1
|
Balduzzi E, Yin W, Lambry JC, Myllykallio H, Aleksandrov A. Additive CHARMM Force Field for Pterins and Folates. J Comput Chem 2025; 46:e27548. [PMID: 39710837 DOI: 10.1002/jcc.27548] [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: 05/31/2024] [Revised: 08/06/2024] [Accepted: 11/15/2024] [Indexed: 12/24/2024]
Abstract
Folates comprise a crucial class of biologically active compounds related to folic acid, playing a vital role in numerous enzymatic reactions. One-carbon metabolism, facilitated by the folate cofactor, supports numerous physiological processes, including biosynthesis, amino acid homeostasis, epigenetic maintenance, and redox defense. Folates share a common pterin heterocyclic ring structure capable of undergoing redox reactions and existing in various protonation states. This study aimed to derive molecular mechanics (MM) parameters compatible with the CHARMM36 all-atom additive force field for pterins and biologically important folates, including pterin, biopterin, and folic acid. Three redox forms were considered: oxidized, dihydrofolate, and tetrahydrofolate states. Across all protonation states, a total of 18 folates were parameterized. Partial charges were derived using the CHARMM force field parametrization protocol, based on targeting reference quantum mechanics monohydrate interactions, electrostatic potential, and dipole moment. Bonded terms were parameterized using one-dimensional adiabatic potential energy surface scans, and two-dimensional scans to parametrize in-ring torsions associated with the puckering states of dihydropterin and tetrahydropterin. The quality of the model was demonstrated through simulations of three protein complexes using optimized and initial parameters. These simulations underscored the significantly enhanced performance of the folate model developed in this study compared to the initial model without optimization in reproducing structural properties of folate-protein complexes. Overall, the presented MM model will be valuable for modeling folates in various redox states and serve as a starting point for parameterizing other folate derivatives.
Collapse
Affiliation(s)
- Elsa Balduzzi
- Laboratoire d'Optique et Biosciences (CNRS UMR7645, INSERM U1182), Ecole Polytechnique, Institut polytechnique de Paris, Palaiseau, France
| | - Wenlu Yin
- Laboratoire d'Optique et Biosciences (CNRS UMR7645, INSERM U1182), Ecole Polytechnique, Institut polytechnique de Paris, Palaiseau, France
| | - Jean-Christophe Lambry
- Laboratoire d'Optique et Biosciences (CNRS UMR7645, INSERM U1182), Ecole Polytechnique, Institut polytechnique de Paris, Palaiseau, France
| | - Hannu Myllykallio
- Laboratoire d'Optique et Biosciences (CNRS UMR7645, INSERM U1182), Ecole Polytechnique, Institut polytechnique de Paris, Palaiseau, France
| | - Alexey Aleksandrov
- Laboratoire d'Optique et Biosciences (CNRS UMR7645, INSERM U1182), Ecole Polytechnique, Institut polytechnique de Paris, Palaiseau, France
| |
Collapse
|
2
|
Ayoub G. Neurodevelopment of Autism: Critical Periods, Stress and Nutrition. Cells 2024; 13:1968. [PMID: 39682717 DOI: 10.3390/cells13231968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disability that presents significant challenges in communication and behavior. ASD prevalence exceeds 2% among eight-year-old children and is at similar levels globally. We propose that critical periods during fetal development and early postnatal years establish the conditions for either neurotypical development or the emergence of autism through mechanisms that influence immune function or delay neuronal development. One critical period is characterized by the requirement for folate, a crucial methyl donor needed for DNA regulation. Insufficient folate availability has been linked to the risk of developing ASD. Another critical period may be affected by oxidative stress or inflammation of the fetal brain, potentially due to inadequate microglial immunity, which can lead to CNS inflammatory changes that disrupt typical neurodevelopment. We suggest that early supplementation with reduced folate and taurine during both the fetal and postnatal stages may be effective in mitigating the severity of ASD symptoms by promoting neurotypical development through these critical neurodevelopmental periods.
Collapse
Affiliation(s)
- George Ayoub
- Psychology, Santa Barbara City College, Santa Barbara, CA 93109, USA
| |
Collapse
|
3
|
James RL, Sisserson T, Cai Z, Dumas ME, Inge LJ, Ranger-Moore J, Mason A, Sloss CM, McArthur K. Development of an FRα Companion Diagnostic Immunohistochemical Assay for Mirvetuximab Soravtansine. Arch Pathol Lab Med 2024; 148:1226-1233. [PMID: 38282564 DOI: 10.5858/arpa.2023-0149-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/17/2023] [Indexed: 01/30/2024]
Abstract
CONTEXT.— Folate receptor-α (FRα, encoded by the FOLR1 gene) is overexpressed in several solid tumor types, including epithelial ovarian cancer (EOC), making it an attractive biomarker and target for FRα-based therapy in ovarian cancer. OBJECTIVE.— To describe the development, analytic verification, and clinical performance of the VENTANA FOLR1 Assay (Ventana Medical Systems Inc) in EOC. DESIGN.— We used industry standard studies to establish the analytic verification of the VENTANA FOLR1 Assay. Furthermore, the VENTANA FOLR1 Assay was used in the ImmunoGen Inc-sponsored SORAYA study to select patients for treatment with mirvetuximab soravtansine (MIRV) in platinum-resistant EOC. RESULTS.— The VENTANA FOLR1 Assay is highly reproducible, demonstrated by a greater than 98% overall percent agreement (OPA) for repeatability and intermediate precision studies, greater than 93% OPA for interreader and greater than 96% for intrareader studies, and greater than 90% OPA across all observations in the interlaboratory reproducibility study. The performance of the VENTANA FOLR1 Assay in the SORAYA study was evaluated by the overall staining acceptability rate, which was calculated using the number of patient specimens that were tested with the VENTANA FOLR1 Assay that had an evaluable result. In the SORAYA trial, data in patients who received MIRV demonstrated clinically meaningful efficacy, and the overall staining acceptability rate of the assay was 98.4%, demonstrating that the VENTANA FOLR1 Assay is safe and effective for selecting patients who may benefit from MIRV. Together, these data showed that the assay is highly reliable, consistently producing evaluable results in the clinical setting. CONCLUSIONS.— The VENTANA FOLR1 Assay is a robust and reproducible assay for detecting FRα expression and identifying a patient population that derived clinically meaningful benefit from MIRV in the SORAYA study.
Collapse
Affiliation(s)
- Racheal L James
- From the Departments of Assay Development (James, Sisserson), Clinical Development & Medical Affairs (Cai, Dumas, Inge, Ranger-Moore, Mason), and Oncology (McArthur), Personalized Healthcare Solutions, Roche Tissue Diagnostics, Tucson, Arizona
| | - Taryn Sisserson
- From the Departments of Assay Development (James, Sisserson), Clinical Development & Medical Affairs (Cai, Dumas, Inge, Ranger-Moore, Mason), and Oncology (McArthur), Personalized Healthcare Solutions, Roche Tissue Diagnostics, Tucson, Arizona
| | - Zhuangyu Cai
- From the Departments of Assay Development (James, Sisserson), Clinical Development & Medical Affairs (Cai, Dumas, Inge, Ranger-Moore, Mason), and Oncology (McArthur), Personalized Healthcare Solutions, Roche Tissue Diagnostics, Tucson, Arizona
| | - Megan E Dumas
- From the Departments of Assay Development (James, Sisserson), Clinical Development & Medical Affairs (Cai, Dumas, Inge, Ranger-Moore, Mason), and Oncology (McArthur), Personalized Healthcare Solutions, Roche Tissue Diagnostics, Tucson, Arizona
| | - Landon J Inge
- From the Departments of Assay Development (James, Sisserson), Clinical Development & Medical Affairs (Cai, Dumas, Inge, Ranger-Moore, Mason), and Oncology (McArthur), Personalized Healthcare Solutions, Roche Tissue Diagnostics, Tucson, Arizona
| | - James Ranger-Moore
- From the Departments of Assay Development (James, Sisserson), Clinical Development & Medical Affairs (Cai, Dumas, Inge, Ranger-Moore, Mason), and Oncology (McArthur), Personalized Healthcare Solutions, Roche Tissue Diagnostics, Tucson, Arizona
| | - Albert Mason
- From the Departments of Assay Development (James, Sisserson), Clinical Development & Medical Affairs (Cai, Dumas, Inge, Ranger-Moore, Mason), and Oncology (McArthur), Personalized Healthcare Solutions, Roche Tissue Diagnostics, Tucson, Arizona
| | - Callum M Sloss
- the Department of Translational Sciences, ImmunoGen Inc, Waltham, Massachusetts (Sloss)
| | - Katherine McArthur
- From the Departments of Assay Development (James, Sisserson), Clinical Development & Medical Affairs (Cai, Dumas, Inge, Ranger-Moore, Mason), and Oncology (McArthur), Personalized Healthcare Solutions, Roche Tissue Diagnostics, Tucson, Arizona
| |
Collapse
|
4
|
Weinkove D. Folates, bacteria and ageing: insights from the model organism C. elegans in the study of nutrition and ageing. Proc Nutr Soc 2024:1-5. [PMID: 39439268 PMCID: PMC7617194 DOI: 10.1017/s0029665124004890] [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] [Indexed: 10/25/2024]
Abstract
The relationship between nutrition and ageing is complex. The metabolism and synthesis of micronutrients within the gut microbiome can influence human health but is challenging to study. Furthermore, studying ageing in humans is time-consuming and difficult to control for environmental factors. Studies in model organisms can guide research efforts in this area. This review describes how the nematode Caenorhabditis elegans can be used to study how bacteria and diet influence ageing and inform follow-on studies in humans. It is known that certain bacteria accelerate ageing in C. elegans. This age-accelerating effect is prevented by inhibiting folate synthesis within the bacteria, and we propose that in the human microbiome, certain bacteria also accelerate ageing in a way that can be modulated by interfering with bacterial folate synthesis. Bacterial-derived folates do not promote ageing themselves; rather, ageing is accelerated by bacteria in some way, either through secondary metabolites or other bacterial activity, which is dependent on bacterial folate synthesis. In humans, it may be possible to inhibit bacterial folate synthesis in the human gut while maintaining healthy folate status in the body via food and supplementation. The supplement form of folic acid has a common breakdown product that can be used by bacteria to increase folate synthesis. Thus, supplementation with folic acid may not be good for health in certain circumstances such as in older people or those with an excess of proteobacteria in their microbiome. For these groups, alternative supplement strategies may be a safer way to ensure adequate folate levels.
Collapse
Affiliation(s)
- David Weinkove
- Department of Biosciences, Durham University, DurhamDH1 3LE, UK
| |
Collapse
|
5
|
Blank HM, Hammer SE, Boatright L, Roberts C, Heyden KE, Nagarajan A, Tsuchiya M, Brun M, Johnson CD, Stover PJ, Sitcheran R, Kennedy BK, Adams LG, Kaeberlein M, Field MS, Threadgill DW, Andrews-Polymenis HL, Polymenis M. Late-life dietary folate restriction reduces biosynthesis without compromising healthspan in mice. Life Sci Alliance 2024; 7:e202402868. [PMID: 39043420 PMCID: PMC11266815 DOI: 10.26508/lsa.202402868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 07/25/2024] Open
Abstract
Folate is a vitamin required for cell growth and is present in fortified foods in the form of folic acid to prevent congenital abnormalities. The impact of low-folate status on life-long health is poorly understood. We found that limiting folate levels with the folate antagonist methotrexate increased the lifespan of yeast and worms. We then restricted folate intake in aged mice and measured various health metrics, metabolites, and gene expression signatures. Limiting folate intake decreased anabolic biosynthetic processes in mice and enhanced metabolic plasticity. Despite reduced serum folate levels in mice with limited folic acid intake, these animals maintained their weight and adiposity late in life, and we did not observe adverse health outcomes. These results argue that the effectiveness of folate dietary interventions may vary depending on an individual's age and sex. A higher folate intake is advantageous during the early stages of life to support cell divisions needed for proper development. However, a lower folate intake later in life may result in healthier aging.
