
- Child Neurology, Emma Children's Hospital, Amsterdam Leukodystrophy Center, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands; Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands
| | - Marjo S van der Knaap
- Child Neurology, Emma Children's Hospital, Amsterdam Leukodystrophy Center, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, the Netherlands; Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands.
| |
Collapse
|
2
|
Škapik IP, Giacomelli C, Hahn S, Deinlein H, Gallant P, Diebold M, Biayna J, Hendricks A, Olimski L, Otto C, Kastner C, Wolf E, Schülein-Völk C, Maurus K, Rosenwald A, Schleussner N, Jackstadt RF, Schlegel N, Germer CT, Bushell M, Eilers M, Schmidt S, Wiegering A. Maintenance of p-eIF2α levels by the eIF2B complex is vital for colorectal cancer. EMBO J 2025; 44:2075-2105. [PMID: 40016419 PMCID: PMC11962125 DOI: 10.1038/s44318-025-00381-9] [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: 02/19/2024] [Revised: 01/17/2025] [Accepted: 01/23/2025] [Indexed: 03/01/2025] Open
Abstract
Protein synthesis is an essential process, deregulated in multiple tumor types showing differential dependence on translation factors compared to untransformed tissue. We show that colorectal cancer (CRC) with loss-of-function mutation in the APC tumor suppressor depends on an oncogenic translation program regulated by the ability to sense phosphorylated eIF2α (p-eIF2α). Despite increased protein synthesis rates following APC loss, eIF2α phosphorylation, typically associated with translation inhibition, is enhanced in CRC. Elevated p-eIF2α, and its proper sensing by the decameric eIF2B complex, are essential to balance translation. Knockdown or mutation of eIF2Bα and eIF2Bδ, two eIF2B subunits responsible for sensing p-eIF2α, impairs CRC viability, demonstrating that the eIF2B/p-eIF2α nexus is vital for CRC. Specifically, the decameric eIF2B linked by two eIF2Bα subunits is critical for translating growth-promoting mRNAs which are induced upon APC loss. Depletion of eIF2Bα in APC-deficient murine and patient-derived organoids establishes a therapeutic window, validating eIF2Bα as a target for clinical intervention. In conclusion, we demonstrate how the expression of the oncogenic signature in CRC is crucially controlled at the translational level.
Collapse
Affiliation(s)
- Ivana Paskov Škapik
- Theodor Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, 97080, Würzburg, Germany
- Goethe University Frankfurt, University Hospital, Department of General, Visceral, Transplant and Thoracic Surgery, Frankfurt am Main, Germany
| | - Chiara Giacomelli
- CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Sarah Hahn
- Theodor Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, 97080, Würzburg, Germany
- Goethe University Frankfurt, University Hospital, Department of General, Visceral, Transplant and Thoracic Surgery, Frankfurt am Main, Germany
| | - Hanna Deinlein
- Theodor Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Peter Gallant
- Theodor Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| | - Mathias Diebold
- Theodor Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany
- Institute of Pharmacy and Food Chemistry, University of Würzburg, 97074, Würzburg, Germany
| | - Josep Biayna
- Theodor Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, 97080, Würzburg, Germany
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, 60590, Frankfurt am Main, Germany
| | - Anne Hendricks
- Theodor Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Leon Olimski
- Theodor Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Christoph Otto
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Carolin Kastner
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Elmar Wolf
- Theodor Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany
- Institute of Biochemistry, CAU Kiel, 24118, Kiel, Germany
| | | | - Katja Maurus
- Institute of Pathology, University of Würzburg, 97074, Würzburg, Germany
| | - Andreas Rosenwald
- Institute of Pathology, University of Würzburg, 97074, Würzburg, Germany
| | - Nikolai Schleussner
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, University Heidelberg, 69120, Heidelberg, Germany
- Cancer Progression and Metastasis Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany
| | - Rene-Filip Jackstadt
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Cancer Progression and Metastasis Group, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- German Cancer Consortium (DKTK), DKFZ, Core Center Heidelberg, Heidelberg, Germany
| | - Nicolas Schlegel
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Christoph-Thomas Germer
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, 97080, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Martin Bushell
- CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- School of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Martin Eilers
- Theodor Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, 97080, Würzburg, Germany
| | - Stefanie Schmidt
- Theodor Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany.
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, 97080, Würzburg, Germany.
| | - Armin Wiegering
- Theodor Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany.
- Department of General, Visceral, Transplant, Vascular and Pediatric Surgery, University Hospital Würzburg, 97080, Würzburg, Germany.
- Goethe University Frankfurt, University Hospital, Department of General, Visceral, Transplant and Thoracic Surgery, Frankfurt am Main, Germany.
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, 97080, Würzburg, Germany.
| |
Collapse
|
3
|
Barny LA, Hermanson JN, Garcia SK, Stauffer PE, Plate L. Dissecting Branch-Specific Unfolded Protein Response Activation in Drug-Tolerant BRAF-Mutant Melanoma using Data-Independent Acquisition Mass Spectrometry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644425. [PMID: 40196682 PMCID: PMC11974750 DOI: 10.1101/2025.03.20.644425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Cells rely on the Unfolded Protein Response (UPR) to maintain ER protein homeostasis (proteostasis) when faced with elevated levels of misfolded and aggregated proteins. The UPR is comprised of three main branches-ATF6, IRE1, and PERK-that coordinate the synthesis of proteins involved in folding, trafficking, and degradation of nascent proteins to restore ER function. Dysregulation of the UPR is linked to numerous diseases, including neurodegenerative disorders, cancer, and diabetes. Despite its importance, identifying UPR targets has been challenging due to their heterogeneous induction, which varies by cell type and tissue. Additionally, defining the magnitude and range of UPR-regulated genes is difficult because of intricate temporal regulation, feedback between UPR branches, and extensive cross-talk with other stress-signaling pathways. To comprehensively identify UPR-regulated proteins and determine their branch specificity, we developed a data-independent acquisition (DIA) liquid-chromatography mass spectrometry (LC-MS) pipeline. Our optimized workflow improved identifications of low-abundant UPR proteins and leveraged an automated SP3-based protocol on the Biomek i5 liquid handler for label-free peptide preparation. Using engineered stable cell lines that enable selective pharmacological activation of each UPR branch without triggering global UPR activation, we identified branch-specific UPR proteomic targets. These targets were subsequently applied to investigate proteomic changes in multiple patient-derived BRAF-mutant melanoma cell lines treated with a BRAF inhibitor (PLX4720, i.e., vemurafenib). Our findings revealed differential regulation of the XBP1s branch of the UPR in the BRAF-mutant melanoma cell lines after PLX4720 treatment, likely due to calcium activation, suggesting that the UPR plays a role as a non-genetic mechanism of drug tolerance in melanoma. In conclusion, the validated branch-specific UPR proteomic targets identified in this study provide a robust framework for investigating this pathway across different cell types, drug treatments, and disease conditions in a high-throughput manner.
Collapse
Affiliation(s)
- Lea A Barny
- Chemical and Physical Biology Program, Vanderbilt University Medical Center, Nashville, TN, 37235
| | - Jake N Hermanson
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, 37235
| | - Sarah K Garcia
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235
| | - Philip E Stauffer
- Chemical and Physical Biology Program, Vanderbilt University Medical Center, Nashville, TN, 37235
| | - Lars Plate
- Chemical and Physical Biology Program, Vanderbilt University Medical Center, Nashville, TN, 37235
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, 37235
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232
| |
Collapse
|
4
|
Harris DT, Jan CH. CRISPuRe-seq: pooled screening of barcoded ribonucleoprotein reporters reveals regulation of RNA polymerase III transcription by the integrated stress response via mTOR. Nucleic Acids Res 2025; 53:gkaf062. [PMID: 39921565 PMCID: PMC11806354 DOI: 10.1093/nar/gkaf062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 02/10/2025] Open
Abstract
Genetic screens using CRISPR (Clustered Regularly Interspaced Palindromic Repeats) provide valuable information about gene function. Nearly all pooled screening technologies rely on the cell to link genotype to phenotype, making it challenging to assay mechanistically informative, biochemically defined phenotypes. Here, we present CRISPuRe-seq (CRISPR PuRification), a novel pooled screening strategy that expands the universe of accessible phenotypes through the purification of ribonucleoprotein complexes that link genotypes to expressed RNA barcodes. While screening for regulators of the integrated stress response (ISR), we serendipitously discovered that the ISR represses transfer RNA (tRNA) production under conditions of reduced protein synthesis. This regulation is mediated through inhibition of mTORC1 and corresponding activation of the RNA polymerase III inhibitor MAF1. These data demonstrate that coherent downregulation of tRNA expression and protein synthesis is achieved through cross-talk between the ISR and mTOR, two master integrators of cell state.
Collapse
Affiliation(s)
- David T Harris
- Calico Life Sciences LLC, South San Francisco, CA 94080, United States
| | - Calvin H Jan
- Calico Life Sciences LLC, South San Francisco, CA 94080, United States
| |
Collapse
|
5
|
Alsahlawi Z, Isa HM, Alresias S, Hasan SM, Malalla HA, Ebrahim AK, Ali K. Vanishing White Matter Disease in Children: An Unusual Association, a Novel Mutation, and a Literature Review. Cureus 2024; 16:e73667. [PMID: 39677130 PMCID: PMC11645671 DOI: 10.7759/cureus.73667] [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] [Accepted: 11/08/2024] [Indexed: 12/17/2024] Open
Abstract
Vanishing white matter (VWM) disease is an autosomal recessive disorder caused by mutations in the gene EIF2B encoding the subunits 1-5 of eukaryotic initiation factor 2B. Although rare, with a reported prevalence of 1:80,000 (0.001%), it was considered as one of the most common leukodystrophies. However, the worldwide incidence and prevalence of this disease are not clear. In Bahrain, of 21 patients who were diagnosed with leukodystrophy, two patients were found to have VWM disease accounting for 9.5%. Vaccinations and infections were the trigger factors for this disease to manifest. Rapid neurological deterioration, loss of developmental milestones, and seizure disorders are the main presentations in both patients. Magnetic resonance imaging (MRI) showed the classical radiological changes of demyelination and leukodystrophy. Patient 1 had associated ulcerative colitis, a finding that was not reported before. Patient 1's condition progressed to a vegetative stage, while patient 2 passed away, reflecting the poor disease outcome. In patient 2, a novel homozygous missense mutation was found in the EIF2B3 gene (c.25G>A, p.Ala9Thr). In this report, we present in detail the prevalence of VWM disease among cases with leukodystrophy, patients' characteristics, clinical presentations, radiological findings, associated diseases, genetic results, and clinical outcomes in the main tertiary hospital in Bahrain between 1998 and 2024. Moreover, we conducted a thorough literature review on this rare condition.
Collapse
Affiliation(s)
- Zahra Alsahlawi
- Department of Pediatrics, Arabian Gulf University, Manama, BHR
- Department of Pediatrics, Salmaniya Medical Complex, Manama, BHR
| | - Hasan M Isa
- Department of Pediatrics, Arabian Gulf University, Manama, BHR
- Department of Pediatrics, Salmaniya Medical Complex, Manama, BHR
| | | | | | | | - Ayman K Ebrahim
- Department of Neurology, Salmaniya Medical Complex, Manama, BHR
| | - Khadija Ali
- Department of Pediatrics, Salmaniya Medical Complex, Manama, BHR
| |
Collapse
|
6
|
Thuppanattumadam Ananthasubramanian S, Arunachal G, Padmanabha H, Mahale RR. Adult-Onset EIF2B-Pathies: A Clinical, Imaging and Genetic Profiling with Literature Review. Can J Neurol Sci 2024:1-8. [PMID: 39450483 DOI: 10.1017/cjn.2024.308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
BACKGROUND Vanishing white matter syndrome is one of the leukoencephalopathies caused by recessive mutations in gene EIF2B1-5. Adult-onset EIF2B-pathies (clinical onset after age 16 years) have been reported to be less common. OBJECTIVE Description of the clinical, imaging and genetic profile of adult-onset EIF2B-pathies and comparison of Indian cohort with Asian and European cohorts. METHODS Report of two cases of adult-onset EIF2B-pathies and a comprehensive review of genetically confirmed adult-onset EIF2B-pathies since 2001 from Indian, Asian and European cohorts. RESULTS Two patients were females, with median age at presentation of 25.5 years (24-27 years) and onset at 19 years (18-20 years). The median duration of symptoms was 6.5 years (6-7 years). Both had cerebellar ataxia, spasticity, cognitive impairment and bladder involvement. Brain magnetic resonance imaging (MRI) showed leukoencephalopathy with rarefaction in both patients and corpus callosum involvement in one patient. Genetics showed homozygous missense variant in the EIF2B3 gene in both patients. The Indian cohort of seven patients had similar clinical and radiological features and common variants in EIF2B3 (n = 4). The Asian cohort had 24 cases, and the European cohort had 61 cases with similar clinical features, radiological features and common variants in EIF2B5. CONCLUSION Adult-onset EIF2B-pathies have a distinct clinical profile of female predominance with cerebellar ataxia, spasticity and cognitive decline as the commonest triad of clinical manifestations and leukoencephalopathy with rarefaction on brain MRI. Variants in EIF2B5 were common in the Asian and European cohorts and EIF2B3 in the Indian cohort.
Collapse
Affiliation(s)
| | - Gautham Arunachal
- Department of Human Genetics, National Institute of Mental Health and Neurosciences, Bengaluru, KA, India
| | - Hansashree Padmanabha
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, KA, India
| | - Rohan Ramachandra Mahale
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, KA, India
| |
Collapse
|
7
|
Vignard V, Baruteau AE, Toutain B, Mercier S, Isidor B, Redon R, Schott JJ, Küry S, Bézieau S, Monsoro-Burq AH, Ebstein F. Exploring the origins of neurodevelopmental proteasomopathies associated with cardiac malformations: are neural crest cells central to certain pathological mechanisms? Front Cell Dev Biol 2024; 12:1370905. [PMID: 39071803 PMCID: PMC11272537 DOI: 10.3389/fcell.2024.1370905] [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: 01/15/2024] [Accepted: 06/05/2024] [Indexed: 07/30/2024] Open
Abstract
Neurodevelopmental proteasomopathies constitute a recently defined class of rare Mendelian disorders, arising from genomic alterations in proteasome-related genes. These alterations result in the dysfunction of proteasomes, which are multi-subunit protein complexes essential for maintaining cellular protein homeostasis. The clinical phenotype of these diseases manifests as a syndromic association involving impaired neural development and multisystem abnormalities, notably craniofacial anomalies and malformations of the cardiac outflow tract (OFT). These observations suggest that proteasome loss-of-function variants primarily affect specific embryonic cell types which serve as origins for both craniofacial structures and the conotruncal portion of the heart. In this hypothesis article, we propose that neural crest cells (NCCs), a highly multipotent cell population, which generates craniofacial skeleton, mesenchyme as well as the OFT of the heart, in addition to many other derivatives, would exhibit a distinctive vulnerability to protein homeostasis perturbations. Herein, we introduce the diverse cellular compensatory pathways activated in response to protein homeostasis disruption and explore their potential implications for NCC physiology. Altogether, the paper advocates for investigating proteasome biology within NCCs and their early cranial and cardiac derivatives, offering a rationale for future exploration and laying the initial groundwork for therapeutic considerations.
Collapse
Affiliation(s)
- Virginie Vignard
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, Nantes, France
| | - Alban-Elouen Baruteau
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, Nantes, France
- CHU Nantes, Department of Pediatric Cardiology and Pediatric Cardiac Surgery, FHU PRECICARE, Nantes Université, Nantes, France
- Nantes Université, CHU Nantes, INSERM, CIC FEA 1413, Nantes, France
| | - Bérénice Toutain
- Nantes Université, CNRS, INSERM, l’institut du thorax, Nantes, France
| | - Sandra Mercier
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, Nantes, France
- CHU Nantes, Service de Génétique Médicale, Nantes Université, Nantes, France
| | - Bertrand Isidor
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, Nantes, France
- CHU Nantes, Service de Génétique Médicale, Nantes Université, Nantes, France
| | - Richard Redon
- Nantes Université, CNRS, INSERM, l’institut du thorax, Nantes, France
| | | | - Sébastien Küry
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, Nantes, France
- CHU Nantes, Service de Génétique Médicale, Nantes Université, Nantes, France
| | - Stéphane Bézieau
- Nantes Université, CHU Nantes, CNRS, INSERM, l’institut du thorax, Nantes, France
- CHU Nantes, Service de Génétique Médicale, Nantes Université, Nantes, France
| | - Anne H. Monsoro-Burq
- Faculté des Sciences d'Orsay, CNRS, UMR 3347, INSERM, Université Paris-Saclay, Orsay, France
- Institut Curie, PSL Research University, CNRS, UMR 3347, INSERM, Orsay, France
- Institut Universitaire de France, Paris, France
| | - Frédéric Ebstein
- Nantes Université, CNRS, INSERM, l’institut du thorax, Nantes, France
| |
Collapse
|
8
|
Albacete MAP, Simão GN, Lourenço CM, Dos Santos AC. Vanishing white matter disease: imaging, clinical and molecular correlation in Brazilian families. Neuroradiology 2024:10.1007/s00234-024-03405-z. [PMID: 38886214 DOI: 10.1007/s00234-024-03405-z] [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: 11/27/2023] [Accepted: 06/09/2024] [Indexed: 06/20/2024]
Abstract
PURPOSE To characterize Vanishing White Matter Disease (VWM) cases from a Brazilian University Tertiary hospital, focusing on brain magnetic resonance image (MRI) aspects, clinical and molecular data. METHODS Medical records and brain MRI of 13 genetically confirmed VWM patients were reviewed. Epidemiological data such as age at symptom onset, gender and main symptoms were analyzed, along with genetic mutations and MRI characteristics, such as the distribution of white matter lesions and atrophy. RESULTS The majority of patients were female, with the age of symptom onset ranging from 1 year and 6 months to 40 years. All mutations were identified in the EIF2B5 gene, the most prevalent being c.338G > A (p.Arg113His), and a novel mutation related to the disease was discovered, c.1051G > A (p.Gly351Ser). Trauma or infection were significant triggers. The most frequent symptoms were ataxia and limb spasticity. All MRI scans displayed deep white matter involvement, cystic degeneration, with U-fibers relatively spared and a predilection for the frontoparietal region. Lesions in the corpus callosum and posterior fossa were present in all patients. Follow-up exams revealed the evolution of white matter lesions and cerebral atrophy, which correlated with clinical deterioration. CONCLUSIONS VWM affects various age groups, with a significant clinical and genetic variability. A novel mutation associated with the disease is highlighted. MRI reveals a typical pattern of white matter involvement, characterized by diffuse lesions in the periventricular and deep regions, with subsequent extension to the subcortical areas, accompanied by cystic degeneration, and plays a crucial role in diagnosis and follow-up.
