1
|
Chern T, Tong X, Bauer WG, Quispe-Parra DJ, Gao X, Gerner-Mauro KN, Poché RA. New genetic tools to define the pathophysiology of inborn errors of cobalamin metabolism impacting mammalian development. Differentiation 2025; 143:100868. [PMID: 40411980 DOI: 10.1016/j.diff.2025.100868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 04/30/2025] [Accepted: 05/15/2025] [Indexed: 05/27/2025]
Abstract
The congenital, autosomal recessive disorder combined methylmalonic acidemia and homocystinuria - cblC type, is the most common inborn error of cobalamin (vitamin B12) metabolism. In its early onset form, cblC profoundly impacts fetal development of the central nervous system, hematopoietic system, and other tissues. Previously, mutations in the MMACHC gene, which encodes a protein required for the intracellular trafficking and enzymatic processing of free cobalamin into active coenzyme forms, were found to cause cblC. These coenzymes are required in two metabolic pathways which produce either succinyl-CoA in the mitochondria or methionine in the cytosol. However, due to a lack of sufficient animal models, the exact pathophysiology of cblC remains unknown. Moreover, there is evidence to suggest that MMACHC may have roles outside of cobalamin metabolism and that cobalamin itself may be required for additional, unknown metabolic pathways. Here, we report the generation and characterization of three new mouse lines aimed at further defining the role of MMACHC and cobalamin in mammalian development. CRISPR/Cas9 genome editing was used to develop an HA-tagged version of Mmachc, which will aid in affinity purification and spatiotemporal localization of the MMACHC protein. To clarify which metabolic perturbations downstream of Mmachc loss give rise to tissue-specific developmental defects, we also created floxed alleles for both methionine synthase (Mtr) and methylmalonyl-CoA mutase (Mmut), which are the only known cobalamin dependent enzymes in mammals. In total, these new mouse models significantly expand upon the repertoire of genetic reagents to clarify the pathophysiology of cblC as well as define both the canonical and hypothesized noncanonical roles of MMACHC in mammalian development.
Collapse
Affiliation(s)
- Tiffany Chern
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xuefei Tong
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - William G Bauer
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA; Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - David J Quispe-Parra
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA; Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xia Gao
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, 1100 Bates Ave., Houston, TX, 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Kamryn N Gerner-Mauro
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ross A Poché
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA; Development, Disease Models and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA; Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA; Cancer and Cell Biology Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
2
|
Golabi M, Kazemi D, Chadeganipour AS, Fouladseresht H, Sullman MJM, Ghezelbash B, Dastgerdi AY, Eskandari N. The Role of Cobalamin in Multiple Sclerosis: An Update. Inflammation 2025; 48:485-500. [PMID: 38902541 DOI: 10.1007/s10753-024-02075-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 06/22/2024]
Abstract
Multiple sclerosis (MS) is a neurodegenerative condition that results in axonal and permanent damage to the central nervous system, necessitating healing owing to autoimmune reactions and persistent neuroinflammation. Antioxidant and anti-inflammatory drugs are essential for the management of oxidative stress and neuroinflammation. Additionally, multivitamin supplementation, particularly vitamin B12 (cobalamin), may be beneficial for neuronal protection. Although there is no documented connection between vitamin B12 deficiency and MS, researchers have explored its potential as a metabolic cause. This review highlights the therapeutic benefits of cobalamin (Cbl) in patients with MS.
Collapse
Affiliation(s)
- Marjan Golabi
- Applied Physiology Research Center, Cardiovascular Research Institute, Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Danial Kazemi
- Student Research Committee, Isfahan University of Medical Science, Isfahan, Iran
| | | | - Hamed Fouladseresht
- Applied Physiology Research Center, Cardiovascular Research Institute, Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mark J M Sullman
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
- Department of Social Sciences, University of Nicosia, Nicosia, Cyprus
| | - Behrooz Ghezelbash
- Laboratory Hematology and Blood Banking, School of Allied Medical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ava Yeganegi Dastgerdi
- Department of Cell and Molecular Biology, Falavarjan Branch, Islamic Azad University of Science, Isfahan, Iran
| | - Nahid Eskandari
- Applied Physiology Research Center, Cardiovascular Research Institute, Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
3
|
Yousfi N, Mousseaux C, Hamza A, Laville P, Mille M, Philippe N, Dancer M, Bouder C, Luque Y, Rafat C, Mesnard L. Genomic analysis of adult thrombotic microangiopathies in less than 3 days: from rapid to fast genomics to treatment. Blood 2024; 144:2266-2269. [PMID: 39197070 DOI: 10.1182/blood.2024024976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/07/2024] [Accepted: 08/07/2024] [Indexed: 08/30/2024] Open
Abstract
ABSTRACT Using nanopore sequencing, we showed the feasibility and impact of rapid genomic screening for managing thrombotic microangiopathies in 18 prospective cases, achieving diagnoses in <3 days. We compared the results with standard exome sequencing, cost efficiency, and complement blockade initiation.
Collapse
Affiliation(s)
- Nadhir Yousfi
- Unité Mixte de Recherche S1155, INSERM, Sorbonne Université, Paris, France
| | - Cyril Mousseaux
- Unité Mixte de Recherche S1155, INSERM, Sorbonne Université, Paris, France
- Service de Soins Intensifs Néphrologiques et Rein Aigu, Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, Paris, France
- Faculté de Médecine, Sorbonne Université, Paris, France
- French Intensive Care Renal Network, Lyon, France
| | - Abderaouf Hamza
- Department de Génétique, Institut Curie, Paris Science et Lettres Université, Paris, France
| | - Pierre Laville
- Unité Mixte de Recherche S1155, INSERM, Sorbonne Université, Paris, France
- Eurofins Biomnis Laboratory, Lyon, France
| | | | | | - Marine Dancer
- Department de Génétique, Institut Curie, Paris Science et Lettres Université, Paris, France
| | - Christophe Bouder
- Institut des Systèmes Intélligents et de la Robotique, Centre National de la Recheque Scientifique, Sorbonne Université, Paris, France
| | - Yosu Luque
- Unité Mixte de Recherche S1155, INSERM, Sorbonne Université, Paris, France
- Service de Soins Intensifs Néphrologiques et Rein Aigu, Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, Paris, France
- Faculté de Médecine, Sorbonne Université, Paris, France
- French Intensive Care Renal Network, Lyon, France
| | - Cédric Rafat
- Unité Mixte de Recherche S1155, INSERM, Sorbonne Université, Paris, France
- Service de Soins Intensifs Néphrologiques et Rein Aigu, Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, Paris, France
- French Intensive Care Renal Network, Lyon, France
| | - Laurent Mesnard
- Unité Mixte de Recherche S1155, INSERM, Sorbonne Université, Paris, France
- Service de Soins Intensifs Néphrologiques et Rein Aigu, Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, Paris, France
- Faculté de Médecine, Sorbonne Université, Paris, France
- French Intensive Care Renal Network, Lyon, France
- Institut des Sciences du Calcul et des Données, Sorbonne Université, Paris, France
- Centre National Référence des Micro-Angiopathies Thrombotiques, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, Paris, France
| |
Collapse
|
4
|
Mucha P, Kus F, Cysewski D, Smolenski RT, Tomczyk M. Vitamin B 12 Metabolism: A Network of Multi-Protein Mediated Processes. Int J Mol Sci 2024; 25:8021. [PMID: 39125597 PMCID: PMC11311337 DOI: 10.3390/ijms25158021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/12/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
The water-soluble vitamin, vitamin B12, also known as cobalamin, plays a crucial role in cellular metabolism, particularly in DNA synthesis, methylation, and mitochondrial functionality. Its deficiency can lead to hematological and neurological disorders; however, the manifestation of these clinical outcomes is relatively late. It leads to difficulties in the early diagnosis of vitamin B12 deficiency. A prolonged lack of vitamin B12 may have severe consequences including increased morbidity to neurological and cardiovascular diseases. Beyond inadequate dietary intake, vitamin B12 deficiency might be caused by insufficient bioavailability, blood transport disruptions, or impaired cellular uptake and metabolism. Despite nearly 70 years of knowledge since the isolation and characterization of this vitamin, there are still gaps in understanding its metabolic pathways. Thus, this review aims to compile current knowledge about the crucial proteins necessary to efficiently accumulate and process vitamin B12 in humans, presenting these systems as a multi-protein network. The epidemiological consequences, diagnosis, and treatment of vitamin B12 deficiency are also highlighted. We also discuss clinical warnings of vitamin B12 deficiency based on the ongoing test of specific moonlighting proteins engaged in vitamin B12 metabolic pathways.
Collapse
Affiliation(s)
- Patryk Mucha
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland; (P.M.); (F.K.); (R.T.S.)
| | - Filip Kus
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland; (P.M.); (F.K.); (R.T.S.)
- Laboratory of Protein Biochemistry, Intercollegiate Faculty of Biotechnology of University of Gdansk and Medical University of Gdansk, 80-307 Gdansk, Poland
| | - Dominik Cysewski
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland;
| | - Ryszard T. Smolenski
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland; (P.M.); (F.K.); (R.T.S.)
| | - Marta Tomczyk
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland; (P.M.); (F.K.); (R.T.S.)
| |
Collapse
|
5
|
Robea MA, Ilie OD, Nicoara MN, Solcan G, Romila LE, Ureche D, Ciobica A. Vitamin B 12 Ameliorates Pesticide-Induced Sociability Impairment in Zebrafish ( Danio rerio): A Prospective Controlled Intervention Study. Animals (Basel) 2024; 14:405. [PMID: 38338046 PMCID: PMC10854844 DOI: 10.3390/ani14030405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 01/18/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Constant exposure to a variety of environmental factors has become increasingly problematic. A variety of illnesses are initiated or aided by the presence of certain perturbing factors. In the case of autism spectrum disorder, the environmental component plays an important part in determining the overall picture. Moreover, the lack of therapies to relieve existing symptoms complicates the fight against this condition. As a result, animal models have been used to make biomedical research easier and more suited for disease investigations. The current study used zebrafish as an animal model to mimic a real-life scenario: acute exposure to an increased dose of pesticides, followed by prospective intervention-based therapy with vitamin B12 (vit. B12). It is known that vit. B12 is involved in brain function nerve tissue, and red blood cell formation. Aside from this, the role of vit. B12 in the redox processes is recognized for its help against free radicals. To investigate the effect of vit. B12, fish were divided into four different groups and exposed to a pesticide mixture (600 μg L-1 fipronil + 600 μg L-1 pyriproxyfen) and 0.24 μg L-1 vit. B12 for 14 days. The impact of the compounds was assessed daily with EthoVision XT 11.5 software for behavioral observations, especially for sociability, quantified by the social interaction test. In addition, at the end of the study, the activities of superoxide dismutase (SOD), glutathione peroxidase (GPx), and malondialdehyde (MDA) were measured. The results showed significant improvements in locomotor activity parameters and a positive influence of the vitamin on sociability. Regarding the state of oxidative stress, high activity was found for SOD and GPx in the case of vit. B12, while fish exposed to the mixture of pesticides and vit. B12 had a lower level of MDA. In conclusion, the study provides new data about the effect of vit. B12 in zebrafish, highlighting the potential use of vitamin supplementation to maintain and support the function of the organism.