Collapse
Affiliation(s)
- Heidi M Blank
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, USA
| | - Staci E Hammer
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, USA
| | - Laurel Boatright
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, USA
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Courtney Roberts
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, USA
| | - Katarina E Heyden
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Aravindh Nagarajan
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
- Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX, USA
| | - Mitsuhiro Tsuchiya
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Marcel Brun
- Texas A&M Agrilife Research, Genomics and Bioinformatics Service, College Station, TX, USA
| | - Charles D Johnson
- Texas A&M Agrilife Research, Genomics and Bioinformatics Service, College Station, TX, USA
| | - Patrick J Stover
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, USA
- Institute for Advancing Health Through Agriculture, Texas A&M University, College Station, TX, USA
- Department of Nutrition, Texas A&M University, College Station, TX, USA
| | - Raquel Sitcheran
- Department of Cell Biology and Genetics, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
| | - Brian K Kennedy
- Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - L Garry Adams
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M, College Station, TX, USA
| | - Matt Kaeberlein
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Optispan, Inc., Seattle, WA, USA
| | - Martha S Field
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - David W Threadgill
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, USA
- Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX, USA
- Department of Nutrition, Texas A&M University, College Station, TX, USA
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, TX, USA
| | - Helene L Andrews-Polymenis
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University Health Science Center, Bryan, TX, USA
- Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX, USA
| | - Michael Polymenis
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, USA
- Interdisciplinary Program in Genetics, Texas A&M University, College Station, TX, USA
- Institute for Advancing Health Through Agriculture, Texas A&M University, College Station, TX, USA
| |
Collapse
|
6
|
Alampi JD, Lanphear BP, MacFarlane AJ, Oulhote Y, Braun JM, Muckle G, Arbuckle TE, Ashley-Martin J, Hu JM, Chen A, McCandless LC. Combined Exposure to Folate and Lead during Pregnancy and Autistic-Like Behaviors among Canadian Children from the MIREC Pregnancy and Birth Cohort. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:107003. [PMID: 39412272 PMCID: PMC11481933 DOI: 10.1289/ehp14479] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 08/29/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND Folic acid (FA) supplementation may attenuate the associations between gestational exposure to certain chemicals and autism or autistic-like behaviors, but to our knowledge, this has not been assessed for lead. OBJECTIVES We examined whether the relationship between gestational blood-lead levels (BLLs) and autistic-like behaviors was modified by gestational plasma total folate concentrations, FA supplementation, and maternal methylenetetrahydrofolate reductase (MTHFR) 677C>T genotype. METHODS We used data from the Maternal-Infant Research on Environmental Chemicals study (2008-2011), a Canadian pregnancy and birth cohort study. Childhood autistic-like behaviors were documented in 601 children 3-4 y of age with the Social Responsiveness Scale-2 (SRS-2), where higher scores denote more autistic-like behaviors. We measured BLLs and plasma total folate concentrations during the first and third trimesters of pregnancy. We also estimated gestational FA supplementation via surveys and genotyped the maternal MTHFR 677C>T single nucleotide polymorphism (SNP). We estimated the confounder-adjusted associations between log 2 -transformed BLLs and SRS-2 scores by two indicators of folate exposure and maternal MTHFR 677C>T genotype using linear regression. RESULTS Third-trimester BLLs were associated with increased SRS-2 scores [β a d j = 3.3 ; 95% confidence interval (CI): 1.1, 5.5] among participants with low (< 10 th percentile), third-trimester, plasma total folate concentrations, but BLL-SRS-2 associations were null (β a d j = - 0.3 ; 95% CI: - 1.2 , 0.5) among those in the middle category (≥ 10 th and < 80 th percentiles) (p-interaction < 0.001 ). FA supplementation also attenuated these associations. Both folate indicators modified first-trimester BLL-SRS-2 associations, but to a lesser extent. Third-trimester BLL-SRS-2 associations were slightly stronger among participants who were homozygous for the T (minor) allele of the MTHFR 677C>T SNP (β a d j = 0.9 ; 95% CI: - 1.2 , 3.1) than those without the T allele (β a d j = - 0.3 ; 95% CI: - 1.3 , 0.7), but the difference was not statistically significant (p -interaction = 0.28 ). DISCUSSION Folate may modify the associations between gestational lead exposure and childhood autistic-like behaviors, suggesting that it mitigates the neurotoxic effects of prenatal lead exposure. https://doi.org/10.1289/EHP14479.
Collapse
Affiliation(s)
- Joshua D. Alampi
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Bruce P. Lanphear
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Amanda J. MacFarlane
- Texas A&M Agriculture, Food, and Nutrition Evidence Center, Fort Worth, Texas, USA
| | - Youssef Oulhote
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Joseph M. Braun
- Department of Epidemiology, Brown University, Providence, Rhode Island, USA
| | - Gina Muckle
- Centre Hospitalier Universitaire de Québec Research Centre and School of Psychology, Laval University, Québec City, Québec, Canada
| | - Tye E. Arbuckle
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario, Canada
| | - Jillian Ashley-Martin
- Environmental Health Science and Research Bureau, Healthy Environments and Consumer Safety Branch, Health Canada, Ottawa, Ontario, Canada
| | - Janice M.Y. Hu
- Faculty of Health Sciences, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Aimin Chen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
7
|
Long H, Liu M, Rao Z, Guan S, Chen X, Huang X, Cao L, Han R. RNA-Seq-Based Transcriptome Analysis of Chinese Cordyceps Aqueous Extracts Protective Effect against Adriamycin-Induced mpc5 Cell Injury. Int J Mol Sci 2024; 25:10352. [PMID: 39408685 PMCID: PMC11476491 DOI: 10.3390/ijms251910352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/17/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Pharmacogenomic analysis based on drug transcriptome characteristics is widely used to identify mechanisms of action. The purpose of this study was to elucidate the molecular mechanism of protective effect against adriamycin (ADM)-induced mpc5 cell injury of Chinese cordyceps aqueous extracts (WCCs) by a systematic transcriptomic analysis. The phytochemicals of WCCs were analyzed via the "phenol-sulfuric acid method", high-performance liquid chromatography (HPLC), and HPLC-mass spectrometry (MS). We analyzed the drug-reaction transcriptome profiles of mpc5 cell after treating them with WCCs. RNA-seq analysis revealed that WCCs alleviated ADM-induced mpc5 cell injury via restoring the expression of certain genes to normal level mainly in the one-carbon pool by the folate pathway, followed by the relaxin, apelin, PI3K-Akt, and nucleotide-binding, oligomerization domain (NOD)-like receptor signaling pathway, enhancing DNA synthesis and repair, cell proliferation, fibrosis reduction, and immune regulation. Otherwise, WCCs also modulated the proliferation and survival of the mpc5 cell by regulating metabolic pathways, and partially restores the expression of genes related to human disease pathways. These findings provide an innovative understanding of the molecular mechanism of the protective effect of WCCs on ADM-induced mpc5 cell injury at the molecular transcription level, and Mthfd2, Dhfr, Atf4, Creb5, Apln, and Serpine1, etc., may be potential novel targets for treating nephrotic syndrome.
Collapse
Affiliation(s)
- Hailin Long
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China; (H.L.); (M.L.); (Z.R.); (X.H.); (L.C.)
| | - Mengzhen Liu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China; (H.L.); (M.L.); (Z.R.); (X.H.); (L.C.)
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Zhongchen Rao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China; (H.L.); (M.L.); (Z.R.); (X.H.); (L.C.)
| | - Shanyue Guan
- Instrumental Analysis and Research Center, Sun Yat-sen University, Guangzhou 510275, China;
| | - Xiaotian Chen
- Center for Industrial Analysis and Testing, Guangdong Academy of Sciences, Guangzhou 510650, China;
| | - Xiaoting Huang
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China; (H.L.); (M.L.); (Z.R.); (X.H.); (L.C.)
| | - Li Cao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China; (H.L.); (M.L.); (Z.R.); (X.H.); (L.C.)
| | - Richou Han
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510260, China; (H.L.); (M.L.); (Z.R.); (X.H.); (L.C.)
| |
Collapse
|
8
|
Suehs BA, Gatlin DM, Wu G. Glycine nutrition and biochemistry from an aquaculture perspective. Anim Front 2024; 14:17-23. [PMID: 39246842 PMCID: PMC11377068 DOI: 10.1093/af/vfae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/10/2024] Open
Affiliation(s)
- Blaine A Suehs
- Department of Ecology and Conservation Biology, Texas A&M University, College Station, TX 77843, USA
| | - Delbert M Gatlin
- Department of Ecology and Conservation Biology, Texas A&M University, College Station, TX 77843, USA
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
9
|
Lee CM, Hwang Y, Kim M, Park YC, Kim H, Fang S. PHGDH: a novel therapeutic target in cancer. Exp Mol Med 2024; 56:1513-1522. [PMID: 38945960 PMCID: PMC11297271 DOI: 10.1038/s12276-024-01268-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/18/2024] [Accepted: 04/01/2024] [Indexed: 07/02/2024] Open
Abstract
Serine is a key contributor to the generation of one-carbon units for DNA synthesis during cellular proliferation. In addition, it plays a crucial role in the production of antioxidants that prevent abnormal proliferation and stress in cancer cells. In recent studies, the relationship between cancer metabolism and the serine biosynthesis pathway has been highlighted. In this context, 3-phosphoglycerate dehydrogenase (PHGDH) is notable as a key enzyme that functions as the primary rate-limiting enzyme in the serine biosynthesis pathway, facilitating the conversion of 3-phosphoglycerate to 3-phosphohydroxypyruvate. Elevated PHGDH activity in diverse cancer cells is mediated through genetic amplification, posttranslational modification, increased transcription, and allosteric regulation. Ultimately, these characteristics allow PHGDH to not only influence the growth and progression of cancer but also play an important role in metastasis and drug resistance. Consequently, PHGDH has emerged as a crucial focal point in cancer research. In this review, the structural aspects of PHGDH and its involvement in one-carbon metabolism are investigated, and PHGDH is proposed as a potential therapeutic target in diverse cancers. By elucidating how PHGDH expression promotes cancer growth, the goal of this review is to provide insight into innovative treatment strategies. This paper aims to reveal how PHGDH inhibitors can overcome resistance mechanisms, contributing to the development of effective cancer treatments.
Collapse
Affiliation(s)
- Chae Min Lee
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yeseong Hwang
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Minki Kim
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ye-Chan Park
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyeonhui Kim
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sungsoon Fang
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Department of Biomedical Sciences, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Chronic Intractable Disease for Systems Medicine Research Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Severance Institute for Vascular and Metabolic Research, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
10
|
Gülersoy E, Balıkçı C, Şahan A, Günal İ, Atlı MO. NMR-based metabolomic investigation of dogs with acute flaccid paralysis due to tick paralysis. Vet Med Sci 2024; 10:e1528. [PMID: 38952268 PMCID: PMC11217601 DOI: 10.1002/vms3.1528] [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: 02/13/2024] [Revised: 05/20/2024] [Accepted: 06/10/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Acute flaccid paralysis (AFP) is a complex clinical syndrome with various aetiologies. If untreated, AFP may lead to death due to failure of respiratory muscles. Tick paralysis, which is a noninfectious neurologic syndrome of AFP, occurs following tick attachment, engorgement, and injection of tick saliva toxins. There is no specific diagnostic test for tick paralysis, and mortality increases as definitive diagnosis is delayed. Although metabolomic investigation of tick saliva was conducted, there is a lack of research on metabolomic evaluation of hosts affected by tick paralysis. OBJECTIVES Thus, the aim of this study is to investigate metabolomic changes in serum samples of dogs with tick paralysis due to Rhipicephalus sanguineus using NMR-based metabolomics and to identify potential diagnostic/prognostic markers. MATERIALS AND METHODS Forty dogs infested with R. sanguineus, with clinical findings compatible with AFP and with a confirmed tick paralysis diagnosis ex juvantibus, constituted the Paralysis Group. Ten healthy dogs, which were admitted either for vaccination and/or check-up purposes, constituted the Control Group. After the confirmation tick paralysis, medical history, vaccination and nutritional status, body surface area and estimated tick numbers of all the dogs were noted. Physical examination included body temperature, heart and respiratory rate, capillary refill time evaluation and Modified Glasgow Coma Scale calculation. Serum samples were extracted from venous blood samples of all the dogs and were prepared for NMR analysis, and NMR-based metabolomics identification and quantification were performed. RESULTS NMR-based serum metabolomics of the present study revealed distinct up/down-regulated expressions, presenting a promising avenue. Moreover, it was observed that energy metabolism and especially liver functions were impaired in dogs with tick paralysis, and not only the respiratory system but also the kidneys were affected. CONCLUSION It was concluded that the present approach may help to better understand the pathological mechanisms developing in cases of AFP due to tick paralysis.
Collapse
Affiliation(s)
- Erdem Gülersoy
- Veterinary FacultyDepartment of Internal MedicineHarran UniversityŞanlıurfaTurkey
| | - Canberk Balıkçı
- Veterinary FacultyDepartment of Internal MedicineHarran UniversityŞanlıurfaTurkey
| | - Adem Şahan
- Veterinary FacultyDepartment of Internal MedicineHarran UniversityŞanlıurfaTurkey
| | - İsmail Günal
- Veterinary FacultyDepartment of Internal MedicineHarran UniversityŞanlıurfaTurkey
| | - Mehmet Osman Atlı
- Veterinary FacultyDepartment of Reproduction and Artificial InseminationHarran UniversityŞanlıurfaTurkey
| |
Collapse
|
11
|
Yu Y, Martins LM. Mitochondrial One-Carbon Metabolism and Alzheimer's Disease. Int J Mol Sci 2024; 25:6302. [PMID: 38928008 PMCID: PMC11203557 DOI: 10.3390/ijms25126302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 06/28/2024] Open
Abstract
Mitochondrial one-carbon metabolism provides carbon units to several pathways, including nucleic acid synthesis, mitochondrial metabolism, amino acid metabolism, and methylation reactions. Late-onset Alzheimer's disease is the most common age-related neurodegenerative disease, characterised by impaired energy metabolism, and is potentially linked to mitochondrial bioenergetics. Here, we discuss the intersection between the molecular pathways linked to both mitochondrial one-carbon metabolism and Alzheimer's disease. We propose that enhancing one-carbon metabolism could promote the metabolic processes that help brain cells cope with Alzheimer's disease-related injuries. We also highlight potential therapeutic avenues to leverage one-carbon metabolism to delay Alzheimer's disease pathology.
Collapse
Affiliation(s)
- Yizhou Yu
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| | - L. Miguel Martins
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge CB2 1QR, UK
| |
Collapse
|
12
|
Pszczołowska M, Walczak K, Misków W, Antosz K, Batko J, Karska J, Leszek J. Molecular cross-talk between long COVID-19 and Alzheimer's disease. GeroScience 2024; 46:2885-2899. [PMID: 38393535 PMCID: PMC11009207 DOI: 10.1007/s11357-024-01096-1] [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: 11/22/2023] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
The long COVID (coronavirus disease), a multisystemic condition following severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection, is one of the widespread problems. Some of its symptoms affect the nervous system and resemble symptoms of Alzheimer's disease (AD)-a neurodegenerative condition caused by the accumulation of amyloid beta and hyperphosphorylation of tau proteins. Multiple studies have found dependence between these two conditions. Patients with Alzheimer's disease have a greater risk of SARS-CoV-2 infection due to increased levels of angiotensin-converting enzyme 2 (ACE2), and the infection itself promotes amyloid beta generation which enhances the risk of AD. Also, the molecular pathways are alike-misregulations in folate-mediated one-carbon metabolism, a deficit of Cq10, and disease-associated microglia. Medical imaging in both of these diseases shows a decrease in the volume of gray matter, global brain size reduction, and hypometabolism in the parahippocampal gyrus, thalamus, and cingulate cortex. In some studies, a similar approach to applied medication can be seen, including the use of amino adamantanes and phenolic compounds of rosemary. The significance of these connections and their possible application in medical practice still needs further study but there is a possibility that they will help to better understand long COVID.