Collapse
Affiliation(s)
- Marianna Angelo Palmejani Albacete
- Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto - FMRP- USP, R. Ten. Catão Roxo, 3900 - Vila Monte Alegre, Ribeirão Preto, SP, 14015-010, Brazil.
| | - Gustavo Novelino Simão
- Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto - FMRP- USP, R. Ten. Catão Roxo, 3900 - Vila Monte Alegre, Ribeirão Preto, SP, 14015-010, Brazil
| | - Charles Marques Lourenço
- Neurogenetics Unit - Inborn Errors of Metabolism Clinics, National Reference Center for Rare Diseases, Faculdade de Medicina de São José Do Rio Preto, Av. Brigadeiro Faria Lima, - 5416 - Vila São Pedro, São José Do Rio Preto, SP, 15090-000, Brazil
| | - Antonio Carlos Dos Santos
- Hospital das Clínicas da Faculdade de Medicina de Ribeirão Preto - FMRP- USP, R. Ten. Catão Roxo, 3900 - Vila Monte Alegre, Ribeirão Preto, SP, 14015-010, Brazil
| |
Collapse
|
9
|
Gui M, He M, Qin L. Adult-onset leukoencephalopathy with vanishing white matter with compound heterozygous EIF2B3 gene variants. BMC Neurol 2024; 24:201. [PMID: 38872124 PMCID: PMC11170766 DOI: 10.1186/s12883-024-03721-0] [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: 03/02/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024] Open
Abstract
BACKGROUND Leukoencephalopathy with vanishing white matter (VWM) is an autosomal recessive disorder affecting the white matter of the brain. It typically manifests during childhood, with clinical features including sudden and severe neurological deterioration triggered by stressors such as febrile illness, minor head trauma, or stressful events. Adult-onset cases of VWM are exceptionally uncommon. CASE PRESENTATION In this case, we present an adult patient who exhibited late-onset progressive VWM characterized by ataxia, postural instability, cognitive impairment, and emotional disturbances. Comprehensive screening for endocrine, metabolic, tumor, and immunologic disorders yielded normal or negative results. Brain imaging revealed diffuse and confluent hyperintensity in the white matter on T2-weighted images, along with periventricular cavitations. Genetic testing confirmed the diagnosis of VWM, identifying two heterozygous variants in the eukaryotic translation initiation factor 2B subunit γ (EIF2B3) gene: a pathogenic variant, c.1037 T > C (p.I346T), and a variant of undetermined significance, c.22A > T (p.M8L). Upon a 2-year follow-up, the patient's symptoms deteriorated rapidly following a COVID-19 infection. CONCLUSIONS In conclusion, we have presented a case of classical adult-onset VWM. Since there are no cures or definitive treatments for the disease, it's extremely important to focus on early diagnosis and the prevention of stressors to avoid acute deterioration.
Collapse
Affiliation(s)
- Meilin Gui
- Department of Neurology, The Second Xiangya Hospital, Central South University, 139# Renmin Road, Changsha, Hunan, 410011, China
| | - Miao He
- Department of Neurology, The Second Xiangya Hospital, Central South University, 139# Renmin Road, Changsha, Hunan, 410011, China
| | - Lixia Qin
- Department of Neurology, The Second Xiangya Hospital, Central South University, 139# Renmin Road, Changsha, Hunan, 410011, China.
- National Clinical Research Center On Mental Disorders, Changsha, Hunan, China.
| |
Collapse
|
10
|
Pei Y, Liu S, Wang L, Chen C, Hu M, Xue Y, Guan D, Xie L, Liao H, Zhou J, Zhang H. Design, Synthesis, and Biological Evaluation of Eukaryotic Initiation Factor 2B (eIF2B) Activators. ChemMedChem 2024; 19:e202300716. [PMID: 38426720 DOI: 10.1002/cmdc.202300716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/02/2024]
Abstract
The eukaryotic initiation factor 2B (eIF2B) is a key regulator in protein-regulated signaling pathways and is closely related to the function of the central nervous system. Modulating eIF2B could retard the process of neurodegenerative diseases, including Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and vanishing white matter disease (VWM) et al. Here, we designed and synthesized a series of novel eIF2B activators containing oxadiazole fragments. The activating effects of compounds on eIF2B were investigated through testing the inhibition of ATF4 expression. Of all the targeted compounds, compounds 21 and 29 exhibited potent inhibition on ATF4 expression with IC50 values of 32.43 nM and 47.71 nM, respectively, which were stronger than that of ISRIB (IC50=67.90 nM). ATF4 mRNA assay showed that these two compounds could restore ATF4 mRNA to normal levels in thapsigargin-stimulated HeLa cells. Protein Translation assay showed that both compounds were effective in restoring protein synthesis. Compound potency assay showed that both compounds had similar potency to ISRIB with EC50 values of 5.844 and 37.70 nM. Cytotoxicity assay revealed that compounds 21 and 29 had low toxicity and were worth further investigation.
Collapse
Affiliation(s)
- Yifeng Pei
- Center for Drug Discovery, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Sentao Liu
- Center for Drug Discovery, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Lixun Wang
- Center for Drug Discovery, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Chao Chen
- Center for Drug Discovery, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Mengqiu Hu
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Yi Xue
- Center for Drug Discovery, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Dezhong Guan
- Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Lingfeng Xie
- Center for Drug Discovery, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Hong Liao
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing, China
| | - Jinpei Zhou
- Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, PR China
| | - Huibin Zhang
- Center for Drug Discovery, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, 210009, PR China
| |
Collapse
|
11
|
Lu HJ, Koju N, Sheng R. Mammalian integrated stress responses in stressed organelles and their functions. Acta Pharmacol Sin 2024; 45:1095-1114. [PMID: 38267546 PMCID: PMC11130345 DOI: 10.1038/s41401-023-01225-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 12/30/2023] [Indexed: 01/26/2024]
Abstract
The integrated stress response (ISR) triggered in response to various cellular stress enables mammalian cells to effectively cope with diverse stressful conditions while maintaining their normal functions. Four kinases (PERK, PKR, GCN2, and HRI) of ISR regulate ISR signaling and intracellular protein translation via mediating the phosphorylation of eukaryotic translation initiation factor 2 α (eIF2α) at Ser51. Early ISR creates an opportunity for cells to repair themselves and restore homeostasis. This effect, however, is reversed in the late stages of ISR. Currently, some studies have shown the non-negligible impact of ISR on diseases such as ischemic diseases, cognitive impairment, metabolic syndrome, cancer, vanishing white matter, etc. Hence, artificial regulation of ISR and its signaling with ISR modulators becomes a promising therapeutic strategy for relieving disease symptoms and improving clinical outcomes. Here, we provide an overview of the essential mechanisms of ISR and describe the ISR-related pathways in organelles including mitochondria, endoplasmic reticulum, Golgi apparatus, and lysosomes. Meanwhile, the regulatory effects of ISR modulators and their potential application in various diseases are also enumerated.
Collapse
Affiliation(s)
- Hao-Jun Lu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Nirmala Koju
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
12
|
Yazaki J, Yamanashi T, Nemoto S, Kobayashi A, Han YW, Hasegawa T, Iwase A, Ishikawa M, Konno R, Imami K, Kawashima Y, Seita J. Mapping adipocyte interactome networks by HaloTag-enrichment-mass spectrometry. Biol Methods Protoc 2024; 9:bpae039. [PMID: 38884001 PMCID: PMC11180226 DOI: 10.1093/biomethods/bpae039] [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: 05/01/2024] [Revised: 05/19/2024] [Accepted: 05/28/2024] [Indexed: 06/18/2024] Open
Abstract
Mapping protein interaction complexes in their natural state in vivo is arguably the Holy Grail of protein network analysis. Detection of protein interaction stoichiometry has been an important technical challenge, as few studies have focused on this. This may, however, be solved by artificial intelligence (AI) and proteomics. Here, we describe the development of HaloTag-based affinity purification mass spectrometry (HaloMS), a high-throughput HaloMS assay for protein interaction discovery. The approach enables the rapid capture of newly expressed proteins, eliminating tedious conventional one-by-one assays. As a proof-of-principle, we used HaloMS to evaluate the protein complex interactions of 17 regulatory proteins in human adipocytes. The adipocyte interactome network was validated using an in vitro pull-down assay and AI-based prediction tools. Applying HaloMS to probe adipocyte differentiation facilitated the identification of previously unknown transcription factor (TF)-protein complexes, revealing proteome-wide human adipocyte TF networks and shedding light on how different pathways are integrated.
Collapse
Affiliation(s)
- Junshi Yazaki
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
- Faculty of Agriculture, Laboratory for Genome Biology, Setsunan University, Osaka, 573-0101, Japan
| | - Takashi Yamanashi
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
- Medical Data Deep Learning Team, Advanced Data Science Project, RIKEN Information R&D and Strategy Headquarters, RIKEN, Tokyo, 103-0027, Japan
- School of Integrative and Global Majors, University of Tsukuba, Tsukuba, 305-8577, Japan
| | - Shino Nemoto
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Atsuo Kobayashi
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Yong-Woon Han
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Tomoko Hasegawa
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Akira Iwase
- Cell Function Research Team, RIKEN Center for Sustainable Resource Science, Yokohama, 230-0045, Japan
| | - Masaki Ishikawa
- Department of Applied Genomics, Technology Development Team, Kazusa DNA Research Institute, Kisarazu, 292-0818, Japan
| | - Ryo Konno
- Department of Applied Genomics, Technology Development Team, Kazusa DNA Research Institute, Kisarazu, 292-0818, Japan
| | - Koshi Imami
- Proteome Homeostasis Research Unit, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
| | - Yusuke Kawashima
- Department of Applied Genomics, Technology Development Team, Kazusa DNA Research Institute, Kisarazu, 292-0818, Japan
| | - Jun Seita
- Laboratory for Integrative Genomics, RIKEN Center for Integrative Medical Sciences, Yokohama, 230-0045, Japan
- Medical Data Deep Learning Team, Advanced Data Science Project, RIKEN Information R&D and Strategy Headquarters, RIKEN, Tokyo, 103-0027, Japan
- School of Integrative and Global Majors, University of Tsukuba, Tsukuba, 305-8577, Japan
| |
Collapse
|
13
|
Man JHK, Zarekiani P, Mosen P, de Kok M, Debets DO, Breur M, Altelaar M, van der Knaap MS, Bugiani M. Proteomic dissection of vanishing white matter pathogenesis. Cell Mol Life Sci 2024; 81:234. [PMID: 38789799 PMCID: PMC11126554 DOI: 10.1007/s00018-024-05258-4] [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: 09/18/2023] [Revised: 03/30/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024]
Abstract
Vanishing white matter (VWM) is a leukodystrophy caused by biallelic pathogenic variants in eukaryotic translation initiation factor 2B. To date, it remains unclear which factors contribute to VWM pathogenesis. Here, we investigated the basis of VWM pathogenesis using the 2b5ho mouse model. We first mapped the temporal proteome in the cerebellum, corpus callosum, cortex, and brainstem of 2b5ho and wild-type (WT) mice. Protein changes observed in 2b5ho mice were then cross-referenced with published proteomic datasets from VWM patient brain tissue to define alterations relevant to the human disease. By comparing 2b5ho mice with their region- and age-matched WT counterparts, we showed that the proteome in the cerebellum and cortex of 2b5ho mice was already dysregulated prior to pathology development, whereas proteome changes in the corpus callosum only occurred after pathology onset. Remarkably, protein changes in the brainstem were transient, indicating that a compensatory mechanism might occur in this region. Importantly, 2b5ho mouse brain proteome changes reflect features well-known in VWM. Comparison of the 2b5ho mouse and VWM patient brain proteomes revealed shared changes. These could represent changes that contribute to the disease or even drive its progression in patients. Taken together, we show that the 2b5ho mouse brain proteome is affected in a region- and time-dependent manner. We found that the 2b5ho mouse model partly replicates the human disease at the protein level, providing a resource to study aspects of VWM pathogenesis by highlighting alterations from early to late disease stages, and those that possibly drive disease progression.
Collapse
Affiliation(s)
- Jodie H K Man
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Molecular and Cellular Mechanisms, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Parand Zarekiani
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Peter Mosen
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, The Netherlands
- Netherlands Proteomics Center, Utrecht, The Netherlands
| | - Mike de Kok
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Donna O Debets
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, The Netherlands
- Netherlands Proteomics Center, Utrecht, The Netherlands
| | - Marjolein Breur
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Molecular and Cellular Mechanisms, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, The Netherlands
- Netherlands Proteomics Center, Utrecht, The Netherlands
| | - Marjo S van der Knaap
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Molecular and Cellular Mechanisms, Amsterdam Neuroscience, Amsterdam, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Marianna Bugiani
- Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
14
|
Böck D, Revers IM, Bomhof ASJ, Hillen AEJ, Boeijink C, Kissling L, Egli S, Moreno-Mateos MA, van der Knaap MS, van Til NP, Schwank G. In vivo base editing of a pathogenic Eif2b5 variant improves vanishing white matter phenotypes in mice. Mol Ther 2024; 32:1328-1343. [PMID: 38454603 PMCID: PMC11081866 DOI: 10.1016/j.ymthe.2024.03.009] [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: 10/17/2023] [Revised: 02/05/2024] [Accepted: 03/05/2024] [Indexed: 03/09/2024] Open
Abstract
Vanishing white matter (VWM) is a fatal leukodystrophy caused by recessive mutations in subunits of the eukaryotic translation initiation factor 2B. Currently, there are no effective therapies for VWM. Here, we assessed the potential of adenine base editing to correct human pathogenic VWM variants in mouse models. Using adeno-associated viral vectors, we delivered intein-split adenine base editors into the cerebral ventricles of newborn VWM mice, resulting in 45.9% ± 5.9% correction of the Eif2b5R191H variant in the cortex. Treatment slightly increased mature astrocyte populations and partially recovered the integrated stress response (ISR) in female VWM animals. This led to notable improvements in bodyweight and grip strength in females; however, locomotor disabilities were not rescued. Further molecular analyses suggest that more precise editing (i.e., lower rates of bystander editing) as well as more efficient delivery of the base editors to deep brain regions and oligodendrocytes would have been required for a broader phenotypic rescue. Our study emphasizes the potential, but also identifies limitations, of current in vivo base-editing approaches for the treatment of VWM or other leukodystrophies.
Collapse
Affiliation(s)
- Desirée Böck
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland
| | - Ilma M Revers
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam and Amsterdam Neuroscience, Cellular & Molecular Mechanisms, 1105AZ Amsterdam, the Netherlands
| | - Anastasia S J Bomhof
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam and Amsterdam Neuroscience, Cellular & Molecular Mechanisms, 1105AZ Amsterdam, the Netherlands
| | - Anne E J Hillen
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam and Amsterdam Neuroscience, Cellular & Molecular Mechanisms, 1105AZ Amsterdam, the Netherlands
| | - Claire Boeijink
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam and Amsterdam Neuroscience, Cellular & Molecular Mechanisms, 1105AZ Amsterdam, the Netherlands
| | - Lucas Kissling
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland
| | - Sabina Egli
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland
| | - Miguel A Moreno-Mateos
- Andalusian Center for Developmental Biology (CABD), Pablo de Olavide University/CSIC/Junta de Andalucía, 41013 Seville, Spain; Department of Molecular Biology and Biochemical Engineering, Pablo de Olavide University, 41013 Seville, Spain
| | - Marjo S van der Knaap
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam and Amsterdam Neuroscience, Cellular & Molecular Mechanisms, 1105AZ Amsterdam, the Netherlands; Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081HV Amsterdam, the Netherlands
| | - Niek P van Til
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam and Amsterdam Neuroscience, Cellular & Molecular Mechanisms, 1105AZ Amsterdam, the Netherlands; Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, 1081HV Amsterdam, the Netherlands.
| | - Gerald Schwank
- Institute of Pharmacology and Toxicology, University of Zurich, 8057 Zurich, Switzerland.
| |
Collapse
|
15
|
Schoenmakers DH, van Beelen I, Voermans MMC, Perik D, Stellingwerff MD, Wolf NI, Berkhof J, van der Knaap MS. Adaptive behavior assessed by Vineland-3 as comprehensive outcome measure in vanishing white matter. Ann Clin Transl Neurol 2024; 11:650-661. [PMID: 38217081 DOI: 10.1002/acn3.51985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/20/2023] [Accepted: 12/19/2023] [Indexed: 01/14/2024] Open
Abstract
OBJECTIVES Investigate the results and usability of the Vineland-3 as an outcome measure in vanishing white matter patients. METHODS A cross-sectional investigation of the Vineland-3 based on interviews with caregivers, the Health Utilities Index, and the modified Rankin Scale in 64 vanishing white matter patients. RESULTS Adaptive behavior measured with the Vineland-3 is impaired in the vast majority of vanishing white matter patients and significantly impacts daily life. Typically, the daily living skills domain is most severely affected and the socialization domain is the least affected. Based on the metric properties and the clinical relevance, the standard scores for the daily living skills domain and Adaptive Behavior Composite have the best properties to be used as an outcome measure. INTERPRETATION The Vineland-3 appears to be a useful outcome measure to explore and quantify complex cognitive, behavioral, and psychiatric impairments affecting daily functioning in vanishing white matter. Further research should address the longitudinal evaluation of this tool and its additional value to standard neuropsychological and clinical examination.
