1
|
Büchler LR, Blomgren LKM, Bürer C, Zanotelli VRT, Froese DS. Evidence for interaction of 5,10-methylenetetrahydrofolate reductase (MTHFR) with methylenetetrahydrofolate dehydrogenase (MTHFD1) and general control nonderepressible 1 (GCN1). Biochimie 2025; 230:138-146. [PMID: 39571719 DOI: 10.1016/j.biochi.2024.11.010] [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: 08/22/2024] [Revised: 11/07/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024]
Abstract
5,10-Methylenetetrahydrofolate reductase (MTHFR) is a folate cycle enzyme required for the intracellular synthesis of methionine. MTHFR was previously shown to be partially phosphorylated at 16 residues, which was abrogated by conversion of threonine 34 to alanine (T34A) or truncation of the first 37 amino acids (i.e. expression of amino acids 38-656), and promoted by methionine supplementation. Here, we over-expressed wild-type MTHFR (MTFHRWT), as well as the variants MTHFRT34A and MTHFR38-656 in 293T cells to provide further insights into these mechanisms. We demonstrate that following incubation in high methionine conditions (100-1000 μM) MTHFRWT is almost completely phosphorylated, but in methionine restricted conditions (0-10 μM) phosphorylation is reduced, while MTHFRT34A always remains unphosphorylated. Following affinity purification coupled mass spectrometry of an empty vector, MTHFRWT, MTHFRT34A and MTHFR38-656 in three separate experiments, we identified 134 proteins consistently pulled-down by all three MTHFR protein variants, of which 5 were indicated to be likely true interactors (SAINT prediction threshold of 0.95 and 2 fold-change). Amongst these were the folate cycle enzyme methylenetetrahydrofolate dehydrogenase (MTHFD1) and the amino acid starvation sensor general control nonderepressible 1 (GCN1). Immunoprecipitation-immunoblotting of MTHFRWT replicated interaction with both proteins. An AlphaFold 3 generated model of the MTHFR-MTHFD1 interaction places the MTHFD1 dehydrogenase/cyclohydrolase domain in direct contact with the MTHFR catalytic domain, suggesting their interaction may facilitate direct delivery of methylenetetrahydrofolate. Overall, we confirm methionine availability increases MTHFR phosphorylation, and identified potential interaction of MTHFR with MTHFD1 and GCN1.
Collapse
Affiliation(s)
- Linda R Büchler
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Linnea K M Blomgren
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Céline Bürer
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Vito R T Zanotelli
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - D Sean Froese
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
2
|
Sultan E, Pati D, Kumar S, Sahu BB. Arabidopsis METHYLENETETRAHYDROFOLATE REDUCTASE 2 functions independently of PENETRATION 2 during primary immunity against rice blast. JOURNAL OF EXPERIMENTAL BOTANY 2025; 76:1032-1048. [PMID: 39450434 DOI: 10.1093/jxb/erae435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
Non-host resistance (NHR) is the most durable and robust form of innate immunity, with a surge of interest in its role in crop improvement. Of the NHR genes identified against rice blast, a devastating disease caused by Magnaporthe oryzae, Arabidopsis PEN2 is indispensable for pre-penetration resistance to M. oryzae, while a consortium of genes orchestrates post-penetration resistance via lesser known mechanisms. We identified M. oryzae-susceptible mosA (mthfr2 pen2-3) from a randomly mutagenized Arabidopsis pen2-3 population using forward genetics. Analysis of T-DNA-inserted mthfr2 lines and pen2-3-complemented mosA lines revealed that MTHFR2-dependent resistance to M. oryzae is independent of PEN2. MTHFR2-defective plants exhibited higher accumulation of reactive oxygen species and expression of salicylic acid-dependent defense markers. MTHFR2-ligand docking revealed that A55V non-synonymous substitution in mosA altered ligand binding efficiency. This further affected the metabolomic profile of mosA, effectively allowing in vitro germination and development of M. oryzae conidia. Moreover, the loss-of-function mutation in mthfr2 (involved in the 1C metabolic pathway) potentiated mosA immunity against Pst DC3000. In conclusion, our findings showed that MTHFR2 is a positive modulator of NHR against M. oryzae. This work documents another layer of conserved yet divergent metabolomic defense in Arabidopsis regulated by folate-mediated 1C metabolism that has the potential to revolutionize crop improvement.