Collapse
Affiliation(s)
| | - Kamil Walczak
- Faculty of Medicine, Wrocław Medical University, Wrocław, Poland
| | - Weronika Misków
- Faculty of Medicine, Wrocław Medical University, Wrocław, Poland
| | - Katarzyna Antosz
- Faculty of Medicine, Wrocław Medical University, Wrocław, Poland
| | - Joanna Batko
- Faculty of Medicine, Wrocław Medical University, Wrocław, Poland
| | - Julia Karska
- Clinic of Psychiatry, Department of Psychiatry, Medical Department, Wrocław Medical University, Wrocław, Poland
| | - Jerzy Leszek
- Clinic of Psychiatry, Department of Psychiatry, Medical Department, Wrocław Medical University, Wrocław, Poland
| |
Collapse
|
13
|
Tsukamoto M, Hishida A, Tamura T, Nagayoshi M, Okada R, Kubo Y, Kato Y, Hamajima N, Nishida Y, Shimanoe C, Ibusuki R, Shibuya K, Takashima N, Nakamura Y, Kusakabe M, Nakamura Y, Koyanagi YN, Oze I, Nishiyama T, Suzuki S, Watanabe I, Matsui D, Otonari J, Ikezaki H, Katsuura-Kamano S, Arisawa K, Kuriki K, Nakatochi M, Momozawa Y, Takeuchi K, Wakai K, Matsuo K. GWAS of Folate Metabolism With Gene-environment Interaction Analysis Revealed the Possible Role of Lifestyles in the Control of Blood Folate Metabolites in Japanese: The J-MICC Study. J Epidemiol 2024; 34:228-237. [PMID: 37517992 PMCID: PMC10999522 DOI: 10.2188/jea.je20220341] [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: 01/09/2023] [Accepted: 06/30/2023] [Indexed: 08/01/2023] Open
Abstract
BACKGROUND The present genome-wide association study (GWAS) aimed to reveal the genetic loci associated with folate metabolites, as well as to detect related gene-environment interactions in Japanese. METHODS We conducted the GWAS of plasma homocysteine (Hcy), folic acid (FA), and vitamin B12 (VB12) levels in the Japan Multi-Institutional Collaborative Cohort (J-MICC) Study participants who joined from 2005 to 2012, and also estimated gene-environment interactions. In the replication phase, we used data from the Yakumo Study conducted in 2009. In the discovery phase, data of 2,263 participants from four independent study sites of the J-MICC Study were analyzed. In the replication phase, data of 573 participants from the Yakumo Study were analyzed. RESULTS For Hcy, MTHFR locus on chr 1, NOX4 on chr 11, CHMP1A on chr 16, and DPEP1 on chr 16 reached genome-wide significance (P < 5 × 10-8). MTHFR also associated with FA, and FUT2 on chr 19 associated with VB12. We investigated gene-environment interactions in both studies and found significant interactions between MTHFR C677T and ever drinking, current drinking, and physical activity >33% on Hcy (β = 0.039, 0.038 and -0.054, P = 0.018, 0.021 and <0.001, respectively) and the interaction of MTHFR C677T with ever drinking on FA (β = 0.033, P = 0.048). CONCLUSION The present GWAS revealed the folate metabolism-associated genetic loci and gene-environment interactions with drinking and physical activity in Japanese, suggesting the possibility of future personalized cardiovascular disease prevention.
Collapse
Affiliation(s)
- Mineko Tsukamoto
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Asahi Hishida
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Tamura
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mako Nagayoshi
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Rieko Okada
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoko Kubo
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasufumi Kato
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Nobuyuki Hamajima
- Department of Healthcare Administration, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuichiro Nishida
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Saga, Japan
| | | | - Rie Ibusuki
- Department of International Island and Community Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Kenichi Shibuya
- Department of International Island and Community Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Naoyuki Takashima
- Department of Public Health, Shiga University of Medical Science, Otsu, Japan
| | - Yasuyuki Nakamura
- Department of Public Health, Shiga University of Medical Science, Otsu, Japan
| | - Miho Kusakabe
- Cancer Prevention Center, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Yohko Nakamura
- Cancer Prevention Center, Chiba Cancer Center Research Institute, Chiba, Japan
| | - Yuriko N. Koyanagi
- Division of Cancer Information and Control, Department of Preventive Medicine, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Isao Oze
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Takeshi Nishiyama
- Department of Public Health, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Sadao Suzuki
- Department of Public Health, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Isao Watanabe
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Daisuke Matsui
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Jun Otonari
- Department of Psychosomatic Medicine, Kyushu University Graduate School of Medical Sciences, Faculty of Medical Sciences, Fukuoka, Japan
| | - Hiroaki Ikezaki
- Department of Comprehensive General Internal Medicine, Kyushu University Graduate School of Medical Sciences, Faculty of Medical Sciences, Fukuoka, Japan
| | - Sakurako Katsuura-Kamano
- Department of Preventive Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Kokichi Arisawa
- Laboratory of Public Health, Division of Nutritional Sciences, School of Food and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan
| | - Kiyonori Kuriki
- Laboratory of Public Health, Division of Nutritional Sciences, School of Food and Nutritional Sciences, University of Shizuoka, Shizuoka, Japan
| | - Masahiro Nakatochi
- Public Health Informatics Unit, Department of Integrated Health Sciences, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukihide Momozawa
- Laboratory for Genotyping Development, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kenji Takeuchi
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of International and Community Oral Health, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Kenji Wakai
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of Epidemiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
14
|
Rork AM, Bala AS, Renner T. Dynamic evolution of the mTHF gene family associated with primary metabolism across life. BMC Genomics 2024; 25:432. [PMID: 38693486 PMCID: PMC11064299 DOI: 10.1186/s12864-024-10159-8] [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: 05/03/2023] [Accepted: 02/25/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND The folate cycle of one-carbon (C1) metabolism, which plays a central role in the biosynthesis of nucleotides and amino acids, demonstrates the significance of metabolic adaptation. We investigated the evolutionary history of the methylenetetrahydrofolate dehydrogenase (mTHF) gene family, one of the main drivers of the folate cycle, across life. RESULTS Through comparative genomic and phylogenetic analyses, we found that several lineages of Archaea lacked domains vital for folate cycle function such as the mTHF catalytic and NAD(P)-binding domains of FolD. Within eukaryotes, the mTHF gene family diversified rapidly. For example, several duplications have been observed in lineages including the Amoebozoa, Opisthokonta, and Viridiplantae. In a common ancestor of Opisthokonta, FolD and FTHFS underwent fusion giving rise to the gene MTHFD1, possessing the domains of both genes. CONCLUSIONS Our evolutionary reconstruction of the mTHF gene family associated with a primary metabolic pathway reveals dynamic evolution, including gene birth-and-death, gene fusion, and potential horizontal gene transfer events and/or amino acid convergence.
Collapse
Affiliation(s)
- Adam M Rork
- Department of Entomology, The Pennsylvania State University, University Park, Pennsylvania, 16802, USA.
- Department of Entomology, Purdue University, West Lafayette, Indiana, 47907, USA.
| | - Arthi S Bala
- Department of Entomology, The Pennsylvania State University, University Park, Pennsylvania, 16802, USA
- Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC, 20007, USA
- School of Medicine, Georgetown University, Washington, DC, 20007, USA
| | - Tanya Renner
- Department of Entomology, The Pennsylvania State University, University Park, Pennsylvania, 16802, USA.
| |
Collapse
|
15
|
Bookey N, Drago P, Leung KY, Hughes L, MacCooey A, Ozaki M, Henry M, De Castro SCP, Doykov I, Heywood WE, Mills K, Murphy MM, Cavallé-Busquets P, Campbell S, Burtenshaw D, Meleady P, Cahill PA, Greene NDE, Parle-McDermott A. The Differential Translation Capabilities of the Human DHFR2 Gene Indicates a Developmental and Tissue-Specific Endogenous Protein of Low Abundance. Mol Cell Proteomics 2024; 23:100718. [PMID: 38224738 PMCID: PMC10884974 DOI: 10.1016/j.mcpro.2024.100718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/05/2024] [Accepted: 01/12/2024] [Indexed: 01/17/2024] Open
Abstract
A functional role has been ascribed to the human dihydrofolate reductase 2 (DHFR2) gene based on the enzymatic activity of recombinant versions of the predicted translated protein. However, the in vivo function is still unclear. The high amino acid sequence identity (92%) between DHFR2 and its parental homolog, DHFR, makes analysis of the endogenous protein challenging. This paper describes a targeted mass spectrometry proteomics approach in several human cell lines and tissue types to identify DHFR2-specific peptides as evidence of its translation. We show definitive evidence that the DHFR2 activity in the mitochondria is in fact mediated by DHFR, and not DHFR2. Analysis of Ribo-seq data and an experimental assessment of ribosome association using a sucrose cushion showed that the two main Ensembl annotated mRNA isoforms of DHFR2, 201 and 202, are differentially associated with the ribosome. This indicates a functional role at both the RNA and protein level. However, we were unable to detect DHFR2 protein at a detectable level in most cell types examined despite various RNA isoforms of DHFR2 being relatively abundant. We did detect a DHFR2-specific peptide in embryonic heart, indicating that the protein may have a specific role during embryogenesis. We propose that the main functionality of the DHFR2 gene in adult cells is likely to arise at the RNA level.
Collapse
Affiliation(s)
- Niamh Bookey
- School of Biotechnology, Dublin City University, Dublin, Ireland; DCU Life Sciences Institute, Dublin City University, Dublin, Ireland.
| | - Paola Drago
- School of Biotechnology, Dublin City University, Dublin, Ireland; DCU Life Sciences Institute, Dublin City University, Dublin, Ireland
| | - Kit-Yi Leung
- Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Linda Hughes
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Aoife MacCooey
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Mari Ozaki
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Michael Henry
- DCU Life Sciences Institute, Dublin City University, Dublin, Ireland
| | - Sandra C P De Castro
- Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Ivan Doykov
- Translational Mass Spectrometry Research Group, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Wendy E Heywood
- Translational Mass Spectrometry Research Group, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Kevin Mills
- Translational Mass Spectrometry Research Group, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Michelle M Murphy
- Area of Preventive Medicine and Public Health, Department of Basic Medical Sciences, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, IISPV and CIBERobn (Instituto de Salud Carlos III), Reus, Spain
| | - Pere Cavallé-Busquets
- Area of Obstetrics, Hospital Universitari Sant Joan de Reus, IISPV and CIBERobn (Instituto de Salud Carlos III), Reus, Spain
| | - Susan Campbell
- Sheffield Hallam University, Department of Biosciences and Chemistry, Sheffield, UK
| | | | - Paula Meleady
- School of Biotechnology, Dublin City University, Dublin, Ireland; DCU Life Sciences Institute, Dublin City University, Dublin, Ireland
| | - Paul A Cahill
- School of Biotechnology, Dublin City University, Dublin, Ireland
| | - Nicholas D E Greene
- Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Anne Parle-McDermott
- School of Biotechnology, Dublin City University, Dublin, Ireland; DCU Life Sciences Institute, Dublin City University, Dublin, Ireland.
| |
Collapse
|
16
|
Bashiri G, Bulloch EMM, Bramley WR, Davidson M, Stuteley SM, Young PG, Harris PWR, Naqvi MSH, Middleditch MJ, Schmitz M, Chang WC, Baker EN, Squire CJ. Poly-γ-glutamylation of biomolecules. Nat Commun 2024; 15:1310. [PMID: 38346985 PMCID: PMC10861534 DOI: 10.1038/s41467-024-45632-1] [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: 06/15/2023] [Accepted: 01/24/2024] [Indexed: 02/15/2024] Open
Abstract
Poly-γ-glutamate tails are a distinctive feature of archaeal, bacterial, and eukaryotic cofactors, including the folates and F420. Despite decades of research, key mechanistic questions remain as to how enzymes successively add glutamates to poly-γ-glutamate chains while maintaining cofactor specificity. Here, we show how poly-γ-glutamylation of folate and F420 by folylpolyglutamate synthases and γ-glutamyl ligases, non-homologous enzymes, occurs via processive addition of L-glutamate onto growing γ-glutamyl chain termini. We further reveal structural snapshots of the archaeal γ-glutamyl ligase (CofE) in action, crucially including a bulged-chain product that shows how the cofactor is retained while successive glutamates are added to the chain terminus. This bulging substrate model of processive poly-γ-glutamylation by terminal extension is arguably ubiquitous in such biopolymerisation reactions, including addition to folates, and demonstrates convergent evolution in diverse species from archaea to humans.
Collapse
Affiliation(s)
- Ghader Bashiri
- School of Biological Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.
- Maurice Wilkins Center for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.
| | - Esther M M Bulloch
- School of Biological Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
- Maurice Wilkins Center for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - William R Bramley
- School of Biological Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Madison Davidson
- Department of Chemistry, North Carolina State University, Raleigh, NC, 27695, USA
| | - Stephanie M Stuteley
- School of Biological Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
- Maurice Wilkins Center for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Paul G Young
- School of Biological Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
- Maurice Wilkins Center for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Paul W R Harris
- School of Biological Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
- Maurice Wilkins Center for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Muhammad S H Naqvi
- School of Biological Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Martin J Middleditch
- School of Biological Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Michael Schmitz
- School of Chemical Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Wei-Chen Chang
- Department of Chemistry, North Carolina State University, Raleigh, NC, 27695, USA
| | - Edward N Baker
- School of Biological Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
- Maurice Wilkins Center for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Christopher J Squire
- School of Biological Sciences, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.
- Maurice Wilkins Center for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.
| |
Collapse
|
17
|
Blank HM, Hammer SE, Boatright L, Roberts C, Heyden KE, Nagarajan A, Tsuchiya M, Brun M, Johnson CD, Stover PJ, Sitcheran R, Kennedy BK, Adams LG, Kaeberlein M, Field MS, Threadgill DW, Andrews-Polymenis HL, Polymenis M. Late-life dietary folate restriction reduces biosynthetic processes without compromising healthspan in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.12.575290. [PMID: 38260683 PMCID: PMC10802571 DOI: 10.1101/2024.01.12.575290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Folate is a vitamin required for cell growth and is present in fortified foods in the form of folic acid to prevent congenital abnormalities. The impact of low folate status on life-long health is poorly understood. We found that limiting folate levels with the folate antagonist methotrexate increased the lifespan of yeast and worms. We then restricted folate intake in aged mice and measured various health metrics, metabolites, and gene expression signatures. Limiting folate intake decreased anabolic biosynthetic processes in mice and enhanced metabolic plasticity. Despite reduced serum folate levels in mice with limited folic acid intake, these animals maintained their weight and adiposity late in life, and we did not observe adverse health outcomes. These results argue that the effectiveness of folate dietary interventions may vary depending on an individual's age and sex. A higher folate intake is advantageous during the early stages of life to support cell divisions needed for proper development. However, a lower folate intake later in life may result in healthier aging.