Collapse
Affiliation(s)
- Daphne H Schoenmakers
- Department of Child Neurology, Emma's Children's Hospital, Amsterdam UMC location Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands
- Medicine for Society, Platform at Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Irene van Beelen
- Department of Child Neurology, Emma's Children's Hospital, Amsterdam UMC location Vrije Universiteit, Amsterdam, The Netherlands
| | - Marije M C Voermans
- Department of Child Neurology, Emma's Children's Hospital, Amsterdam UMC location Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands
| | - Denise Perik
- Department of Medical Psychology, Amsterdam UMC location Vrije Universiteit, Amsterdam, The Netherlands
| | - Menno D Stellingwerff
- Department of Child Neurology, Emma's Children's Hospital, Amsterdam UMC location Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands
| | - Nicole I Wolf
- Department of Child Neurology, Emma's Children's Hospital, Amsterdam UMC location Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands
| | - Johannes Berkhof
- Department of Epidemiology and Data Science, Amsterdam UMC location Vrije Universiteit, Amsterdam, The Netherlands
| | - Marjo S van der Knaap
- Department of Child Neurology, Emma's Children's Hospital, Amsterdam UMC location Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Haakonsen DL, Heider M, Ingersoll AJ, Vodehnal K, Witus SR, Uenaka T, Wernig M, Rapé M. Stress response silencing by an E3 ligase mutated in neurodegeneration. Nature 2024; 626:874-880. [PMID: 38297121 PMCID: PMC10881396 DOI: 10.1038/s41586-023-06985-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 12/15/2023] [Indexed: 02/02/2024]
Abstract
Stress response pathways detect and alleviate adverse conditions to safeguard cell and tissue homeostasis, yet their prolonged activation induces apoptosis and disrupts organismal health1-3. How stress responses are turned off at the right time and place remains poorly understood. Here we report a ubiquitin-dependent mechanism that silences the cellular response to mitochondrial protein import stress. Crucial to this process is the silencing factor of the integrated stress response (SIFI), a large E3 ligase complex mutated in ataxia and in early-onset dementia that degrades both unimported mitochondrial precursors and stress response components. By recognizing bifunctional substrate motifs that equally encode protein localization and stability, the SIFI complex turns off a general stress response after a specific stress event has been resolved. Pharmacological stress response silencing sustains cell survival even if stress resolution failed, which underscores the importance of signal termination and provides a roadmap for treating neurodegenerative diseases caused by mitochondrial import defects.
Collapse
Affiliation(s)
- Diane L Haakonsen
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California at Berkeley, Berkeley, CA, USA
| | - Michael Heider
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA
| | - Andrew J Ingersoll
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA
| | - Kayla Vodehnal
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Samuel R Witus
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA
- Howard Hughes Medical Institute, University of California at Berkeley, Berkeley, CA, USA
| | - Takeshi Uenaka
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Marius Wernig
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Rapé
- Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, USA.
- Howard Hughes Medical Institute, University of California at Berkeley, Berkeley, CA, USA.
- California Institute for Quantitative Biosciences (QB3), University of California at Berkeley, Berkeley, CA, USA.
| |
Collapse
|
17
|
Amiri M, Kiniry SJ, Possemato AP, Mahmood N, Basiri T, Dufour CR, Tabatabaei N, Deng Q, Bellucci MA, Harwalkar K, Stokes MP, Giguère V, Kaufman RJ, Yamanaka Y, Baranov PV, Tahmasebi S, Sonenberg N. Impact of eIF2α phosphorylation on the translational landscape of mouse embryonic stem cells. Cell Rep 2024; 43:113615. [PMID: 38159280 PMCID: PMC10962698 DOI: 10.1016/j.celrep.2023.113615] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/24/2023] [Accepted: 12/06/2023] [Indexed: 01/03/2024] Open
Abstract
The integrated stress response (ISR) is critical for cell survival under stress. In response to diverse environmental cues, eIF2α becomes phosphorylated, engendering a dramatic change in mRNA translation. The activation of ISR plays a pivotal role in the early embryogenesis, but the eIF2-dependent translational landscape in pluripotent embryonic stem cells (ESCs) is largely unexplored. We employ a multi-omics approach consisting of ribosome profiling, proteomics, and metabolomics in wild-type (eIF2α+/+) and phosphorylation-deficient mutant eIF2α (eIF2αA/A) mouse ESCs (mESCs) to investigate phosphorylated (p)-eIF2α-dependent translational control of naive pluripotency. We show a transient increase in p-eIF2α in the naive epiblast layer of E4.5 embryos. Absence of eIF2α phosphorylation engenders an exit from naive pluripotency following 2i (two chemical inhibitors of MEK1/2 and GSK3α/β) withdrawal. p-eIF2α controls translation of mRNAs encoding proteins that govern pluripotency, chromatin organization, and glutathione synthesis. Thus, p-eIF2α acts as a key regulator of the naive pluripotency gene regulatory network.
Collapse
Affiliation(s)
- Mehdi Amiri
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Stephen J Kiniry
- School of Biochemistry and Cell Biology, University College Cork, T12 XF62 Cork, Ireland
| | | | - Niaz Mahmood
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Tayebeh Basiri
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Catherine R Dufour
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Negar Tabatabaei
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA
| | - Qiyun Deng
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Michael A Bellucci
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Keerthana Harwalkar
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada; Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada
| | - Matthew P Stokes
- Cell Signaling Technology, Inc., 3 Trask Lane, Danvers, MA 01923, USA
| | - Vincent Giguère
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Randal J Kaufman
- Degenerative Diseases Program, Center for Genetic Disorders and Aging Research, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Yojiro Yamanaka
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada; Department of Human Genetics, McGill University, Montreal, QC H3A 0C7, Canada
| | - Pavel V Baranov
- School of Biochemistry and Cell Biology, University College Cork, T12 XF62 Cork, Ireland
| | - Soroush Tahmasebi
- Department of Pharmacology and Regenerative Medicine, University of Illinois College of Medicine, Chicago, IL 60612, USA.
| | - Nahum Sonenberg
- Department of Biochemistry, McGill University, Montreal, QC H3A 1A3, Canada; Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada.
| |
Collapse
|
18
|
Zhang SX, Wang JJ, Starr CR, Lee EJ, Park KS, Zhylkibayev A, Medina A, Lin JH, Gorbatyuk M. The endoplasmic reticulum: Homeostasis and crosstalk in retinal health and disease. Prog Retin Eye Res 2024; 98:101231. [PMID: 38092262 PMCID: PMC11056313 DOI: 10.1016/j.preteyeres.2023.101231] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/19/2023]
Abstract
The endoplasmic reticulum (ER) is the largest intracellular organelle carrying out a broad range of important cellular functions including protein biosynthesis, folding, and trafficking, lipid and sterol biosynthesis, carbohydrate metabolism, and calcium storage and gated release. In addition, the ER makes close contact with multiple intracellular organelles such as mitochondria and the plasma membrane to actively regulate the biogenesis, remodeling, and function of these organelles. Therefore, maintaining a homeostatic and functional ER is critical for the survival and function of cells. This vital process is implemented through well-orchestrated signaling pathways of the unfolded protein response (UPR). The UPR is activated when misfolded or unfolded proteins accumulate in the ER, a condition known as ER stress, and functions to restore ER homeostasis thus promoting cell survival. However, prolonged activation or dysregulation of the UPR can lead to cell death and other detrimental events such as inflammation and oxidative stress; these processes are implicated in the pathogenesis of many human diseases including retinal disorders. In this review manuscript, we discuss the unique features of the ER and ER stress signaling in the retina and retinal neurons and describe recent advances in the research to uncover the role of ER stress signaling in neurodegenerative retinal diseases including age-related macular degeneration, inherited retinal degeneration, achromatopsia and cone diseases, and diabetic retinopathy. In some chapters, we highlight the complex interactions between the ER and other intracellular organelles focusing on mitochondria and illustrate how ER stress signaling regulates common cellular stress pathways such as autophagy. We also touch upon the integrated stress response in retinal degeneration and diabetic retinopathy. Finally, we provide an update on the current development of pharmacological agents targeting the UPR response and discuss some unresolved questions and knowledge gaps to be addressed by future research.
Collapse
Affiliation(s)
- Sarah X Zhang
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States.
| | - Josh J Wang
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Christopher R Starr
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Eun-Jin Lee
- Department of Ophthalmology and Byers Eye Institute, Stanford University, Stanford, CA, United States; VA Palo Alto Healthcare System, Palo Alto, CA, United States; Department of Pathology, Stanford University, Stanford, CA, United States
| | - Karen Sophia Park
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Assylbek Zhylkibayev
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andy Medina
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Jonathan H Lin
- Department of Ophthalmology and Byers Eye Institute, Stanford University, Stanford, CA, United States; VA Palo Alto Healthcare System, Palo Alto, CA, United States; Department of Pathology, Stanford University, Stanford, CA, United States
| | - Marina Gorbatyuk
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
19
|
van der Knaap MS, Bugiani M, Abbink TEM. Vanishing white matter. HANDBOOK OF CLINICAL NEUROLOGY 2024; 204:77-94. [PMID: 39322396 DOI: 10.1016/b978-0-323-99209-1.00015-6] [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
"Vanishing white matter" (VWM) is a leukodystrophy caused by autosomal recessive pathogenic variants in the genes encoding the subunits of eukaryotic initiation factor 2B (eIF2B). Disease onset and disease course are extremely variable. Onset varies from the antenatal period until senescence. The age of onset is predictive of disease severity. VWM is characterized by chronic neurologic deterioration and, additionally, episodes of rapid and major neurologic decline, provoked by stresses such as febrile infections and minor head trauma. The disease is dominated by degeneration of the white matter of the central nervous system due to dysfunction of oligodendrocytes and in particular astrocytes. Organs other than the brain are rarely affected, with the exception of the ovaries. The reason for the selective vulnerability of the white matter of the central nervous system and, less consistently, the ovaries is poorly understood. eIF2B is a central regulatory factor in the integrated stress response (ISR). Genetic variants decrease eIF2B activity and thereby cause constitutive activation of the ISR downstream of eIF2B. Strikingly, the ISR is specifically activated in astrocytes. Modulation of eIF2B activity and ISR activation in VWM mouse models impacts disease severity, revealing eIF2B-regulated pathways as potential druggable targets.
Collapse
Affiliation(s)
- Marjo S van der Knaap
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Center, and Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Vrije Universiteit, Amsterdam, The Netherlands.
| | - Marianna Bugiani
- Department of Pathology, Amsterdam Leukodystrophy Center, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Truus E M Abbink
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam UMC, Amsterdam, The Netherlands; Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Vrije Universiteit, Amsterdam, The Netherlands
| |
Collapse
|
20
|
Guo R, Rippert A, Cook EB, Alves CAP, Bird LM, Izumi K. Expansion of clinical and variant spectrum of EEF2-related neurodevelopmental disorder: Report of two additional cases. Am J Med Genet A 2023; 191:2602-2609. [PMID: 37159414 PMCID: PMC10527330 DOI: 10.1002/ajmg.a.63230] [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: 01/31/2023] [Revised: 04/14/2023] [Accepted: 04/24/2023] [Indexed: 05/11/2023]
Abstract
Eukaryotic translation elongation factor 2 (eEF2), encoded by the gene EEF2, is an essential factor involved in the elongation phase of protein translation. A specific heterozygous missense variant (p.P596H) in EEF2 was originally identified in association with autosomal dominant adult-onset spinocerebellar ataxia-26 (SCA26). More recently, additional heterozygous missense variants in this gene have been described to cause a novel, childhood-onset neurodevelopmental disorder with benign external hydrocephalus. Herein, we report two unrelated individuals with a similar gene-disease correlation to support this latter observation. Patient 1 is a 7-year-old male with a previously reported, de novo missense variant (p.V28M) who has motor and speech delay, autism spectrum disorder, failure to thrive with relative macrocephaly, unilateral microphthalmia with coloboma and eczema. Patient 2 is a 4-year-old female with a novel de novo nonsense variant (p.Q145X) with motor and speech delay, hypotonia, macrocephaly with benign ventricular enlargement, and keratosis pilaris. These additional cases help to further expand the genotypic and phenotypic spectrum of this newly described EEF2-related neurodevelopmental syndrome.
Collapse
Affiliation(s)
- Rose Guo
- Division of Human Genetics, Children’s Hospital of Philadelphia, Pennsylvania, Pennsylvania, USA
| | - Alyssa Rippert
- Division of Human Genetics, Children’s Hospital of Philadelphia, Pennsylvania, Pennsylvania, USA
| | - Edward B Cook
- Division of Human Genetics, Children’s Hospital of Philadelphia, Pennsylvania, Pennsylvania, USA
| | - Cesar Augusto P Alves
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Lynne M Bird
- Department of Pediatrics, University of California San Diego; Division of Dysmorphology/Genetics, Rady Children’s Hospital San Diego, San Diego, California, USA
| | - Kosuke Izumi
- Division of Human Genetics, Children’s Hospital of Philadelphia, Pennsylvania, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Current affiliation: Division of Genetics and Metabolism, Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
21
|
Schoenmakers DH, Leferink PS, Vanderver A, Bonkowsky JL, Krägeloh-Mann I, Bernard G, Bertini E, Fatemi A, Fogel BL, Wolf NI, Skwirut D, Buck A, Holberg B, Saunier-Vivar EF, Rauner R, Dekker H, van Bokhoven P, Stellingwerff MD, Berkhof J, van der Knaap MS. Core protocol development for phase 2/3 clinical trials in the leukodystrophy vanishing white matter: a consensus statement by the VWM consortium and patient advocates. BMC Neurol 2023; 23:305. [PMID: 37592248 PMCID: PMC10433679 DOI: 10.1186/s12883-023-03354-9] [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: 01/05/2023] [Accepted: 08/02/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND The leukodystrophy "Vanishing White Matter" (VWM) is an orphan disease with neurological decline and high mortality. Currently, VWM has no approved treatments, but advances in understanding pathophysiology have led to identification of promising therapies. Several investigational medicinal products are either in or about to enter clinical trial phase. Clinical trials in VWM pose serious challenges, as VWM has an episodic disease course; disease phenotype is highly heterogeneous and predictable only for early onset; and study power is limited by the small patient numbers. To address these challenges and accelerate therapy delivery, the VWM Consortium, a group of academic clinicians with expertise in VWM, decided to develop a core protocol to function as a template for trials, to improve trial design and facilitate sharing of control data, while permitting flexibility regarding other trial details. Overall aims of the core protocol are to collect safety, tolerability, and efficacy data for treatment assessment and marketing authorization. METHODS To develop the core protocol, the VWM Consortium designated a committee, including clinician members of the VWM Consortium, family and patient group advocates, and experts in statistics, clinical trial design and alliancing with industries. We drafted three age-specific protocols, to stratify into more homogeneous patient groups, of ages ≥ 18 years, ≥ 6 to < 18 years and < 6 years. We chose double-blind, randomized, placebo-controlled design for patients aged ≥ 6 years; and open-label non-randomized natural-history-controlled design for patients < 6 years. The protocol describes study populations, age-specific endpoints, inclusion and exclusion criteria, study schedules, sample size determinations, and statistical considerations. DISCUSSION The core protocol provides a shared uniformity across trials, enables a pool of shared controls, and reduces the total number of patients necessary per trial, limiting the number of patients on placebo. All VWM clinical trials are suggested to adhere to the core protocol. Other trial components such as choice of primary outcome, pharmacokinetics, pharmacodynamics, and biomarkers are flexible and unconstrained by the core protocol. Each sponsor is responsible for their trial execution, while the control data are handled by a shared research organization. This core protocol benefits the efficiency of parallel and consecutive trials in VWM, and we hope accelerates time to availability of treatments for VWM. TRIAL REGISTRATION NA. From a scientific and ethical perspective, it is strongly recommended that all interventional trials using this core protocol are registered in a clinical trial register.