Collapse
Affiliation(s)
- Madalina Andreea Robea
- Doctoral School of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, Bd. Carol I, 20A, 700505 Iasi, Romania;
| | - Ovidiu Dumitru Ilie
- Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, University Street No. 16, 700115 Iasi, Romania;
| | - Mircea Nicusor Nicoara
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, Bd. Carol I, 20A, 700505 Iasi, Romania;
- Doctoral School of Geosciences, Faculty of Geography and Geology, “Alexandru Ioan Cuza” University of Iasi, 700505 Iasi, Romania
| | - Gheorghe Solcan
- Internal Medicine Clinic, Faculty of Veterinary Medicine, Ion Ionescu de la Brad Iasi University of Life Sciences, 700489 Iasi, Romania;
| | | | - Dorel Ureche
- Department of Biology, Ecology and Environmental Protection, Faculty of Sciences, University “Vasile Alecsandri“ of Bacau, Calea Marasesti Street, No. 157, 600115 Bacau, Romania
| | - Alin Ciobica
- Department of Biology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iasi, Bd. Carol I, 20A, 700505 Iasi, Romania;
- Academy of Romanian Scientists, 54, Independence Street, Sector 5, 050094 Bucharest, Romania
- Center of Biomedical Research, Romanian Academy, Iasi Branch, Teodor Codrescu 2, 700481 Iasi, Romania
| |
Collapse
|
6
|
Mathew AR, Di Matteo G, La Rosa P, Barbati SA, Mannina L, Moreno S, Tata AM, Cavallucci V, Fidaleo M. Vitamin B12 Deficiency and the Nervous System: Beyond Metabolic Decompensation-Comparing Biological Models and Gaining New Insights into Molecular and Cellular Mechanisms. Int J Mol Sci 2024; 25:590. [PMID: 38203763 PMCID: PMC10778862 DOI: 10.3390/ijms25010590] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/16/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Vitamin B12 (VitB12) is a micronutrient and acts as a cofactor for fundamental biochemical reactions: the synthesis of succinyl-CoA from methylmalonyl-CoA and biotin, and the synthesis of methionine from folic acid and homocysteine. VitB12 deficiency can determine a wide range of diseases, including nervous system impairments. Although clinical evidence shows a direct role of VitB12 in neuronal homeostasis, the molecular mechanisms are yet to be characterized in depth. Earlier investigations focused on exploring the biochemical shifts resulting from a deficiency in the function of VitB12 as a coenzyme, while more recent studies propose a broader mechanism, encompassing changes at the molecular/cellular levels. Here, we explore existing study models employed to investigate the role of VitB12 in the nervous system, including the challenges inherent in replicating deficiency/supplementation in experimental settings. Moreover, we discuss the potential biochemical alterations and ensuing mechanisms that might be modified at the molecular/cellular level (such as epigenetic modifications or changes in lysosomal activity). We also address the role of VitB12 deficiency in initiating processes that contribute to nervous system deterioration, including ROS accumulation, inflammation, and demyelination. Consequently, a complex biological landscape emerges, requiring further investigative efforts to grasp the intricacies involved and identify potential therapeutic targets.
Collapse
Affiliation(s)
- Aimee Rachel Mathew
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (A.R.M.); (A.M.T.)
| | - Giacomo Di Matteo
- Department of Chemistry and Technology of Drugs, Sapienza University of Rome, 00185 Rome, Italy; (G.D.M.); (L.M.)
| | - Piergiorgio La Rosa
- Division of Neuroscience, Department of Psychology, Sapienza University of Rome, 00185 Rome, Italy;
- European Center for Brain Research, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| | - Saviana Antonella Barbati
- Departmental Faculty of Medicine and Surgery, UniCamillus-Saint Camillus International University of Health Sciences, 00131 Rome, Italy;
| | - Luisa Mannina
- Department of Chemistry and Technology of Drugs, Sapienza University of Rome, 00185 Rome, Italy; (G.D.M.); (L.M.)
| | - Sandra Moreno
- Department of Science, University Roma Tre, 00146 Rome, Italy;
- Laboratory of Neurodevelopment, Neurogenetics and Neuromolecular Biology, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy
| | - Ada Maria Tata
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (A.R.M.); (A.M.T.)
- Research Centre of Neurobiology “Daniel Bovet”, Sapienza University of Rome, 00185 Rome, Italy
| | - Virve Cavallucci
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy;
- Fondazione Policlinico Universitario “A. Gemelli” IRCCS, 00168 Rome, Italy
| | - Marco Fidaleo
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy; (A.R.M.); (A.M.T.)
- Research Center for Nanotechnology Applied to Engineering (CNIS), Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
7
|
Antony P, Baby B, Ali A, Vijayan R, Al Jasmi F. Interaction of Glutathione with MMACHC Arginine-Rich Pocket Variants Associated with Cobalamin C Disease: Insights from Molecular Modeling. Biomedicines 2023; 11:3217. [PMID: 38137438 PMCID: PMC10740964 DOI: 10.3390/biomedicines11123217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 11/20/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Methylmalonic aciduria and homocystinuria type C protein (MMACHC) is required by the body to metabolize cobalamin (Cbl). Due to its complex structure and cofactor forms, Cbl passes through an extensive series of absorptive and processing steps before being delivered to mitochondrial methyl malonyl-CoA mutase and cytosolic methionine synthase. Depending on the cofactor attached, MMACHC performs either flavin-dependent reductive decyanation or glutathione (GSH)-dependent dealkylation. The alkyl groups of Cbl have to be removed in the presence of GSH to produce intermediates that can later be converted into active cofactor forms. Pathogenic mutations in the GSH binding site, such as R161Q, R161G, R206P, R206W, and R206Q, have been reported to cause Cbl diseases. The impact of these variations on MMACHC's structure and how it affects GSH and Cbl binding at the molecular level is poorly understood. To better understand the molecular basis of this interaction, mutant structures involving the MMACHC-MeCbl-GSH complex were generated using in silico site-directed point mutations and explored using molecular dynamics (MD) simulations. The results revealed that mutations in the key arginine residues disrupt GSH binding by breaking the interactions and reducing the free energy of binding of GSH. Specifically, variations at position 206 appeared to produce weaker GSH binding. The lowered binding affinity for GSH in the variant structures could impact metabolic pathways involving Cbl and its trafficking.
Collapse
Affiliation(s)
- Priya Antony
- Department of Biology, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Bincy Baby
- Department of Biology, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Amanat Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Ranjit Vijayan
- Department of Biology, College of Science, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- The Big Data Analytics Center, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| | - Fatma Al Jasmi
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
- Department of Pediatrics, Tawam Hospital, Al Ain P.O. Box 15258, United Arab Emirates
| |
Collapse
|
8
|
Mascarenhas R, Guha A, Li Z, Ruetz M, An S, Seravalli J, Banerjee R. Cobalt-Sulfur Coordination Chemistry Drives B 12 Loading onto Methionine Synthase. J Am Chem Soc 2023:10.1021/jacs.3c07941. [PMID: 37916782 PMCID: PMC11063128 DOI: 10.1021/jacs.3c07941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Cobalt-sulfur (Co-S) coordination is labile to both oxidation and reduction chemistry and is rarely seen in nature. Cobalamin (or vitamin B12) is an essential cobalt-containing organometallic cofactor in mammals and is escorted via an intricate network of chaperones to a single cytoplasmic target, methionine synthase. In this study, we report that the human cobalamin trafficking protein, MMADHC, exploits the chemical lability of Co-S coordination for cofactor off-loading onto methionine synthase. Cys-261 on MMADHC serves as the β-axial ligand to cobalamin. Complex formation between MMADHC and methionine synthase is signaled by loss of the lower axial nitrogen ligand, leading to five-coordinate thiolato-cobalamin. Nucleophilic displacement by the vicinal thiolate, Cys-262, completes cofactor transfer to methionine synthase and release of a cysteine disulfide-containing MMADHC. The physiological relevance of this mechanism is supported by clinical variants of MMADHC, which impair cofactor binding and off-loading, explaining the molecular basis of the associated homocystinuria.
Collapse
Affiliation(s)
- Romila Mascarenhas
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Arkajit Guha
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Zhu Li
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Markus Ruetz
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Sojin An
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Javier Seravalli
- Department of Biological Chemistry, University of Nebraska-Lincoln, Lincoln, Nebraska 68588, United States
| | - Ruma Banerjee
- Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
9
|
Mascarenhas R, Guha A, Li Z, Ruetz M, An S, Seravalli J, Banerjee R. Cobalt-sulfur coordination chemistry drives B 12 loading onto methionine synthase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.25.550549. [PMID: 37546824 PMCID: PMC10402061 DOI: 10.1101/2023.07.25.550549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Cobalt-sulfur (Co-S) coordination is labile to both oxidation and reduction chemistry and is rarely seen in Nature. Cobalamin (or vitamin B 12 ) is an essential cobalt-containing organometallic cofactor in mammals, and is escorted via an intricate network of chaperones to a single cytoplasmic target, methionine synthase. In this study, we report that the human cobalamin trafficking protein, MMADHC, exploits the chemical lability of Co-S coordination, for cofactor off-loading onto methionine synthase. Cys-261 on MMADHC serves as the β-axial ligand to cobalamin. Complex formation between MMADHC and methionine synthase is signaled by loss of the lower axial nitrogen ligand, leading to five-coordinate thiolato-cobalamin. Nucleophilic displacement by the vicinal thiolate, Cys-262, completes cofactor transfer to methionine synthase and release of a cysteine disulfide-containing MMADHC. The physiological relevance of this mechanism is supported by clinical variants of MMADHC, which impair cofactor binding and off-loading, explaining the molecular basis of the associated homocystinuria.