Collapse
Affiliation(s)
- Heidi M. Blank
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, United States
| | - Staci E. Hammer
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, United States
| | - Laurel Boatright
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, United States
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University Health Science Center, Bryan, United States
| | - Courtney Roberts
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, United States
| | - Katarina E. Heyden
- Division of Nutritional Sciences, Cornell University, Ithaca, United States
| | - Aravindh Nagarajan
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University Health Science Center, Bryan, United States
- Interdisciplinary Program in Genetics, Texas A&M University, College Station, United States
| | - Mitsuhiro Tsuchiya
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, United States
| | - Marcel Brun
- Texas A&M Agrilife Research, Genomics and Bioinformatics Service, College Station, United States
| | - Charles D. Johnson
- Texas A&M Agrilife Research, Genomics and Bioinformatics Service, College Station, United States
| | - Patrick J. Stover
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, United States
- Institute for Advancing Health through Agriculture, Texas A&M University, College Station, United States
- Department of Nutrition, Texas A&M University, College Station, United States
| | - Raquel Sitcheran
- Department of Cell Biology and Genetics, School of Medicine, Texas A&M University Health Science Center, Bryan, United States
| | - Brian K. Kennedy
- Departments of Biochemistry and Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Centre for Healthy Ageing, National University of Singapore, National University Health System, Singapore, Singapore
| | - L. Garry Adams
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M, College Station, Texas, USA
| | - Matt Kaeberlein
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, United States
- Optispan, Inc., Seattle, United States
| | - Martha S. Field
- Division of Nutritional Sciences, Cornell University, Ithaca, United States
| | - David W. Threadgill
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, United States
- Interdisciplinary Program in Genetics, Texas A&M University, College Station, United States
- Department of Nutrition, Texas A&M University, College Station, United States
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, United States
| | - Helene L. Andrews-Polymenis
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University Health Science Center, Bryan, United States
- Interdisciplinary Program in Genetics, Texas A&M University, College Station, United States
| | - Michael Polymenis
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, United States
- Interdisciplinary Program in Genetics, Texas A&M University, College Station, United States
- Institute for Advancing Health through Agriculture, Texas A&M University, College Station, United States
| |
Collapse
|
18
|
Derakhshan M, Kessler NJ, Hellenthal G, Silver MJ. Metastable epialleles in humans. Trends Genet 2024; 40:52-68. [PMID: 38000919 DOI: 10.1016/j.tig.2023.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 11/26/2023]
Abstract
First identified in isogenic mice, metastable epialleles (MEs) are loci where the extent of DNA methylation (DNAm) is variable between individuals but correlates across tissues derived from different germ layers within a given individual. This property, termed systemic interindividual variation (SIV), is attributed to stochastic methylation establishment before germ layer differentiation. Evidence suggests that some putative human MEs are sensitive to environmental exposures in early development. In this review we introduce key concepts pertaining to human MEs, describe methods used to identify MEs in humans, and review their genomic features. We also highlight studies linking DNAm at putative human MEs to early environmental exposures and postnatal (including disease) phenotypes.
Collapse
Affiliation(s)
- Maria Derakhshan
- London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK
| | - Noah J Kessler
- Department of Genetics, University of Cambridge, Cambridge CB2 3EH, UK
| | | | - Matt J Silver
- London School of Hygiene and Tropical Medicine, London WC1E 7HT, UK; Medical Research Council (MRC) Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Banjul, The Gambia.
| |
Collapse
|
19
|
Xu X, Zhu J, Fang L, Zou Z, Yuan J, Peng M, Yu G, Wu D, Liu Y, Tang J. Exome sequencing identified novel variants in three Chinese patients with 5,10-methenyltetrahydrofolate synthetase deficiency. Front Genet 2023; 14:1236849. [PMID: 37795244 PMCID: PMC10545881 DOI: 10.3389/fgene.2023.1236849] [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: 07/11/2023] [Accepted: 08/25/2023] [Indexed: 10/06/2023] Open
Abstract
5,10-methenyltetrahydrofolate synthetase (MTHFS) deficiency is a folate metabolism disorder known as a rare autosomal recessive neurodevelopmental disorder (MIM: #618367). With central nervous system involvements, it is mainly characterized by developmental delay, epilepsy, microcephaly, hypertonia, and cranial nerves involvement. Here, we report three new cases with MTHFS deficiency from two non-consanguineous Chinese families. All patients showed white matter dysplasia and global developmental delay, of which only patient 1 and 2 manifested tonic-clonic seizures. Moreover, patient 2 had severe eczema and patient 3 had recurrent diarrhea. Both phenotypic features are firstly found in MTHFS deficiency. Trio whole-exome sequencing and sanger sequencing were used to identify four novel variants, p.Y169Tfs*17, p.S53F, c.117+1delG, and p.E61G in the MTHFS gene. The identification of four novel pathogenic variants and varied clinical features in three affected patients expands the genotype and phenotype spectrum of MTHFS deficiency. We also reviewed all cases of MTHFS deficiency that had previously been reported. The experience of diagnosis and treatment from these cases provides us a more comprehensive understanding of this rare disease.
Collapse
Affiliation(s)
- Xiaoyan Xu
- Pediatric Neurorehabilitation Center, Pediatric Department, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jing Zhu
- Pediatric Neurorehabilitation Center, Pediatric Department, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Liwei Fang
- Pediatric Neurorehabilitation Center, Pediatric Department, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhuo Zou
- Department of Rehabilitation, Kunming Children’s Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - Jingjing Yuan
- Pediatric Neurorehabilitation Center, Pediatric Department, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Min Peng
- Chigene (Beijing) Translational Medical Research Center Co, Ltd, Beijing, China
| | - Guoliang Yu
- Chigene (Beijing) Translational Medical Research Center Co, Ltd, Beijing, China
| | - De Wu
- Pediatric Neurorehabilitation Center, Pediatric Department, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yun Liu
- Department of Rehabilitation, Kunming Children’s Hospital, Kunming Medical University, Kunming, Yunnan, China
| | - Jiulai Tang
- Pediatric Neurorehabilitation Center, Pediatric Department, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
20
|
Petersen JM, Smith-Webb RS, Shaw GM, Carmichael SL, Desrosiers TA, Nestoridi E, Darling AM, Parker SE, Politis MD, Yazdy MM, Werler MM. Periconceptional intakes of methyl donors and other micronutrients involved in one-carbon metabolism may further reduce the risk of neural tube defects in offspring: a United States population-based case-control study of women meeting the folic acid recommendations. Am J Clin Nutr 2023; 118:720-728. [PMID: 37661108 PMCID: PMC10624769 DOI: 10.1016/j.ajcnut.2023.05.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/23/2023] [Accepted: 05/31/2023] [Indexed: 09/05/2023] Open
Abstract
BACKGROUND Neural tube defects (NTDs) still occur among some women who consume 400 μg of folic acid for prevention. It has been hypothesized that intakes of methyl donors and other micronutrients involved in one-carbon metabolism may further protect against NTDs. OBJECTIVES To investigate whether intakes of vitamin B6, vitamin B12, choline, betaine, methionine, thiamine, riboflavin, and zinc, individually or in combination, were associated with NTD risk reduction in offspring of women meeting the folic acid recommendations. METHODS Data were from the National Birth Defects Prevention Study (United States population-based, case-control). We restricted deliveries between 1999 and 2011 with daily periconceptional folic acid supplementation or estimated dietary folate equivalents ≥400 μg. NTD cases were live births, stillbirths, or terminations affected by spina bifida, anencephaly, or encephalocele (n = 1227). Controls were live births without a major birth defect (n = 7095). We categorized intake of each micronutrient as higher or lower based on a combination of diet (estimated from a food frequency questionnaire) and periconceptional vitamin supplementation. We estimated NTD associations for higher compared with lower intake of each micronutrient, individually and in combination, expressed as odds ratios (ORs) and 95% confidence intervals (CIs), adjusted for age, race/ethnicity, education, and study center. RESULTS NTD associations with each micronutrient were weak to modest. Greater NTD reductions were observed with concurrent higher-amount intakes of multiple micronutrients. For instance, NTD odds were ∼50% lower among participants with ≥4 micronutrients with higher-amount intakes than among participants with ≤1 micronutrient with higher-amount intake (adjusted OR: 0.53; 95% CI: 0.33, 0.86). The strongest reduction occurred with concurrent higher-amount intakes of vitamin B6, vitamin B12, choline, betaine, and methionine (adjusted OR: 0.26; 95% CI: 0.09, 0.77) compared with ≤1 micronutrient with higher-amount intake. CONCLUSIONS Our findings support that NTD prevention, in the context of folic acid fortification, could be augmented with intakes of methyl donors and other micronutrients involved in folate metabolism.
Collapse
Affiliation(s)
- Julie M Petersen
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, United States; Massachusetts Center for Birth Defects Research and Prevention, Massachusetts Department of Public Health, Boston, MA, United States.
| | - Rashida S Smith-Webb
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, United States
| | - Gary M Shaw
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Suzan L Carmichael
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States; Center for Population Health Sciences, Stanford University School of Medicine, Stanford, CA, United States
| | - Tania A Desrosiers
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Eirini Nestoridi
- Massachusetts Center for Birth Defects Research and Prevention, Massachusetts Department of Public Health, Boston, MA, United States
| | - Anne Marie Darling
- Massachusetts Center for Birth Defects Research and Prevention, Massachusetts Department of Public Health, Boston, MA, United States
| | - Samantha E Parker
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, United States
| | - Maria D Politis
- Arkansas Center for Birth Defects Research and Prevention, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Mahsa M Yazdy
- Massachusetts Center for Birth Defects Research and Prevention, Massachusetts Department of Public Health, Boston, MA, United States
| | - Martha M Werler
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, United States
| |
Collapse
|
21
|
Cuevas-López B, Romero-Ramirez EI, García-Arroyo FE, Tapia E, León-Contreras JC, Silva-Palacios A, Roldán FJ, Campos ONM, Hernandez-Esquivel L, Marín-Hernández A, Gonzaga-Sánchez JG, Hernández-Pando R, Pedraza-Chaverri J, Sánchez-Lozada LG, Aparicio-Trejo OE. NAC Pre-Administration Prevents Cardiac Mitochondrial Bioenergetics, Dynamics, Biogenesis, and Redox Alteration in Folic Acid-AKI-Induced Cardio-Renal Syndrome Type 3. Antioxidants (Basel) 2023; 12:1592. [PMID: 37627587 PMCID: PMC10451243 DOI: 10.3390/antiox12081592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
The incidence of kidney disease is increasing worldwide. Acute kidney injury (AKI) can strongly favor cardio-renal syndrome (CRS) type 3 development. However, the mechanism involved in CRS development is not entirely understood. In this sense, mitochondrial impairment in both organs has become a central axis in CRS physiopathology. This study aimed to elucidate the molecular mechanisms associated with cardiac mitochondrial impairment and its role in CRS development in the folic acid-induced AKI (FA-AKI) model. Our results showed that 48 h after FA-AKI, the administration of N-acetyl-cysteine (NAC), a mitochondrial glutathione regulator, prevented the early increase in inflammatory and cell death markers and oxidative stress in the heart. This was associated with the ability of NAC to protect heart mitochondrial bioenergetics, principally oxidative phosphorylation (OXPHOS) and membrane potential, through complex I activity and the preservation of glutathione balance, thus preventing mitochondrial dynamics shifting to fission and the decreases in mitochondrial biogenesis and mass. Our data show, for the first time, that mitochondrial bioenergetics impairment plays a critical role in the mechanism that leads to heart damage. Furthermore, NAC heart mitochondrial preservation during an AKI event can be a valuable strategy to prevent CRS type 3 development.