Collapse
Affiliation(s)
- Daphne H Schoenmakers
- Department of Child Neurology, Emma's Children's Hospital, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Platform "Medicine for Society", Amsterdam UMC Location University of Amsterdam, Amsterdam, The Netherlands
| | - Prisca S Leferink
- IXA Neuroscience, Amsterdam Neuroscience, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Adeline Vanderver
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Joshua L Bonkowsky
- Division of Pediatric Neurology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah, USA
- Primary Children's Hospital, Intermountain Healthcare, Salt Lake City, Utah, USA
| | - Ingeborg Krägeloh-Mann
- Department of Developmental and Child Neurology, Social Pediatrics, University Children's Hospital Tübingen, Tübingen, Germany
| | - Geneviève Bernard
- Departments of Neurology and Neurosurgery, Pediatrics and Human Genetics, McGill University; Department Specialized Medicine, Division of Medical Genetics, McGill University Health Center, Montreal, Canada
- Child Health and Human Development Program, Research Institute of the McGill University Health Center, Montreal, Canada
| | - Enrico Bertini
- Research Unit of Neuromuscular and Neurodegenerative Diseases, Translational Pediatrics and Clinical Genetics Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Ali Fatemi
- Kennedy Krieger Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Brent L Fogel
- Los Angeles David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Nicole I Wolf
- Department of Child Neurology, Emma's Children's Hospital, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands
| | - Donna Skwirut
- United Leukodystrophy Foundation, DeKalb Illinois, USA
- VWM Families Foundation, Greenwhich, CT, USA
| | | | | | - Elise F Saunier-Vivar
- Research Department, European Leukodystrophies Association International and European Leukodystrophies Association France, Paris, France
| | - Robert Rauner
- United Leukodystrophy Foundation, DeKalb Illinois, USA
| | - Hanka Dekker
- Vereniging Volwassenen, Kinderen en Stofwisselingsziekten, Zwolle, The Netherlands
| | - Pieter van Bokhoven
- IXA Neuroscience, Amsterdam Neuroscience, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Menno D Stellingwerff
- Department of Child Neurology, Emma's Children's Hospital, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands
| | - Johannes Berkhof
- Department of Epidemiology and Data Science, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Marjo S van der Knaap
- Department of Child Neurology, Emma's Children's Hospital, Amsterdam UMC Location Vrije Universiteit, Amsterdam, The Netherlands.
- Amsterdam Leukodystrophy Center, Amsterdam Neuroscience, Cellular & Molecular Mechanisms, Amsterdam, The Netherlands.
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, The Netherlands.
| |
Collapse
|
22
|
Man JHK, van Gelder CAGH, Breur M, Molenaar D, Abbink T, Altelaar M, Bugiani M, van der Knaap MS. Regional vulnerability of brain white matter in vanishing white matter. Acta Neuropathol Commun 2023; 11:103. [PMID: 37349783 PMCID: PMC10286497 DOI: 10.1186/s40478-023-01599-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/03/2023] [Indexed: 06/24/2023] Open
Abstract
Vanishing white matter (VWM) is a leukodystrophy that primarily manifests in young children. In this disease, the brain white matter is differentially affected in a predictable pattern with telencephalic brain areas being most severely affected, while others remain allegedly completely spared. Using high-resolution mass spectrometry-based proteomics, we investigated the proteome patterns of the white matter in the severely affected frontal lobe and normal appearing pons in VWM and control cases to identify molecular bases underlying regional vulnerability. By comparing VWM patients to controls, we identified disease-specific proteome patterns. We showed substantial changes in both the VWM frontal and pons white matter at the protein level. Side-by-side comparison of brain region-specific proteome patterns further revealed regional differences. We found that different cell types were affected in the VWM frontal white matter than in the pons. Gene ontology and pathway analyses identified involvement of region specific biological processes, of which pathways involved in cellular respiratory metabolism were overarching features. In the VWM frontal white matter, proteins involved in glycolysis/gluconeogenesis and metabolism of various amino acids were decreased compared to controls. By contrast, in the VWM pons white matter, we found a decrease in proteins involved in oxidative phosphorylation. Taken together, our data show that brain regions are affected in parallel in VWM, but to different degrees. We found region-specific involvement of different cell types and discovered that cellular respiratory metabolism is likely to be differentially affected across white matter regions in VWM. These region-specific changes help explain regional vulnerability to pathology in VWM.
Collapse
Affiliation(s)
- Jodie H K Man
- Department of Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, VU University Amsterdam, Amsterdam, 1081 HV, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, Amsterdam, 1081 HV, The Netherlands
- Molecular and Cellular Mechanisms, Amsterdam Neuroscience, Amsterdam, 1081 HV, The Netherlands
| | - Charlotte A G H van Gelder
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, 3584 CS, The Netherlands
- Netherlands Proteomics Center, Utrecht, 3584 CS, The Netherlands
| | - Marjolein Breur
- Department of Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, VU University Amsterdam, Amsterdam, 1081 HV, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, Amsterdam, 1081 HV, The Netherlands
- Molecular and Cellular Mechanisms, Amsterdam Neuroscience, Amsterdam, 1081 HV, The Netherlands
| | - Douwe Molenaar
- Department of Systems Bioinformatics, VU University Amsterdam, Amsterdam, 1081 HV, The Netherlands
| | - Truus Abbink
- Department of Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, VU University Amsterdam, Amsterdam, 1081 HV, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, Amsterdam, 1081 HV, The Netherlands
- Molecular and Cellular Mechanisms, Amsterdam Neuroscience, Amsterdam, 1081 HV, The Netherlands
| | - Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Utrecht, 3584 CS, The Netherlands
- Netherlands Proteomics Center, Utrecht, 3584 CS, The Netherlands
| | - Marianna Bugiani
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, Amsterdam, 1081 HV, The Netherlands.
- Molecular and Cellular Mechanisms, Amsterdam Neuroscience, Amsterdam, 1081 HV, The Netherlands.
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, 1081 HV, The Netherlands.
| | - Marjo S van der Knaap
- Department of Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, VU University Amsterdam, Amsterdam, 1081 HV, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, Amsterdam, 1081 HV, The Netherlands
- Molecular and Cellular Mechanisms, Amsterdam Neuroscience, Amsterdam, 1081 HV, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, Amsterdam, 1081 HV, The Netherlands
| |
Collapse
|
23
|
Imanishi R, Nakau K, Shimada S, Oka H, Takeguchi R, Tanaka R, Sugiyama T, Nii M, Okamoto T, Nagaya K, Makita Y, Yanagi K, Kaname T, Takahashi S. A novel HECW2 variant in an infant with congenital long QT syndrome. Hum Genome Var 2023; 10:17. [PMID: 37280227 PMCID: PMC10244414 DOI: 10.1038/s41439-023-00245-w] [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: 04/14/2023] [Revised: 05/04/2023] [Accepted: 05/11/2023] [Indexed: 06/08/2023] Open
Abstract
Pathogenic variants of HECW2 have been reported in cases of neurodevelopmental disorder with hypotonia, seizures, and absent language (NDHSAL; OMIM #617268). A novel HECW2 variant (NM_001348768.2:c.4343 T > C,p.Leu1448Ser) was identified in an NDHSAL infant with severe cardiac comorbidities. The patient presented with fetal tachyarrhythmia and hydrops and was postnatally diagnosed with long QT syndrome. This study provides evidence that HECW2 pathogenic variants can cause long QT syndrome along with neurodevelopmental disorders.
Collapse
Affiliation(s)
- Rina Imanishi
- Department of Pediatrics, Asahikawa Medical University, Hokkaido, Japan
| | - Kouichi Nakau
- Department of Pediatrics, Asahikawa Medical University, Hokkaido, Japan
| | - Sorachi Shimada
- Department of Pediatrics, Asahikawa Medical University, Hokkaido, Japan
| | - Hideharu Oka
- Department of Pediatrics, Asahikawa Medical University, Hokkaido, Japan
| | - Ryo Takeguchi
- Department of Pediatrics, Asahikawa Medical University, Hokkaido, Japan
| | - Ryosuke Tanaka
- Department of Pediatrics, Asahikawa Medical University, Hokkaido, Japan
| | - Tatsutoshi Sugiyama
- Division of Neonatology, Center for Maternity and Infant Care, Asahikawa Medical University Hospital, Hokkaido, Japan
| | - Mitsumaro Nii
- Division of Neonatology, Center for Maternity and Infant Care, Asahikawa Medical University Hospital, Hokkaido, Japan
| | - Toshio Okamoto
- Division of Neonatology, Center for Maternity and Infant Care, Asahikawa Medical University Hospital, Hokkaido, Japan
| | - Ken Nagaya
- Division of Neonatology, Center for Maternity and Infant Care, Asahikawa Medical University Hospital, Hokkaido, Japan
| | - Yoshio Makita
- Department of Genetic Counseling, Asahikawa Medical University Hospital, Hokkaido, Japan
| | - Kumiko Yanagi
- Department of Genome Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Tadashi Kaname
- Department of Genome Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Satoru Takahashi
- Department of Pediatrics, Asahikawa Medical University, Hokkaido, Japan.
| |
Collapse
|
24
|
Burgunder JM. Mechanisms underlying phenotypic variation in neurogenetic disorders. Nat Rev Neurol 2023:10.1038/s41582-023-00811-4. [PMID: 37202496 DOI: 10.1038/s41582-023-00811-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2023] [Indexed: 05/20/2023]
Abstract
Neurological diseases associated with pathogenic variants in a specific gene, or even with a specific pathogenic variant, can show profound phenotypic variation with regard to symptom presentation, age at onset and disease course. Highlighting examples from a range of neurogenetic disorders, this Review explores emerging mechanisms that are involved in this variability, including environmental, genetic and epigenetic factors that influence the expressivity and penetrance of pathogenic variants. Environmental factors, some of which can potentially be modified to prevent disease, include trauma, stress and metabolic changes. Dynamic patterns of pathogenic variants might explain some of the phenotypic variations, for example, in the case of disorders caused by DNA repeat expansions such as Huntington disease (HD). An important role for modifier genes has also been identified in some neurogenetic disorders, including HD, spinocerebellar ataxia and X-linked dystonia-parkinsonism. In other disorders, such as spastic paraplegia, the basis for most of the phenotypic variability remains unclear. Epigenetic factors have been implicated in disorders such as SGCE-related myoclonus-dystonia and HD. Knowledge of the mechanisms underlying phenotypic variation is already starting to influence management strategies and clinical trials for neurogenetic disorders.
Collapse
|
25
|
Stellingwerff MD, van de Wiel MA, van der Knaap MS. Radiological correlates of episodes of acute decline in the leukodystrophy vanishing white matter. Neuroradiology 2023; 65:855-863. [PMID: 36574026 DOI: 10.1007/s00234-022-03097-3] [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: 09/12/2022] [Accepted: 11/25/2022] [Indexed: 12/28/2022]
Abstract
PURPOSE Patients with vanishing white matter (VWM) experience unremitting chronic neurological decline and stress-provoked episodes of rapid, partially reversible decline. Cerebral white matter abnormalities are progressive, without improvement, and are therefore unlikely to be related to the episodes. We determined which radiological findings are related to episodic decline. METHODS MRI scans of VWM patients were retrospectively analyzed. Patients were grouped into A (never episodes) and B (episodes). Signal abnormalities outside the cerebral white matter were rated as absent, mild, or severe. A sum score was developed with abnormalities only seen in group B. The temporal relationship between signal abnormalities and episodes was determined by subdividing scans into those made before, less than 3 months after, and more than 3 months after onset of an episode. RESULTS Five hundred forty-three examinations of 298 patients were analyzed. Mild and severe signal abnormalities in the caudate nucleus, putamen, globus pallidus, thalamus, midbrain, medulla oblongata, and severe signal abnormalities in the pons were only seen in group B. The sum score, constructed with these abnormalities, depended on the timing of the scan (χ2(2, 400) = 22.8; p < .001): it was least often abnormal before, most often abnormal with the highest value shortly after, and lower longer than 3 months after an episode. CONCLUSION In VWM, signal abnormalities in brainstem, thalamus, and basal ganglia are related to episodic decline and can improve. Knowledge of the natural MRI history in VWM is important for clinical interpretation of MRI findings and crucial in therapy trials.
Collapse
Affiliation(s)
- Menno D Stellingwerff
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam and Amsterdam Neuroscience, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Mark A van de Wiel
- Department of Epidemiology and Data Science, and Amsterdam School of Public Health, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Marjo S van der Knaap
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam and Amsterdam Neuroscience, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, The Netherlands.
| |
Collapse
|
26
|
Storkebaum E, Rosenblum K, Sonenberg N. Messenger RNA Translation Defects in Neurodegenerative Diseases. N Engl J Med 2023; 388:1015-1030. [PMID: 36920757 DOI: 10.1056/nejmra2215795] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/16/2023]
Affiliation(s)
- Erik Storkebaum
- From the Molecular Neurobiology Laboratory, Donders Center for Neuroscience, Donders Institute for Brain, Cognition, and Behavior, and the Faculty of Science, Radboud University, Nijmegen, the Netherlands (E.S.); the Sagol Department of Neurobiology, Faculty of Natural Sciences, and the Center for Genetic Manipulation in the Brain, University of Haifa, Haifa, Israel (K.R.); and the Department of Biochemistry and Goodman Cancer Institute, McGill University, Montreal (N.S.)
| | - Kobi Rosenblum
- From the Molecular Neurobiology Laboratory, Donders Center for Neuroscience, Donders Institute for Brain, Cognition, and Behavior, and the Faculty of Science, Radboud University, Nijmegen, the Netherlands (E.S.); the Sagol Department of Neurobiology, Faculty of Natural Sciences, and the Center for Genetic Manipulation in the Brain, University of Haifa, Haifa, Israel (K.R.); and the Department of Biochemistry and Goodman Cancer Institute, McGill University, Montreal (N.S.)
| | - Nahum Sonenberg
- From the Molecular Neurobiology Laboratory, Donders Center for Neuroscience, Donders Institute for Brain, Cognition, and Behavior, and the Faculty of Science, Radboud University, Nijmegen, the Netherlands (E.S.); the Sagol Department of Neurobiology, Faculty of Natural Sciences, and the Center for Genetic Manipulation in the Brain, University of Haifa, Haifa, Israel (K.R.); and the Department of Biochemistry and Goodman Cancer Institute, McGill University, Montreal (N.S.)
| |
Collapse
|
27
|
Leucoencefalopatie ereditarie e leucodistrofie dell’adulto. Neurologia 2022. [DOI: 10.1016/s1634-7072(22)47096-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
|
28
|
Man JHK, van Gelder CAGH, Breur M, Okkes D, Molenaar D, van der Sluis S, Abbink T, Altelaar M, van der Knaap MS, Bugiani M. Cortical Pathology in Vanishing White Matter. Cells 2022; 11:cells11223581. [PMID: 36429009 PMCID: PMC9688115 DOI: 10.3390/cells11223581] [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: 07/25/2022] [Revised: 09/24/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022] Open
Abstract
Vanishing white matter (VWM) is classified as a leukodystrophy with astrocytes as primary drivers in its pathogenesis. Magnetic resonance imaging has documented the progressive thinning of cortices in long-surviving patients. Routine histopathological analyses, however, have not yet pointed to cortical involvement in VWM. Here, we provide a comprehensive analysis of the VWM cortex. We employed high-resolution-mass-spectrometry-based proteomics and immunohistochemistry to gain insight into possible molecular disease mechanisms in the cortices of VWM patients. The proteome analysis revealed 268 differentially expressed proteins in the VWM cortices compared to the controls. A majority of these proteins formed a major protein interaction network. A subsequent gene ontology analysis identified enrichment for terms such as cellular metabolism, particularly mitochondrial activity. Importantly, some of the proteins with the most prominent changes in expression were found in astrocytes, indicating cortical astrocytic involvement. Indeed, we confirmed that VWM cortical astrocytes exhibit morphological changes and are less complex in structure than control cells. Our findings also suggest that these astrocytes are immature and not reactive. Taken together, we provide insights into cortical involvement in VWM, which has to be taken into account when developing therapeutic strategies.
Collapse
Affiliation(s)
- Jodie H. K. Man
- Department of Child Neurology, Emma Children’s Hospital, Amsterdam University Medical Centers, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
- Molecular and Cellular Mechanisms, Amsterdam Neuroscience, 1081 HV Amsterdam, The Netherlands
| | - Charlotte A. G. H. van Gelder
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, 3584 CS Utrecht, The Netherlands
- Netherlands Proteomics Center, 3584 CS Utrecht, The Netherlands
| | - Marjolein Breur
- Department of Child Neurology, Emma Children’s Hospital, Amsterdam University Medical Centers, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
- Molecular and Cellular Mechanisms, Amsterdam Neuroscience, 1081 HV Amsterdam, The Netherlands
| | - Daniel Okkes
- Department of Child Neurology, Emma Children’s Hospital, Amsterdam University Medical Centers, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
- Molecular and Cellular Mechanisms, Amsterdam Neuroscience, 1081 HV Amsterdam, The Netherlands
| | - Douwe Molenaar
- Department of Systems Bioinformatics, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Sophie van der Sluis
- Department of Child and Adolescent Psychology and Psychiatry, Complex Trait Genetics, Amsterdam Neuroscience, VU University Medical Center, 1081 HV Amsterdam, The Netherlands
| | - Truus Abbink
- Department of Child Neurology, Emma Children’s Hospital, Amsterdam University Medical Centers, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
- Molecular and Cellular Mechanisms, Amsterdam Neuroscience, 1081 HV Amsterdam, The Netherlands
| | - Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, 3584 CS Utrecht, The Netherlands
- Netherlands Proteomics Center, 3584 CS Utrecht, The Netherlands
| | - Marjo S. van der Knaap
- Department of Child Neurology, Emma Children’s Hospital, Amsterdam University Medical Centers, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
- Molecular and Cellular Mechanisms, Amsterdam Neuroscience, 1081 HV Amsterdam, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Marianna Bugiani
- Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
- Molecular and Cellular Mechanisms, Amsterdam Neuroscience, 1081 HV Amsterdam, The Netherlands
- Department of Pathology, Amsterdam University Medical Centers, 1081 HV Amsterdam, The Netherlands
- Correspondence: ; Tel.: +31-6-48517239
| |
Collapse
|
29
|
Martin S, Allan KC, Pinkard O, Sweet T, Tesar PJ, Coller J. Oligodendrocyte differentiation alters tRNA modifications and codon optimality-mediated mRNA decay. Nat Commun 2022; 13:5003. [PMID: 36008413 PMCID: PMC9411196 DOI: 10.1038/s41467-022-32766-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 08/15/2022] [Indexed: 11/08/2022] Open
Abstract
Oligodendrocytes are specialized cells that confer neuronal myelination in the central nervous system. Leukodystrophies associated with oligodendrocyte deficits and hypomyelination are known to result when a number of tRNA metabolism genes are mutated. Thus, for unknown reasons, oligodendrocytes may be hypersensitive to perturbations in tRNA biology. In this study, we survey the tRNA transcriptome in the murine oligodendrocyte cell lineage and find that specific tRNAs are hypomodified in oligodendrocytes within or near the anticodon compared to oligodendrocyte progenitor cells (OPCs). This hypomodified state may be the result of differential expression of key modification enzymes during oligodendrocyte differentiation. Moreover, we observe a concomitant relationship between tRNA hypomodification and tRNA decoding potential; observing oligodendrocyte specific alterations in codon optimality-mediated mRNA decay and ribosome transit. Our results reveal that oligodendrocytes naturally maintain a delicate, hypersensitized tRNA/mRNA axis. We suggest this axis is a potential mediator of pathology in leukodystrophies and white matter disease when further insult to tRNA metabolism is introduced.