Collapse
Affiliation(s)
- Eram Sultan
- Department of Life Science, National Institute of Technology Rourkela, Odisha 769008, India
| | - Debasish Pati
- Department of Life Science, National Institute of Technology Rourkela, Odisha 769008, India
| | - Sanjeev Kumar
- Indian Agricultural Statistics Research Institute (ICAR-IASRI), Library Avenue, Pusa, New Delhi 110012, India
| | - Binod Bihari Sahu
- Department of Life Science, National Institute of Technology Rourkela, Odisha 769008, India
| |
Collapse
|
3
|
Singh J, Wilkins G, Goodman-Vincent E, Chishti S, Bonilla Guerrero R, McFadden L, Zahavi Z, Santosh P. Co-Occurring Methylenetetrahydrofolate Reductase ( MTHFR) rs1801133 and rs1801131 Genotypes as Associative Genetic Modifiers of Clinical Severity in Rett Syndrome. Brain Sci 2024; 14:624. [PMID: 39061365 PMCID: PMC11275218 DOI: 10.3390/brainsci14070624] [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: 05/17/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
AIM Remethylation disorders such as 5,10-methylenetetrahydrofolate reductase (MTHFR) deficiency reduce the remethylation of homocysteine to methionine. The resulting hyperhomocysteinemia can lead to serious neurological consequences and multisystem toxicity. The role of MTHFR genotypes has not been investigated in patients with Rett Syndrome (RTT). In this study, we sought to assess the impact of co-occurring MTHFR genotypes on symptom profiles in RTT. METHOD Using pharmacogenomic (PGx) testing, the MTHFR genetic polymorphisms rs1801133 (c.665C>T mutation) and rs1801131 (c.1286A>C mutation) were determined in 65 patients (18.7 years ± 12.1 [mean ± standard deviation]) with RTT as part of routine clinical care within the Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, a National and Specialist Child and Adolescent Mental Health Service (CAMHS) in the UK. The clinical severity of patients was assessed using the RTT-anchored Clinical Global Impression Scale (RTT-CGI). RESULTS The clinical severity symptom distribution varied between the homozygous and heterozygous MTHFR rs1801133 and rs1801131 genotypes. Those with the homozygous genotype had a narrower spread of severity scores across several domains (language and communication, ambulation, hand-use and eye contact clinical domains). Patients with the homozygous genotype had statistically significantly greater CGI-Severity scores than individuals with a non-homozygous MTHFR genotype (Z = -2.44, p = 0.015). When comparing the ratings of moderately impaired (4), markedly impaired (5), severely impaired (6) and extremely impaired (7), individuals with the homozygous MTHFR genotype were more impaired than those with the non-homozygous MTHFR genotype (Z = -2.06, p = 0.039). There was no statistically significant difference in the number of prescribed anti-epileptic drugs between the genotypes. CONCLUSIONS Our findings show that in those with a pathogenic RTT genetic variant, co-occurring homozygotic MTHFR rs1801133 and rs1801131 polymorphisms may act as associative genetic modifiers of clinical severity in a subset of patients. Profiling of rs1801133 and rs1801131 in RTT may therefore be useful, especially for high-risk patients who may be at the most risk from symptom deterioration.
Collapse
Affiliation(s)
- Jatinder Singh
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK; (J.S.); (G.W.); (E.G.-V.); (S.C.); (L.M.)
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD), South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
| | - Georgina Wilkins
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK; (J.S.); (G.W.); (E.G.-V.); (S.C.); (L.M.)
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD), South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
| | - Ella Goodman-Vincent
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK; (J.S.); (G.W.); (E.G.-V.); (S.C.); (L.M.)
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD), South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
| | - Samiya Chishti
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK; (J.S.); (G.W.); (E.G.-V.); (S.C.); (L.M.)
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD), South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
| | | | - Leighton McFadden
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK; (J.S.); (G.W.); (E.G.-V.); (S.C.); (L.M.)
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD), South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
| | - Zvi Zahavi
- Myogenes Limited, Borehamwood WD6 4PJ, UK;
| | - Paramala Santosh
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK; (J.S.); (G.W.); (E.G.-V.); (S.C.); (L.M.)
- Centre for Interventional Paediatric Psychopharmacology and Rare Diseases (CIPPRD), South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK
- Centre for Interventional Paediatric Psychopharmacology (CIPP) Rett Centre, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
| |
Collapse
|
4
|
Blomgren LKM, Huber M, Mackinnon SR, Bürer C, Baslé A, Yue WW, Froese DS, McCorvie TJ. Dynamic inter-domain transformations mediate the allosteric regulation of human 5, 10-methylenetetrahydrofolate reductase. Nat Commun 2024; 15:3248. [PMID: 38622112 PMCID: PMC11018872 DOI: 10.1038/s41467-024-47174-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 03/22/2024] [Indexed: 04/17/2024] Open
Abstract
5,10-methylenetetrahydrofolate reductase (MTHFR) commits folate-derived one-carbon units to generate the methyl-donor S-adenosyl-L-methionine (SAM). Eukaryotic MTHFR appends to the well-conserved catalytic domain (CD) a unique regulatory domain (RD) that confers feedback inhibition by SAM. Here we determine the cryo-electron microscopy structures of human MTHFR bound to SAM and its demethylated product S-adenosyl-L-homocysteine (SAH). In the active state, with the RD bound to a single SAH, the CD is flexible and exposes its active site for catalysis. However, in the inhibited state the RD pocket is remodelled, exposing a second SAM-binding site that was previously occluded. Dual-SAM bound MTHFR demonstrates a substantially rearranged inter-domain linker that reorients the CD, inserts a loop into the active site, positions Tyr404 to bind the cofactor FAD, and blocks substrate access. Our data therefore explain the long-distance regulatory mechanism of MTHFR inhibition, underpinned by the transition between dual-SAM and single-SAH binding in response to cellular methylation status.
Collapse
Affiliation(s)
- Linnea K M Blomgren
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, University of Zürich, Zürich, CH-8032, Switzerland
| | - Melanie Huber
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, University of Zürich, Zürich, CH-8032, Switzerland
| | - Sabrina R Mackinnon
- Biosciences Institute, The Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Céline Bürer
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, University of Zürich, Zürich, CH-8032, Switzerland
| | - Arnaud Baslé
- Biosciences Institute, The Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Wyatt W Yue
- Biosciences Institute, The Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
- Centre for Medicines Discovery, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, OX3 7DQ, UK.
| | - D Sean Froese
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, University of Zürich, Zürich, CH-8032, Switzerland.
| | - Thomas J McCorvie
- Biosciences Institute, The Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
5
|
Sito H, Tan SC. Genetic polymorphisms as potential pharmacogenetic biomarkers for platinum-based chemotherapy in non-small cell lung cancer. Mol Biol Rep 2024; 51:102. [PMID: 38217759 DOI: 10.1007/s11033-023-08915-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/08/2023] [Indexed: 01/15/2024]
Abstract
Platinum-based chemotherapy (PBC) is a widely used treatment for various solid tumors, including non-small cell lung cancer (NSCLC). However, its efficacy is often compromised by the emergence of drug resistance in patients. There is growing evidence that genetic variations may influence the susceptibility of NSCLC patients to develop resistance to PBC. Here, we provide a comprehensive overview of the mechanisms underlying platinum drug resistance and highlight the important role that genetic polymorphisms play in this process. This paper discussed the genetic variants that regulate DNA repair, cellular movement, drug transport, metabolic processing, and immune response, with a focus on their effects on response to PBC. The potential applications of these genetic polymorphisms as predictive indicators in clinical practice are explored, as are the challenges associated with their implementation.