Collapse
|
10
|
McCorvie TJ, Ferreira D, Yue WW, Froese DS. The complex machinery of human cobalamin metabolism. J Inherit Metab Dis 2023; 46:406-420. [PMID: 36680553 DOI: 10.1002/jimd.12593] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
Vitamin B12 (cobalamin, Cbl) is required as a cofactor by two human enzymes, 5-methyltetrahydrofolate-homocysteine methyltransferase (MTR) and methylmalonyl-CoA mutase (MMUT). Within the body, a vast array of transporters, enzymes and chaperones are required for the generation and delivery of these cofactor forms. How they perform these functions is dictated by the structure and interactions of the proteins involved, the molecular bases of which are only now being elucidated. In this review, we highlight recent insights into human Cbl metabolism and address open questions in the field by employing a protein structure and interactome based perspective. We discuss how three very similar proteins-haptocorrin, intrinsic factor and transcobalamin-exploit slight structural differences and unique ligand receptor interactions to effect selective Cbl absorption and internalisation. We describe recent advances in the understanding of how endocytosed Cbl is transported across the lysosomal membrane and the implications of the recently solved ABCD4 structure. We detail how MMACHC and MMADHC cooperate to modify and target cytosolic Cbl to the client enzymes MTR and MMUT using ingenious modifications to an ancient nitroreductase fold, and how MTR and MMUT link with their accessory enzymes to sustainably harness the supernucleophilic potential of Cbl. Finally, we provide an outlook on how future studies may combine structural and interactome based approaches and incorporate knowledge of post-translational modifications to bring further insights.
Collapse
Affiliation(s)
- Thomas J McCorvie
- Biosciences Institute, The Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - Douglas Ferreira
- Biosciences Institute, The Medical School, Newcastle University, Newcastle upon Tyne, UK
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Wyatt W Yue
- Biosciences Institute, The Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - D Sean Froese
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, University of Zürich, Zürich, Switzerland
| |
Collapse
|
11
|
Comparative Transcriptomics and Methylomics Reveal Adaptive Responses of Digestive and Metabolic Genes to Dietary Shift in Giant and Red Pandas. Genes (Basel) 2022; 13:genes13081446. [PMID: 36011357 PMCID: PMC9407821 DOI: 10.3390/genes13081446] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/01/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
Both the giant panda (Ailuropoda melanoleuca) and red panda (Ailurus fulgens) belong to the order Carnivora, but have changed their dietary habits to eating bamboo exclusively. The convergent evolution characteristics of their morphology, genome and gut flora have been found in the two pandas. However, the research on the convergent adaptation of their digestion and metabolism to the bamboo diet, mediated by the dietary shift of the two pandas at the gene-expression and epigenetic regulation levels, is still lacking. We therefore used RNA sequencing among five species (two pandas and three non-herbivore mammals) and bisulfite sequencing among three species (two pandas and a carnivore ferret) to sequence key digestion and metabolism tissues (stomach and small intestine). Our results provide evidence that the convergent differentially expressed genes (related to carbohydrate utilization, bile secretion, Lys and Arg metabolism, vitamin B12 utilization and cyanide detoxification) of the two pandas are adaptive responses to the bamboo diet containing low lipids, low Lys and Arg, low vitamin B12 and high cyanide. We also profiled the genome-wide methylome maps of giant panda, red panda and ferret, and the results indicated that the promoter methylation of the two pandas may regulate digestive and metabolic genes to adapt to sudden environmental changes, and then, transmit genetic information to future generations to evolve into bamboo eaters. Taken together, our study provides new insights into the molecular mechanisms of the dietary shift and the adaptation to a strict bamboo diet in both pandas using comparative transcriptomics and methylomics.
Collapse
|
12
|
Esser AJ, Mukherjee S, Dereven‘kov IA, Makarov SV, Jacobsen DW, Spiekerkoetter U, Hannibal L. Versatile Enzymology and Heterogeneous Phenotypes in Cobalamin Complementation Type C Disease. iScience 2022; 25:104981. [PMID: 36105582 PMCID: PMC9464900 DOI: 10.1016/j.isci.2022.104981] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Nutritional deficiency and genetic errors that impair the transport, absorption, and utilization of vitamin B12 (B12) lead to hematological and neurological manifestations. The cblC disease (cobalamin complementation type C) is an autosomal recessive disorder caused by mutations and epi-mutations in the MMACHC gene and the most common inborn error of B12 metabolism. Pathogenic mutations in MMACHC disrupt enzymatic processing of B12, an indispensable step before micronutrient utilization by the two B12-dependent enzymes methionine synthase (MS) and methylmalonyl-CoA mutase (MUT). As a result, patients with cblC disease exhibit plasma elevation of homocysteine (Hcy, substrate of MS) and methylmalonic acid (MMA, degradation product of methylmalonyl-CoA, substrate of MUT). The cblC disorder manifests early in childhood or in late adulthood with heterogeneous multi-organ involvement. This review covers current knowledge on the cblC disease, structure–function relationships of the MMACHC protein, the genotypic and phenotypic spectra in humans, experimental disease models, and promising therapies.
Collapse
|
13
|
Wiedemann A, Oussalah A, Lamireau N, Théron M, Julien M, Mergnac JP, Augay B, Deniaud P, Alix T, Frayssinoux M, Feillet F, Guéant JL. Clinical, phenotypic and genetic landscape of case reports with genetically proven inherited disorders of vitamin B 12 metabolism: A meta-analysis. Cell Rep Med 2022; 3:100670. [PMID: 35764087 PMCID: PMC9381384 DOI: 10.1016/j.xcrm.2022.100670] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 11/22/2021] [Accepted: 06/02/2022] [Indexed: 10/31/2022]
Abstract
Inherited disorders of B12 metabolism produce a broad spectrum of manifestations, with limited knowledge of the influence of age and the function of related genes. We report a meta-analysis on 824 patients with a genetically proven diagnosis of an inherited disorder of vitamin B12 metabolism. Gene clusters and age categories are associated with patients' manifestations. The "cytoplasmic transport" cluster is associated with neurological and ophthalmological manifestations, the "mitochondrion" cluster with hypotonia, acute metabolic decompensation, and death, and the "B12 availability" and "remethylation" clusters with anemia and cytopenia. Hypotonia, EEG abnormalities, nystagmus, and strabismus are predominant in the younger patients, while neurological manifestations, such as walking difficulties, peripheral neuropathy, pyramidal syndrome, cerebral atrophy, psychiatric disorders, and thromboembolic manifestations, are predominant in the older patients. These results should prompt systematic checking of markers of vitamin B12 status, including homocysteine and methylmalonic acid, when usual causes of these manifestations are discarded in adult patients.
Collapse
Affiliation(s)
- Arnaud Wiedemann
- Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Faculty of Medicine of Nancy, University of Lorraine, INSERM UMR_S 1256, 54000 Nancy, France; Department of Pediatrics, University Hospital of Nancy, 54000 Nancy, France; Reference Center for Inborn Errors of Metabolism (ORPHA67872), University Hospital of Nancy, 54000 Nancy, France
| | - Abderrahim Oussalah
- Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Faculty of Medicine of Nancy, University of Lorraine, INSERM UMR_S 1256, 54000 Nancy, France; Reference Center for Inborn Errors of Metabolism (ORPHA67872), University Hospital of Nancy, 54000 Nancy, France; Department of Molecular Medicine, Division of Biochemistry, Molecular Biology, Nutrition, and Metabolism, University Hospital of Nancy, 54000 Nancy, France
| | - Nathalie Lamireau
- Department of Pediatrics, University Hospital of Nancy, 54000 Nancy, France
| | - Maurane Théron
- Department of Pediatrics, University Hospital of Nancy, 54000 Nancy, France
| | - Melissa Julien
- Department of Molecular Medicine, Division of Biochemistry, Molecular Biology, Nutrition, and Metabolism, University Hospital of Nancy, 54000 Nancy, France
| | | | - Baptiste Augay
- Department of Pediatrics, University Hospital of Nancy, 54000 Nancy, France
| | - Pauline Deniaud
- Department of Pediatrics, University Hospital of Nancy, 54000 Nancy, France
| | - Tom Alix
- Department of Molecular Medicine, Division of Biochemistry, Molecular Biology, Nutrition, and Metabolism, University Hospital of Nancy, 54000 Nancy, France
| | - Marine Frayssinoux
- Department of Molecular Medicine, Division of Biochemistry, Molecular Biology, Nutrition, and Metabolism, University Hospital of Nancy, 54000 Nancy, France
| | - François Feillet
- Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Faculty of Medicine of Nancy, University of Lorraine, INSERM UMR_S 1256, 54000 Nancy, France; Department of Pediatrics, University Hospital of Nancy, 54000 Nancy, France; Reference Center for Inborn Errors of Metabolism (ORPHA67872), University Hospital of Nancy, 54000 Nancy, France
| | - Jean-Louis Guéant
- Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Faculty of Medicine of Nancy, University of Lorraine, INSERM UMR_S 1256, 54000 Nancy, France; Reference Center for Inborn Errors of Metabolism (ORPHA67872), University Hospital of Nancy, 54000 Nancy, France; Department of Molecular Medicine, Division of Biochemistry, Molecular Biology, Nutrition, and Metabolism, University Hospital of Nancy, 54000 Nancy, France.
| |
Collapse
|
14
|
Passantino R, Mangione MR, Ortore MG, Costa MA, Provenzano A, Amenitsch H, Sabbatella R, Alfano C, Martorana V, Vilasi S. Investigation on a MMACHC mutant from cblC disease: The c.394C>T variant. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2022; 1870:140793. [PMID: 35618206 DOI: 10.1016/j.bbapap.2022.140793] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 06/15/2023]
Abstract
The cblC disease is an inborn disorder of the vitamin B12 (cobalamin, Cbl) metabolism characterized by methylmalonic aciduria and homocystinuria. The clinical consequences of this disease are devastating and, even when early treated with current therapies, the affected children manifest symptoms involving vision, growth, and learning. The illness is caused by mutations in the gene codifying for MMACHC, a 282aa protein that transports and transforms the different Cbl forms. Here we present data on the structural properties of the truncated protein p.R132X resulting from the c.394C > T mutation that, along with c.271dupA and c.331C > T, is among the most common mutations in cblC. Although missing part of the Cbl binding domain, p.R132X is associated to late-onset symptoms and, therefore, it is supposed to retain residual function. However, to our knowledge structural-functional studies on c.394C > T mutant aimed at verifying this hypothesis are still lacking. By using a biophysical approach including Circular Dichroism, fluorescence, Small Angle X-ray Scattering, and Molecular Dynamics, we show that the mutant protein MMACHC-R132X retains secondary structure elements and remains compact in solution, partly preserving its binding affinity for Cbl. Insights on the fragile stability of MMACHC-R132X-Cbl are provided.