Collapse
Affiliation(s)
- Sophie Martin
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Kevin C Allan
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Otis Pinkard
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Thomas Sweet
- Center for Proteomics and Bioinformatics, Department of Nutrition, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Paul J Tesar
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Jeff Coller
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Biology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
30
|
Pandey S, Shen K, Lee SH, Shen YAA, Wang Y, Otero-García M, Kotova N, Vito ST, Laufer BI, Newton DF, Rezzonico MG, Hanson JE, Kaminker JS, Bohlen CJ, Yuen TJ, Friedman BA. Disease-associated oligodendrocyte responses across neurodegenerative diseases. Cell Rep 2022; 40:111189. [PMID: 36001972 DOI: 10.1016/j.celrep.2022.111189] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 05/17/2022] [Accepted: 07/20/2022] [Indexed: 12/29/2022] Open
Abstract
Oligodendrocyte dysfunction has been implicated in the pathogenesis of neurodegenerative diseases, so understanding oligodendrocyte activation states would shed light on disease processes. We identify three distinct activation states of oligodendrocytes from single-cell RNA sequencing (RNA-seq) of mouse models of Alzheimer's disease (AD) and multiple sclerosis (MS): DA1 (disease-associated1, associated with immunogenic genes), DA2 (disease-associated2, associated with genes influencing survival), and IFN (associated with interferon response genes). Spatial analysis of disease-associated oligodendrocytes (DAOs) in the cuprizone model reveals that DA1 and DA2 are established outside of the lesion area during demyelination and that DA1 repopulates the lesion during remyelination. Independent meta-analysis of human single-nucleus RNA-seq datasets reveals that the transcriptional responses of MS oligodendrocytes share features with mouse models. In contrast, the oligodendrocyte activation signature observed in human AD is largely distinct from those observed in mice. This catalog of oligodendrocyte activation states (http://research-pub.gene.com/OligoLandscape/) will be important to understand disease progression and develop therapeutic interventions.
Collapse
Affiliation(s)
- Shristi Pandey
- Department of OMNI Bioinformatics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Kimberle Shen
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Seung-Hye Lee
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Yun-An A Shen
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Yuanyuan Wang
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | | - Stephen T Vito
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Benjamin I Laufer
- Department of OMNI Bioinformatics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Dwight F Newton
- Department of OMNI Bioinformatics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA; Roche Global IT Solution Centre, Hoffman-La Roche Canada, 7070 Mississauga Road, Mississauga, ON, Canada
| | - Mitchell G Rezzonico
- Department of OMNI Bioinformatics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jesse E Hanson
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Joshua S Kaminker
- Department of OMNI Bioinformatics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Christopher J Bohlen
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Tracy J Yuen
- Department of Neuroscience, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Brad A Friedman
- Department of OMNI Bioinformatics, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
31
|
Ren Y, Yu X, Chen B, Tang H, Niu S, Wang X, Pan H, Zhang Z. Genotypic and phenotypic characteristics of juvenile/adult onset vanishing white matter: a series of 14 Chinese patients. Neurol Sci 2022; 43:4961-4977. [DOI: 10.1007/s10072-022-06011-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/14/2022] [Indexed: 11/29/2022]
|
32
|
Tian Y, Liu Q, Zhou Y, Chen XY, Pan Y, Xu H, Yang Z. Identification of a Novel Heterozygous Mutation in the EIF2B4 Gene Associated With Vanishing White Matter Disease. Front Bioeng Biotechnol 2022; 10:901452. [PMID: 35860328 PMCID: PMC9289103 DOI: 10.3389/fbioe.2022.901452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 05/23/2022] [Indexed: 12/02/2022] Open
Abstract
Vanishing white matter disease (VWM) is one of the most common childhood inherited leukoencephalopathies with autosomal recessive inheritance. Mutations in five genes, EIF2B1-5, have been identified as the major cause of VWM. In this study, a targeted gene capture sequencing panel comprising 160 known pathogenic genes associated with leukoencephalopathies was performed in a large Han Chinese family affected by adult-onset VWM, and a novel heterozygous missense mutation (c.1337G > A [p. R446H]) in EIF2B4 (NM_001034116.2) was detected. Further functional studies in HEK 293 cells showed dramatically reduced EIF2Bδ protein levels in the mutated group compared with the wild-type group. This study revealed that a heterozygous missense mutation (c.1337G > A [p. R446H]) in EIF2B4 was potentially associated with the adult-onset mild phenotype of VWM. In contrast to previous reports, autosomal dominant inheritance was also observed in adult-onset VWM.
Collapse
Affiliation(s)
- Yun Tian
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Qiong Liu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Yafang Zhou
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xiao-Yu Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yongcheng Pan
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Hunan Province in Neurodegenerative Disorders, Central South University, Changsha, China
| | - Hongwei Xu
- Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhuanyi Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Zhuanyi Yang,
| |
Collapse
|
33
|
Wongkittichote P, Mar SS, McKinstry RC, Nguyen H. Case Report: A Novel EIF2B3 Pathogenic Variant in Central Nervous System Hypomyelination/Vanishing White Matter. Front Genet 2022; 13:893057. [PMID: 35783294 PMCID: PMC9247212 DOI: 10.3389/fgene.2022.893057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/11/2022] [Indexed: 11/23/2022] Open
Abstract
Leukodystrophies are a group of heterogeneous disorders affecting brain myelin. Among those, childhood ataxia with central nervous system hypomyelination/vanishing white matter (CACH/VWM) is one of the more common inherited leukodystrophies. Pathogenic variants in one of the genes encoding five subunits of EIF2B are associated with CACH/VWM. Herein, we presented a case of CACH/VWM who developed ataxia following a minor head injury. Brain magnetic resonance imaging showed extensive white matter signal abnormality. Diagnosis of CACH/VWM was confirmed by the presence of compound heterozygous variants in EIF2B3: the previously known pathogenic variant c c.260C>T (p.Ala87Val) and the novel variant c.673C>T (p.Arg225Trp). Based on the American College of Medical Genetics (ACMG) recommendations, we classified p.Arg225Trp as likely pathogenic. We report a novel variant in a patient with CACH/VWM and highlight the importance of genetic testing in patients with leukodystrophies.
Collapse
Affiliation(s)
- Parith Wongkittichote
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, St Louis, MO, United States
| | - Soe Soe Mar
- Division of Pediatric Neurology, Department of Neurology, Washington University School of Medicine, St Louis, MO, United States
| | - Robert C. McKinstry
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, United States
| | - Hoanh Nguyen
- Division of Genetics and Genomic Medicine, Department of Pediatrics, Washington University School of Medicine, St Louis, MO, United States
- *Correspondence: Hoanh Nguyen,
| |
Collapse
|
34
|
Kong F, Zheng H, Liu X, Lin S, Wang J, Guo Z. Association Between Late-Onset Leukoencephalopathy With Vanishing White Matter and Compound Heterozygous EIF2B5 Gene Mutations: A Case Report and Review of the Literature. Front Neurol 2022; 13:813032. [PMID: 35785335 PMCID: PMC9243765 DOI: 10.3389/fneur.2022.813032] [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: 11/11/2021] [Accepted: 05/11/2022] [Indexed: 11/16/2022] Open
Abstract
Leukoencephalopathy with vanishing white matter (LVWM) is an autosomal recessive disease. Ovarioleukodystrophy is defined as LVWM in females showing signs or symptoms of gradual ovarian failure. We present a 38-year-old female with ovarioleukodystrophy who showed status epilepticus, gait instability, slurred speech, abdominal tendon hyperreflexia, and ovarian failure. Abnormal EEG, characteristic magnetic resonance, and unreported EIF2B5 compound heterozygous mutations [c.1016G>A (p.R339Q) and c.1157G>A (p.G386D)] were found. Furthermore, the present report summarizes 20 female patients with adult-onset ovarioleukodystrophy and EIF2B5 gene mutations. In conclusion, a new genetic locus for LVWM was discovered. Compared with previous cases, mutations at different EIF2B5 sites might have different clinical manifestations and obvious clinical heterogeneity.
Collapse
Affiliation(s)
- Fanxin Kong
- Encephalopathy and Psychology Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
- *Correspondence: Fanxin Kong
| | - Haotao Zheng
- Encephalopathy and Psychology Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Xuan Liu
- Encephalopathy and Psychology Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Songjun Lin
- Encephalopathy and Psychology Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Jianjun Wang
- Encephalopathy and Psychology Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
- Jianjun Wang
| | - Zhouke Guo
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, China
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| |
Collapse
|
35
|
Emerging roles of endoplasmic reticulum proteostasis in brain development. Cells Dev 2022; 170:203781. [DOI: 10.1016/j.cdev.2022.203781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 04/12/2022] [Accepted: 04/20/2022] [Indexed: 11/21/2022]
|
36
|
The role of eIF2 phosphorylation in cell and organismal physiology: new roles for well-known actors. Biochem J 2022; 479:1059-1082. [PMID: 35604373 DOI: 10.1042/bcj20220068] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 02/06/2023]
Abstract
Control of protein synthesis (mRNA translation) plays key roles in shaping the proteome and in many physiological, including homeostatic, responses. One long-known translational control mechanism involves phosphorylation of initiation factor, eIF2, which is catalysed by any one of four protein kinases, which are generally activated in response to stresses. They form a key arm of the integrated stress response (ISR). Phosphorylated eIF2 inhibits eIF2B (the protein that promotes exchange of eIF2-bound GDP for GTP) and thus impairs general protein synthesis. However, this mechanism actually promotes translation of certain mRNAs by virtue of specific features they possess. Recent work has uncovered many previously unknown features of this regulatory system. Several studies have yielded crucial insights into the structure and control of eIF2, including that eIF2B is regulated by several metabolites. Recent studies also reveal that control of eIF2 and the ISR helps determine organismal lifespan and surprising roles in sensing mitochondrial stresses and in controlling the mammalian target of rapamycin (mTOR). The latter effect involves an unexpected role for one of the eIF2 kinases, HRI. Phosphoproteomic analysis identified new substrates for another eIF2 kinase, Gcn2, which senses the availability of amino acids. Several genetic disorders arise from mutations in genes for eIF2α kinases or eIF2B (i.e. vanishing white matter disease, VWM and microcephaly, epileptic seizures, microcephaly, hypogenitalism, diabetes and obesity, MEHMO). Furthermore, the eIF2-mediated ISR plays roles in cognitive decline associated with Alzheimer's disease. New findings suggest potential therapeutic value in interfering with the ISR in certain settings, including VWM, for example by using compounds that promote eIF2B activity.
Collapse
|
37
|
Boone M, Wang L, Lawrence RE, Frost A, Walter P, Schoof M. A point mutation in the nucleotide exchange factor eIF2B constitutively activates the integrated stress response by allosteric modulation. eLife 2022; 11:e76171. [PMID: 35416150 PMCID: PMC9132573 DOI: 10.7554/elife.76171] [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: 12/08/2021] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
In eukaryotic cells, stressors reprogram the cellular proteome by activating the integrated stress response (ISR). In its canonical form, stress-sensing kinases phosphorylate the eukaryotic translation initiation factor eIF2 (eIF2-P), which ultimately leads to reduced levels of ternary complex required for initiation of mRNA translation. Previously we showed that translational control is primarily exerted through a conformational switch in eIF2's nucleotide exchange factor, eIF2B, which shifts from its active A-State conformation to its inhibited I-State conformation upon eIF2-P binding, resulting in reduced nucleotide exchange on eIF2 (Schoof et al. 2021). Here, we show functionally and structurally how a single histidine to aspartate point mutation in eIF2B's β subunit (H160D) mimics the effects of eIF2-P binding by promoting an I-State like conformation, resulting in eIF2-P independent activation of the ISR. These findings corroborate our previously proposed A/I-State model of allosteric ISR regulation.
Collapse
Affiliation(s)
- Morgane Boone
- Howard Hughes Medical Institute, University of California at San FranciscoSan FranciscoUnited States
- Department of Biochemistry and Biophysics, University of California at San FranciscoSan FranciscoUnited States
| | - Lan Wang
- Howard Hughes Medical Institute, University of California at San FranciscoSan FranciscoUnited States
- Department of Biochemistry and Biophysics, University of California at San FranciscoSan FranciscoUnited States
| | - Rosalie E Lawrence
- Howard Hughes Medical Institute, University of California at San FranciscoSan FranciscoUnited States
- Department of Biochemistry and Biophysics, University of California at San FranciscoSan FranciscoUnited States
| | - Adam Frost
- Department of Biochemistry and Biophysics, University of California at San FranciscoSan FranciscoUnited States
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | - Peter Walter
- Howard Hughes Medical Institute, University of California at San FranciscoSan FranciscoUnited States
- Department of Biochemistry and Biophysics, University of California at San FranciscoSan FranciscoUnited States
| | - Michael Schoof
- Howard Hughes Medical Institute, University of California at San FranciscoSan FranciscoUnited States
- Department of Biochemistry and Biophysics, University of California at San FranciscoSan FranciscoUnited States
| |
Collapse
|
38
|
van der Knaap MS, Bonkowsky JL, Vanderver A, Schiffmann R, Krägeloh-Mann I, Bertini E, Bernard G, Fatemi SA, Wolf NI, Saunier-Vivar E, Rauner R, Dekker H, van Bokhoven P, van de Ven P, Leferink PS. Therapy Trial Design in Vanishing White Matter: An Expert Consortium Opinion. Neurol Genet 2022; 8:e657. [PMID: 35128050 PMCID: PMC8811717 DOI: 10.1212/nxg.0000000000000657] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 12/21/2021] [Indexed: 01/04/2023]
Abstract
Vanishing white matter (VWM) is a leukodystrophy caused by recessive variants in the genes EIF2B1-EIF2B5. It is characterized by chronic neurologic deterioration with superimposed stress-provoked episodes of rapid decline. Disease onset spans from the antenatal period through senescence. Age at onset predicts disease evolution for patients with early onset, whereas disease evolution is unpredictable for later onset; patients with infantile and early childhood onset consistently have severe disease with rapid neurologic decline and often early death, whereas patients with later onset have highly variable disease. VWM is rare, but likely underdiagnosed, particularly in adults. Apart from measures to prevent stressors that could provoke acute deteriorations, only symptomatic care is currently offered. With increased insight into VWM disease mechanisms, opportunities for treatment have emerged. EIF2B1-EIF2B5 encode the 5-subunit eukaryotic initiation factor 2B complex, which is essential for translation of mRNAs into proteins and is a principal regulator of the integrated stress response (ISR). ISR deregulation is central to VWM pathology. Targeting components of the ISR has proven beneficial in mutant VWM mouse models, and several drugs are now in clinical development. However, clinical trials in VWM pose considerable challenges: low numbers of known patients with VWM, unpredictable disease course for patients with onset after early childhood, absence of intermediate biomarkers, and novel first-in-human molecular targets. Given these challenges and considering the critical need to offer therapies, we have formulated recommendations for enhanced diagnosis, drug trial setup, and patient selection, based on our expert evaluation of molecular, laboratory, and clinical data.