Collapse
Affiliation(s)
- Hilary Sito
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| | - Shing Cheng Tan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| |
Collapse
|
6
|
Li J, Nian Y, Liu J, Yang M, Jin Y, Kang X, Xu H, Shang Z, Lin W. Identification of a Potential Antimycobacterial Drug Sensitizer Targeting a Flavin-Independent Methylenetetrahydrofolate Reductase. ACS OMEGA 2023; 8:38406-38417. [PMID: 37867661 PMCID: PMC10586308 DOI: 10.1021/acsomega.3c05021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/19/2023] [Indexed: 10/24/2023]
Abstract
The increasing antibiotic resistance of Mycobacterium tuberculosis and pathogenic nontuberculosis mycobacteria highlights the urgent need for new prevention and treatment strategies. Recently, the cocrystal structure of a Mycobacterium smegmatis flavin-independent 5,10-methylenetetrahydrofolate reductase (MsmMTHFR) that binds with a reduced nicotinamide adenine dinucleotide (NADH) has been well-determined, providing a structural basis for the screening of antimycobacterial leads targeting MsmMTHFR, a new enzyme involved in tetrahydrofolic acid (THF) biosynthesis. In this study, we identified compound AB131 as a promising candidate that fits well into the NADH binding pocket of MsmMTHFR through virtual screening. We discovered that AB131 and its derivatives (13 and 14) can sensitize the antimycobacterial activity of the antitubercular drug para-aminosalicyclic acid (PAS) by 2-5-fold against various species of mycobacteria. Although the compounds themselves do not exhibit any antimycobacterial activity, the high binding affinity of AB131 with MsmMTHFR or Rv2172c was evaluated by microscale thermophoresis analysis. Additionally, we predicted and validated the key residues (V115, V117, P118, and R163) of MsmMTHFR that are involved in the interaction with AB131 by using molecular docking and mutagenesis analysis. These findings offer a potential exploitable target for developing potent and specific antimycobacterial drug sensitizers.
Collapse
Affiliation(s)
- Jiacong Li
- Department
of Pathogen Biology, School of Medicine & Holistic Integrative
Medicine, Nanjing University of Chinese
Medicine, 210023 Nanjing, China
- School
of Pharmacy, Nanjing University of Chinese
Medicine, 210023 Nanjing, China
- State
Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 200237 Shanghai, China
| | - Yong Nian
- School
of Pharmacy, Nanjing University of Chinese
Medicine, 210023 Nanjing, China
| | - Jian Liu
- School
of Pharmacy, Nanjing University of Chinese
Medicine, 210023 Nanjing, China
| | - Mingxia Yang
- Department
of Pathogen Biology, School of Medicine & Holistic Integrative
Medicine, Nanjing University of Chinese
Medicine, 210023 Nanjing, China
- The
Center for Microbes, Development and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, 200031 Shanghai, China
| | - Yuanling Jin
- Department
of Pathogen Biology, School of Medicine & Holistic Integrative
Medicine, Nanjing University of Chinese
Medicine, 210023 Nanjing, China
| | - Xiaoman Kang
- CAS
Key Laboratory of Synthetic Biology, Centre
of Excellence for Molecular Plant Sciences, Chinese Academy of Sciences, 200032 Shanghai, China
| | - Haodong Xu
- School
of Pharmacy, Nanjing University of Chinese
Medicine, 210023 Nanjing, China
| | - Zhuo Shang
- School of
Pharmaceutical Sciences, Shandong University, 250100 Jinan, China
| | - Wei Lin
- Department
of Pathogen Biology, School of Medicine & Holistic Integrative
Medicine, Nanjing University of Chinese
Medicine, 210023 Nanjing, China
- School
of Pharmacy, Nanjing University of Chinese
Medicine, 210023 Nanjing, China
- State
Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 200237 Shanghai, China
- Jiangsu
Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, 210023 Nanjing, China
| |
Collapse
|
7
|
Ahmed S, Akbar F, DeBerardinis RJ, Ni M, Afroze B. Evaluation of the clinical, biochemical, and genetic presentation of neonatal and adult-onset 5,10-methylene tetrahydrofolate reductase (MTHFR) deficiency in patients from Pakistan. J Pediatr Endocrinol Metab 2023; 36:761-771. [PMID: 37440674 DOI: 10.1515/jpem-2023-0083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023]
Abstract
OBJECTIVES To study the biochemical, clinical and molecular characteristics of 5,10- methylenetetrahydrofolate reductase (MTHFR) deficiency in Pakistani patients from a single center. METHODS Medical charts, urine organic acid chromatograms, plasma methionine and Hcys levels, and molecular testing results of MTHFR gene of patients presenting at the Biochemical Genetics Clinic, AKUH from 2016 to 2022 were reviewed. RESULTS Neonatal MTHFR deficiency was found in five patients. The median (IQR) age of symptom onset and diagnosis were 18 (8.5-22) and 26 (16.5-31) days. The median lag between symptom onset and diagnosis was 8 (4.5-12.5) days. The median age of treatment initiation and duration of treatment were 26 (16.5-49) and 32 (25.5-54) days. The most common clinical features were lethargy, poor feeding, and seizures. The MTHFR gene sequencing revealed homozygous variants p.K510K, p.R567*, and p.R157W. Renal insufficiency manifesting as elevated serum creatinine and responding to betaine therapy was noted in one patient. This has not been previously reported in neonatal MTHFR deficiency and may reflect engagement of alternate pathways of remethylation. Adult onset MTHFR deficiency was found in six patients, with a heterogeneous neurological presentation. The median lag between symptoms onset and diagnosis was 7 (3-11) years. MTHFR gene sequencing revealed homozygous variant p.A195V in five patients from one family and p.G261V in the other. Two of the five reported variants are novel that include p.R157W and p.G261V. CONCLUSIONS Eleven patients of this rare disorder from a single center indicate the need for clinical awareness and appropriate biochemical evaluation to ensure optimal outcomes.