Collapse
Affiliation(s)
- Rosa Passantino
- Biophysics Institute, National Research Council, Palermo 90143, Italy
| | | | - Maria Grazia Ortore
- Dept. Life and Environmental Sciences, Marche Polytechnic University, Ancona 60131, Italy
| | | | | | | | | | | | | | - Silvia Vilasi
- Biophysics Institute, National Research Council, Palermo 90143, Italy.
| |
Collapse
|
15
|
Kiessling E, Peters F, Ebner LJ, Merolla L, Samardzija M, Baumgartner MR, Grimm C, Froese DS. HIF1 and DROSHA are involved in MMACHC repression in hypoxia. Biochim Biophys Acta Gen Subj 2022; 1866:130175. [DOI: 10.1016/j.bbagen.2022.130175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 05/03/2022] [Accepted: 05/23/2022] [Indexed: 11/25/2022]
|
16
|
Watkins D, Rosenblatt DS. Inherited defects of cobalamin metabolism. VITAMINS AND HORMONES 2022; 119:355-376. [PMID: 35337626 DOI: 10.1016/bs.vh.2022.01.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Cobalamin (vitamin B12) is required for activity of the enzymes methylmalonyl-CoA mutase and methionine synthase in human cells. Inborn errors affecting cobalamin uptake or metabolism are characterized by accumulation of the substrates for these enzymes, methylmalonic acid and homocysteine, in blood and urine. Inborn errors affecting synthesis of the adenosylcobalamin coenzyme required by methylmalonyl-CoA mutase (cblA and cblB) result in isolated methylmalonic aciduria; inborn errors affecting synthesis of the methylcobalamin coenzyme required by methionine synthase (cblE and cblG) result in isolated homocystinuria. Combined methylmalonic aciduria and homocystinuria is seen in patients with impaired intestinal cobalamin absorption (intrinsic factor deficiency, Imerslund-Gräsbeck syndrome) and with defects affecting synthesis of both cobalamin coenzymes (cblC, cblD, cblF and cblJ). A series of disorders caused by pathogenic variant mutations affecting gene regulators (transcription factors) of the MMACHC gene have recently been described (HCFC1 [cblX disorder] and deficiencies of THAP11, and ZNF143 [the cblK disorder]).
Collapse
Affiliation(s)
- David Watkins
- Department of Human Genetics, McGill University, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
| | - David S Rosenblatt
- Department of Human Genetics, McGill University, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| |
Collapse
|
17
|
Zhu J, Wan S, Zhao X, Zhu B, Lv Y, Jiang H. Acute Lymphoblastic Leukemia in Combined Methylmalonic Acidemia and Homocysteinemia (cblC Type): A Case Report and Literature Review. Front Genet 2022; 13:856552. [PMID: 35495149 PMCID: PMC9048794 DOI: 10.3389/fgene.2022.856552] [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: 01/17/2022] [Accepted: 03/21/2022] [Indexed: 11/25/2022] Open
Abstract
Background: Methylmalonic acidemia (MMA) can display many clinical manifestations, among which acute lymphoblastic leukemia (ALL) has not been reported, and congenital heart disease (CHD) is also rare. Case presentation: We report an MMA case with ALL and CHD in a 5.5-year-old girl. With developmental delay and local brain atrophy in MRI, she was diagnosed with cerebral palsy at 9 months old. Rehabilitation was performed since then. This time she was admitted to hospital because of weakness and widespread bleeding spots. ALL-L2 (pre-B-cell) was confirmed by bone marrow morphology and immunophenotyping. Echocardiography showed patent foramen ovale. The girl was treated with VDLD and CAML chemotherapy, during which she developed seizures, edema and renal insufficiency. Decrease of muscle strength was also found in physical examination. Screening for inherited metabolic disorders showed significantly elevated levels of methylmalonate-2, acetylcarnitine (C2), propionylcarnitine (C3), C3/C2 and homocysteine. Gene analysis revealed a compound heterozygous mutaion in MMACHC (NM_015,560): c.80A > G (p.Gln27Arg) and c.609G > A (p.Trp203*). CblC type MMA was diagnosed. Intramuscular injection of cyanocobalamin and intravenous L-carnitine treatment were applied. The edema vanished gradually, and chemotherapy of small dosage of vindesine was given intermittently when condition permitted. 2 months later, muscle strength of both lower limbs were significantly improved to nearly grade 5. The levels of methylmalonic acid and homocysteine were improved. Conclusion: Metabolic disease screening and gene analysis are very necessary for diseases with complex clinical symptoms. ALL can be a rare manifestation for MMA. Synopsis: We report a case of methylmalonic acidemia with acute lymphoblastic leukemia and congenital heart disease, which uncovered the importance of genetic testing and metabolic diseases screening in patients with multiple systemic organ involvement.
Collapse
Affiliation(s)
- Jun Zhu
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, China
| | - Shuisen Wan
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, China
| | - Xueqi Zhao
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, China
| | - Binlu Zhu
- Department of Pediatrics, West China Second University Hospital, Chengdu, China
| | - Yuan Lv
- Department of Gynecology and Obstetrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hongkun Jiang
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, China
- *Correspondence: Hongkun Jiang,
| |
Collapse
|
18
|
Chern T, Achilleos A, Tong X, Hill MC, Saltzman AB, Reineke LC, Chaudhury A, Dasgupta SK, Redhead Y, Watkins D, Neilson JR, Thiagarajan P, Green JBA, Malovannaya A, Martin JF, Rosenblatt DS, Poché RA. Mutations in Hcfc1 and Ronin result in an inborn error of cobalamin metabolism and ribosomopathy. Nat Commun 2022; 13:134. [PMID: 35013307 PMCID: PMC8748873 DOI: 10.1038/s41467-021-27759-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 12/13/2021] [Indexed: 12/26/2022] Open
Abstract
Combined methylmalonic acidemia and homocystinuria (cblC) is the most common inborn error of intracellular cobalamin metabolism and due to mutations in Methylmalonic Aciduria type C and Homocystinuria (MMACHC). Recently, mutations in the transcriptional regulators HCFC1 and RONIN (THAP11) were shown to result in cellular phenocopies of cblC. Since HCFC1/RONIN jointly regulate MMACHC, patients with mutations in these factors suffer from reduced MMACHC expression and exhibit a cblC-like disease. However, additional de-regulated genes and the resulting pathophysiology is unknown. Therefore, we have generated mouse models of this disease. In addition to exhibiting loss of Mmachc, metabolic perturbations, and developmental defects previously observed in cblC, we uncovered reduced expression of target genes that encode ribosome protein subunits. We also identified specific phenotypes that we ascribe to deregulation of ribosome biogenesis impacting normal translation during development. These findings identify HCFC1/RONIN as transcriptional regulators of ribosome biogenesis during development and their mutation results in complex syndromes exhibiting aspects of both cblC and ribosomopathies. Combined methylmalonic acidemia (MMA) and hyperhomocysteinemias are inborn errors of vitamin B12 metabolism, and mutations in the transcriptional regulators HCFC1 and RONIN (THAP11) underlie some forms of these disorders. Here the authors generated mouse models of a human syndrome due to mutations in RONIN (THAP11) and HCFC1, and show that this syndrome is both an inborn error of vitamin B12 metabolism and displays some features of ribosomopathy.
Collapse
Affiliation(s)
- Tiffany Chern
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA.,Graduate Program in Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Annita Achilleos
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Basic and Clinical Sciences, University of Nicosia Medical School, Nicosia, Cyprus.
| | - Xuefei Tong
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Matthew C Hill
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA.,Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Alexander B Saltzman
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Lucas C Reineke
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Arindam Chaudhury
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Swapan K Dasgupta
- Department of Pathology, Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, 77030, USA
| | - Yushi Redhead
- The Francis Crick Institute, London, NW1 1AT, UK.,Centre for Craniofacial Biology and Regeneration, King's College London, London, SE1 9RT, UK
| | - David Watkins
- Division of Medical Genetics, Department of Specialized Medicine, McGill University Health Centre, Montreal, QC, Canada.,Division of Medical Biochemistry, Department of Specialized Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Joel R Neilson
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA.,Graduate Program in Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA.,Development, Disease Models and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Perumal Thiagarajan
- Department of Pathology, Center for Translational Research on Inflammatory Diseases, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, 77030, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jeremy B A Green
- Centre for Craniofacial Biology and Regeneration, King's College London, London, SE1 9RT, UK
| | - Anna Malovannaya
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - James F Martin
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA.,Graduate Program in Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA.,Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA.,Development, Disease Models and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA.,Texas Heart Institute, Houston, TX, 77030, USA
| | - David S Rosenblatt
- Division of Medical Genetics, Department of Specialized Medicine, McGill University Health Centre, Montreal, QC, Canada.,Division of Medical Biochemistry, Department of Specialized Medicine, McGill University Health Centre, Montreal, QC, Canada.,Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Ross A Poché
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA. .,Graduate Program in Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA. .,Graduate Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Development, Disease Models and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
19
|
Kiessling E, Nötzli S, Todorova V, Forny M, Baumgartner MR, Samardzija M, Krijt J, Kožich V, Grimm C, Froese DS. Absence of MMACHC in peripheral retinal cells does not lead to an ocular phenotype in mice. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166201. [PMID: 34147638 DOI: 10.1016/j.bbadis.2021.166201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/25/2021] [Accepted: 06/08/2021] [Indexed: 01/10/2023]
Abstract
Combined methylmalonic aciduria with homocystinuria (cblC type) is a rare disease caused by mutations in the MMACHC gene. MMACHC encodes an enzyme crucial for intracellular vitamin B12 metabolism, leading to the accumulation of toxic metabolites e.g. methylmalonic acid (MMA) and homocysteine (Hcy), and secondary disturbances in folate and one-carbon metabolism when not fully functional. Patients with cblC deficiency often present in the neonatal or early childhood period with a severe multisystem pathology, which comprises a broad spectrum of treatment-resistant ophthalmological phenotypes, including retinal degeneration, impaired vision, and vascular changes. To examine the potential function of MMACHC in the retina and how its loss may impact disease, we performed gene expression studies in human and mouse, which showed that local expression of MMACHC in the retina and retinal pigment epithelium is relatively stable over time. To study whether functional MMACHC is required for retinal function and tissue integrity, we generated a transgenic mouse lacking Mmachc expression in cells of the peripheral retina. Characterization of this mouse revealed accumulation of cblC disease related metabolites, including MMA and the folate-dependent purine synthesis intermediates AICA-riboside and SAICA-riboside in the retina. Nevertheless, fundus appearance, morphology, vasculature, and cellular composition of the retina, as well as ocular function, remained normal in mice up to 6 or 12 months of age. Our data indicates that peripheral retinal neurons do not require intrinsic expression of Mmachc for survival and function and questions whether a local MMACHC deficiency is responsible for the retinal phenotypes in patients.