Collapse
Affiliation(s)
- Belén Cuevas-López
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| | - Edgar Ignacio Romero-Ramirez
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| | - Fernando E. García-Arroyo
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| | - Edilia Tapia
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| | - Juan Carlos León-Contreras
- Experimental Pathology Section, National Institute of Medical Sciences and Nutrition “Salvador Zubirán”, Mexico City 14000, Mexico; (J.C.L.-C.); (R.H.-P.)
| | - Alejandro Silva-Palacios
- Department of Cardiovascular Biomedicine, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico;
| | - Francisco-Javier Roldán
- Outpatient Department, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico;
| | - Omar Noel Medina Campos
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City 04510, Mexico; (O.N.M.C.); (J.P.-C.)
| | - Luz Hernandez-Esquivel
- Department of Biochemistry, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (L.H.-E.); (A.M.-H.)
| | - Alvaro Marín-Hernández
- Department of Biochemistry, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (L.H.-E.); (A.M.-H.)
| | - José Guillermo Gonzaga-Sánchez
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| | - Rogelio Hernández-Pando
- Experimental Pathology Section, National Institute of Medical Sciences and Nutrition “Salvador Zubirán”, Mexico City 14000, Mexico; (J.C.L.-C.); (R.H.-P.)
| | - José Pedraza-Chaverri
- Department of Biology, Faculty of Chemistry, National Autonomous University of Mexico, Mexico City 04510, Mexico; (O.N.M.C.); (J.P.-C.)
| | - Laura Gabriela Sánchez-Lozada
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| | - Omar Emiliano Aparicio-Trejo
- Department of Cardio-Renal Physiology, National Institute of Cardiology Ignacio Chávez, Mexico City 14080, Mexico; (B.C.-L.); (E.I.R.-R.); (F.E.G.-A.); (E.T.); (J.G.G.-S.); (L.G.S.-L.)
| |
Collapse
|
22
|
Ren H, Wang K, Liu Z, Zhong X, Liang M, Liao Y. Effect of Low Dietary Folate on Mouse Spermatogenesis and Spindle Assembly Checkpoint Dysfunction May Contribute to Folate Deficiency-Induced Chromosomal Instability in Cultured Mouse Spermatogonia. DNA Cell Biol 2023; 42:515-525. [PMID: 37289823 DOI: 10.1089/dna.2023.0035] [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] [Indexed: 06/10/2023] Open
Abstract
Folate, as the initial substrate in one-carbon metabolism, is involved in the synthesis of important substances such as DNA, RNA, and protein. Folate deficiency (FD) is associated with male subfertility and impaired spermatogenesis, yet the underlying mechanisms are poorly understood. In the present study, we established an animal model of FD to investigate the effect of FD on spermatogenesis. GC-1 spermatogonia were used as a model to investigate the effect of FD on proliferation, viability, and chromosomal instability (CIN). Furthermore, we explored the expression of core genes and proteins of spindle assembly checkpoint (SAC), a signaling cascade ensuring accurate chromosome segregation and preventing CIN during mitosis. Cells were maintained in medium containing 0, 20, 200, or 2000 nM folate for 14 days. CIN was measured by using a cytokinesis-blocked micronucleus cytome assay. We found that sperm counts decreased significantly (p < 0.001) and the rate of sperm with defects in the head increased significantly (p < 0.05) in FD diet mice. We also found, relative to the folate-sufficient conditions (2000 nM), cells cultured with 0, 20, or 200 nM folate exhibited delayed growth and increased apoptosis in an inverse dose-dependent manner. FD (0, 20, or 200 nM) significantly induced CIN (p < 0.001, p < 0.001, and p < 0.05, respectively). Moreover, FD significantly and inverse dose dependently increased the mRNA and protein expression of several key SAC-related genes. The results indicate that FD impairs SAC activity, which contributes to mitotic aberrations and CIN. These findings establish a novel association between FD and SAC dysfunction. Thus, FD-impaired spermatogenesis may be partly due to genomic instability and proliferation inhibition of spermatogonia.
Collapse
Affiliation(s)
- Huanhuan Ren
- School of Life Science, Bengbu Medical College, Bengbu, China
- Department of Clinical Laboratory, Fuyang Fifth People's Hospital, Fuyang, China
| | - Kaixian Wang
- School of Life Science, Bengbu Medical College, Bengbu, China
| | - Zirui Liu
- School of Life Science, Bengbu Medical College, Bengbu, China
| | - Xuansheng Zhong
- School of Life Science, Bengbu Medical College, Bengbu, China
| | - Meng Liang
- School of Life Science, Bengbu Medical College, Bengbu, China
| | - Yaping Liao
- School of Life Science, Bengbu Medical College, Bengbu, China
| |
Collapse
|
23
|
Botté ES, Bennett H, Engelberts JP, Thomas T, Bell JJ, Webster NS, Luter HM. Future ocean conditions induce necrosis, microbial dysbiosis and nutrient cycling imbalance in the reef sponge Stylissa flabelliformis. ISME COMMUNICATIONS 2023; 3:53. [PMID: 37311801 PMCID: PMC10264452 DOI: 10.1038/s43705-023-00247-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/22/2023] [Accepted: 04/14/2023] [Indexed: 06/15/2023]
Abstract
Oceans are rapidly warming and acidifying in the context of climate change, threatening sensitive marine biota including coral reef sponges. Ocean warming (OW) and ocean acidification (OA) can impact host health and associated microbiome, but few studies have investigated these effects, which are generally studied in isolation, on a specific component of the holobiont. Here we present a comprehensive view of the consequences of simultaneous OW and OA for the tropical sponge Stylissa flabelliformis. We found no interactive effect on the host health or microbiome. Furthermore, OA (pH 7.6 versus pH 8.0) had no impact, while OW (31.5 °C versus 28.5 °C) caused tissue necrosis, as well as dysbiosis and shifts in microbial functions in healthy tissue of necrotic sponges. Major taxonomic shifts included a complete loss of archaea, reduced proportions of Gammaproteobacteria and elevated relative abundances of Alphaproteobacteria. OW weakened sponge-microbe interactions, with a reduced capacity for nutrient exchange and phagocytosis evasion, indicating lower representations of stable symbionts. The potential for microbially-driven nitrogen and sulphur cycling was reduced, as was amino acid metabolism. Crucially, the dysbiosis annihilated the potential for ammonia detoxification, possibly leading to accumulation of toxic ammonia, nutrient imbalance, and host tissue necrosis. Putative defence against reactive oxygen species was greater at 31.5 °C, perhaps as microorganisms capable of resisting temperature-driven oxidative stress were favoured. We conclude that healthy symbiosis in S. flabelliformis is unlikely to be disrupted by future OA but will be deeply impacted by temperatures predicted for 2100 under a "business-as-usual" carbon emission scenario.
Collapse
Affiliation(s)
- Emmanuelle S Botté
- Centre for Marine Science and Innovation, School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, New South Wales, Australia.
- Australian Institute of Marine Science, Townsville, Queensland, Australia.
| | - Holly Bennett
- Australian Institute of Marine Science, Townsville, Queensland, Australia
- Victoria University of Wellington, Wellington, New Zealand
- Cawthron Institute, Nelson, New Zealand
| | - J Pamela Engelberts
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Queensland, Australia
| | - Torsten Thomas
- Centre for Marine Science and Innovation, School of Biological, Earth and Environmental Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - James J Bell
- Victoria University of Wellington, Wellington, New Zealand
| | - Nicole S Webster
- Australian Institute of Marine Science, Townsville, Queensland, Australia
- Australian Centre for Ecogenomics, School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Queensland, Australia
- Australian Antarctic Division, Hobart, Tasmania, Australia
| | - Heidi M Luter
- Australian Institute of Marine Science, Townsville, Queensland, Australia.
| |
Collapse
|
24
|
Pilesi E, Angioli C, Graziani C, Parroni A, Contestabile R, Tramonti A, Vernì F. A gene-nutrient interaction between vitamin B6 and serine hydroxymethyltransferase (SHMT) affects genome integrity in Drosophila. J Cell Physiol 2023. [PMID: 37183313 DOI: 10.1002/jcp.31033] [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: 02/13/2023] [Revised: 04/06/2023] [Accepted: 04/18/2023] [Indexed: 05/16/2023]
Abstract
Pyridoxal 5'-phosphate (PLP), the catalytically active form of vitamin B6, participates as a cofactor to one carbon (1C) pathway that produces precursors for DNA metabolism. The concerted action of PLP-dependent serine hydroxymethyltransferase (SHMT) and thymidylate synthase (TS) leads to the biosynthesis of thymidylate (dTMP), which plays an essential function in DNA synthesis and repair. PLP deficiency causes chromosome aberrations (CABs) in Drosophila and human cells, rising the hypothesis that an altered 1C metabolism may be involved. To test this hypothesis, we used Drosophila as a model system and found, firstly, that in PLP deficient larvae SHMT activity is reduced by 40%. Second, we found that RNAi-induced SHMT depletion causes chromosome damage rescued by PLP supplementation and strongly exacerbated by PLP depletion. RNAi-induced TS depletion causes severe chromosome damage, but this is only slightly enhanced by PLP depletion. dTMP supplementation rescues CABs in both PLP-deficient and PLP-proficient SHMTRNAi . Altogether these data suggest that a reduction of SHMT activity caused by PLP deficiency contributes to chromosome damage by reducing dTMP biosynthesis. In addition, our work brings to light a gene-nutrient interaction between SHMT decreased activity and PLP deficiency impacting on genome stability that may be translated to humans.
Collapse
Affiliation(s)
- Eleonora Pilesi
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Chiara Angioli
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Claudio Graziani
- Department of Biochemical Sciences "A. Rossi Fanelli", Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Alessia Parroni
- Department of Biochemical Sciences "A. Rossi Fanelli", Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
- Institute of Molecular Biology and Pathology, National Research Council (IBPM-CNR), Rome, Italy
| | - Roberto Contestabile
- Department of Biochemical Sciences "A. Rossi Fanelli", Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Angela Tramonti
- Department of Biochemical Sciences "A. Rossi Fanelli", Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
- Institute of Molecular Biology and Pathology, National Research Council (IBPM-CNR), Rome, Italy
| | - Fiammetta Vernì
- Department of Biology and Biotechnology "Charles Darwin", Sapienza University of Rome, Rome, Italy
| |
Collapse
|
25
|
Freitas-Costa NC, Andrade P, Normando P, Salvatte Nunes KS, Raymundo CE, Ribeiro de Castro IR, Maria de Aquino Lacerda E, Farias DR, Kac G. Association of development quotient with nutritional status of vitamins B6, B12, and folate in 6-59-month-old children: Results from the Brazilian National Survey on Child Nutrition (ENANI-2019). Am J Clin Nutr 2023:S0002-9165(23)47384-4. [PMID: 37105522 DOI: 10.1016/j.ajcnut.2023.04.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/28/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Vitamins B6, B12, and folate are essential for the formation and maintenance of the human brain, but studies evaluating these vitamins with early childhood development (ECD) in children under five are limited and controversial. OBJECTIVE To evaluate the association between vitamins B6, B12, and folate concentrations/status and ECD. METHODS Data regarding 6,520 children aged 6-59 months (from the Brazilian National Survey on Child Nutrition [ENANI-2019]) were analyzed. ECD was assessed using the Survey of Well-being of Young Children's milestones questionnaire. Vitamin B6 concentration (nmol/L) was classified according to the tertile of the distribution and with the cutoff <20 nmol/L. Folate concentrations >45.3 nmol/L were classified as high, and vitamin B12 <150 pmol/L as deficient. The graded response model was used to estimate developmental age, and the developmental quotient (DQ) was calculated as the developmental age divided by chronological age. Multiple linear regression models were adjusted for confounders. RESULTS The DQ mean (95% confidence interval) for Brazilian children was 0.99 (0.97-1.01). Children aged 6-23 months (1.13 [1.10-1.16]) had a higher DQ mean than those aged 24-35 (0.99 [0.95-1.03]) and 36-59 months (0.89 [0.86-0.92]). Child age was inversely associated with DQ (β=-0.007; p<0.001). An interaction between child age and vitamin B12 deficiency in the DQ (β=-0.005; p<0.001) indicated that, in children aged 36-59 months, the DQ was markedly lower in children with B12 deficiency than in those without B12 deficiency. Vitamin B6 concentrations were directly associated with the DQ (β=0.0004; p=0.031) among children aged 24-59 months in the adjusted model. No association was observed between folate status and DQ. CONCLUSIONS In Brazil, the DQ is lower among older children, and those with vitamin B12 deficiency. Vitamin B6 status was directly associated with the DQ in children aged 24-59 months.
Collapse
Affiliation(s)
- Nathalia Cristina Freitas-Costa
- Nutritional Epidemiology Observatory, Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro Andrade
- Institute of Applied Economic Research, Rio de Janeiro, Brazil
| | - Paula Normando
- Nutritional Epidemiology Observatory, Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Keronlainy Silva Salvatte Nunes
- Nutritional Epidemiology Observatory, Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos Eduardo Raymundo
- Institute of Collective Health, School of Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Elisa Maria de Aquino Lacerda
- Nutritional Epidemiology Observatory, Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Dayana Rodrigues Farias
- Nutritional Epidemiology Observatory, Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gilberto Kac
- Nutritional Epidemiology Observatory, Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
26
|
Wu HHL, McDonnell T, Chinnadurai R. Physiological Associations between Vitamin B Deficiency and Diabetic Kidney Disease. Biomedicines 2023; 11:biomedicines11041153. [PMID: 37189771 DOI: 10.3390/biomedicines11041153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/24/2023] [Accepted: 04/10/2023] [Indexed: 05/17/2023] Open
Abstract
The number of people living with chronic kidney disease (CKD) is growing as our global population continues to expand. With aging, diabetes, and cardiovascular disease being major harbingers of kidney disease, the number of people diagnosed with diabetic kidney disease (DKD) has grown concurrently. Poor clinical outcomes in DKD could be influenced by an array of factors-inadequate glycemic control, obesity, metabolic acidosis, anemia, cellular senescence, infection and inflammation, cognitive impairment, reduced physical exercise threshold, and, importantly, malnutrition contributing to protein-energy wasting, sarcopenia, and frailty. Amongst the various causes of malnutrition in DKD, the metabolic mechanisms of vitamin B (B1 (Thiamine), B2 (Riboflavin), B3 (Niacin/Nicotinamide), B5 (Pantothenic Acid), B6 (Pyridoxine), B8 (Biotin), B9 (Folate), and B12 (Cobalamin)) deficiency and its clinical impact has garnered greater scientific interest over the past decade. There remains extensive debate on the biochemical intricacies of vitamin B metabolic pathways and how their deficiencies may affect the development of CKD, diabetes, and subsequently DKD, and vice-versa. Our article provides a review of updated evidence on the biochemical and physiological properties of the vitamin B sub-forms in normal states, and how vitamin B deficiency and defects in their metabolic pathways may influence CKD/DKD pathophysiology, and in reverse how CKD/DKD progression may affect vitamin B metabolism. We hope our article increases awareness of vitamin B deficiency in DKD and the complex physiological associations that exist between vitamin B deficiency, diabetes, and CKD. Further research efforts are needed going forward to address the knowledge gaps on this topic.