Collapse
Affiliation(s)
- Sibtain Ahmed
- Section of Chemical Pathology, Department of Pathology and Laboratory Medicine, Aga Khan University, Karachi, Pakistan
| | - Fizza Akbar
- Department of Paediatrics & Child Health, Aga Khan University Hospital, Karachi, Pakistan
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute at UT Southwestern, Dallas, TX, USA
- Howard Hughes Medical Institute, UT Southwestern, Dallas, TX, USA
| | - Min Ni
- Children's Medical Center Research Institute at UT Southwestern, Dallas, TX, USA
| | - Bushra Afroze
- Department of Paediatrics & Child Health, Aga Khan University Hospital, Karachi, Pakistan
| |
Collapse
|
8
|
Li J, Yang M, Li W, Lu C, Feng D, Shang Z, Wang C, Lin W. Structural and functional characterization of a mycobacterial methylenetetrahydrofolate reductase utilizing NADH as the exclusive cofactor. Biochem J 2023; 480:1129-1146. [PMID: 37435857 DOI: 10.1042/bcj20230138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/05/2023] [Accepted: 07/12/2023] [Indexed: 07/13/2023]
Abstract
5,10-Methylenetetraydrofolate reductase (MTHFR) is a key enzyme in folate metabolism. MSMEG_6649, a non-canonical MTHFR from Mycobacterium smegmatis, was previously reported as a monomeric protein lacking the flavin coenzyme. However, the structural basis for its unique flavin-independent catalytic mechanism remains poorly understood. Here, we determined the crystal structures of apo MTHFR MSMEG_6649 and its complex with NADH from M. smegmatis. Structural analysis revealed that the groove formed by the loops 4 and 5 of non-canonical MSMEG_6649 interacting with FAD was significantly larger than that of canonical MTHFR. Meanwhile, the NADH-binding site in MSMEG_6649 is highly similar to the FAD binding site in canonical MTHFR, suggesting that NADH plays the same role (immediate hydride donor for methylenetetraydrofolate) as FAD in the catalytic reaction. Using biochemical analysis, molecular modeling, and site-directed mutagenesis, the critical residues participating in the binding of NADH and the substrate 5,10-methylenetetrahydrofolate as well as the product 5-methyltetrahydrofolate were identified and validated. Taken together, this work not only provides a good starting point for understanding the potential catalytic mechanism for MSMEG_6649, but also identifies an exploitable target for the development of anti-mycobacterial drugs.
Collapse
Affiliation(s)
- Jiacong Li
- Department of Pathogen Biology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Mingxia Yang
- The Center for Microbes, Development and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Weijia Li
- Department of Pathogen Biology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chujie Lu
- Department of Pathogen Biology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Deyu Feng
- The Center for Microbes, Development and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Zhuo Shang
- School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - Chengyuan Wang
- The Center for Microbes, Development and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Wei Lin
- Department of Pathogen Biology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
9
|
Barretta F, Uomo F, Fecarotta S, Albano L, Crisci D, Verde A, Fisco MG, Gallo G, Dottore Stagna D, Pricolo MR, Alagia M, Terrone G, Rossi A, Parenti G, Ruoppolo M, Mazzaccara C, Frisso G. Contribution of Genetic Test to Early Diagnosis of Methylenetetrahydrofolate Reductase (MTHFR) Deficiency: The Experience of a Reference Center in Southern Italy. Genes (Basel) 2023; 14:genes14050980. [PMID: 37239340 DOI: 10.3390/genes14050980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND the deficiency of 5,10-Methylenetetrahydrofolate reductase (MTHFR) constitutes a rare and severe metabolic disease and is included in most expanded newborn screening (NBS) programs worldwide. Patients with severe MTHFR deficiency develop neurological disorders and premature vascular disease. Timely diagnosis through NBS allows early treatment, resulting in improved outcomes. METHODS we report the diagnostic yield of genetic testing for MTHFR deficiency diagnosis, in a reference Centre of Southern Italy between 2017 and 2022. MTHFR deficiency was suspected in four newborns showing hypomethioninemia and hyperhomocysteinemia; otherwise, one patient born in pre-screening era showed clinical symptoms and laboratory signs that prompted to perform genetic testing for MTHFR deficiency. RESULTS molecular analysis of the MTHFR gene revealed a genotype compatible with MTHFR deficiency in two NBS-positive newborns and in the symptomatic patient. This allowed for promptly beginning the adequate metabolic therapy. CONCLUSIONS our results strongly support the need for genetic testing to quickly support the definitive diagnosis of MTHFR deficiency and start therapy. Furthermore, our study extends knowledge of the molecular epidemiology of MTHFR deficiency by identifying a novel mutation in the MTHFR gene.