Collapse
Affiliation(s)
- Marjo S. van der Knaap
- From the Department of Pediatric Neurology (M.S.v.d.K., N.I.W.), Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers; Amsterdam Neuroscience (M.S.v.d.K., N.I.W.); Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands; Division of Pediatric Neurology (J.L.B.), Department of Pediatrics, University of Utah School of Medicine; Primary Children's Hospital (J.L.B.), Intermountain Healthcare, Salt Lake City, UT; Division of Neurology (A.V.), Children's Hospital of Philadelphia; Department of Neurology (A.V.), Perelman School of Medicine, University of Pennsylvania, PA; 4D Molecular Therapeutics (R.S.), Emeryville, CA; Department of Developmental and Child Neurology (I.K.-M.), Social Pediatrics, University Children's Hospital Tübingen, Germany; Department of Neuroscience (E.B.), Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy; Departments of Neurology and Neurosurgery (G.B.), Pediatrics and Human Genetics, McGill University; Department Specialized Medicine (G.B.), Division of Medical Genetics, McGill University Health Center; Child Health and Human Development Program (G.B.), Research Institute of the McGill University Health Center, Montreal, Canada; Kennedy Krieger Institute (S.A.F.), Johns Hopkins University, Baltimore, MD; Research Department (E.S.-V.), European Leukodystrophies Association International and European Leukodystrophies Association France, Paris, France; United Leukodystrophy Foundation (R.R.), DeKalb, IL; Vereniging Volwassenen, Kinderen en Stofwisselingsziekten (H.D.), Zwolle, the Netherlands; Industry Alliance Office (P.v.B., P.S.L.), Amsterdam Neuroscience, Amsterdam University Medical Centers; and Department of Epidemiology and Data Science (P.v.d.V.), Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, the Netherlands
| | - Joshua L. Bonkowsky
- From the Department of Pediatric Neurology (M.S.v.d.K., N.I.W.), Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers; Amsterdam Neuroscience (M.S.v.d.K., N.I.W.); Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands; Division of Pediatric Neurology (J.L.B.), Department of Pediatrics, University of Utah School of Medicine; Primary Children's Hospital (J.L.B.), Intermountain Healthcare, Salt Lake City, UT; Division of Neurology (A.V.), Children's Hospital of Philadelphia; Department of Neurology (A.V.), Perelman School of Medicine, University of Pennsylvania, PA; 4D Molecular Therapeutics (R.S.), Emeryville, CA; Department of Developmental and Child Neurology (I.K.-M.), Social Pediatrics, University Children's Hospital Tübingen, Germany; Department of Neuroscience (E.B.), Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy; Departments of Neurology and Neurosurgery (G.B.), Pediatrics and Human Genetics, McGill University; Department Specialized Medicine (G.B.), Division of Medical Genetics, McGill University Health Center; Child Health and Human Development Program (G.B.), Research Institute of the McGill University Health Center, Montreal, Canada; Kennedy Krieger Institute (S.A.F.), Johns Hopkins University, Baltimore, MD; Research Department (E.S.-V.), European Leukodystrophies Association International and European Leukodystrophies Association France, Paris, France; United Leukodystrophy Foundation (R.R.), DeKalb, IL; Vereniging Volwassenen, Kinderen en Stofwisselingsziekten (H.D.), Zwolle, the Netherlands; Industry Alliance Office (P.v.B., P.S.L.), Amsterdam Neuroscience, Amsterdam University Medical Centers; and Department of Epidemiology and Data Science (P.v.d.V.), Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, the Netherlands
| | - Adeline Vanderver
- From the Department of Pediatric Neurology (M.S.v.d.K., N.I.W.), Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers; Amsterdam Neuroscience (M.S.v.d.K., N.I.W.); Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands; Division of Pediatric Neurology (J.L.B.), Department of Pediatrics, University of Utah School of Medicine; Primary Children's Hospital (J.L.B.), Intermountain Healthcare, Salt Lake City, UT; Division of Neurology (A.V.), Children's Hospital of Philadelphia; Department of Neurology (A.V.), Perelman School of Medicine, University of Pennsylvania, PA; 4D Molecular Therapeutics (R.S.), Emeryville, CA; Department of Developmental and Child Neurology (I.K.-M.), Social Pediatrics, University Children's Hospital Tübingen, Germany; Department of Neuroscience (E.B.), Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy; Departments of Neurology and Neurosurgery (G.B.), Pediatrics and Human Genetics, McGill University; Department Specialized Medicine (G.B.), Division of Medical Genetics, McGill University Health Center; Child Health and Human Development Program (G.B.), Research Institute of the McGill University Health Center, Montreal, Canada; Kennedy Krieger Institute (S.A.F.), Johns Hopkins University, Baltimore, MD; Research Department (E.S.-V.), European Leukodystrophies Association International and European Leukodystrophies Association France, Paris, France; United Leukodystrophy Foundation (R.R.), DeKalb, IL; Vereniging Volwassenen, Kinderen en Stofwisselingsziekten (H.D.), Zwolle, the Netherlands; Industry Alliance Office (P.v.B., P.S.L.), Amsterdam Neuroscience, Amsterdam University Medical Centers; and Department of Epidemiology and Data Science (P.v.d.V.), Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, the Netherlands
| | - Raphael Schiffmann
- From the Department of Pediatric Neurology (M.S.v.d.K., N.I.W.), Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers; Amsterdam Neuroscience (M.S.v.d.K., N.I.W.); Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands; Division of Pediatric Neurology (J.L.B.), Department of Pediatrics, University of Utah School of Medicine; Primary Children's Hospital (J.L.B.), Intermountain Healthcare, Salt Lake City, UT; Division of Neurology (A.V.), Children's Hospital of Philadelphia; Department of Neurology (A.V.), Perelman School of Medicine, University of Pennsylvania, PA; 4D Molecular Therapeutics (R.S.), Emeryville, CA; Department of Developmental and Child Neurology (I.K.-M.), Social Pediatrics, University Children's Hospital Tübingen, Germany; Department of Neuroscience (E.B.), Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy; Departments of Neurology and Neurosurgery (G.B.), Pediatrics and Human Genetics, McGill University; Department Specialized Medicine (G.B.), Division of Medical Genetics, McGill University Health Center; Child Health and Human Development Program (G.B.), Research Institute of the McGill University Health Center, Montreal, Canada; Kennedy Krieger Institute (S.A.F.), Johns Hopkins University, Baltimore, MD; Research Department (E.S.-V.), European Leukodystrophies Association International and European Leukodystrophies Association France, Paris, France; United Leukodystrophy Foundation (R.R.), DeKalb, IL; Vereniging Volwassenen, Kinderen en Stofwisselingsziekten (H.D.), Zwolle, the Netherlands; Industry Alliance Office (P.v.B., P.S.L.), Amsterdam Neuroscience, Amsterdam University Medical Centers; and Department of Epidemiology and Data Science (P.v.d.V.), Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, the Netherlands
| | - Ingeborg Krägeloh-Mann
- From the Department of Pediatric Neurology (M.S.v.d.K., N.I.W.), Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers; Amsterdam Neuroscience (M.S.v.d.K., N.I.W.); Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands; Division of Pediatric Neurology (J.L.B.), Department of Pediatrics, University of Utah School of Medicine; Primary Children's Hospital (J.L.B.), Intermountain Healthcare, Salt Lake City, UT; Division of Neurology (A.V.), Children's Hospital of Philadelphia; Department of Neurology (A.V.), Perelman School of Medicine, University of Pennsylvania, PA; 4D Molecular Therapeutics (R.S.), Emeryville, CA; Department of Developmental and Child Neurology (I.K.-M.), Social Pediatrics, University Children's Hospital Tübingen, Germany; Department of Neuroscience (E.B.), Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy; Departments of Neurology and Neurosurgery (G.B.), Pediatrics and Human Genetics, McGill University; Department Specialized Medicine (G.B.), Division of Medical Genetics, McGill University Health Center; Child Health and Human Development Program (G.B.), Research Institute of the McGill University Health Center, Montreal, Canada; Kennedy Krieger Institute (S.A.F.), Johns Hopkins University, Baltimore, MD; Research Department (E.S.-V.), European Leukodystrophies Association International and European Leukodystrophies Association France, Paris, France; United Leukodystrophy Foundation (R.R.), DeKalb, IL; Vereniging Volwassenen, Kinderen en Stofwisselingsziekten (H.D.), Zwolle, the Netherlands; Industry Alliance Office (P.v.B., P.S.L.), Amsterdam Neuroscience, Amsterdam University Medical Centers; and Department of Epidemiology and Data Science (P.v.d.V.), Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, the Netherlands
| | - Enrico Bertini
- From the Department of Pediatric Neurology (M.S.v.d.K., N.I.W.), Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers; Amsterdam Neuroscience (M.S.v.d.K., N.I.W.); Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands; Division of Pediatric Neurology (J.L.B.), Department of Pediatrics, University of Utah School of Medicine; Primary Children's Hospital (J.L.B.), Intermountain Healthcare, Salt Lake City, UT; Division of Neurology (A.V.), Children's Hospital of Philadelphia; Department of Neurology (A.V.), Perelman School of Medicine, University of Pennsylvania, PA; 4D Molecular Therapeutics (R.S.), Emeryville, CA; Department of Developmental and Child Neurology (I.K.-M.), Social Pediatrics, University Children's Hospital Tübingen, Germany; Department of Neuroscience (E.B.), Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy; Departments of Neurology and Neurosurgery (G.B.), Pediatrics and Human Genetics, McGill University; Department Specialized Medicine (G.B.), Division of Medical Genetics, McGill University Health Center; Child Health and Human Development Program (G.B.), Research Institute of the McGill University Health Center, Montreal, Canada; Kennedy Krieger Institute (S.A.F.), Johns Hopkins University, Baltimore, MD; Research Department (E.S.-V.), European Leukodystrophies Association International and European Leukodystrophies Association France, Paris, France; United Leukodystrophy Foundation (R.R.), DeKalb, IL; Vereniging Volwassenen, Kinderen en Stofwisselingsziekten (H.D.), Zwolle, the Netherlands; Industry Alliance Office (P.v.B., P.S.L.), Amsterdam Neuroscience, Amsterdam University Medical Centers; and Department of Epidemiology and Data Science (P.v.d.V.), Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, the Netherlands
| | - Genevieve Bernard
- From the Department of Pediatric Neurology (M.S.v.d.K., N.I.W.), Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers; Amsterdam Neuroscience (M.S.v.d.K., N.I.W.); Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands; Division of Pediatric Neurology (J.L.B.), Department of Pediatrics, University of Utah School of Medicine; Primary Children's Hospital (J.L.B.), Intermountain Healthcare, Salt Lake City, UT; Division of Neurology (A.V.), Children's Hospital of Philadelphia; Department of Neurology (A.V.), Perelman School of Medicine, University of Pennsylvania, PA; 4D Molecular Therapeutics (R.S.), Emeryville, CA; Department of Developmental and Child Neurology (I.K.-M.), Social Pediatrics, University Children's Hospital Tübingen, Germany; Department of Neuroscience (E.B.), Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy; Departments of Neurology and Neurosurgery (G.B.), Pediatrics and Human Genetics, McGill University; Department Specialized Medicine (G.B.), Division of Medical Genetics, McGill University Health Center; Child Health and Human Development Program (G.B.), Research Institute of the McGill University Health Center, Montreal, Canada; Kennedy Krieger Institute (S.A.F.), Johns Hopkins University, Baltimore, MD; Research Department (E.S.-V.), European Leukodystrophies Association International and European Leukodystrophies Association France, Paris, France; United Leukodystrophy Foundation (R.R.), DeKalb, IL; Vereniging Volwassenen, Kinderen en Stofwisselingsziekten (H.D.), Zwolle, the Netherlands; Industry Alliance Office (P.v.B., P.S.L.), Amsterdam Neuroscience, Amsterdam University Medical Centers; and Department of Epidemiology and Data Science (P.v.d.V.), Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, the Netherlands
| | - Seyed Ali Fatemi
- From the Department of Pediatric Neurology (M.S.v.d.K., N.I.W.), Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers; Amsterdam Neuroscience (M.S.v.d.K., N.I.W.); Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands; Division of Pediatric Neurology (J.L.B.), Department of Pediatrics, University of Utah School of Medicine; Primary Children's Hospital (J.L.B.), Intermountain Healthcare, Salt Lake City, UT; Division of Neurology (A.V.), Children's Hospital of Philadelphia; Department of Neurology (A.V.), Perelman School of Medicine, University of Pennsylvania, PA; 4D Molecular Therapeutics (R.S.), Emeryville, CA; Department of Developmental and Child Neurology (I.K.-M.), Social Pediatrics, University Children's Hospital Tübingen, Germany; Department of Neuroscience (E.B.), Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy; Departments of Neurology and Neurosurgery (G.B.), Pediatrics and Human Genetics, McGill University; Department Specialized Medicine (G.B.), Division of Medical Genetics, McGill University Health Center; Child Health and Human Development Program (G.B.), Research Institute of the McGill University Health Center, Montreal, Canada; Kennedy Krieger Institute (S.A.F.), Johns Hopkins University, Baltimore, MD; Research Department (E.S.-V.), European Leukodystrophies Association International and European Leukodystrophies Association France, Paris, France; United Leukodystrophy Foundation (R.R.), DeKalb, IL; Vereniging Volwassenen, Kinderen en Stofwisselingsziekten (H.D.), Zwolle, the Netherlands; Industry Alliance Office (P.v.B., P.S.L.), Amsterdam Neuroscience, Amsterdam University Medical Centers; and Department of Epidemiology and Data Science (P.v.d.V.), Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, the Netherlands
| | - Nicole I. Wolf
- From the Department of Pediatric Neurology (M.S.v.d.K., N.I.W.), Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers; Amsterdam Neuroscience (M.S.v.d.K., N.I.W.); Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands; Division of Pediatric Neurology (J.L.B.), Department of Pediatrics, University of Utah School of Medicine; Primary Children's Hospital (J.L.B.), Intermountain Healthcare, Salt Lake City, UT; Division of Neurology (A.V.), Children's Hospital of Philadelphia; Department of Neurology (A.V.), Perelman School of Medicine, University of Pennsylvania, PA; 4D Molecular Therapeutics (R.S.), Emeryville, CA; Department of Developmental and Child Neurology (I.K.-M.), Social Pediatrics, University Children's Hospital Tübingen, Germany; Department of Neuroscience (E.B.), Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy; Departments of Neurology and Neurosurgery (G.B.), Pediatrics and Human Genetics, McGill University; Department Specialized Medicine (G.B.), Division of Medical Genetics, McGill University Health Center; Child Health and Human Development Program (G.B.), Research Institute of the McGill University Health Center, Montreal, Canada; Kennedy Krieger Institute (S.A.F.), Johns Hopkins University, Baltimore, MD; Research Department (E.S.-V.), European Leukodystrophies Association International and European Leukodystrophies Association France, Paris, France; United Leukodystrophy Foundation (R.R.), DeKalb, IL; Vereniging Volwassenen, Kinderen en Stofwisselingsziekten (H.D.), Zwolle, the Netherlands; Industry Alliance Office (P.v.B., P.S.L.), Amsterdam Neuroscience, Amsterdam University Medical Centers; and Department of Epidemiology and Data Science (P.v.d.V.), Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, the Netherlands
| | - Elise Saunier-Vivar
- From the Department of Pediatric Neurology (M.S.v.d.K., N.I.W.), Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers; Amsterdam Neuroscience (M.S.v.d.K., N.I.W.); Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands; Division of Pediatric Neurology (J.L.B.), Department of Pediatrics, University of Utah School of Medicine; Primary Children's Hospital (J.L.B.), Intermountain Healthcare, Salt Lake City, UT; Division of Neurology (A.V.), Children's Hospital of Philadelphia; Department of Neurology (A.V.), Perelman School of Medicine, University of Pennsylvania, PA; 4D Molecular Therapeutics (R.S.), Emeryville, CA; Department of Developmental and Child Neurology (I.K.-M.), Social Pediatrics, University Children's Hospital Tübingen, Germany; Department of Neuroscience (E.B.), Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy; Departments of Neurology and Neurosurgery (G.B.), Pediatrics and Human Genetics, McGill University; Department Specialized Medicine (G.B.), Division of Medical Genetics, McGill University Health Center; Child Health and Human Development Program (G.B.), Research Institute of the McGill University Health Center, Montreal, Canada; Kennedy Krieger Institute (S.A.F.), Johns Hopkins University, Baltimore, MD; Research Department (E.S.-V.), European Leukodystrophies Association International and European Leukodystrophies Association France, Paris, France; United Leukodystrophy Foundation (R.R.), DeKalb, IL; Vereniging Volwassenen, Kinderen en Stofwisselingsziekten (H.D.), Zwolle, the Netherlands; Industry Alliance Office (P.v.B., P.S.L.), Amsterdam Neuroscience, Amsterdam University Medical Centers; and Department of Epidemiology and Data Science (P.v.d.V.), Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, the Netherlands
| | - Robert Rauner
- From the Department of Pediatric Neurology (M.S.v.d.K., N.I.W.), Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers; Amsterdam Neuroscience (M.S.v.d.K., N.I.W.); Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands; Division of Pediatric Neurology (J.L.B.), Department of Pediatrics, University of Utah School of Medicine; Primary Children's Hospital (J.L.B.), Intermountain Healthcare, Salt Lake City, UT; Division of Neurology (A.V.), Children's Hospital of Philadelphia; Department of Neurology (A.V.), Perelman School of Medicine, University of Pennsylvania, PA; 4D Molecular Therapeutics (R.S.), Emeryville, CA; Department of Developmental and Child Neurology (I.K.-M.), Social Pediatrics, University Children's Hospital Tübingen, Germany; Department of Neuroscience (E.B.), Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy; Departments of Neurology and Neurosurgery (G.B.), Pediatrics and Human Genetics, McGill University; Department Specialized Medicine (G.B.), Division of Medical Genetics, McGill University Health Center; Child Health and Human Development Program (G.B.), Research Institute of the McGill University Health Center, Montreal, Canada; Kennedy Krieger Institute (S.A.F.), Johns Hopkins University, Baltimore, MD; Research Department (E.S.-V.), European Leukodystrophies Association International and European Leukodystrophies Association France, Paris, France; United Leukodystrophy Foundation (R.R.), DeKalb, IL; Vereniging Volwassenen, Kinderen en Stofwisselingsziekten (H.D.), Zwolle, the Netherlands; Industry Alliance Office (P.v.B., P.S.L.), Amsterdam Neuroscience, Amsterdam University Medical Centers; and Department of Epidemiology and Data Science (P.v.d.V.), Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, the Netherlands
| | - Hanka Dekker