Collapse
Affiliation(s)
- Henry H L Wu
- Renal Research Laboratory, Kolling Institute of Medical Research, Royal North Shore Hospital, The University of Sydney, Sydney, NSW 2065, Australia
| | - Thomas McDonnell
- Department of Renal Medicine, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK
| | - Rajkumar Chinnadurai
- Department of Renal Medicine, Northern Care Alliance NHS Foundation Trust, Salford M6 8HD, UK
- Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M1 7HR, UK
| |
Collapse
|
27
|
Nanjireddy PM, Olejniczak SH, Buxbaum NP. Targeting of chimeric antigen receptor T cell metabolism to improve therapeutic outcomes. Front Immunol 2023; 14:1121565. [PMID: 36999013 PMCID: PMC10043186 DOI: 10.3389/fimmu.2023.1121565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/17/2023] [Indexed: 03/16/2023] Open
Abstract
Genetically engineered chimeric antigen receptor (CAR) T cells can cure patients with cancers that are refractory to standard therapeutic approaches. To date, adoptive cell therapies have been less effective against solid tumors, largely due to impaired homing and function of immune cells within the immunosuppressive tumor microenvironment (TME). Cellular metabolism plays a key role in T cell function and survival and is amenable to manipulation. This manuscript provides an overview of known aspects of CAR T metabolism and describes potential approaches to manipulate metabolic features of CAR T to yield better anti-tumor responses. Distinct T cell phenotypes that are linked to cellular metabolism profiles are associated with improved anti-tumor responses. Several steps within the CAR T manufacture process are amenable to interventions that can generate and maintain favorable intracellular metabolism phenotypes. For example, co-stimulatory signaling is executed through metabolic rewiring. Use of metabolic regulators during CAR T expansion or systemically in the patient following adoptive transfer are described as potential approaches to generate and maintain metabolic states that can confer improved in vivo T cell function and persistence. Cytokine and nutrient selection during the expansion process can be tailored to yield CAR T products with more favorable metabolic features. In summary, improved understanding of CAR T cellular metabolism and its manipulations have the potential to guide the development of more effective adoptive cell therapies.
Collapse
Affiliation(s)
- Priyanka Maridhi Nanjireddy
- Department of Pediatric Oncology, Pediatric Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
- Immunology Department, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Scott H. Olejniczak
- Immunology Department, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Nataliya Prokopenko Buxbaum
- Department of Pediatrics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
- *Correspondence: Nataliya Prokopenko Buxbaum,
| |
Collapse
|
28
|
Hammer SE, Polymenis M. One-carbon metabolic enzymes are regulated during cell division and make distinct contributions to the metabolome and cell cycle progression in Saccharomyces cerevisiae. G3 (BETHESDA, MD.) 2023; 13:6983127. [PMID: 36627750 PMCID: PMC9997564 DOI: 10.1093/g3journal/jkad005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 01/12/2023]
Abstract
Enzymes of one-carbon (1C) metabolism play pivotal roles in proliferating cells. They are involved in the metabolism of amino acids, nucleotides, and lipids and the supply of all cellular methylations. However, there is limited information about how these enzymes are regulated during cell division and how cell cycle kinetics are affected in several loss-of-function mutants of 1C metabolism. Here, we report that the levels of the S. cerevisiae enzymes Ade17p and Cho2p, involved in the de novo synthesis of purines and phosphatidylcholine (PC), respectively, are cell cycle-regulated. Cells lacking Ade17p, Cho2p, or Shm2p (an enzyme that supplies 1C units from serine) have distinct alterations in size homeostasis and cell cycle kinetics. Loss of Ade17p leads to a specific delay at START, when cells commit to a new round of cell division, while loss of Shm2p has broader effects, reducing growth rate. Furthermore, the inability to synthesize PC de novo in cho2Δ cells delays START and reduces the coherence of nuclear elongation late in the cell cycle. Loss of Cho2p also leads to profound metabolite changes. Besides the expected changes in the lipidome, cho2Δ cells have reduced levels of amino acids, resembling cells shifted to poorer media. These results reveal the different ways that 1C metabolism allocates resources to affect cell proliferation at multiple cell cycle transitions.
Collapse
Affiliation(s)
- Staci E Hammer
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| | - Michael Polymenis
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
29
|
Stark M, Levin M, Ulitsky I, Assaraf YG. Folylpolyglutamate synthetase mRNA G-quadruplexes regulate its cell protrusion localization and enhance a cancer cell invasive phenotype upon folate repletion. BMC Biol 2023; 21:13. [PMID: 36721160 PMCID: PMC9889130 DOI: 10.1186/s12915-023-01525-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 01/23/2023] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Folates are crucial for the biosynthesis of nucleotides and amino acids, essential for cell proliferation and development. Folate deficiency induces DNA damage, developmental defects, and tumorigenicity. The obligatory enzyme folylpolyglutamate synthetase (FPGS) mediates intracellular folate retention via cytosolic and mitochondrial folate polyglutamylation. Our previous paper demonstrated the association of the cytosolic FPGS (cFPGS) with the cytoskeleton and various cell protrusion proteins. Based on these recent findings, the aim of the current study was to investigate the potential role of cFPGS at cell protrusions. RESULTS Here we uncovered a central role for two G-quadruplex (GQ) motifs in the 3'UTR of FPGS mediating the localization of cFPGS mRNA and protein at cell protrusions. Using the MBSV6-loop reporter system and fluorescence microscopy, we demonstrate that following folate deprivation, cFPGS mRNA is retained in the endoplasmic reticulum, whereas upon 15 min of folate repletion, this mRNA is rapidly translocated to cell protrusions in a 3'UTR- and actin-dependent manner. The actin dependency of this folate-induced mRNA translocation is shown by treatment with Latrunculin B and inhibitors of the Ras homolog family member A (RhoA) pathway. Upon folate repletion, the FPGS 3'UTR GQs induce an amoeboid/mesenchymal hybrid cell phenotype during migration and invasion through a collagen gel matrix. Targeted disruption of the 3'UTR GQ motifs by introducing point mutations or masking them by antisense oligonucleotides abrogated cell protrusion targeting of cFPGS mRNA. CONCLUSIONS Collectively, the GQ motifs within the 3'UTR of FPGS regulate its transcript and protein localization at cell protrusions in response to a folate cue, inducing cancer cell invasive phenotype. These novel findings suggest that the 3'UTR GQ motifs of FPGS constitute an attractive druggable target aimed at inhibition of cancer invasion and metastasis.
Collapse
Affiliation(s)
- Michal Stark
- grid.6451.60000000121102151The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, 3200003 Haifa, Israel
| | - May Levin
- grid.6451.60000000121102151The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, 3200003 Haifa, Israel ,grid.507132.2Present address: May Levin, MeMed Diagnostics Ltd, Tirat Carmel, Israel
| | - Igor Ulitsky
- grid.13992.300000 0004 0604 7563Department of Immunology and Regenerative Biology and Department of Molecular Neuroscience, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Yehuda G. Assaraf
- grid.6451.60000000121102151The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, 3200003 Haifa, Israel
| |
Collapse
|
30
|
Gök V, Erdem Ş, Haliloğlu Y, Bişgin A, Belkaya S, Başaran KE, Canatan MF, Özcan A, Yılmaz E, Acıpayam C, Karakükcü M, Canatan H, Per H, Patıroğlu T, Eken A, Ünal E. Immunodeficiency associated with a novel functionally defective variant of SLC19A1 benefits from folinic acid treatment. Genes Immun 2023; 24:12-20. [PMID: 36517554 DOI: 10.1038/s41435-022-00191-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
Insufficient dietary folate intake, hereditary malabsorption, or defects in folate transport may lead to combined immunodeficiency (CID). Although loss of function mutations in the major intestinal folate transporter PCFT/SLC46A1 was shown to be associated with CID, the evidence for pathogenic variants of RFC/SLC19A1 resulting in immunodeficiency was lacking. We report two cousins carrying a homozygous pathogenic variant c.1042 G > A, resulting in p.G348R substitution who showed symptoms of immunodeficiency associated with defects of folate transport. SLC19A1 expression by peripheral blood mononuclear cells (PBMC) was quantified by real-time qPCR and immunostaining. T cell proliferation, methotrexate resistance, NK cell cytotoxicity, Treg cells and cytokine production by T cells were examined by flow cytometric assays. Patients were treated with and benefited from folinic acid. Studies revealed normal NK cell cytotoxicity, Treg cell counts, and naive-memory T cell percentages. Although SLC19A1 mRNA and protein expression were unaltered, remarkably, mitogen induced-T cell proliferation was significantly reduced at suboptimal folic acid and supraoptimal folinic acid concentrations. In addition, patients' PBMCs were resistant to methotrexate-induced apoptosis supporting a functionally defective SLC19A1. This study presents the second pathogenic SLC19A1 variant in the literature, providing the first experimental evidence that functionally defective variants of SLC19A1 may present with symptoms of immunodeficiency.
Collapse
Affiliation(s)
- Veysel Gök
- Department of Pediatrics, Division of Pediatric Hematology & Oncology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Şerife Erdem
- Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Türkiye.,Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Yeşim Haliloğlu
- Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Türkiye.,Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Atıl Bişgin
- Department of Medical Genetics, Faculty of Medicine, Çukurova University, Adana, Türkiye
| | - Serkan Belkaya
- Department of Molecular Biology and Genetics, Faculty of Science, Bilkent University, Ankara, Türkiye
| | - Kemal Erdem Başaran
- Department of Physiology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | | | - Alper Özcan
- Department of Pediatrics, Division of Pediatric Hematology & Oncology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Ebru Yılmaz
- Department of Pediatrics, Division of Pediatric Hematology & Oncology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Can Acıpayam
- Department of Pediatrics, Division of Pediatric Hematology & Oncology, Faculty of Medicine, Sütçü İmam University, Kahramanmaraş, Türkiye
| | - Musa Karakükcü
- Department of Pediatrics, Division of Pediatric Hematology & Oncology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Halit Canatan
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Hüseyin Per
- Department of Pediatrics, Division of Pediatric Neurology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Türkan Patıroğlu
- Department of Pediatrics, Division of Pediatric Hematology & Oncology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye.,Department of Pediatrics, Division of Pediatric Immunology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye
| | - Ahmet Eken
- Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Türkiye. .,Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye.
| | - Ekrem Ünal
- Department of Pediatrics, Division of Pediatric Hematology & Oncology, Faculty of Medicine, Erciyes University, Kayseri, Türkiye. .,Genome and Stem Cell Center (GENKOK), Erciyes University, Kayseri, Türkiye. .,Department of Blood Banking and Transfusion Medicine, Health Science Institution, Erciyes University, Kayseri, Türkiye.
| |
Collapse
|
31
|
Relationship between Maternal Vitamin D Levels and Adverse Outcomes. Nutrients 2022; 14:nu14204230. [PMID: 36296914 PMCID: PMC9610169 DOI: 10.3390/nu14204230] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/28/2022] [Accepted: 10/08/2022] [Indexed: 11/23/2022] Open
Abstract
Vitamin D (VD), a fat-soluble vitamin, has a variety of functions that are important for growth and development, including regulation of cell differentiation and apoptosis, immune system development, and brain development. As such, VD status during pregnancy is critical for maternal health, fetal skeletal growth, and optimal pregnancy outcomes. Studies have confirmed that adverse pregnancy outcomes, such as preeclampsia, low birth weight, neonatal hypocalcemia, poor postnatal growth, skeletal fragility, and increased incidence of autoimmune diseases, can be associated with low VD levels during pregnancy and infancy. Thus, there is growing interest in the role of VD during pregnancy. This review summarizes the potential adverse health outcomes of maternal VD status during pregnancy for both mother and offspring (gestational diabetes mellitus, hypertensive gestational hypertension, intrauterine growth restriction, miscarriage, stillbirth, and preterm birth) and discusses the underlying mechanisms (regulation of cytokine pathways, immune system processing, internal secretion, placental function, etc.) of VD in regulating each of the outcomes. This review aims to provide a basis for public health intervention strategies to reduce the incidence of adverse pregnancies.
Collapse
|
32
|
Xiao Y, Yu Y, Xie L, Li K, Guo X, Li G, Liu J, Li G, Hu J. A genome-wide association study of folates in sweet corn kernels. FRONTIERS IN PLANT SCIENCE 2022; 13:1004455. [PMID: 36247547 PMCID: PMC9562826 DOI: 10.3389/fpls.2022.1004455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/02/2022] [Indexed: 06/10/2023]
Abstract
Folate is commonly synthesized in natural plants and is an essential water-soluble vitamin of great importance inhuman health. Although the key genes involved in folate biosynthesis and transformation pathways have been identified in plants, the genetic architecture of folate in sweet corn kernels remain largely unclear. In this study, an association panel of 295 inbred lines of sweet corn was constructed. Six folate derivatives were quantified in sweet corn kernels at 20 days after pollination and a total of 95 loci were identified for eight folate traits using a genome-wide association study. A peak GWAS signal revealed that natural variation in ZmFCL, encoding a 5-formyltetrahydrofolate cyclo-ligase, accounted for 30.12% of phenotypic variation in 5-FTHF content. Further analysis revealed that two adjacent SNPs on the second exon resulting in an AA-to-GG in the gene and an Asn-to-Gly change in the protein could be the causative variant influencing 5-FTHF content. Meanwhile, 5-FTHF content was negatively correlated with ZmFCL expression levels in the population. These results extend our knowledge regarding the genetic basis of folate and provide molecular markers for the optimization of folate levels in sweet corn kernels.