Collapse
Affiliation(s)
- Ferdinando Barretta
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
- CEINGE Advanced Biotechnologies Franco Salvatore, 80131 Naples, Italy
| | - Fabiana Uomo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Simona Fecarotta
- Metabolic Diseases Unit, Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, 80131 Naples, Italy
| | - Lucia Albano
- CEINGE Advanced Biotechnologies Franco Salvatore, 80131 Naples, Italy
| | - Daniela Crisci
- CEINGE Advanced Biotechnologies Franco Salvatore, 80131 Naples, Italy
| | - Alessandra Verde
- Metabolic Diseases Unit, Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, 80131 Naples, Italy
| | | | - Giovanna Gallo
- CEINGE Advanced Biotechnologies Franco Salvatore, 80131 Naples, Italy
| | - Daniela Dottore Stagna
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | | | - Marianna Alagia
- Metabolic Diseases Unit, Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, 80131 Naples, Italy
| | - Gaetano Terrone
- Metabolic Diseases Unit, Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, 80131 Naples, Italy
| | - Alessandro Rossi
- Metabolic Diseases Unit, Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, 80131 Naples, Italy
| | - Giancarlo Parenti
- Metabolic Diseases Unit, Department of Translational Medical Science, Section of Pediatrics, University of Naples Federico II, 80131 Naples, Italy
| | - Margherita Ruoppolo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
- CEINGE Advanced Biotechnologies Franco Salvatore, 80131 Naples, Italy
| | - Cristina Mazzaccara
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
- CEINGE Advanced Biotechnologies Franco Salvatore, 80131 Naples, Italy
| | - Giulia Frisso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
- CEINGE Advanced Biotechnologies Franco Salvatore, 80131 Naples, Italy
| |
Collapse
|
10
|
Lu Y, Zhao S, He X, Yang H, Wang X, Miao C, Liu H, Zhang X. Novel compound heterozygous mutations of MTHFR Gene in a Chinese family with homocystinuria due to MTHFR deficiency. BMC Med Genomics 2022; 15:271. [PMID: 36567323 PMCID: PMC9790122 DOI: 10.1186/s12920-022-01408-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 12/05/2022] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Homocystinuria due to methylenetetrahydrofolate reductase (MTHFR) deficiency is a rare autosomal recessive disorder. The purpose of this study is to expand the mutation site of the MTHFR gene and provide genetic counseling for this family. METHODS A couple came to our hospital for pre-pregnancy genetic counseling. We collected the family history and detailed clinical information, then performed whole-exome sequencing, and analyzed the pathogenicity of the candidate mutations. RESULTS We found that the father of the proband had homocystinuria, the proband and his brother had low blood methionine levels at birth, and the brain MRI showed brain dysplasia. The third fetus was found to have a broadened triangle of the bilateral ventricle at 19 weeks of pregnancy. The compound heterozygous variants of c.602 A > C (p.His201Pro) and c.1316T > C (p.Leu439Pro) of the MTHFR gene in the first three fetuses were found by whole-exome sequencing. The heterozygous c.602 A > C variant of the MTHFR gene is a novel missense variant that has been submitted to the ClinVar with Variation ID 992,662. CONCLUSION In consideration of the clinical phenotype, family history, and result of genetic testing, we speculated that both patients may have homocystinuria due to MTHFR deficiency. Homocystinuria due to MTHFR deficiency caused by compound heterozygous mutations composed of the MTHFR gene in this family may be associated with cerebral atrophy and cerebral dysplasia. The novel compound heterozygous mutations broaden the mutation spectrum of the MTHFR gene and enhance the application of genetic counseling and carrier screening in rare diseases.
Collapse
Affiliation(s)
- Yitong Lu
- Xi’an People’s Hospital (Xi’an Fourth Hospital), Xi’an, China
| | - Shaozhi Zhao
- Xi’an People’s Hospital (Xi’an Fourth Hospital), Xi’an, China
| | - Xiaohui He
- Xi’an People’s Hospital (Xi’an Fourth Hospital), Xi’an, China
| | - Hua Yang
- Xi’an People’s Hospital (Xi’an Fourth Hospital), Xi’an, China
| | - Xiaolei Wang
- Xi’an People’s Hospital (Xi’an Fourth Hospital), Xi’an, China
| | - Chen Miao
- Xi’an People’s Hospital (Xi’an Fourth Hospital), Xi’an, China
| | - Hongwei Liu
- Xi’an People’s Hospital (Xi’an Fourth Hospital), Xi’an, China
| | - Xinwen Zhang
- Xi’an People’s Hospital (Xi’an Fourth Hospital), Xi’an, China
| |
Collapse
|
11
|
Sun H, Song K, Zhou Y, Ding JF, Tu B, Yang JJ, Sha JM, Zhao JY, Zhang Y, Tao H. MTHFR epigenetic derepression protects against diabetes cardiac fibrosis. Free Radic Biol Med 2022; 193:330-341. [PMID: 36279972 DOI: 10.1016/j.freeradbiomed.2022.10.304] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/17/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Diabetes cardiac fibrosis is associated with altered DNA methylation of fibrogenic genes; however, the underlying mechanisms remain unclear. OBJECTIVES In this study, we investigate the critical role of DNA methylation aberration-associated suppression of MTHFR in diabetes cardiac fibrosis, and the protective effects of folate on diabetes cardiac fibrosis, using cultured cells, animal models, and clinical samples. METHODS AND RESULTS Herein, we report that DNA methylation repression of MTHFR, critically involved in diabetes cardiac fibrosis, mediates the significant protective effects of folate in a mouse model of diabetes cardiac fibrosis induced by STZ. Heart MTHFR expression was markedly suppressed in diabetes cardiac fibrosis patients and mice, accompanied by increased DNMT3A and MTHFR promoter methylation. Knockdown of DNMT3A demethylated MTHFR promoter, recovered the MTHFR loss, and alleviated the diabetes cardiac fibrosis pathology and cardiac fibroblasts pyroptosis. Mechanistically, DNMT3A epigenetically repressed MTHFR expression via methylation of the promoter. Interestingly, folate supplementation can rescue the effect of MTHFR loss in diabetes cardiac fibrosis, suggesting that inactivation of MTHFR through epigenetics is a critical mediator of diabetes cardiac fibrosis. CONCLUSIONS The current study identifies that MTHFR repression due to aberrant DNMT3A elevation and subsequent MTHFR promoter hypermethylation is likely an important epigenetic feature of diabetes cardiac fibrosis, and folate supplementation protects against diabetes cardiac fibrosis.