Collapse
Affiliation(s)
- Eva Kiessling
- Lab for Retinal Cell Biology, Dept. Ophthalmology, University Hospital Zurich, University of Zürich, Switzerland
| | - Sarah Nötzli
- Lab for Retinal Cell Biology, Dept. Ophthalmology, University Hospital Zurich, University of Zürich, Switzerland
| | - Vyara Todorova
- Lab for Retinal Cell Biology, Dept. Ophthalmology, University Hospital Zurich, University of Zürich, Switzerland
| | - Merima Forny
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, University of Zürich, Switzerland
| | - Matthias R Baumgartner
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, University of Zürich, Switzerland
| | - Marijana Samardzija
- Lab for Retinal Cell Biology, Dept. Ophthalmology, University Hospital Zurich, University of Zürich, Switzerland
| | - Jakub Krijt
- Dept. of Pediatrics and Inherited Metabolic Disorders, Charles University-First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - Viktor Kožich
- Dept. of Pediatrics and Inherited Metabolic Disorders, Charles University-First Faculty of Medicine and General University Hospital, Prague, Czech Republic
| | - Christian Grimm
- Lab for Retinal Cell Biology, Dept. Ophthalmology, University Hospital Zurich, University of Zürich, Switzerland.
| | - D Sean Froese
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, University of Zürich, Switzerland.
| |
Collapse
|
20
|
Sobczyńska-Malefora A, Delvin E, McCaddon A, Ahmadi KR, Harrington DJ. Vitamin B 12 status in health and disease: a critical review. Diagnosis of deficiency and insufficiency - clinical and laboratory pitfalls. Crit Rev Clin Lab Sci 2021; 58:399-429. [PMID: 33881359 DOI: 10.1080/10408363.2021.1885339] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Vitamin B12 (cobalamin) is an essential cofactor for two metabolic pathways. It is obtained principally from food of animal origin. Cobalamin becomes bioavailable through a series of steps pertaining to its release from dietary protein, intrinsic factor-mediated absorption, haptocorrin or transcobalamin-mediated transport, cellular uptake, and two enzymatic conversions (via methionine synthase and methylmalonyl-CoA-mutase) into cofactor forms: methylcobalamin and adenosylcobalamin. Vitamin B12 deficiency can masquerade as a multitude of illnesses, presenting different perspectives from the point of view of the hematologist, neurologist, gastroenterologist, general physician, or dietician. Increased physician vigilance and heightened patient awareness often account for its early presentation, and testing sometimes occurs during a phase of vitamin B12 insufficiency before the main onset of the disease. The chosen test often depends on its availability rather than on the diagnostic performance and sensitivity to irrelevant factors interfering with vitamin B12 markers. Although serum B12 is still the most commonly used and widely available test, diagnostics by holotranscobalamin, serum methylmalonic acid, and plasma homocysteine measurements have grown in the last several years in routine practice. The lack of a robust absorption test, coupled with compromised sensitivity and specificity of other tests (intrinsic factor and gastric parietal cell antibodies), hinders determination of the cause for depleted B12 status. This can lead to incorrect supplementation regimes and uncertainty regarding later treatment. This review discusses currently available knowledge on vitamin B12, informs the reader about the pitfalls of tests for assessing its deficiency, reviews B12 status in various populations at different disease stages, and provides recommendations for interpretation, treatment, and associated risks. Future directions for diagnostics of B12 status and health interventions are also discussed.
Collapse
Affiliation(s)
- Agata Sobczyńska-Malefora
- The Nutristasis Unit, Viapath, St. Thomas' Hospital, London, UK.,Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Edgard Delvin
- Sainte-Justine UHC Research Centre, Montreal, Canada.,Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, Canada
| | | | - Kourosh R Ahmadi
- Department of Nutrition & Metabolism, School of Biosciences and Medicine, University of Surrey, Guildford, UK
| | - Dominic J Harrington
- The Nutristasis Unit, Viapath, St. Thomas' Hospital, London, UK.,Faculty of Life Sciences and Medicine, King's College London, London, UK
| |
Collapse
|
21
|
Ching-López A, Martinez-Gonzalez LJ, Arrabal L, Sáiz J, Gavilán Á, Barbas C, Lorente JA, Roldán S, Sánchez MJ, Gutierrez-Ríos P. Combined Genome, Transcriptome and Metabolome Analysis in the Diagnosis of Childhood Cerebellar Ataxia. Int J Mol Sci 2021; 22:2990. [PMID: 33804237 PMCID: PMC8002209 DOI: 10.3390/ijms22062990] [Citation(s) in RCA: 5] [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: 02/26/2021] [Revised: 03/11/2021] [Accepted: 03/13/2021] [Indexed: 12/30/2022] Open
Abstract
Ataxia in children is a common clinical sign of numerous neurological disorders consisting of impaired coordination of voluntary muscle movement. Its most common form, cerebellar ataxia, describes a heterogeneous array of neurologic conditions with uncountable causes broadly divided as acquired or genetic. Numerous genetic disorders are associated with chronic progressive ataxia, which complicates clinical management, particularly on the diagnostic stage. Advances in omics technologies enable improvements in clinical practice and research, so we proposed a multi-omics approach to aid in the genetic diagnosis and molecular elucidation of an undiagnosed infantile condition of chronic progressive cerebellar ataxia. Using whole-exome sequencing, RNA-seq, and untargeted metabolomics, we identified three clinically relevant mutations (rs141471029, rs191582628 and rs398124292) and an altered metabolic profile in our patient. Two POLR1C diagnostic variants already classified as pathogenic were found, and a diagnosis of hypomyelinating leukodystrophy was achieved. A mutation on the MMACHC gene, known to be associated with methylmalonic aciduria and homocystinuria cblC type, was also found. Additionally, preliminary metabolome analysis revealed alterations in our patient's amino acid, fatty acid and carbohydrate metabolism. Our findings provided a definitive genetic diagnosis reinforcing the association between POLR1C mutations and hypomyelinating leukodystrophy and highlighted the relevance of multi-omics approaches to the disease.
Collapse
Affiliation(s)
- Ana Ching-López
- CIBER Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain;
- Andalusian School of Public Health (EASP), 18080 Granada, Spain
| | - Luis Javier Martinez-Gonzalez
- GENYO, Centre for Genomics and Oncological Research, Pfizer, University of Granada, Andalusian Regional Government, PTS Granada, 18016 Granada, Spain; (L.J.M.-G.); (J.A.L.)
| | - Luisa Arrabal
- Pediatric Neurology Department, Hospital Virgen de las Nieves, 18014 Granada, Spain; (L.A.); (S.R.)
| | - Jorge Sáiz
- Centre for Metabolomics and Bioanalysis (CEMBIO), Chemistry and Biochemistry Department, Pharmacy Faculty, Universidad San Pablo-CEU, 28925 Madrid, Spain; (J.S.); (C.B.)
| | - Ángela Gavilán
- Institute of Biomedicine of Seville (IBIS), 41013 Seville, Spain;
| | - Coral Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO), Chemistry and Biochemistry Department, Pharmacy Faculty, Universidad San Pablo-CEU, 28925 Madrid, Spain; (J.S.); (C.B.)
| | - Jose Antonio Lorente
- GENYO, Centre for Genomics and Oncological Research, Pfizer, University of Granada, Andalusian Regional Government, PTS Granada, 18016 Granada, Spain; (L.J.M.-G.); (J.A.L.)
- Laboratory of Genetic Identification, Legal Medicine and Toxicology Department, Faculty of Medicine-PTS, University of Granada, 18016 Granada, Spain
| | - Susana Roldán
- Pediatric Neurology Department, Hospital Virgen de las Nieves, 18014 Granada, Spain; (L.A.); (S.R.)
| | - Maria José Sánchez
- CIBER Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain;
- Andalusian School of Public Health (EASP), 18080 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain;
| | | |
Collapse
|
22
|
Torices L, de las Heras J, Arango-Lasprilla JC, Cortés JM, Nunes-Xavier CE, Pulido R. MMADHC premature termination codons in the pathogenesis of cobalamin D disorder: Potential of translational readthrough reconstitution. Mol Genet Metab Rep 2021; 26:100710. [PMID: 33552904 PMCID: PMC7847965 DOI: 10.1016/j.ymgmr.2021.100710] [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: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 11/21/2022] Open
Abstract
Mutations in the MMADHC gene cause cobalamin D disorder (cblD), an autosomal recessive inborn disease with defects in intracellular cobalamin (cbl, vitamin B12) metabolism. CblD patients present methylmalonic aciduria (MMA), homocystinuria (HC), or combined MMA/HC, and usually suffer developmental delay and cognitive deficits. The most frequent MMADHC genetic alterations associated with disease generate MMADHC truncated proteins, in many cases due to mutations that create premature termination codons (PTC). In this study, we have performed a comprehensive and global characterization of MMADHC protein variants generated by all annotated MMADHC PTC mutations in cblD patients, and analyzed the potential of inducible translational PTC readthrough to reconstitute MMADHC biosynthesis. MMADHC protein truncation caused by disease-associated PTC differentially affected the alternative usage of translation initiation sites, protein abundance, and subcellular localization of MMADHC. Aminoglycoside compounds induced translational PTC readthrough of MMADHC truncated variants, allowing the biosynthesis of full-length MMADHC in a PTC-specific manner. Our results suggest that translational PTC readthrough-based interventions could complement current therapies for cblD patients carrying specific MMADHC PTC mutations.