Collapse
Affiliation(s)
- Yingni Xiao
- Crops Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Provincial Key Laboratory of Crop Genetic Improvement, Guangzhou, China
| | - Yongtao Yu
- Crops Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Provincial Key Laboratory of Crop Genetic Improvement, Guangzhou, China
| | - Lihua Xie
- Crops Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Provincial Key Laboratory of Crop Genetic Improvement, Guangzhou, China
| | - Kun Li
- Crops Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Provincial Key Laboratory of Crop Genetic Improvement, Guangzhou, China
| | - Xinbo Guo
- School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Guangyu Li
- Crops Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Provincial Key Laboratory of Crop Genetic Improvement, Guangzhou, China
| | - Jianhua Liu
- Crops Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Provincial Key Laboratory of Crop Genetic Improvement, Guangzhou, China
| | - Gaoke Li
- Crops Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Provincial Key Laboratory of Crop Genetic Improvement, Guangzhou, China
| | - Jianguang Hu
- Crops Research Institute, Guangdong Academy of Agricultural Sciences/Guangdong Provincial Key Laboratory of Crop Genetic Improvement, Guangzhou, China
| |
Collapse
|
33
|
You Y, Doi Y, Maeda N, Masuo S, Takeshita N, Takaya N. Carboxypeptidase G and pterin deaminase metabolic pathways degrade folic acid in Variovorax sp. F1. BMC Microbiol 2022; 22:225. [PMID: 36167524 PMCID: PMC9513972 DOI: 10.1186/s12866-022-02643-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 09/15/2022] [Indexed: 12/05/2022] Open
Abstract
Background Folic acid (FA) is a synthetic vitamin (B9) and the oxidized form of a metabolic cofactor that is essential for life. Although the biosynthetic mechanisms of FA are established, its environmental degradation mechanism has not been fully elucidated. The present study aimed to identify bacteria in soil that degrade FA and the mechanisms involved. Results We isolated the soil bacterium Variovorax sp. F1 from sampled weed rhizospheres in a grassland and investigated its FA degradation mechanism. Cultured Variovorax sp. F1 rapidly degraded FA to pteroic acid (PA), indicating that FA hydrolysis to PA and glutamate. We cloned the carboxypeptidase G (CPG) gene and found widely distributed paralogs within the Variovorax genus. Recombinant CPG preferred FA and deaminofolic acid as substrates, indicating its involvement in FA degradation by Variovorax. Prolonged culture of Variovorax sp. F1 resulted in decreased rates of deaminofolic acid (DFA) and deaminopteroic acid (DPA) accumulation. This indicated that the deamination reaction also comprised a route of FA degradation. We also identified an F1 gene that was orthologous to the pterin deaminase gene (Arad3529) of Agrobacterium radiobacter. The encoded protein deaminated FA and PA to DFA and DPA, which was consistent with the deamination activity of FA and PA in bacterial cell-free extracts. Conclusion We discovered that the two enzymes required for FA degradation pathways in isolates of Variovorax sp. F1 comprise CPG and pterin deaminase, and that DFA and PA are intermediates in the generation of DPA. Supplementary Information The online version contains supplementary material available at 10.1186/s12866-022-02643-6.
Collapse
Affiliation(s)
- Yungmi You
- Faculty of Life and Environmental Sciences, Microbiology Research Center for Sustainability, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572, Japan
| | - Yuki Doi
- Faculty of Life and Environmental Sciences, Microbiology Research Center for Sustainability, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572, Japan
| | - Norifumi Maeda
- Faculty of Life and Environmental Sciences, Microbiology Research Center for Sustainability, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572, Japan
| | - Shunsuke Masuo
- Faculty of Life and Environmental Sciences, Microbiology Research Center for Sustainability, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572, Japan
| | - Norio Takeshita
- Faculty of Life and Environmental Sciences, Microbiology Research Center for Sustainability, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572, Japan
| | - Naoki Takaya
- Faculty of Life and Environmental Sciences, Microbiology Research Center for Sustainability, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8572, Japan.
| |
Collapse
|
34
|
Weber N, Kouřimská L, Kulma M, Petříčková D, Seufert F, Rychlik M. Folate contents in insects as promising food components quantified by stable isotope dilution. Front Nutr 2022; 9:970255. [PMID: 36159461 PMCID: PMC9499261 DOI: 10.3389/fnut.2022.970255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 08/11/2022] [Indexed: 11/13/2022] Open
Abstract
Concerning the increasing population, edible insects are of growing interest due to several advantages such as sustainable production and as a source of high-quality nutrients. One of the less studied nutrients are folates, in the context of insects is folates, which play an important role in human metabolism. In the article, we describe how six different insect species are reared and extracted for five common folate vitamers by high-performance liquid chromatography interfaced with tandem mass spectrometry (LC-MS/MS). For this purpose, house crickets (Acheta domesticus—adults), Jamaican field crickets (Gryllus assimilis—adults), discoid cockroaches (Blaberus discoidalis—nymphs), migratory locusts (Locusta migratoria—adults), mealworms (Tenebrio molitor), and lesser mealworms (Alphitobius diaperinus) were investigated. The total folate content differs between 59.1 ± 6.50 and 143 ± 11.1 μg/100 g. Also, the feed, which was adapted to the needs of the insects and differed for some species, was extracted for their total folate content. The four different feed compositions (rapeseed, chicken feed, bramble leaves, and a mix of chicken feed, wheat bran, and carrot/apples) showed a folate content of about 100 μg/100 g, except for hay, where the content was 300 μg/100 g. In general, the insect folate content is comparable to other food and seems to be a promising source of folates. However, the amount of consumption needed to meet the requirement must also be considered. Moreover, the effect of different influencing factors is not yet entirely clear and needs further investigation.
Collapse
Affiliation(s)
- Nadine Weber
- Chair of Analytical Food Chemistry, Research Department Life Science Engineering, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- *Correspondence: Nadine Weber,
| | - Lenka Kouřimská
- Department of Microbiology, Nutrition and Dietetics, Czech University of Life Sciences Prague, Praha-Suchdol, Czechia
| | - Martin Kulma
- Department of Zoology and Fisheries, Czech University of Life Sciences Prague, Praha-Suchdol, Czechia
| | - Dora Petříčková
- Department of Microbiology, Nutrition and Dietetics, Czech University of Life Sciences Prague, Praha-Suchdol, Czechia
| | - Franziska Seufert
- Chair of Analytical Food Chemistry, Research Department Life Science Engineering, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Michael Rychlik
- Chair of Analytical Food Chemistry, Research Department Life Science Engineering, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, St Lucia, QLD, Australia
| |
Collapse
|
35
|
Achreja A, Yu T, Mittal A, Choppara S, Animasahun O, Nenwani M, Wuchu F, Meurs N, Mohan A, Jeon JH, Sarangi I, Jayaraman A, Owen S, Kulkarni R, Cusato M, Weinberg F, Kweon HK, Subramanian C, Wicha MS, Merajver SD, Nagrath S, Cho KR, DiFeo A, Lu X, Nagrath D. Metabolic collateral lethal target identification reveals MTHFD2 paralogue dependency in ovarian cancer. Nat Metab 2022; 4:1119-1137. [PMID: 36131208 DOI: 10.1038/s42255-022-00636-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/09/2022] [Indexed: 11/08/2022]
Abstract
Recurrent loss-of-function deletions cause frequent inactivation of tumour suppressor genes but often also involve the collateral deletion of essential genes in chromosomal proximity, engendering dependence on paralogues that maintain similar function. Although these paralogues are attractive anticancer targets, no methodology exists to uncover such collateral lethal genes. Here we report a framework for collateral lethal gene identification via metabolic fluxes, CLIM, and use it to reveal MTHFD2 as a collateral lethal gene in UQCR11-deleted ovarian tumours. We show that MTHFD2 has a non-canonical oxidative function to provide mitochondrial NAD+, and demonstrate the regulation of systemic metabolic activity by the paralogue metabolic pathway maintaining metabolic flux compensation. This UQCR11-MTHFD2 collateral lethality is confirmed in vivo, with MTHFD2 inhibition leading to complete remission of UQCR11-deleted ovarian tumours. Using CLIM's machine learning and genome-scale metabolic flux analysis, we elucidate the broad efficacy of targeting MTHFD2 despite distinct cancer genetic profiles co-occurring with UQCR11 deletion and irrespective of stromal compositions of tumours.
Collapse
Affiliation(s)
- Abhinav Achreja
- Laboratory for Systems Biology of Human Diseases, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Tao Yu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Anjali Mittal
- Laboratory for Systems Biology of Human Diseases, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Srinadh Choppara
- Laboratory for Systems Biology of Human Diseases, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Olamide Animasahun
- Laboratory for Systems Biology of Human Diseases, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Minal Nenwani
- Laboratory for Systems Biology of Human Diseases, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Fulei Wuchu
- Laboratory for Systems Biology of Human Diseases, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Noah Meurs
- Laboratory for Systems Biology of Human Diseases, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Aradhana Mohan
- Laboratory for Systems Biology of Human Diseases, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jin Heon Jeon
- Laboratory for Systems Biology of Human Diseases, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Itisam Sarangi
- Laboratory for Systems Biology of Human Diseases, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Anusha Jayaraman
- Laboratory for Systems Biology of Human Diseases, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
| | - Sarah Owen
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Reva Kulkarni
- Laboratory for Systems Biology of Human Diseases, University of Michigan, Ann Arbor, MI, USA
- Department of Electrical and Computer Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Michele Cusato
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Frank Weinberg
- Hematology and Oncology, University of Illinois, Chicago, IL, USA
| | - Hye Kyong Kweon
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Chitra Subramanian
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Max S Wicha
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Sofia D Merajver
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Sunitha Nagrath
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Kathleen R Cho
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Analisa DiFeo
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | - Xiongbin Lu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
- Melvin & Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA.
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Deepak Nagrath
- Laboratory for Systems Biology of Human Diseases, University of Michigan, Ann Arbor, MI, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI, USA.
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
36
|
Crider KS, Qi YP, Yeung LF, Mai CT, Head Zauche L, Wang A, Daniels K, Williams JL. Folic Acid and the Prevention of Birth Defects: 30 Years of Opportunity and Controversies. Annu Rev Nutr 2022; 42:423-452. [PMID: 35995050 PMCID: PMC9875360 DOI: 10.1146/annurev-nutr-043020-091647] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
For three decades, the US Public Health Service has recommended that all persons capable of becoming pregnant consume 400 μg/day of folic acid (FA) to prevent neural tube defects (NTDs). The neural tube forms by 28 days after conception. Fortification can be an effective NTD prevention strategy in populations with limited access to folic acid foods and/or supplements. This review describes the status of mandatory FA fortification among countries that fortify (n = 71) and the research describing the impact of those programs on NTD rates (up to 78% reduction), blood folate concentrations [red blood cell folate concentrations increased ∼1.47-fold (95% CI, 1.27, 1.70) following fortification], and other health outcomes. Across settings, high-quality studies such as those with randomized exposures (e.g., randomized controlled trials, Mendelian randomization studies) are needed to elucidate interactions of FA with vitamin B12 as well as expanded biomarker testing.
Collapse
Affiliation(s)
- Krista S Crider
- Neural Tube Defects Surveillance and Prevention Team, Infant Outcomes Monitoring, Research, and Prevention Branch, Division of Birth Defects and Infant Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia, USA;
| | - Yan Ping Qi
- Neural Tube Defects Surveillance and Prevention Team, Infant Outcomes Monitoring, Research, and Prevention Branch, Division of Birth Defects and Infant Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia, USA;
| | - Lorraine F Yeung
- Neural Tube Defects Surveillance and Prevention Team, Infant Outcomes Monitoring, Research, and Prevention Branch, Division of Birth Defects and Infant Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia, USA;
| | - Cara T Mai
- Neural Tube Defects Surveillance and Prevention Team, Infant Outcomes Monitoring, Research, and Prevention Branch, Division of Birth Defects and Infant Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia, USA;
| | - Lauren Head Zauche
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, USA
| | - Arick Wang
- Neural Tube Defects Surveillance and Prevention Team, Infant Outcomes Monitoring, Research, and Prevention Branch, Division of Birth Defects and Infant Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia, USA;
| | | | - Jennifer L Williams
- Neural Tube Defects Surveillance and Prevention Team, Infant Outcomes Monitoring, Research, and Prevention Branch, Division of Birth Defects and Infant Disorders, National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia, USA;
| |
Collapse
|
37
|
Chi WY, Hsiao TH, Lee GH, Su IH, Chen BH, Tang MJ, Fu TF. The cooperative interplay among inflammation, necroptosis and YAP pathway contributes to the folate deficiency-induced liver cells enlargement. Cell Mol Life Sci 2022; 79:397. [PMID: 35790616 PMCID: PMC11073448 DOI: 10.1007/s00018-022-04425-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/03/2022]
Abstract
Change in cell size may bring in profound impact to cell function and survival, hence the integrity of the organs consisting of those cells. Nevertheless, how cell size is regulated remains incompletely understood. We used the fluorescent zebrafish transgenic line Tg-GGH/LR that displays inducible folate deficiency (FD) and hepatomegaly upon FD induction as in vivo model. We found that FD caused hepatocytes enlargement and increased liver stiffness, which could not be prevented by nucleotides supplementations. Both in vitro and in vivo studies indicated that RIPK3/MLKL-dependent necroptotic pathway and Hippo signaling interactively participated in this FD-induced hepatocytic enlargement in a dual chronological and cooperative manner. FD also induced hepatic inflammation, which convenes a dialog of positive feedback loop between necroptotic and Hippo pathways. The increased MMP13 expression in response to FD elevated TNFα level and further aggravated the hepatocyte enlargement. Meanwhile, F-actin was circumferentially re-allocated at the edge under cell membrane in response to FD. Our results substantiate the interplay among intracellular folate status, pathways regulation, inflammatory responses, actin cytoskeleton and cell volume control, which can be best observed with in vivo platform. Our data also support the use of this Tg-GGH/LR transgenic line for the mechanistical and therapeutic research for the pathologic conditions related to cell size alteration.