Collapse
Affiliation(s)
- He Sun
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China
| | - Kai Song
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China
| | - Yang Zhou
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China
| | - Ji-Fei Ding
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China; Department of Cardiothoracic Surgery, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, Jiangsu Province, PR China
| | - Bin Tu
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China
| | - Jing-Jing Yang
- Department of Clinical Pharmacy, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China
| | - Ji-Ming Sha
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China
| | - Jian-Yuan Zhao
- Institute for Developmental and Regenerative Cardiovascular Medicine, MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, PR China.
| | - Ye Zhang
- Department of Anesthesiology, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China.
| | - Hui Tao
- Department of Cardiothoracic Surgery, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China; Department of Anesthesiology, The Second Hospital of Anhui Medical University, Hefei, 230601, PR China.
| |
Collapse
|
12
|
Shifting landscapes of human MTHFR missense-variant effects. Am J Hum Genet 2021; 108:1283-1300. [PMID: 34214447 PMCID: PMC8322931 DOI: 10.1016/j.ajhg.2021.05.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 05/18/2021] [Indexed: 12/20/2022] Open
Abstract
Most rare clinical missense variants cannot currently be classified as pathogenic or benign. Deficiency in human 5,10-methylenetetrahydrofolate reductase (MTHFR), the most common inherited disorder of folate metabolism, is caused primarily by rare missense variants. Further complicating variant interpretation, variant impacts often depend on environment. An important example of this phenomenon is the MTHFR variant p.Ala222Val (c.665C>T), which is carried by half of all humans and has a phenotypic impact that depends on dietary folate. Here we describe the results of 98,336 variant functional-impact assays, covering nearly all possible MTHFR amino acid substitutions in four folinate environments, each in the presence and absence of p.Ala222Val. The resulting atlas of MTHFR variant effects reveals many complex dependencies on both folinate and p.Ala222Val. MTHFR atlas scores can distinguish pathogenic from benign variants and, among individuals with severe MTHFR deficiency, correlate with age of disease onset. Providing a powerful tool for understanding structure-function relationships, the atlas suggests a role for a disordered loop in retaining cofactor at the active site and identifies variants that enable escape of inhibition by S-adenosylmethionine. Thus, a model based on eight MTHFR variant effect maps illustrates how shifting landscapes of environment- and genetic-background-dependent missense variation can inform our clinical, structural, and functional understanding of MTHFR deficiency.
Collapse
|
13
|
Bezerra GA, Holenstein A, Foster WR, Xie B, Hicks KG, Bürer C, Lutz S, Mukherjee A, Sarkar D, Bhattacharya D, Rutter J, Talukdar A, Brown PJ, Luo M, Shi L, Froese DS, Yue WW. Identification of small molecule allosteric modulators of 5,10-methylenetetrahydrofolate reductase (MTHFR) by targeting its unique regulatory domain. Biochimie 2021; 183:100-107. [PMID: 33476699 PMCID: PMC8040968 DOI: 10.1016/j.biochi.2021.01.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 01/12/2021] [Accepted: 01/12/2021] [Indexed: 12/26/2022]
Abstract
The folate and methionine cycles, constituting one-carbon metabolism, are critical pathways for cell survival. Intersecting these two cycles, 5,10-methylenetetrahydrofolate reductase (MTHFR) directs one-carbon units from the folate to methionine cycle, to be exclusively used for methionine and S-adenosylmethionine (AdoMet) synthesis. MTHFR deficiency and upregulation result in diverse disease states, rendering it an attractive drug target. The activity of MTHFR is inhibited by the binding of AdoMet to an allosteric regulatory domain distal to the enzyme's active site, which we have previously identified to constitute a novel fold with a druggable pocket. Here, we screened 162 AdoMet mimetics using differential scanning fluorimetry, and identified 4 compounds that stabilized this regulatory domain. Three compounds were sinefungin analogues, closely related to AdoMet and S-adenosylhomocysteine (AdoHcy). The strongest thermal stabilisation was provided by (S)-SKI-72, a potent inhibitor originally developed for protein arginine methyltransferase 4 (PRMT4). Using surface plasmon resonance, we confirmed that (S)-SKI-72 binds MTHFR via its allosteric domain with nanomolar affinity. Assay of MTHFR activity in the presence of (S)-SKI-72 demonstrates inhibition of purified enzyme with sub-micromolar potency and endogenous MTHFR from HEK293 cell lysate in the low micromolar range, both of which are lower than AdoMet. Nevertheless, unlike AdoMet, (S)-SKI-72 is unable to completely abolish MTHFR activity, even at very high concentrations. Combining binding assays, kinetic characterization and compound docking, this work indicates the regulatory domain of MTHFR can be targeted by small molecules and presents (S)-SKI-72 as an excellent candidate for development of MTHFR inhibitors.
Collapse
Affiliation(s)
- Gustavo A Bezerra
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, OX3 7DQ, UK
| | - Alexander Holenstein
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, University of Zürich, Switzerland
| | - William R Foster
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, OX3 7DQ, UK
| | - Bing Xie
- Computational Chemistry and Molecular Biophysics Unit, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, USA
| | - Kevin G Hicks
- Department of Biochemistry, University of Utah School of Medicine, USA
| | - Céline Bürer
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, University of Zürich, Switzerland
| | - Seraina Lutz
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, University of Zürich, Switzerland
| | - Ayan Mukherjee
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, WB, India
| | - Dipika Sarkar
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, WB, India
| | - Debomita Bhattacharya
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, WB, India
| | - Jared Rutter
- Department of Biochemistry, University of Utah School of Medicine, USA
| | - Arindam Talukdar
- Department of Organic and Medicinal Chemistry, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata, 700032, WB, India
| | - Peter J Brown
- Structural Genomics Consortium, University of Toronto, Toronto, Ontario, M5G 1L7, Canada
| | - Minkui Luo
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Program of Pharmacology, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Lei Shi
- Computational Chemistry and Molecular Biophysics Unit, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, USA
| | - D Sean Froese
- Division of Metabolism and Children's Research Center, University Children's Hospital Zürich, University of Zürich, Switzerland.