Collapse
Affiliation(s)
- Leire Torices
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Javier de las Heras
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Division of Pediatric Metabolism (CIBER-ER), Cruces University Hospital, Barakaldo, Spain
- Department of Pediatrics, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Juan Carlos Arango-Lasprilla
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Department of Cell Biology and Histology, University of the Basque Country (UPV/EHU), Leioa, Spain
- Ikerbasque, The Basque Foundation for Science, 48013 Bilbao, Spain
| | - Jesús M. Cortés
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Department of Cell Biology and Histology, University of the Basque Country (UPV/EHU), Leioa, Spain
- Ikerbasque, The Basque Foundation for Science, 48013 Bilbao, Spain
| | - Caroline E. Nunes-Xavier
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Institute of Cancer Research, Oslo University Hospital Radiumhospitalet, N-0424 Oslo, Norway
| | - Rafael Pulido
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Ikerbasque, The Basque Foundation for Science, 48013 Bilbao, Spain
| |
Collapse
|
23
|
Wingert V, Mukherjee S, Esser AJ, Behringer S, Tanimowo S, Klenzendorf M, Derevenkov IA, Makarov SV, Jacobsen DW, Spiekerkoetter U, Hannibal L. Thiolatocobalamins repair the activity of pathogenic variants of the human cobalamin processing enzyme CblC. Biochimie 2020; 183:108-125. [PMID: 33190793 DOI: 10.1016/j.biochi.2020.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/08/2020] [Accepted: 10/19/2020] [Indexed: 01/08/2023]
Abstract
Thiolatocobalamins are a class of cobalamins comprised of naturally occurring and synthetic ligands. Glutathionylcobalamin (GSCbl) occurs naturally in mammalian cells, and also as an intermediate in the glutathione-dependent dealkylation of methylcobalamin (MeCbl) to form cob(I)alamin by pure recombinant CblC from C. elegans. Glutathione-driven deglutathionylation of GSCbl was demonstrated both in mammalian as well as in C. elegans CblC. Dethiolation is orders of magnitude faster than dealkylation of Co-C bonded cobalamins, which motivated us to investigate two synthetic thiolatocobalamins as substrates to repair the enzymatic activity of pathogenic CblC variants in humans. We report the synthesis and kinetic characterization of cysteaminylcobalamin (CyaCbl) and 2-mercaptopropionylglycinocobalamin (MpgCbl). Both CyaCbl and MpgCbl were obtained in high purity (90-95%) and yield (78-85%). UV-visible spectral properties agreed with those reported for other thiolatocobalamins with absorbance maxima observed at 372 nm and 532 nm. Both CyaCbl and MpgCbl bound to wild type human recombinant CblC inducing spectral blue-shifts characteristic of the respective base-on to base-off transitions. Addition of excess glutathione (GSH) resulted in rapid elimination of the β-ligand to give aquacobalamin (H2OCbl) as the reaction product under aerobic conditions. Further, CyaCbl and MpgCbl underwent spontaneous dethiolation thereby repairing the loss of activity of pathogenic variants of human CblC, namely R161G and R161Q. We posit that thiolatocobalamins could be exploited therapeutically for the treatment of inborn errors of metabolism that impair processing of dietary and supplemental cobalamin forms. While these disorders are targets for newborn screening in some countries, there is currently no effective treatment available to patients.
Collapse
Affiliation(s)
- Victoria Wingert
- Laboratory of Clinical Biochemistry and Metabolism, Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center - University of Freiburg, 79106, Freiburg, Germany
| | - Srijan Mukherjee
- Laboratory of Clinical Biochemistry and Metabolism, Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center - University of Freiburg, 79106, Freiburg, Germany
| | - Anna J Esser
- Laboratory of Clinical Biochemistry and Metabolism, Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center - University of Freiburg, 79106, Freiburg, Germany
| | - Sidney Behringer
- Laboratory of Clinical Biochemistry and Metabolism, Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center - University of Freiburg, 79106, Freiburg, Germany
| | - Segun Tanimowo
- Laboratory of Clinical Biochemistry and Metabolism, Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center - University of Freiburg, 79106, Freiburg, Germany
| | - Melissa Klenzendorf
- Laboratory of Clinical Biochemistry and Metabolism, Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center - University of Freiburg, 79106, Freiburg, Germany; Faculty of Biology, University of Freiburg Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Ilia A Derevenkov
- Department of Food Chemistry, Ivanovo State University of Chemistry and Technology, Ivanovo, Russian Federation
| | - Sergei V Makarov
- Department of Food Chemistry, Ivanovo State University of Chemistry and Technology, Ivanovo, Russian Federation
| | - Donald W Jacobsen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, USA
| | - Ute Spiekerkoetter
- Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center - University of Freiburg, 79106, Freiburg, Germany
| | - Luciana Hannibal
- Laboratory of Clinical Biochemistry and Metabolism, Department of General Pediatrics, Adolescent Medicine and Neonatology, Faculty of Medicine, Medical Center - University of Freiburg, 79106, Freiburg, Germany.
| |
Collapse
|
24
|
Li Z, Mascarenhas R, Twahir UT, Kallon A, Deb A, Yaw M, Penner-Hahn J, Koutmos M, Warncke K, Banerjee R. An Interprotein Co-S Coordination Complex in the B 12-Trafficking Pathway. J Am Chem Soc 2020; 142:16334-16345. [PMID: 32871076 DOI: 10.1021/jacs.0c06590] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The CblC and CblD chaperones are involved in early steps in the cobalamin trafficking pathway. Cobalamin derivatives entering the cytoplasm are converted by CblC to a common cob(II)alamin intermediate via glutathione-dependent alkyltransferase or reductive elimination activities. Cob(II)alamin is subsequently converted to one of two biologically active alkylcobalamins by downstream chaperones. The function of CblD has been elusive although it is known to form a complex with CblC under certain conditions. Here, we report that CblD provides a sulfur ligand to cob(II)alamin bound to CblC, forming an interprotein coordination complex that rapidly oxidizes to thiolato-cob(III)alamin. Cysteine scanning mutagenesis and EPR spectroscopy identified Cys-261 on CblD as the sulfur donor. The unusual interprotein Co-S bond was characterized by X-ray absorption spectroscopy and visualized in the crystal structure of the human CblD thiolato-cob(III)alamin complex. Our study provides insights into how cobalamin coordination chemistry could be utilized for cofactor translocation in the trafficking pathway.
Collapse
Affiliation(s)
- Zhu Li
- Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, Michigan 48109-0600, United States
| | - Romila Mascarenhas
- Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, Michigan 48109-0600, United States
| | - Umar T Twahir
- Department of Physics, Emory University, Atlanta, Georgia 30322-2430, United States
| | - Albert Kallon
- Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, Michigan 48109-0600, United States
| | - Aniruddha Deb
- Departments of Chemistry and Biophysics, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Madeline Yaw
- Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, Michigan 48109-0600, United States
| | - James Penner-Hahn
- Departments of Chemistry and Biophysics, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Markos Koutmos
- Departments of Chemistry and Biophysics, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Kurt Warncke
- Department of Physics, Emory University, Atlanta, Georgia 30322-2430, United States
| | - Ruma Banerjee
- Department of Biological Chemistry, University of Michigan Medical Center, Ann Arbor, Michigan 48109-0600, United States
| |
Collapse
|
25
|
Chern T, Achilleos A, Tong X, Hsu CW, Wong L, Poché RA. Mouse models to study the pathophysiology of combined methylmalonic acidemia and homocystinuria, cblC type. Dev Biol 2020; 468:1-13. [PMID: 32941884 DOI: 10.1016/j.ydbio.2020.09.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023]
Abstract
Combined methylmalonic acidemia and homocystinuria, cblC type, is the most common inherited disorder of cobalamin metabolism and is characterized by severe fetal developmental defects primarily impacting the central nervous system, hematopoietic system, and heart. CblC was previously shown to be due to mutations in the MMACHC gene, which encodes a protein thought to function in intracellular cobalamin trafficking and biosynthesis of adenosylcobalamin (AdoCbl) and methylcobalamin (MeCbl). These coenzymes are required for the production of succinyl-CoA and methionine, respectively. However, it is currently unclear whether additional roles for MMACHC exist outside of cobalamin metabolism. Furthermore, due to a lack of sufficient animal models, the exact pathophysiology of cblC remains unknown. Here, we report the generation and characterization of two new mouse models to study the role of MMACHC in vivo. CRISPR/Cas9 genome editing was used to develop a Mmachc floxed allele (Mmachcflox/flox), which we validated as a conditional null. For a gain-of-function approach, we generated a transgenic mouse line that over-expresses functional Mmachc (Mmachc-OE+/tg) capable of rescuing Mmachc homozygous mutant lethality. Surprisingly, our data also suggest that these mice may exhibit a partially penetrant maternal-effect rescue, which might have implications for in utero therapeutic interventions to treat cblC. Both the Mmachcflox/flox and Mmachc-OE+/tg mouse models will be valuable resources for understanding the biological roles of MMACHC in a variety of tissue contexts and allow for deeper understanding of the pathophysiology of cblC.
Collapse
Affiliation(s)
- Tiffany Chern
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Annita Achilleos
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xuefei Tong
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chih-Wei Hsu
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Leeyean Wong
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ross A Poché
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; Graduate Program in Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; Development, Disease Models and Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA; Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
26
|
Castro VL, Quintana AM. The role of HCFC1 in syndromic and non-syndromic intellectual disability. ACTA ACUST UNITED AC 2020; 8. [PMID: 34164576 DOI: 10.18103/mra.v8i6.2122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mutations in the HCFC1 gene are associated with cases of syndromic (cblX) and non-syndromic intellectual disability. Syndromic individuals present with severe neurological defects including intractable epilepsy, facial dysmorphia, and intellectual disability. Non-syndromic individuals have also been described and implicate a role for HCFC1 during brain development. The penetrance of phenotypes and the presence of an overall syndrome is associated with the location of the mutation within the HCFC1 protein. Thus, one could hypothesize that the positioning of HCFC1 mutations lead to different neurological phenotypes that include but are not restricted to intellectual disability. The HCFC1 protein is comprised of multiple domains that function in cellular proliferation/metabolism. Several reports of HCFC1 disease variants have been identified, but a comprehensive review of each variant and its associated phenotypes has not yet been compiled. Here we perform a detailed review of HCFC1 function, model systems, variant location, and accompanying phenotypes to highlight current knowledge and the future status of the field.