- From the Department of Pediatric Neurology (M.S.v.d.K., N.I.W.), Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers; Amsterdam Neuroscience (M.S.v.d.K., N.I.W.); Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands; Division of Pediatric Neurology (J.L.B.), Department of Pediatrics, University of Utah School of Medicine; Primary Children's Hospital (J.L.B.), Intermountain Healthcare, Salt Lake City, UT; Division of Neurology (A.V.), Children's Hospital of Philadelphia; Department of Neurology (A.V.), Perelman School of Medicine, University of Pennsylvania, PA; 4D Molecular Therapeutics (R.S.), Emeryville, CA; Department of Developmental and Child Neurology (I.K.-M.), Social Pediatrics, University Children's Hospital Tübingen, Germany; Department of Neuroscience (E.B.), Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy; Departments of Neurology and Neurosurgery (G.B.), Pediatrics and Human Genetics, McGill University; Department Specialized Medicine (G.B.), Division of Medical Genetics, McGill University Health Center; Child Health and Human Development Program (G.B.), Research Institute of the McGill University Health Center, Montreal, Canada; Kennedy Krieger Institute (S.A.F.), Johns Hopkins University, Baltimore, MD; Research Department (E.S.-V.), European Leukodystrophies Association International and European Leukodystrophies Association France, Paris, France; United Leukodystrophy Foundation (R.R.), DeKalb, IL; Vereniging Volwassenen, Kinderen en Stofwisselingsziekten (H.D.), Zwolle, the Netherlands; Industry Alliance Office (P.v.B., P.S.L.), Amsterdam Neuroscience, Amsterdam University Medical Centers; and Department of Epidemiology and Data Science (P.v.d.V.), Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, the Netherlands
| | - Pieter van Bokhoven
- From the Department of Pediatric Neurology (M.S.v.d.K., N.I.W.), Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers; Amsterdam Neuroscience (M.S.v.d.K., N.I.W.); Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands; Division of Pediatric Neurology (J.L.B.), Department of Pediatrics, University of Utah School of Medicine; Primary Children's Hospital (J.L.B.), Intermountain Healthcare, Salt Lake City, UT; Division of Neurology (A.V.), Children's Hospital of Philadelphia; Department of Neurology (A.V.), Perelman School of Medicine, University of Pennsylvania, PA; 4D Molecular Therapeutics (R.S.), Emeryville, CA; Department of Developmental and Child Neurology (I.K.-M.), Social Pediatrics, University Children's Hospital Tübingen, Germany; Department of Neuroscience (E.B.), Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy; Departments of Neurology and Neurosurgery (G.B.), Pediatrics and Human Genetics, McGill University; Department Specialized Medicine (G.B.), Division of Medical Genetics, McGill University Health Center; Child Health and Human Development Program (G.B.), Research Institute of the McGill University Health Center, Montreal, Canada; Kennedy Krieger Institute (S.A.F.), Johns Hopkins University, Baltimore, MD; Research Department (E.S.-V.), European Leukodystrophies Association International and European Leukodystrophies Association France, Paris, France; United Leukodystrophy Foundation (R.R.), DeKalb, IL; Vereniging Volwassenen, Kinderen en Stofwisselingsziekten (H.D.), Zwolle, the Netherlands; Industry Alliance Office (P.v.B., P.S.L.), Amsterdam Neuroscience, Amsterdam University Medical Centers; and Department of Epidemiology and Data Science (P.v.d.V.), Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, the Netherlands
| | - Peter van de Ven
- From the Department of Pediatric Neurology (M.S.v.d.K., N.I.W.), Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers; Amsterdam Neuroscience (M.S.v.d.K., N.I.W.); Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands; Division of Pediatric Neurology (J.L.B.), Department of Pediatrics, University of Utah School of Medicine; Primary Children's Hospital (J.L.B.), Intermountain Healthcare, Salt Lake City, UT; Division of Neurology (A.V.), Children's Hospital of Philadelphia; Department of Neurology (A.V.), Perelman School of Medicine, University of Pennsylvania, PA; 4D Molecular Therapeutics (R.S.), Emeryville, CA; Department of Developmental and Child Neurology (I.K.-M.), Social Pediatrics, University Children's Hospital Tübingen, Germany; Department of Neuroscience (E.B.), Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy; Departments of Neurology and Neurosurgery (G.B.), Pediatrics and Human Genetics, McGill University; Department Specialized Medicine (G.B.), Division of Medical Genetics, McGill University Health Center; Child Health and Human Development Program (G.B.), Research Institute of the McGill University Health Center, Montreal, Canada; Kennedy Krieger Institute (S.A.F.), Johns Hopkins University, Baltimore, MD; Research Department (E.S.-V.), European Leukodystrophies Association International and European Leukodystrophies Association France, Paris, France; United Leukodystrophy Foundation (R.R.), DeKalb, IL; Vereniging Volwassenen, Kinderen en Stofwisselingsziekten (H.D.), Zwolle, the Netherlands; Industry Alliance Office (P.v.B., P.S.L.), Amsterdam Neuroscience, Amsterdam University Medical Centers; and Department of Epidemiology and Data Science (P.v.d.V.), Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, the Netherlands
| | - Prisca S. Leferink
- From the Department of Pediatric Neurology (M.S.v.d.K., N.I.W.), Amsterdam Leukodystrophy Center, Emma Children's Hospital, Amsterdam University Medical Centers; Amsterdam Neuroscience (M.S.v.d.K., N.I.W.); Department of Functional Genomics (M.S.v.d.K.), Center for Neurogenomics and Cognitive Research, Vrije Universiteit, Amsterdam, the Netherlands; Division of Pediatric Neurology (J.L.B.), Department of Pediatrics, University of Utah School of Medicine; Primary Children's Hospital (J.L.B.), Intermountain Healthcare, Salt Lake City, UT; Division of Neurology (A.V.), Children's Hospital of Philadelphia; Department of Neurology (A.V.), Perelman School of Medicine, University of Pennsylvania, PA; 4D Molecular Therapeutics (R.S.), Emeryville, CA; Department of Developmental and Child Neurology (I.K.-M.), Social Pediatrics, University Children's Hospital Tübingen, Germany; Department of Neuroscience (E.B.), Unit of Neuromuscular and Neurodegenerative Diseases, Laboratory of Molecular Medicine, Genetics and Rare Diseases Research Division, IRCCS Ospedale Pediatrico Bambino Gesù, Rome 00146, Italy; Departments of Neurology and Neurosurgery (G.B.), Pediatrics and Human Genetics, McGill University; Department Specialized Medicine (G.B.), Division of Medical Genetics, McGill University Health Center; Child Health and Human Development Program (G.B.), Research Institute of the McGill University Health Center, Montreal, Canada; Kennedy Krieger Institute (S.A.F.), Johns Hopkins University, Baltimore, MD; Research Department (E.S.-V.), European Leukodystrophies Association International and European Leukodystrophies Association France, Paris, France; United Leukodystrophy Foundation (R.R.), DeKalb, IL; Vereniging Volwassenen, Kinderen en Stofwisselingsziekten (H.D.), Zwolle, the Netherlands; Industry Alliance Office (P.v.B., P.S.L.), Amsterdam Neuroscience, Amsterdam University Medical Centers; and Department of Epidemiology and Data Science (P.v.d.V.), Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam, the Netherlands
| |
Collapse
|
39
|
Structural Consequence of Non-Synonymous Single-Nucleotide Variants in the N-Terminal Domain of LIS1. Int J Mol Sci 2022; 23:ijms23063109. [PMID: 35328531 PMCID: PMC8955593 DOI: 10.3390/ijms23063109] [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: 02/08/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 02/04/2023] Open
Abstract
Disruptive neuronal migration during early brain development causes severe brain malformation. Characterized by mislocalization of cortical neurons, this condition is a result of the loss of function of migration regulating genes. One known neuronal migration disorder is lissencephaly (LIS), which is caused by deletions or mutations of the LIS1 (PAFAH1B1) gene that has been implicated in regulating the microtubule motor protein cytoplasmic dynein. Although this class of diseases has recently received considerable attention, the roles of non-synonymous polymorphisms (nsSNPs) in LIS1 on lissencephaly progression remain elusive. Therefore, the present study employed combined bioinformatics and molecular modeling approach to identify potential damaging nsSNPs in the LIS1 gene and provide atomic insight into their roles in LIS1 loss of function. Using this approach, we identified three high-risk nsSNPs, including rs121434486 (F31S), rs587784254 (W55R), and rs757993270 (W55L) in the LIS1 gene, which are located on the N-terminal domain of LIS1. Molecular dynamics simulation highlighted that all variants decreased helical conformation, increased the intermonomeric distance, and thus disrupted intermonomeric contacts in the LIS1 dimer. Furthermore, the presence of variants also caused a loss of positive electrostatic potential and reduced dimer binding potential. Since self-dimerization is an essential aspect of LIS1 to recruit interacting partners, thus these variants are associated with the loss of LIS1 functions. As a corollary, these findings may further provide critical insights on the roles of LIS1 variants in brain malformation.
Collapse
|
40
|
Korneeva NL. Integrated Stress Response in Neuronal Pathology and in Health. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:S111-S127. [PMID: 35501991 DOI: 10.1134/s0006297922140103] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 10/29/2021] [Accepted: 11/02/2021] [Indexed: 06/14/2023]
Abstract
Neurodegeneration involves progressive pathological loss of a specific population of neurons, glial activation, and dysfunction of myelinating oligodendrocytes leading to cognitive impairment and altered movement, breathing, and senses. Neuronal degeneration is a hallmark of aging, stroke, drug abuse, toxic chemical exposure, viral infection, chronic inflammation, and a variety of neurological diseases. Accumulation of intra- and extracellular protein aggregates is a common characteristic of cell pathologies. Excessive production of reactive oxygen species and nitric oxide, induction of endoplasmic reticulum stress, and accumulation of misfolded protein aggregates have been shown to trigger a defensive mechanism called integrated stress response (ISR). Activation of ISR is important for synaptic plasticity in learning and memory formation. However, sustaining of ISR may lead to the development of neuronal pathologies and altered patterns in behavior and perception.
Collapse
Affiliation(s)
- Nadejda L Korneeva
- Louisiana State University Health Science Center, Shreveport, LA 71103, USA.
| |
Collapse
|
41
|
Lanciotti A, Brignone MS, Macioce P, Visentin S, Ambrosini E. Human iPSC-Derived Astrocytes: A Powerful Tool to Study Primary Astrocyte Dysfunction in the Pathogenesis of Rare Leukodystrophies. Int J Mol Sci 2021; 23:ijms23010274. [PMID: 35008700 PMCID: PMC8745131 DOI: 10.3390/ijms23010274] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 12/11/2022] Open
Abstract
Astrocytes are very versatile cells, endowed with multitasking capacities to ensure brain homeostasis maintenance from brain development to adult life. It has become increasingly evident that astrocytes play a central role in many central nervous system pathologies, not only as regulators of defensive responses against brain insults but also as primary culprits of the disease onset and progression. This is particularly evident in some rare leukodystrophies (LDs) where white matter/myelin deterioration is due to primary astrocyte dysfunctions. Understanding the molecular defects causing these LDs may help clarify astrocyte contribution to myelin formation/maintenance and favor the identification of possible therapeutic targets for LDs and other CNS demyelinating diseases. To date, the pathogenic mechanisms of these LDs are poorly known due to the rarity of the pathological tissue and the failure of the animal models to fully recapitulate the human diseases. Thus, the development of human induced pluripotent stem cells (hiPSC) from patient fibroblasts and their differentiation into astrocytes is a promising approach to overcome these issues. In this review, we discuss the primary role of astrocytes in LD pathogenesis, the experimental models currently available and the advantages, future evolutions, perspectives, and limitations of hiPSC to study pathologies implying astrocyte dysfunctions.
Collapse
Affiliation(s)
- Angela Lanciotti
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy; (A.L.); (M.S.B.); (P.M.)
| | - Maria Stefania Brignone
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy; (A.L.); (M.S.B.); (P.M.)
| | - Pompeo Macioce
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy; (A.L.); (M.S.B.); (P.M.)
| | - Sergio Visentin
- National Center for Research and Preclinical and Clinical Evaluation of Drugs, Istituto Superiore di Sanità, 00169 Rome, Italy;
| | - Elena Ambrosini
- Department of Neuroscience, Istituto Superiore di Sanità, 00169 Rome, Italy; (A.L.); (M.S.B.); (P.M.)
- Correspondence: ; Tel.: +39-064-990-2037
| |
Collapse
|
42
|
Deng J, Zhou L, Zhang J, Chang X, Jiang Y, Wang J, Wu Y. Correlation Between Genotype and Age of Onset in Leukoencephalopathy With Vanishing White Matter. Front Genet 2021; 12:729777. [PMID: 34745209 PMCID: PMC8564072 DOI: 10.3389/fgene.2021.729777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
Purpose: Leukoencephalopathy with vanishing white matter (VWM) is an autosomal recessive leukoencephalopathy caused by mutations in any of the five genes encoding the subunits of eukaryotic translation initiation factor 2B (eIF2B). The severity of the disease varies considerably, and its genotypic-phenotypic correlation is still unclear. Age of onset is the only independent clinical predictor for VWM severity. In this study, the correlation between genotype and age at onset of patients was investigated. Methods: Data were collected from patients with VWM in the available literature reports and from those diagnosed in Peking University First Hospital. The age of onset was divided into early-onset (≤4 years) and late-onset type (>4 years) for the analysis of the correlation between genotype and age of onset in patients with VWM. Results: A total of 341 patients were included, 281 were reported in 87 available articles and 60 were diagnosed in our center. A total of 180 different mutations were found, among which 86.1% were missense. The gene (EIF2B1-5) in which the mutation located, and the number of null alleles were not associated with age of onset in these patients. Certain mutations such as eIF2Bε[Arg195His] and eIF2Bε[Arg269Gln] that were predicted to have a serious influence on eIF2B structure were related to earlier age of onset. EIF2Bγ[Ala87Val] which was predicted to have a minimal influence on eIF2B structure, was related to later age of onset. Whereas eIF2Bβ[Glu213Gly], eIF2Bβ[Gly200Val] and eIF2Bε[Thr91Ala], also predicted having a small effect on the structure of eIF2B, did not show correlation with the age of onset. The onset age of patients with one or biallelic missense mutations located in the catalytic domain or other homologous domains in catalytic subunits (eIF2Bγ, ε) was earlier than that of patients with biallelic mutations located in the NT domain. Conclusion: The onset age of patients with different genotypes varied greatly. The degree of influence in protein structure of some missense mutations was correlated with phenotypic severity, but the results were not completely consistent. The combined effect of biallelic mutations, the role of regulatory genes, environmental stress and other potential factors on phenotypes need to be further explored.
Collapse
Affiliation(s)
- Jiong Deng
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Ling Zhou
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Jie Zhang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Xuting Chang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Yuwu Jiang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Jingmin Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Ye Wu
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| |
Collapse
|
43
|
Gowda VK, Srinivasan VM, Nagarajan B, Bhat M, Shivappa SK, Benakappa N. Profile of Indian Children with Childhood Ataxia and Central Nervous System Hypomyelination/Vanishing White Matter Disease: A Single Center Experience from Southern India. J Pediatr Genet 2021; 10:205-212. [PMID: 34504724 DOI: 10.1055/s-0040-1714717] [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: 05/10/2020] [Accepted: 06/28/2020] [Indexed: 10/23/2022]
Abstract
Background Childhood ataxia with central nervous system hypomyelination (CACH) is a recently described childhood inherited white matter disease, caused by mutations in any of the five genes encoding eukaryotic translation initiation factor ( eIF2B ). Methods Retrospective review of the charts of children with CACH was performed from January 2014 to March 2020 at tertiary care center from Southern India. Diagnosis was based on magnetic resonance imaging (MRI) criteria or genetic testing. Results Total number of children with CACH enrolled were 18. Male/female ratio was 10:8. Mean age of presentation was 37.11 months (range = 6-144 months). Affected siblings were seen in five (28%) cases. All children had spasticity, ataxia, and diffuse white matter changes with similar signal as cerebrospinal fluid on all pulse sequences on MRI brain. Of the 18 children, only nine are alive. Duration of illness among deceased children was 9.6667 months (range = 2-16 months). Waxing and waning of symptoms were seen in seven cases. Genetic analysis of EIF2B gene was performed in five cases, among which three mutations were novel. Conclusion A diagnosis of childhood ataxia with central nervous system hypomyelination should be considered in patients presenting with acute onset neuroregression following infection or trauma with associated neuroimaging showing classical white matter findings.
Collapse
Affiliation(s)
- Vykuntaraju K Gowda
- Department of Pediatric Neurology, Indira Gandhi Institute of Child Health, Bengaluru, Karnataka, India
| | - Varunvenkat M Srinivasan
- Department of Pediatric Neurology, Indira Gandhi Institute of Child Health, Bengaluru, Karnataka, India
| | - Balamurugan Nagarajan
- Department of Pediatric Neurology, Indira Gandhi Institute of Child Health, Bengaluru, Karnataka, India
| | - Maya Bhat
- Department of Neuroradiology, National Institute of Mental Health and Neurosciences, Bangalore, Karnataka, India
| | - Sanjay K Shivappa
- Department of Pediatric Medicine, Indira Gandhi Institute of Child Health, Bengaluru, Karnataka, India
| | - Naveen Benakappa
- Department of Pediatric Medicine, Indira Gandhi Institute of Child Health, Bengaluru, Karnataka, India
| |
Collapse
|
44
|
English AM, Green KM, Moon SL. A (dis)integrated stress response: Genetic diseases of eIF2α regulators. WILEY INTERDISCIPLINARY REVIEWS-RNA 2021; 13:e1689. [PMID: 34463036 DOI: 10.1002/wrna.1689] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 01/28/2023]
Abstract
The integrated stress response (ISR) is a conserved mechanism by which eukaryotic cells remodel gene expression to adapt to intrinsic and extrinsic stressors rapidly and reversibly. The ISR is initiated when stress-activated protein kinases phosphorylate the major translation initiation factor eukaryotic translation initiation factor 2ɑ (eIF2ɑ), which globally suppresses translation initiation activity and permits the selective translation of stress-induced genes including important transcription factors such as activating transcription factor 4 (ATF4). Translationally repressed messenger RNAs (mRNAs) and noncoding RNAs assemble into cytoplasmic RNA-protein granules and polyadenylated RNAs are concomitantly stabilized. Thus, regulated changes in mRNA translation, stability, and localization to RNA-protein granules contribute to the reprogramming of gene expression that defines the ISR. We discuss fundamental mechanisms of RNA regulation during the ISR and provide an overview of a growing class of genetic disorders associated with mutant alleles of key translation factors in the ISR pathway. This article is categorized under: RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications RNA in Disease and Development > RNA in Disease Translation > Translation Regulation RNA in Disease and Development > RNA in Development.