| | - Wyatt W Yue
- Structural Genomics Consortium, Nuffield Department of Clinical Medicine, University of Oxford, OX3 7DQ, UK.
| |
Collapse
|
14
|
Massadeh S, Umair M, Alaamery M, Alfadhel M. A Novel Homozygous Non-sense Mutation in the Catalytic Domain of MTHFR Causes Severe 5,10-Methylenetetrahydrofolate Reductase Deficiency. Front Neurol 2019; 10:411. [PMID: 31068897 PMCID: PMC6491806 DOI: 10.3389/fneur.2019.00411] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 04/04/2019] [Indexed: 12/12/2022] Open
Abstract
Background: Severe 5,10-methylenetetrahydrofolate reductase (MTHFR) deficiency is a heterogeneous metabolic disorder inherited in an autosomal recessive manner. Pathogenic mutations in MTHFR gene have been associated with severe MTHFR deficiency. The clinical presentation of MTHFR deficiency is highly variable and associated with several neurological anomalies. Methods: Direct whole-exome sequencing (WES) was performed in all the five available individuals from the family, including the affected individual (III-7) using standard procedures. Results: We observed a proband (III-7) with an abnormality in the cerebral white matter, apnoea, and microcephaly. WES analysis identified a novel homozygous non-sense mutation (c.154C>T; p.Arg52*) in MTHFR gene that segregated with the disease phenotype within the family. Conclusion: We identified a novel non-sense mutation in MTHFR gene in a single Egyptian family with severe MTHFR deficiency. The present investigation is clinically important, as it adds to the growing list of MTHFR mutations, which might help in genetic counseling of families of affected children and proper genotype-phenotype correlation.
Collapse
Affiliation(s)
- Salam Massadeh
- Developmental Medicine Department, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.,Joint Centers of Excellence Program, KACST-BWH/Harvard Center of Excellence for Biomedicine, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Muhammad Umair
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Manal Alaamery
- Developmental Medicine Department, King Abdullah International Medical Research Center, King Saud Bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.,Joint Centers of Excellence Program, KACST-BWH/Harvard Center of Excellence for Biomedicine, King Abdulaziz City for Science and Technology (KACST), Riyadh, Saudi Arabia
| | - Majid Alfadhel
- Medical Genomics Research Department, King Abdullah International Medical Research Center (KAIMRC), King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia.,Division of Genetics, Department of Pediatrics, King Abdullah Specialized Children's Hospital, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| |
Collapse
|
15
|
Structural basis for the regulation of human 5,10-methylenetetrahydrofolate reductase by phosphorylation and S-adenosylmethionine inhibition. Nat Commun 2018; 9:2261. [PMID: 29891918 PMCID: PMC5995969 DOI: 10.1038/s41467-018-04735-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 05/16/2018] [Indexed: 11/08/2022] Open
Abstract
The folate and methionine cycles are crucial for biosynthesis of lipids, nucleotides and proteins, and production of the methyl donor S-adenosylmethionine (SAM). 5,10-methylenetetrahydrofolate reductase (MTHFR) represents a key regulatory connection between these cycles, generating 5-methyltetrahydrofolate for initiation of the methionine cycle, and undergoing allosteric inhibition by its end product SAM. Our 2.5 Å resolution crystal structure of human MTHFR reveals a unique architecture, appending the well-conserved catalytic TIM-barrel to a eukaryote-only SAM-binding domain. The latter domain of novel fold provides the predominant interface for MTHFR homo-dimerization, positioning the N-terminal serine-rich phosphorylation region near the C-terminal SAM-binding domain. This explains how MTHFR phosphorylation, identified on 11 N-terminal residues (16 in total), increases sensitivity to SAM binding and inhibition. Finally, we demonstrate that the 25-amino-acid inter-domain linker enables conformational plasticity and propose it to be a key mediator of SAM regulation. Together, these results provide insight into the molecular regulation of MTHFR. The human enzyme MTHFR links the folate and methionine cycles, which are essential for the biosynthesis of nucleotides and proteins. Here, the authors present the crystal structure and biochemical analysis of human MTHFR, providing molecular insights into its function and regulation in higher eukaryotes.
Collapse
|
16
|
García-Giménez JL, Romá-Mateo C, Pérez-Machado G, Peiró-Chova L, Pallardó FV. Role of glutathione in the regulation of epigenetic mechanisms in disease. Free Radic Biol Med 2017; 112:36-48. [PMID: 28705657 DOI: 10.1016/j.freeradbiomed.2017.07.008] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 06/29/2017] [Accepted: 07/06/2017] [Indexed: 12/14/2022]
Abstract
Epigenetics is a rapidly growing field that studies gene expression modifications not involving changes in the DNA sequence. Histone H3, one of the basic proteins in the nucleosomes that make up chromatin, is S-glutathionylated in mammalian cells and tissues, making Gamma-L-glutamyl-L-cysteinylglycine, glutathione (GSH), a physiological antioxidant and second messenger in cells, a new post-translational modifier of the histone code that alters the structure of the nucleosome. However, the role of GSH in the epigenetic mechanisms likely goes beyond a mere structural function. Evidence supports the hypothesis that there is a link between GSH metabolism and the control of epigenetic mechanisms at different levels (i.e., substrate availability, enzymatic activity for DNA methylation, changes in the expression of microRNAs, and participation in the histone code). However, little is known about the molecular pathways by which GSH can control epigenetic events. Studying mutations in enzymes involved in GSH metabolism and the alterations of the levels of cofactors affecting epigenetic mechanisms appears challenging. However, the number of diseases induced by aberrant epigenetic regulation is growing, so elucidating the intricate network between GSH metabolism, oxidative stress and epigenetics could shed light on how their deregulation contributes to the development of neurodegeneration, cancer, metabolic pathologies and many other types of diseases.