Collapse
Affiliation(s)
- Victoria L Castro
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, 79968
| | - Anita M Quintana
- Department of Biological Sciences, The University of Texas at El Paso, El Paso, TX, 79968
| |
Collapse
|
27
|
Haijes HA, van Hasselt PM, Jans JJM, Verhoeven-Duif NM. Pathophysiology of propionic and methylmalonic acidemias. Part 2: Treatment strategies. J Inherit Metab Dis 2019; 42:745-761. [PMID: 31119742 DOI: 10.1002/jimd.12128] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 05/18/2019] [Accepted: 05/21/2019] [Indexed: 12/31/2022]
Abstract
Despite realizing increased survival rates for propionic acidemia (PA) and methylmalonic acidemia (MMA) patients, the current therapeutic regimen is inadequate for preventing or treating the devastating complications that still can occur. The elucidation of pathophysiology of these complications allows us to evaluate and rethink treatment strategies. In this review we display and discuss potential therapy targets and we give a systematic overview on current, experimental and unexplored treatment strategies in order to provide insight in what we have to offer PA and MMA patients, now and in the future. Evidence on the effectiveness of treatment strategies is often scarce, since none were tested in randomized clinical trials. This raises concerns, since even the current consensus on best practice treatment for PA and MMA is not without controversy. To attain substantial improvements in overall outcome, gene, mRNA or enzyme replacement therapy is most promising since permanent reduction of toxic metabolites allows for a less strict therapeutic regime. Hereby, both mitochondrial-associated and therapy induced complications can theoretically be prevented. However, the road from bench to bedside is long, as it is challenging to design a drug that is delivered to the mitochondria of all tissues that require enzymatic activity, including the brain, without inducing any off-target effects. To improve survival rate and quality of life of PA and MMA patients, there is a need for systematic (re-)evaluation of accepted and potential treatment strategies, so that we can better determine who will benefit when and how from which treatment strategy.
Collapse
Affiliation(s)
- Hanneke A Haijes
- Section Metabolic Diagnostics, Department of Biomedical Genetics, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
- Section Metabolic Diseases, Department of Child Health, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Peter M van Hasselt
- Section Metabolic Diseases, Department of Child Health, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Judith J M Jans
- Section Metabolic Diagnostics, Department of Biomedical Genetics, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Nanda M Verhoeven-Duif
- Section Metabolic Diagnostics, Department of Biomedical Genetics, Centre for Molecular Medicine, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
28
|
Froese DS, Fowler B, Baumgartner MR. Vitamin B 12 , folate, and the methionine remethylation cycle-biochemistry, pathways, and regulation. J Inherit Metab Dis 2019; 42:673-685. [PMID: 30693532 DOI: 10.1002/jimd.12009] [Citation(s) in RCA: 238] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/27/2018] [Accepted: 10/19/2018] [Indexed: 12/16/2022]
Abstract
Vitamin B12 (cobalamin, Cbl) is a nutrient essential to human health. Due to its complex structure and dual cofactor forms, Cbl undergoes a complicated series of absorptive and processing steps before serving as cofactor for the enzymes methylmalonyl-CoA mutase and methionine synthase. Methylmalonyl-CoA mutase is required for the catabolism of certain (branched-chain) amino acids into an anaplerotic substrate in the mitochondrion, and dysfunction of the enzyme itself or in production of its cofactor adenosyl-Cbl result in an inability to successfully undergo protein catabolism with concomitant mitochondrial energy disruption. Methionine synthase catalyzes the methyl-Cbl dependent (re)methylation of homocysteine to methionine within the methionine cycle; a reaction required to produce this essential amino acid and generate S-adenosylmethionine, the most important cellular methyl-donor. Disruption of methionine synthase has wide-ranging implications for all methylation-dependent reactions, including epigenetic modification, but also for the intracellular folate pathway, since methionine synthase uses 5-methyltetrahydrofolate as a one-carbon donor. Folate-bound one-carbon units are also required for deoxythymidine monophosphate and de novo purine synthesis; therefore, the flow of single carbon units to each of these pathways must be regulated based on cellular needs. This review provides an overview on Cbl metabolism with a brief description of absorption and intracellular metabolic pathways. It also provides a description of folate-mediated one-carbon metabolism and its intersection with Cbl at the methionine cycle. Finally, a summary of recent advances in understanding of how both pathways are regulated is presented.
Collapse
Affiliation(s)
- D Sean Froese
- Division of Metabolism and Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Brian Fowler
- Division of Metabolism and Children's Research Center, University Children's Hospital, Zurich, Switzerland
| | - Matthias R Baumgartner
- Division of Metabolism and Children's Research Center, University Children's Hospital, Zurich, Switzerland
| |
Collapse
|
29
|
Huemer M, Baumgartner MR. The clinical presentation of cobalamin-related disorders: From acquired deficiencies to inborn errors of absorption and intracellular pathways. J Inherit Metab Dis 2019; 42:686-705. [PMID: 30761552 DOI: 10.1002/jimd.12012] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 09/25/2018] [Accepted: 09/27/2018] [Indexed: 12/11/2022]
Abstract
This review gives an overview of clinical characteristics, treatment and outcome of nutritional and acquired cobalamin (Cbl; synonym: vitamin B12) deficiencies, inborn errors of Cbl absorption and intracellular trafficking, as well as methylenetetrahydrofolate dehydrogenase (MTHFD1) and methylene tetrahydrofolate reductase (MTHFR) deficiencies, which impair Cbl-dependent remethylation. Acquired and inborn Cbl-related disorders and MTHFR deficiency cause multisystem, often severe disease. Failure to thrive, neurocognitive or psychiatric symptoms, eye disease, bone marrow alterations, microangiopathy and thromboembolic events are characteristic. The recently identified MTHFD1 defect additionally presents with severe immune deficiency. Deficient Cbl-dependent enzymes cause reduced methylation capacity and metabolite toxicity. Further net-effects of perturbed Cbl function or reduced Cbl supply causing oxidative stress, altered cytokine regulation or immune functions are discussed.
Collapse
Affiliation(s)
- Martina Huemer
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, Zürich, Switzerland
- Department of Paediatrics, Landeskrankenhaus Bregenz, Bregenz, Austria
| | - Matthias R Baumgartner
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, Zürich, Switzerland
| |
Collapse
|
30
|
McDonald MK, Fritz JA, Jia D, Scheuchner D, Snyder FF, Stanislaus A, Curle J, Li L, Stabler SP, Allen RH, Mains PE, Gravel RA. Identification of ABC transporters acting in vitamin B 12 metabolism in Caenorhabditis elegans. Mol Genet Metab 2017; 122:160-171. [PMID: 29153845 DOI: 10.1016/j.ymgme.2017.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Revised: 11/10/2017] [Accepted: 11/10/2017] [Indexed: 01/19/2023]
Abstract
Vitamin B12 (cobalamin, Cbl) is a micronutrient essential to human health. Cbl is not utilized as is but must go through complex subcellular and metabolic processing to generate two cofactor forms: methyl-Cbl for methionine synthase, a cytosolic enzyme; and adenosyl-Cbl for methylmalonyl-CoA mutase, a mitochondrial enzyme. Some 10-12 human genes have been identified responsible for the intracellular conversion of Cbl to cofactor forms, including genes that code for ATP-binding cassette (ABC) transporters acting at the lysosomal and plasma membranes. However, the gene for mitochondrial uptake is not known. We hypothesized that ABC transporters should be candidates for other uptake and efflux functions, including mitochondrial transport, and set out to screen ABC transporter mutants for blocks in Cbl utilization using the nematode roundworm Caenorhabditis elegans. Thirty-seven mutant ABC transporters were screened for the excretion of methylmalonic acid (MMA), which should result from loss of Cbl transport into the mitochondria. One mutant, wht-6, showed elevated MMA excretion and reduced [14C]-propionate incorporation, pointing to a functional block in methylmalonyl-CoA mutase. In contrast, the wht-6 mutant appeared to have a normal cytosolic pathway based on analysis of cystathionine excretion, suggesting that cytosolic methionine synthase was functioning properly. Further, the MMA excretion in wht-6 could be partially reversed by including vitamin B12 in the assay medium. The human ortholog of wht-6 is a member of the G family of ABC transporters. We propose wht-6 as a candidate for the transport of Cbl into mitochondria and suggest that a member of the corresponding ABCG family of ABC transporters has this role in humans. Our ABC transporter screen also revealed that mrp-1 and mrp-2 mutants excreted lower MMA than wild type, suggesting they were concentrating intracellular Cbl, consistent with the cellular efflux defect proposed for the mammalian MRP1 ABC transporter.
Collapse
Affiliation(s)
- Megan K McDonald
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, T2N 4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, T2N 4N1, Canada
| | - Julie-Anne Fritz
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, T2N 4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, T2N 4N1, Canada
| | - Dongxin Jia
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, T2N 4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, T2N 4N1, Canada
| | - Deborah Scheuchner
- Department of Medical Genetics, University of Calgary, Calgary, T2N 4N1, Canada
| | - Floyd F Snyder
- Department of Medical Genetics, University of Calgary, Calgary, T2N 4N1, Canada
| | - Avalyn Stanislaus
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Jared Curle
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Liang Li
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada
| | - Sally P Stabler
- Division of Hematology, University of Colorado Denver, Aurora, CO, USA
| | - Robert H Allen
- Division of Hematology, University of Colorado Denver, Aurora, CO, USA
| | - Paul E Mains
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, T2N 4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, T2N 4N1, Canada
| | - Roy A Gravel
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, T2N 4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, T2N 4N1, Canada.