Collapse
Affiliation(s)
- Alyssa M English
- Department of Human Genetics, Center for RNA Biomedicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Katelyn M Green
- Department of Chemistry, Department of Human Genetics, Center for RNA Biomedicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Stephanie L Moon
- Department of Human Genetics, Center for RNA Biomedicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
45
|
Smit WL, de Boer RJ, Meijer BJ, Spaan CN, van Roest M, Koelink PJ, Koster J, Dekker E, Abbink TEM, van der Knaap MS, van den Brink GR, Muncan V, Heijmans J. Translation initiation factor eIF2Bε promotes Wnt-mediated clonogenicity and global translation in intestinal epithelial cells. Stem Cell Res 2021; 55:102499. [PMID: 34399164 DOI: 10.1016/j.scr.2021.102499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/02/2021] [Accepted: 08/05/2021] [Indexed: 11/30/2022] Open
Abstract
Modulation of global mRNA translation, which is essential for intestinal stem cell function, is controlled by Wnt signaling. Loss of tumor supressor APC in stem cells drives adenoma formation through hyperactivion of Wnt signaling and dysregulated translational control. It is unclear whether factors that coordinate global translation in the intestinal epithelium are needed for APC-driven malignant transformation. Here we identified nucleotide exchange factor eIF2Bε as a translation initiation factor involved in Wnt-mediated intestinal epithelial stemness. Using eIF2BεArg191His mice with a homozygous point mutation that leads to dysfunction in the enzymatic activity, we demonstrate that eIF2Bε is involved in small intestinal crypt formation, stemness marker expression, and secreted Paneth cell-derived granule formation. Wnt hyperactivation in ex vivo eIF2BεArg191His organoids, using a GSK3β inhibitor to mimic Apc driven transformation, shows that eIF2Bε is essential for Wnt-mediated clonogenicity and associated increase of the global translational capacity. Finally, we observe high eIF2Bε expression in human colonic adenoma tissues, exposing eIF2Bε as a potential target of CRC stem cells with aberrant Wnt signaling.
Collapse
Affiliation(s)
- W L Smit
- Amsterdam UMC, University of Amsterdam, Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 71, Amsterdam, the Netherlands
| | - R J de Boer
- Amsterdam UMC, University of Amsterdam, Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 71, Amsterdam, the Netherlands
| | - B J Meijer
- Amsterdam UMC, University of Amsterdam, Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 71, Amsterdam, the Netherlands
| | - C N Spaan
- Amsterdam UMC, University of Amsterdam, Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 71, Amsterdam, the Netherlands
| | - M van Roest
- Amsterdam UMC, University of Amsterdam, Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 71, Amsterdam, the Netherlands
| | - P J Koelink
- Amsterdam UMC, University of Amsterdam, Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 71, Amsterdam, the Netherlands
| | - J Koster
- Amsterdam UMC, University of Amsterdam, Department of Oncogenomics, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - E Dekker
- Department of Gastroenterology and Hepatology, Amsterdam University Medical Center, Location Academic Medical Center, Amsterdam, the Netherlands
| | - T E M Abbink
- Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, the Netherlands; Department of Functional Genomics, Amsterdam Neuroscience, VU University, Amsterdam, the Netherlands
| | - M S van der Knaap
- Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - G R van den Brink
- Amsterdam UMC, University of Amsterdam, Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 71, Amsterdam, the Netherlands; Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - V Muncan
- Amsterdam UMC, University of Amsterdam, Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 71, Amsterdam, the Netherlands
| | - J Heijmans
- Amsterdam UMC, University of Amsterdam, Department of Gastroenterology and Hepatology, Tytgat Institute for Liver and Intestinal Research, Meibergdreef 71, Amsterdam, the Netherlands; Amsterdam UMC, University of Amsterdam, Department of Internal Medicine, Meibergdreef 9, Amsterdam, the Netherlands.
| |
Collapse
|
46
|
Mallucci GR, Klenerman D, Rubinsztein DC. Developing Therapies for Neurodegenerative Disorders: Insights from Protein Aggregation and Cellular Stress Responses. Annu Rev Cell Dev Biol 2021; 36:165-189. [PMID: 33021824 DOI: 10.1146/annurev-cellbio-040320-120625] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
As the world's population ages, neurodegenerative disorders are poised to become the commonest cause of death. Despite this, they remain essentially untreatable. Characterized pathologically both by the aggregation of disease-specific misfolded proteins and by changes in cellular stress responses, to date, therapeutic approaches have focused almost exclusively on reducing misfolded protein load-notably amyloid beta (Aβ) in Alzheimer's disease. The repeated failure of clinical trials has led to despondency over the possibility that these disorders will ever be treated. We argue that this is in fact a time for optimism: Targeting various generic stress responses is emerging as an increasingly promising means of modifying disease progression across these disorders. New treatments are approaching clinical trials, while novel means of targeting aggregates could eventually act preventively in early disease.
Collapse
Affiliation(s)
- Giovanna R Mallucci
- UK Dementia Research Institute at the University of Cambridge, Cambridge CB2 0AH, United Kingdom; .,Department of Clinical Neurosciences, University of Cambridge, Cambridge CB2 0AH, United Kingdom
| | - David Klenerman
- UK Dementia Research Institute at the University of Cambridge, Cambridge CB2 0AH, United Kingdom; .,Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, United Kingdom
| | - David C Rubinsztein
- UK Dementia Research Institute at the University of Cambridge, Cambridge CB2 0AH, United Kingdom; .,Cambridge Institute for Medical Research, Cambridge CB2 0XY, United Kingdom
| |
Collapse
|
47
|
Barczykowski AL, Langan TJ, Vanderver A, Jalal K, Carter RL. Death rates in the U.S. due to Leukodystrophies with pediatric forms. Am J Med Genet A 2021; 185:2361-2373. [PMID: 33960638 PMCID: PMC11431180 DOI: 10.1002/ajmg.a.62248] [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: 07/20/2020] [Revised: 03/26/2021] [Accepted: 04/16/2021] [Indexed: 11/08/2022]
Abstract
To use national mortality and state death certificate records to estimate disease specific mortality rates among pediatric and adult populations for 23 leukodystrophies (LDs) with pediatric forms. Additionally, to calculate yearly prevalence and caseload of the most severe LD cases that will eventually result in pediatric death (i.e., pediatric fatality cases). Death certificate records describing cause of death were collected from states based on 10 ICD-10 codes associated with the 23 LDs. Deaths in the U.S. with these codes were distributed into categories based on proportions identified in state death certificate data. Mortality rates, prevalence, and caseload were calculated from resulting expected numbers, population sizes, and average lifetimes. An estimated 1.513 per 1,000,000 0-17 year old's died of these LDs at average age 5.2 years and 0.194 for those ≥18 at an average age of 42.3 years. Prevalence of pediatric fatality cases of these LDs declined from 1999 through 2007 and then remained constant at 6.2 per million children per year through 2012. Epidemiological information, currently lacking for rare diseases, is useful to newborn screening programs, research funding agencies, and care centers for LD patients. Methods used here are generally useful for studying rare diseases.
Collapse
Affiliation(s)
- Amy L. Barczykowski
- Population Health Observatory, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York
- Department of Biostatistics, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York
| | - Thomas J. Langan
- Hunter James Kelly Research Institute, School of Medicine and Biomedical Sciences School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
- Department of Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | - Adeline Vanderver
- The Division of Neurology, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania
- The Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kabir Jalal
- Population Health Observatory, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York
- Department of Biostatistics, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York
| | - Randy L. Carter
- Population Health Observatory, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York
- Department of Biostatistics, School of Public Health and Health Professions, University at Buffalo, Buffalo, New York
| |
Collapse
|
48
|
Maurano M, Snyder JM, Connelly C, Henao-Mejia J, Sidrauski C, Stetson DB. Protein kinase R and the integrated stress response drive immunopathology caused by mutations in the RNA deaminase ADAR1. Immunity 2021; 54:1948-1960.e5. [PMID: 34343497 DOI: 10.1016/j.immuni.2021.07.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/18/2021] [Accepted: 06/30/2021] [Indexed: 10/20/2022]
Abstract
The RNA deaminase ADAR1 is an essential negative regulator of the RNA sensor MDA5, and loss of ADAR1 function triggers inappropriate activation of MDA5 by self-RNAs. Mutations in ADAR, the gene that encodes ADAR1, cause human immune diseases, including Aicardi-Goutières syndrome (AGS). However, the mechanisms of MDA5-dependent disease pathogenesis in vivo remain unknown. Here we generated mice with a single amino acid change in ADAR1 that models the most common human ADAR AGS mutation. These Adar mutant mice developed lethal disease that required MDA5, the RIG-I-like receptor LGP2, type I interferons, and the eIF2α kinase PKR. A small-molecule inhibitor of the integrated stress response (ISR) that acts downstream of eIF2α phosphorylation prevented immunopathology and rescued the mice from mortality. These findings place PKR and the ISR as central components of immunopathology in vivo and identify therapeutic targets for treatment of human diseases associated with the ADAR1-MDA5 axis.
Collapse
Affiliation(s)
- Megan Maurano
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98195, USA; Medical Scientist Training Program, University of Washington School of Medicine, Seattle, WA 98195, USA; Molecular and Cellular Biology Graduate Program, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Jessica M Snyder
- Department of Comparative Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | | | - Jorge Henao-Mejia
- Department of Pathology and Laboratory Medicine, Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; Division of Protective Immunity, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104
| | | | - Daniel B Stetson
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98195, USA.
| |
Collapse
|
49
|
Herrero M, Daw M, Atzmon A, Elroy-Stein O. The Energy Status of Astrocytes Is the Achilles' Heel of eIF2B-Leukodystrophy. Cells 2021; 10:1858. [PMID: 34440627 PMCID: PMC8393801 DOI: 10.3390/cells10081858] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/24/2022] Open
Abstract
Translation initiation factor 2B (eIF2B) is a master regulator of global protein synthesis in all cell types. The mild genetic Eif2b5(R132H) mutation causes a slight reduction in eIF2B enzymatic activity which leads to abnormal composition of mitochondrial electron transfer chain complexes and impaired oxidative phosphorylation. Previous work using primary fibroblasts isolated from Eif2b5(R132H/R132H) mice revealed that owing to increased mitochondrial biogenesis they exhibit normal cellular ATP level. In contrast to fibroblasts, here we show that primary astrocytes isolated from Eif2b5(R132H/R132H) mice are unable to compensate for their metabolic impairment and exhibit chronic state of low ATP level regardless of extensive adaptation efforts. Mutant astrocytes are hypersensitive to oxidative stress and to further energy stress. Moreover, they show migration deficit upon exposure to glucose starvation. The mutation in Eif2b5 prompts reactive oxygen species (ROS)-mediated inferior ability to stimulate the AMP-activated protein kinase (AMPK) axis, due to a requirement to increase the mammalian target of rapamycin complex-1 (mTORC1) signalling in order to enable oxidative glycolysis and generation of specific subclass of ROS-regulating proteins, similar to cancer cells. The data disclose the robust impact of eIF2B on metabolic and redox homeostasis programs in astrocytes and point at their hyper-sensitivity to mutated eIF2B. Thereby, it illuminates the central involvement of astrocytes in Vanishing White Matter Disease (VWMD), a genetic neurodegenerative leukodystrophy caused by homozygous hypomorphic mutations in genes encoding any of the 5 subunits of eIF2B.
Collapse
Affiliation(s)
- Melisa Herrero
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (M.H.); (M.D.); (A.A.)
| | - Maron Daw
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (M.H.); (M.D.); (A.A.)
| | - Andrea Atzmon
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (M.H.); (M.D.); (A.A.)
| | - Orna Elroy-Stein
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (M.H.); (M.D.); (A.A.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
50
|
Stellingwerff MD, Al-Saady ML, van de Brug T, Barkhof F, Pouwels PJW, van der Knaap MS. MRI Natural History of the Leukodystrophy Vanishing White Matter. Radiology 2021; 300:671-680. [PMID: 34184934 DOI: 10.1148/radiol.2021210110] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background In vanishing white matter (VWM), a form of leukodystrophy, earlier onset is associated with faster clinical progression. MRI typically shows rarefaction and cystic destruction of the cerebral white matter. Information on the evolution of VWM according to age at onset is lacking. Purpose To determine whether nature and progression of cerebral white matter abnormalities in VWM differ according to age at onset. Materials and Methods Patients with genetically confirmed VWM were stratified into six groups according to age at onset: younger than 1 year, 1 year to younger than 2 years, 2 years to younger than 4 years, 4 years to younger than 8 years, 8 years to younger than 18 years, and 18 years or older. With institutional review board approval, all available MRI scans obtained between 1985 and 2019 were retrospectively analyzed with three methods: (a) ratio of the width of the lateral ventricles over the skull (ventricle-to-skull ratio [VSR]) was measured to estimate brain atrophy; (b) cerebral white matter was visually scored as percentage normal, hyperintense, rarefied, or cystic on fluid-attenuated inversion recovery (FLAIR) images and converted into a white matter decay score; and (c) the intracranial volume was segmented into normal-appearing white and gray matter, abnormal but structurally present (FLAIR-hyperintense) and rarefied or cystic (FLAIR-hypointense) white matter, and ventricular and extracerebral cerebrospinal fluid (CSF). Multilevel regression analyses with patient as a clustering variable were performed to account for the nested data structure. Results A total of 461 examinations in 270 patients (median age, 7 years [interquartile range, 3-18 years]; 144 female patients) were evaluated; 112 patients had undergone serial imaging. Patients with later onset had higher VSR [F(5) = 8.42; P < .001] and CSF volume [F(5) = 21.7; P < .001] and lower white matter decay score [F(5) = 4.68; P < .001] and rarefied or cystic white matter volume [F(5) = 13.3; P < .001]. Rate of progression of white matter decay scores [b = -1.6, t(109) = -3.9; P < .001] and VSRs [b = -0.05, t (109) = -3.7; P < .001] were lower with later onset. Conclusion A radiologic spectrum based on age at onset exists in vanishing white matter. The earlier the onset, the faster and more cystic the white matter decay, whereas with later onset, white matter atrophy and gliosis predominate. © RSNA, 2021.
Collapse
Affiliation(s)
- Menno D Stellingwerff
- From the Department of Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam 1081 HV, the Netherlands (M.D.S., M.L.A., M.S.v.d.K.); Department of Epidemiology and Data Science, Amsterdam University Medical Centers, Amsterdam, the Netherlands (T.v.d.B.); Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, the Netherlands (F.B., P.J.W.P.); Institutes of Neurology and Health Care Engineering, University College London, London, England (F.B.); and Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands (M.S.v.d.K.)
| | - Murtadha L Al-Saady
- From the Department of Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam 1081 HV, the Netherlands (M.D.S., M.L.A., M.S.v.d.K.); Department of Epidemiology and Data Science, Amsterdam University Medical Centers, Amsterdam, the Netherlands (T.v.d.B.); Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, the Netherlands (F.B., P.J.W.P.); Institutes of Neurology and Health Care Engineering, University College London, London, England (F.B.); and Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands (M.S.v.d.K.)
| | - Tim van de Brug
- From the Department of Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam 1081 HV, the Netherlands (M.D.S., M.L.A., M.S.v.d.K.); Department of Epidemiology and Data Science, Amsterdam University Medical Centers, Amsterdam, the Netherlands (T.v.d.B.); Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, the Netherlands (F.B., P.J.W.P.); Institutes of Neurology and Health Care Engineering, University College London, London, England (F.B.); and Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands (M.S.v.d.K.)
| | - Frederik Barkhof
- From the Department of Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam 1081 HV, the Netherlands (M.D.S., M.L.A., M.S.v.d.K.); Department of Epidemiology and Data Science, Amsterdam University Medical Centers, Amsterdam, the Netherlands (T.v.d.B.); Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, the Netherlands (F.B., P.J.W.P.); Institutes of Neurology and Health Care Engineering, University College London, London, England (F.B.); and Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands (M.S.v.d.K.)
| | - Petra J W Pouwels
- From the Department of Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam 1081 HV, the Netherlands (M.D.S., M.L.A., M.S.v.d.K.); Department of Epidemiology and Data Science, Amsterdam University Medical Centers, Amsterdam, the Netherlands (T.v.d.B.); Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, the Netherlands (F.B., P.J.W.P.); Institutes of Neurology and Health Care Engineering, University College London, London, England (F.B.); and Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands (M.S.v.d.K.)
| | - Marjo S van der Knaap
- From the Department of Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit and Amsterdam Neuroscience, De Boelelaan 1117, Amsterdam 1081 HV, the Netherlands (M.D.S., M.L.A., M.S.v.d.K.); Department of Epidemiology and Data Science, Amsterdam University Medical Centers, Amsterdam, the Netherlands (T.v.d.B.); Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit and Amsterdam Neuroscience, Amsterdam, the Netherlands (F.B., P.J.W.P.); Institutes of Neurology and Health Care Engineering, University College London, London, England (F.B.); and Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, the Netherlands (M.S.v.d.K.)
| |
Collapse
|