Collapse
Affiliation(s)
- José Luis García-Giménez
- Center for Biomedical Network Research on Rare Diseases (CIBERER) Institute of Health Carlos III, Valencia, Spain; Mixed Unit INCLIVA-CIPF Research Institutes, Valencia, Spain; Dept. Physiology, School of Medicine and Dentistry, Universitat de València (UV), Valencia, Spain; Epigenetics Research Platform (CIBERER/UV), Valencia, Spain.
| | - Carlos Romá-Mateo
- Center for Biomedical Network Research on Rare Diseases (CIBERER) Institute of Health Carlos III, Valencia, Spain; Mixed Unit INCLIVA-CIPF Research Institutes, Valencia, Spain; Dept. Physiology, School of Medicine and Dentistry, Universitat de València (UV), Valencia, Spain; Epigenetics Research Platform (CIBERER/UV), Valencia, Spain; Faculty of Biomedicine and Health Sciences, Universidad Europea de Valencia, Valencia, Spain
| | - Gisselle Pérez-Machado
- Dept. Physiology, School of Medicine and Dentistry, Universitat de València (UV), Valencia, Spain; Epigenetics Research Platform (CIBERER/UV), Valencia, Spain
| | | | - Federico V Pallardó
- Center for Biomedical Network Research on Rare Diseases (CIBERER) Institute of Health Carlos III, Valencia, Spain; Mixed Unit INCLIVA-CIPF Research Institutes, Valencia, Spain; Dept. Physiology, School of Medicine and Dentistry, Universitat de València (UV), Valencia, Spain; Epigenetics Research Platform (CIBERER/UV), Valencia, Spain.
| |
Collapse
|
17
|
Nowak I, Bylińska A, Wilczyńska K, Wiśniewski A, Malinowski A, Wilczyński JR, Radwan P, Radwan M, Barcz E, Płoski R, Motak-Pochrzęst H, Banasik M, Sobczyński M, Kuśnierczyk P. The methylenetetrahydrofolate reductase c.c.677 C>T and c.c.1298 A>C polymorphisms in reproductive failures: Experience from an RSA and RIF study on a Polish population. PLoS One 2017; 12:e0186022. [PMID: 29073227 PMCID: PMC5657620 DOI: 10.1371/journal.pone.0186022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 09/22/2017] [Indexed: 12/21/2022] Open
Abstract
Almost 1600 individuals from the Polish population were recruited to this study. Among them 319 were fertile couples, 289 were recurrent spontaneous abortion (RSA) couples, and 131 were in the group of recurrent implantation failure (RIF) following in vitro fertilization. The aim of this study was to evaluate the MTHFR c.c.677 C>T and c.c.1298 A>C polymorphisms’ association with RSA and RIF. We used PCR-RFLP with HinfI (677 C>T) and MboII (1298 A>C) digestion. We observed a protective effect of the female AC genotype (OR = 0.64, p = 0.01) and the C allele (AC+CC genotypes; OR = 0.65, p = 0.009) against RSA. Moreover, 1298 AA/677 CT women were more frequent in RSA (31.14%) and RIF (25.20%) groups in comparison to fertile women (22.88%), although this difference was significant only in the case of RSA (p = 0.022, OR = 1.52). Male combined genotype analysis revealed no association with reproductive failure of their partners. Nevertheless, the female/male combination AA/AC of the 1298 polymorphism was more frequent in RSA couples (p = 0.049, OR = 1.49). However, the significant results became insignificant after Bonferroni correction. In addition, analysis of haplotypes showed significantly higher frequency of the C/C haplotype (1298 C/677 C) in the female control group than in the female RSA group (p = 0.03, OR = 0.77). Moreover, the association between elevated homocysteine (Hcy) level in plasma of RSA and RIF women and MTHFR polymorphisms was investigated but did not reveal significant differences. In conclusion, for clinical practice, it is better to check the homocysteine level in plasma and, if the Hcy level is increased, to recommend patients to take folic acid supplements rather than undergo screening of MTHFR for 1298 A>C and 677 C>T polymorphisms.
Collapse
Affiliation(s)
- Izabela Nowak
- Department of Clinical Immunology, Laboratory of Immunogenetics and Tissue Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
- * E-mail:
| | - Aleksandra Bylińska
- Department of Clinical Immunology, Laboratory of Immunogenetics and Tissue Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Karolina Wilczyńska
- Department of Clinical Immunology, Laboratory of Immunogenetics and Tissue Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Andrzej Wiśniewski
- Department of Clinical Immunology, Laboratory of Immunogenetics and Tissue Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Andrzej Malinowski
- Department of Surgical, Endoscopic and Oncologic Gynecology, Polish Mothers’ Memorial Hospital–Research Institute, Łódź, Poland
| | - Jacek R. Wilczyński
- Department of Gynecology and Gynecologic Oncology, Polish Mothers’ Memorial Hospital–Research Institute, Łódź, Poland
| | - Paweł Radwan
- Department of Reproductive Medicine, Gameta Hospital, Rzgów, Poland
- Biogeno–Regional Science-Technology Center, Podzamcze, Poland
| | - Michał Radwan
- Department of Reproductive Medicine, Gameta Hospital, Rzgów, Poland
| | - Ewa Barcz
- First Chair and Clinic of Obstetrics and Gynecology, Medical University of Warsaw, Warszawa, Poland
| | - Rafał Płoski
- Department of Medical Genetics, Center of Biostructure Research, Medical University of Warsaw, Warszawa, Poland
| | - Hanna Motak-Pochrzęst
- Faculty of Physical Education and Physiotherapy, Opole University of Technology, Opole, Poland
- Obstetric Gynecological Department, District Hospital Strzelce Opolskie, Strzelce Opolskie, Poland
| | | | - Maciej Sobczyński
- Department of Genomics, Faculty of Biotechnology, University of Wrocław, Wrocław, Poland
| | - Piotr Kuśnierczyk
- Department of Clinical Immunology, Laboratory of Immunogenetics and Tissue Immunology, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| |
Collapse
|
18
|
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
|