| |
Collapse
|
31
|
Plessl T, Bürer C, Lutz S, Yue WW, Baumgartner MR, Froese DS. Protein destabilization and loss of protein‐protein interaction are fundamental mechanisms in
cblA
‐type methylmalonic aciduria. Hum Mutat 2017; 38:988-1001. [DOI: 10.1002/humu.23251] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/12/2017] [Accepted: 05/06/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Tanja Plessl
- Division of Metabolism and Children's Research CenterUniversity Children'sHospital Zurich Switzerland
- Zurich Center for Integrative Human PhysiologyUniversity of Zurich Switzerland
| | - Céline Bürer
- Division of Metabolism and Children's Research CenterUniversity Children'sHospital Zurich Switzerland
| | - Seraina Lutz
- Division of Metabolism and Children's Research CenterUniversity Children'sHospital Zurich Switzerland
| | - Wyatt W. Yue
- Structural Genomics ConsortiumNuffield Department of Clinical MedicineUniversity of Oxford Oxford United Kingdom
| | - Matthias R. Baumgartner
- Division of Metabolism and Children's Research CenterUniversity Children'sHospital Zurich Switzerland
- Zurich Center for Integrative Human PhysiologyUniversity of Zurich Switzerland
- radiz – Rare Disease Initiative ZurichClinical Research Priority Program for Rare DiseasesUniversity of ZurichZurich Switzerland
| | - D. Sean Froese
- Division of Metabolism and Children's Research CenterUniversity Children'sHospital Zurich Switzerland
- radiz – Rare Disease Initiative ZurichClinical Research Priority Program for Rare DiseasesUniversity of ZurichZurich Switzerland
| |
Collapse
|
32
|
Fettelschoss V, Burda P, Sagné C, Coelho D, De Laet C, Lutz S, Suormala T, Fowler B, Pietrancosta N, Gasnier B, Bornhauser B, Froese DS, Baumgartner MR. Clinical or ATPase domain mutations in ABCD4 disrupt the interaction between the vitamin B 12-trafficking proteins ABCD4 and LMBD1. J Biol Chem 2017; 292:11980-11991. [PMID: 28572511 DOI: 10.1074/jbc.m117.784819] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 05/24/2017] [Indexed: 01/19/2023] Open
Abstract
Vitamin B12 (cobalamin (Cbl)), in the cofactor forms methyl-Cbl and adenosyl-Cbl, is required for the function of the essential enzymes methionine synthase and methylmalonyl-CoA mutase, respectively. Cbl enters mammalian cells by receptor-mediated endocytosis of protein-bound Cbl followed by lysosomal export of free Cbl to the cytosol and further processing to these cofactor forms. The integral membrane proteins LMBD1 and ABCD4 are required for lysosomal release of Cbl, and mutations in the genes LMBRD1 and ABCD4 result in the cobalamin metabolism disorders cblF and cblJ. We report a new (fifth) patient with the cblJ disorder who presented at 7 days of age with poor feeding, hypotonia, methylmalonic aciduria, and elevated plasma homocysteine and harbored the mutations c.1667_1668delAG [p.Glu556Glyfs*27] and c.1295G>A [p.Arg432Gln] in the ABCD4 gene. Cbl cofactor forms are decreased in fibroblasts from this patient but could be rescued by overexpression of either ABCD4 or, unexpectedly, LMBD1. Using a sensitive live-cell FRET assay, we demonstrated selective interaction between ABCD4 and LMBD1 and decreased interaction when ABCD4 harbored the patient mutations p.Arg432Gln or p.Asn141Lys or when artificial mutations disrupted the ATPase domain. Finally, we showed that ABCD4 lysosomal targeting depends on co-expression of, and interaction with, LMBD1. These data broaden the patient and mutation spectrum of cblJ deficiency, establish a sensitive live-cell assay to detect the LMBD1-ABCD4 interaction, and confirm the importance of this interaction for proper intracellular targeting of ABCD4 and cobalamin cofactor synthesis.
Collapse
Affiliation(s)
- Victoria Fettelschoss
- Division of Metabolism and Children's Research Center, University Children's Hospital, CH-8032 Zurich, Switzerland
| | - Patricie Burda
- Division of Metabolism and Children's Research Center, University Children's Hospital, CH-8032 Zurich, Switzerland
| | - Corinne Sagné
- Neurophotonics Laboratory UMR 8250, Paris Descartes University, CNRS, Sorbonne Paris Cité, F-75006 Paris, France
| | - David Coelho
- UMR-S UL-INSERM U954 Nutrition-Genetics-Environmental Risk Exposure and Reference Centre of Inborn Metabolism Diseases, Medical Faculty of Nancy University and University Hospital Centre, Nancy, France
| | - Corinne De Laet
- Nutrition and Metabolism Unit, Queen Fabiola Children's University Hospital, Free University of Brussels (ULB), 1020 Brussels, Belgium
| | - Seraina Lutz
- Division of Metabolism and Children's Research Center, University Children's Hospital, CH-8032 Zurich, Switzerland
| | - Terttu Suormala
- Division of Metabolism and Children's Research Center, University Children's Hospital, CH-8032 Zurich, Switzerland
| | - Brian Fowler
- Division of Metabolism and Children's Research Center, University Children's Hospital, CH-8032 Zurich, Switzerland
| | - Nicolas Pietrancosta
- CBMIT team, UMR 8601, Paris Descartes University, CNRS, Sorbonne Paris Cité, F-75006 Paris, France
| | - Bruno Gasnier
- Neurophotonics Laboratory UMR 8250, Paris Descartes University, CNRS, Sorbonne Paris Cité, F-75006 Paris, France
| | - Beat Bornhauser
- Department of Oncology, Children's Research Center, University Children's Hospital, CH-8032 Zurich, Switzerland
| | - D Sean Froese
- Division of Metabolism and Children's Research Center, University Children's Hospital, CH-8032 Zurich, Switzerland; Rare Disease Initiative Zurich (radiz), Clinical Research Priority Program for Rare Diseases, University of Zurich, CH-8006 Zurich, Switzerland.
| | - Matthias R Baumgartner
- Division of Metabolism and Children's Research Center, University Children's Hospital, CH-8032 Zurich, Switzerland; Rare Disease Initiative Zurich (radiz), Clinical Research Priority Program for Rare Diseases, University of Zurich, CH-8006 Zurich, Switzerland; Zurich Center for Integrative Human Physiology, University of Zurich, CH-8006 Zurich, Switzerland.
| |
Collapse
|
33
|
Methionine synthase and methionine synthase reductase interact with MMACHC and with MMADHC. Biochim Biophys Acta Mol Basis Dis 2017; 1863:103-112. [DOI: 10.1016/j.bbadis.2016.10.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 10/01/2016] [Accepted: 10/18/2016] [Indexed: 01/02/2023]
|
34
|
Nogueira C, Marcão A, Rocha H, Sousa C, Fonseca H, Valongo C, Vilarinho L. Molecular picture of cobalamin C/D defects before and after newborn screening era. J Med Screen 2016; 24:6-11. [DOI: 10.1177/0969141316641149] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Objective Birth prevalence of Cobalamin (Cbl) C or D defects in Portugal is an estimated 1:85,000, one of the highest worldwide. We compared the genotype/phenotype of patients identified with CblC or CblD before and after the implementation of expanded newborn screening. Methods Twenty-five Portuguese CblC/D patients, 14 symptomatic and 11 identified through screening, were diagnosed using gas chromatography or tandem mass spectrometry. Molecular characterization was performed through the study of MMACHC and MMADHC genes. Results The most common MMACHC mutation, c.271dupA, was present in 100% of MMACHC alleles of all CblC screened patients, in contrast with the 61% identified before expanded newborn screening. All studied cases (except one, who presented a CblD deficiency) presented a CblC defect. More CblC late-onset patients were diagnosed before the introduction of newborn screening than in the post newborn screening era, probably because some early onset patients died without a definitive diagnosis. Conclusion The molecular data found in this cohort contribute to the improvement of screening and diagnosis of Cbl defects and would enable a confirmatory diagnosis of these patients, reducing the need for complex, costly, laborious, and time-consuming biochemical/enzymatic tests.
Collapse
Affiliation(s)
- C Nogueira
- Newborn Screening, Metabolism & Genetics Unit, Human Genetics Department, National Institute of Health Dr Ricardo Jorge, Porto, Portugal
| | - A Marcão
- Newborn Screening, Metabolism & Genetics Unit, Human Genetics Department, National Institute of Health Dr Ricardo Jorge, Porto, Portugal
| | - H Rocha
- Newborn Screening, Metabolism & Genetics Unit, Human Genetics Department, National Institute of Health Dr Ricardo Jorge, Porto, Portugal
| | - C Sousa
- Newborn Screening, Metabolism & Genetics Unit, Human Genetics Department, National Institute of Health Dr Ricardo Jorge, Porto, Portugal
| | - H Fonseca
- Newborn Screening, Metabolism & Genetics Unit, Human Genetics Department, National Institute of Health Dr Ricardo Jorge, Porto, Portugal
| | - C Valongo
- Newborn Screening, Metabolism & Genetics Unit, Human Genetics Department, National Institute of Health Dr Ricardo Jorge, Porto, Portugal
| | - L Vilarinho
- Newborn Screening, Metabolism & Genetics Unit, Human Genetics Department, National Institute of Health Dr Ricardo Jorge, Porto, Portugal
| |
Collapse
|
35
|
Yue WW. From structural biology to designing therapy for inborn errors of metabolism. J Inherit Metab Dis 2016; 39:489-98. [PMID: 27240455 PMCID: PMC4920855 DOI: 10.1007/s10545-016-9923-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 02/09/2016] [Accepted: 02/11/2016] [Indexed: 12/11/2022]
Abstract
At the SSIEM Symposium in Istanbul 2010, I presented an overview of protein structural approaches in the study of inborn errors of metabolism (Yue and Oppermann 2011). Five years on, the field is going strong with new protein structures, uncovered catalytic functions and novel chemical matters for metabolic enzymes, setting the stage for the next generation of drug discovery. This article aims to update on recent advances and lessons learnt on inborn errors of metabolism via the protein-centric approach, citing examples of work from my group, collaborators and co-workers that cover diverse pathways of transsulfuration, cobalamin and glycogen metabolism. Taking into consideration that many inborn errors of metabolism result in the loss of enzyme function, this presentation aims to outline three key principles that guide the design of small molecule therapy in this technically challenging field: (1) integrating structural, biochemical and cell-based data to evaluate the wide spectrum of mutation-driven enzyme defects in stability, catalysis and protein-protein interaction; (2) studying multi-domain proteins and multi-protein complexes as examples from nature, to learn how enzymes are activated by small molecules; (3) surveying different regions of the enzyme, away from its active site, that can be targeted for the design of allosteric activators and inhibitors.
Collapse
Affiliation(s)
- Wyatt W Yue
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, UK.
| |
Collapse
|
36
|
Mass Spectrometry-Based Metabolomic and Proteomic Strategies in Organic Acidemias. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9210408. [PMID: 27403441 PMCID: PMC4923558 DOI: 10.1155/2016/9210408] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 05/15/2016] [Indexed: 12/17/2022]
Abstract
Organic acidemias (OAs) are inherited metabolic disorders caused by deficiency of enzymatic activities in the catabolism of amino acids, carbohydrates, or lipids. These disorders result in the accumulation of mono-, di-, or tricarboxylic acids, generally referred to as organic acids. The OA outcomes can involve different organs and/or systems. Some OA disorders are easily managed if promptly diagnosed and treated, whereas, in others cases, such as propionate metabolism-related OAs (propionic acidemia, PA; methylmalonic acidemia, MMA), neither diet, vitamin therapy, nor liver transplantation appears to prevent multiorgan impairment. Here, we review the recent developments in dissecting molecular bases of OAs by using integration of mass spectrometry- (MS-) based metabolomic and proteomic strategies. MS-based techniques have facilitated the rapid and economical evaluation of a broad spectrum of metabolites in various body fluids, also collected in small samples, like dried blood spots. This approach has enabled the timely diagnosis of OAs, thereby facilitating early therapeutic intervention. Besides providing an overview of MS-based approaches most frequently used to study the molecular mechanisms underlying OA pathophysiology, we discuss the principal challenges of metabolomic and proteomic applications to OAs.
Collapse
|
37
|
|