Collapse
Affiliation(s)
- Wan-Yu Chi
- The Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tsun-Hsien Hsiao
- The Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Gang-Hui Lee
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | - I-Hsiu Su
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | - Bing-Hung Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Center for Biomarkers and Biotech Drugs, Kaohsiung Medical University, Kaohsiung, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Ming-Jer Tang
- International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tzu-Fun Fu
- The Institute of Basic Medical Science, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, No. 1, University Rd., Tainan, 701, Taiwan.
| |
Collapse
|
38
|
Berger MM, Shenkin A, Schweinlin A, Amrein K, Augsburger M, Biesalski HK, Bischoff SC, Casaer MP, Gundogan K, Lepp HL, de Man AME, Muscogiuri G, Pietka M, Pironi L, Rezzi S, Cuerda C. ESPEN micronutrient guideline. Clin Nutr 2022; 41:1357-1424. [PMID: 35365361 DOI: 10.1016/j.clnu.2022.02.015] [Citation(s) in RCA: 230] [Impact Index Per Article: 76.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 02/16/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Trace elements and vitamins, named together micronutrients (MNs), are essential for human metabolism. Recent research has shown the importance of MNs in common pathologies, with significant deficiencies impacting the outcome. OBJECTIVE This guideline aims to provide information for daily clinical nutrition practice regarding assessment of MN status, monitoring, and prescription. It proposes a consensus terminology, since many words are used imprecisely, resulting in confusion. This is particularly true for the words "deficiency", "repletion", "complement", and "supplement". METHODS The expert group attempted to apply the 2015 standard operating procedures (SOP) for ESPEN which focuses on disease. However, this approach could not be applied due to the multiple diseases requiring clinical nutrition resulting in one text for each MN, rather than for diseases. An extensive search of the literature was conducted in the databases Medline, PubMed, Cochrane, Google Scholar, and CINAHL. The search focused on physiological data, historical evidence (published before PubMed release in 1996), and observational and/or randomized trials. For each MN, the main functions, optimal analytical methods, impact of inflammation, potential toxicity, and provision during enteral or parenteral nutrition were addressed. The SOP wording was applied for strength of recommendations. RESULTS There was a limited number of interventional trials, preventing meta-analysis and leading to a low level of evidence. The recommendations underwent a consensus process, which resulted in a percentage of agreement (%): strong consensus required of >90% of votes. Altogether the guideline proposes sets of recommendations for 26 MNs, resulting in 170 single recommendations. Critical MNs were identified with deficiencies being present in numerous acute and chronic diseases. Monitoring and management strategies are proposed. CONCLUSION This guideline should enable addressing suboptimal and deficient status of a bundle of MNs in at-risk diseases. In particular, it offers practical advice on MN provision and monitoring during nutritional support.
Collapse
Affiliation(s)
- Mette M Berger
- Department of Adult Intensive Care, Lausanne University Hospital (CHUV), Lausanne, Switzerland.
| | - Alan Shenkin
- Institute of Aging and Chronic Disease, University of Liverpool, Liverpool, UK.
| | - Anna Schweinlin
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany.
| | - Karin Amrein
- Medical University of Graz, Department of Internal Medicine, Division of Endocrinology and Diabetology, Austria.
| | - Marc Augsburger
- University Centre of Legal Medicine Lausanne-Geneva, Lausanne University Hospital and University of Lausanne, Geneva University Hospital and University of Geneva, Lausanne-Geneva, Switzerland.
| | | | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany.
| | - Michael P Casaer
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Intensive Care Medicine, Leuven, Belgium.
| | - Kursat Gundogan
- Division of Intensive Care Medicine, Department of Internal Medicine, Erciyes University School of Medicine, Kayseri, Turkey.
| | | | - Angélique M E de Man
- Department of Intensive Care Medicine, Research VUmc Intensive Care (REVIVE), Amsterdam Cardiovascular Science (ACS), Amsterdam Infection and Immunity Institute (AI&II), Amsterdam Medical Data Science (AMDS), Amsterdam UMC, Location VUmc, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, the Netherlands.
| | - Giovanna Muscogiuri
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università di Napoli (Federico II), Naples, Italy; United Nations Educational, Scientific and Cultural Organization (UNESCO) Chair for Health Education and Sustainable Development, Federico II, University, Naples, Italy.
| | - Magdalena Pietka
- Pharmacy Department, Stanley Dudrick's Memorial Hospital, Skawina, Poland.
| | - Loris Pironi
- Alma Mater Studiorum - University of Bologna, Department of Medical and Surgical Sciences, Italy; IRCCS Azienda Ospedaliero-Universitaria di Bologna, Centre for Chronic Intestinal Failure - Clinical Nutrition and Metabolism Unit, Italy.
| | - Serge Rezzi
- Swiss Nutrition and Health Foundation (SNHf), Epalinges, Switzerland.
| | - Cristina Cuerda
- Departamento de Medicina, Universidad Complutense de Madrid, Nutrition Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain.
| |
Collapse
|
39
|
Francisco A, Figueira TR, Castilho RF. Mitochondrial NAD(P) + Transhydrogenase: From Molecular Features to Physiology and Disease. Antioxid Redox Signal 2022; 36:864-884. [PMID: 34155914 DOI: 10.1089/ars.2021.0111] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Significance: Proton-translocating NAD(P)+ transhydrogenase, also known as nicotinamide nucleotide transhydrogenase (NNT), catalyzes a reversible reaction coupling the protonmotive force across the inner mitochondrial membrane and hydride (H-, a proton plus two electrons) transfer between the mitochondrial pools of NAD(H) and NADP(H). The forward NNT reaction is a source of NADPH in the mitochondrial matrix, fueling antioxidant and biosynthetic pathways with reductive potential. Despite the greater emphasis given to the net forward reaction, the reverse NNT reaction that oxidizes NADPH also occurs in physiological and pathological conditions. Recent Advances: NNT (dys)function has been linked to various metabolic pathways and disease phenotypes. Most of these findings have been based on spontaneous loss-of-function Nnt mutations found in the C57BL/6J mouse strain (NntC57BL/6J mutation) and disease-causing Nnt mutations in humans. The present review focuses on recent advances based on the mouse NntC57BL/6J mutation. Critical Issues: Most studies associating NNT function with disease phenotypes have been based on comparisons between different strains of inbred mice (with or without the NntC57BL/6J mutation), which creates uncertainties over the actual contribution of NNT in the context of other potential genetic modifiers. Future Directions: Future research might contribute to understanding the role of NNT in pathological conditions and elucidate how NNT regulates physiological signaling through its forward and reverse reactions. The importance of NNT in redox balance and tumor cell proliferation makes it a potential target of new therapeutic strategies for oxidative-stress-mediated diseases and cancer. Antioxid. Redox Signal. 36, 864-884.
Collapse
Affiliation(s)
- Annelise Francisco
- Department of Pathology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| | - Tiago Rezende Figueira
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, Brazil
| | - Roger Frigério Castilho
- Department of Pathology, Faculty of Medical Sciences, University of Campinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
40
|
Cazacu N, Chilom CG, David M, Florescu M. Conformational Changes in the BSA-LT4 Complex Induced by the Presence of Vitamins: Spectroscopic Approach and Molecular Docking. Int J Mol Sci 2022; 23:ijms23084215. [PMID: 35457032 PMCID: PMC9027512 DOI: 10.3390/ijms23084215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/05/2022] [Accepted: 04/08/2022] [Indexed: 11/25/2022] Open
Abstract
Levothyroxine (LT4) is known for its use in various conditions including hypothyroidism. LT4 interaction with serum albumin may be influenced by the presence of vitamins. For this reason, we investigated the effect of vitamin C, vitamin B12, and folic acid on the complex of Bovine Serum Albumin with LT4 (BSA-LT4). UV-Vis spectroscopy was used to monitor the influence of vitamins on the BSA-LT4 complex. Fluorescence spectroscopy revealed a static quenching mechanism of the fluorescence of BSA-LT4 complex by the vitamin C and folic acid and a combined mechanism for vitamin B12. The interaction of vitamin C and folic acid with BSA-LT4 was moderate, while the binding of vitamin B12 was much stronger, extending the storage time of LT4 in blood plasma. Synchronous fluorescence found that the vitamins were closer to the vicinity of Trp than to Tyr and the effect was more pronounced for the binding of vitamin B12. The thermal stability of the BSA-LT4 complex was more evident, but no influence on the stability of BSA-LT4 complex was obtained for vitamin C. Molecular docking studies showed that vitamin C and folic acid bound the same site of the protein, while vitamin B12 bonded to a different site.
Collapse
Affiliation(s)
- Nicoleta Cazacu
- Department of Electricity, Solid Physics and Biophysics, Faculty of Physics, University of Bucharest, Str. Atomistilor No. 405, CP MG-11, Bucuresti-Magurele, 077125 Magurele, Romania; (N.C.); (C.G.C.)
| | - Claudia G. Chilom
- Department of Electricity, Solid Physics and Biophysics, Faculty of Physics, University of Bucharest, Str. Atomistilor No. 405, CP MG-11, Bucuresti-Magurele, 077125 Magurele, Romania; (N.C.); (C.G.C.)
| | - Melinda David
- Department of Fundamental, Prophylactic and Clinical Disciplines, Faculty of Medicine, Transilvania University of Brasov, Str. Universitatii No. 1, Building C, Room CI30, 500068 Brasov, Romania;
| | - Monica Florescu
- Department of Fundamental, Prophylactic and Clinical Disciplines, Faculty of Medicine, Transilvania University of Brasov, Str. Universitatii No. 1, Building C, Room CI30, 500068 Brasov, Romania;
- Correspondence:
| |
Collapse
|
41
|
Lian T, Wang X, Li S, Jiang H, Zhang C, Wang H, Jiang L. Comparative Transcriptome Analysis Reveals Mechanisms of Folate Accumulation in Maize Grains. Int J Mol Sci 2022; 23:ijms23031708. [PMID: 35163628 PMCID: PMC8836222 DOI: 10.3390/ijms23031708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 02/05/2023] Open
Abstract
Previously, the complexity of folate accumulation in the early stages of maize kernel development has been reported, but the mechanisms of folate accumulation are unclear. Two maize inbred lines, DAN3130 and JI63, with different patterns of folate accumulation and different total folate contents in mature kernels were used to investigate the transcriptional regulation of folate metabolism during late stages of kernel formation by comparative transcriptome analysis. The folate accumulation during DAP 24 to mature kernels could be controlled by circumjacent pathways of folate biosynthesis, such as pyruvate metabolism, glutamate metabolism, and serine/glycine metabolism. In addition, the folate variation between these two inbred lines was related to those genes among folate metabolism, such as genes in the pteridine branch, para-aminobenzoate branch, serine/tetrahydrofolate (THF)/5-methyltetrahydrofolate cycle, and the conversion of THF monoglutamate to THF polyglutamate. The findings provided insight into folate accumulation mechanisms during maize kernel formation to promote folate biofortification.
Collapse
Affiliation(s)
- Tong Lian
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (T.L.); (S.L.); (C.Z.)
- Plant Genetics, Gembloux Agro-Bio Tech, University of Liège, 5030 Gembloux, Belgium
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya 572000, China
| | - Xuxia Wang
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China; (X.W.); (H.J.)
| | - Sha Li
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (T.L.); (S.L.); (C.Z.)
| | - Haiyang Jiang
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China; (X.W.); (H.J.)
| | - Chunyi Zhang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (T.L.); (S.L.); (C.Z.)
- Sanya Institute, Hainan Academy of Agricultural Sciences, Sanya 572000, China
| | - Huan Wang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (T.L.); (S.L.); (C.Z.)
- School of Life Sciences, Anhui Agricultural University, Hefei 230036, China; (X.W.); (H.J.)
- National Agricultural Science and Technology Center, Chengdu 610213, China
- Correspondence: (H.W.); (L.J.)
| | - Ling Jiang
- Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100081, China; (T.L.); (S.L.); (C.Z.)
- Correspondence: (H.W.); (L.J.)
| |
Collapse
|
42
|
Huang S, Li Z, Liu M, Zhou M, Weng J, He Y, Jiang Y, Zhang H, Sun H. Reaction-based fluorescent and chemiluminescent probes for formaldehyde detection and imaging. Chem Commun (Camb) 2022; 58:1442-1453. [PMID: 34991152 DOI: 10.1039/d1cc05644a] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Formaldehyde (FA), a reactive carbonyl species, is classified as Group 1 carcinogen by International Agency for Research on Cancer (IARC) in 2004. In addition, clinical studies have implicated that elevated levels of FA have been associated with different kinds of diseases, such as neurodegenerative diseases, diabetes, and chronic liver and heart disorders. However, in addition to the direct inhalation of FA in the environment, most organisms can also produce FA endogenously by demethylases and oxidases during the metabolism of amino acids and xenobiotics. Since FA plays an important role in physiological and pathological processes, developing reliable and efficient methods to monitor FA levels in biological samples is crucial. Reaction-based fluorescent/chemiluminescent probes have provided robust methods for FA detection and real-time visualization in living organisms. In this highlight, we will summarize the major developments in the structure design and applications of FA probes in recent years. Three main strategies for designing FA probes have been discussed and grouped by different reaction mechanisms. In addition, some miscellaneous reaction mechanisms have also been discussed. We also highlight novel applications of these probes in biological systems, which offer powerful tools to discover the diverse functions of FA in physiology and pathology processes.
Collapse
Affiliation(s)
- Shumei Huang
- School of Chemical Engineering and Light Industry and School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, China.
| | - Zejun Li
- School of Chemical Engineering and Light Industry and School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, China. .,Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| | - Minghui Liu
- School of Chemical Engineering and Light Industry and School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, China.
| | - Mengjiao Zhou
- School of Pharmacy, Nantong University, Nantong 226001, China
| | - Jintao Weng
- School of Chemical Engineering and Light Industry and School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, China.
| | - Yong He
- School of Chemical Engineering and Light Industry and School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, China.
| | - Yin Jiang
- School of Chemical Engineering and Light Industry and School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, China.
| | - Huatang Zhang
- School of Chemical Engineering and Light Industry and School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, Guangdong, 510006, China.
| | - Hongyan Sun
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China.,Department of Chemistry and Center of Super-Diamond and Advanced Films (COSDAF), City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, China.
| |
Collapse
|
43
|
OUP accepted manuscript. Nutr Rev 2022; 80:2178-2197. [DOI: 10.1093/nutrit/nuac025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
44
|
Salamoun YM, Polireddy K, Cho YK, Medcalf MR, Funk RS. Methotrexate Disposition, Anti-Folate Activity, and Metabolomic Profiling to Identify Molecular Markers of Disease Activity and Drug Response in the Collagen-Induced Arthritis Mouse Model. Metabolites 2021; 12:metabo12010024. [PMID: 35050146 PMCID: PMC8780148 DOI: 10.3390/metabo12010024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 01/02/2023] Open
Abstract
Methotrexate (MTX) is widely used in the treatment of autoimmune arthritis but is limited by its unpredictable and variable response profile. Currently, no biomarkers exist to predict or monitor early therapeutic responses to MTX. Using a collagen-induced arthritis (CIA) mouse model, this study aimed to identify biochemical pathways and biomarkers associated with MTX efficacy in autoimmune arthritis. Following arthritis disease induction, DBA/1J mice were treated with subcutaneous MTX (20 mg/kg/week) and disease activity was assessed based on disease activity scores (DAS) and paw volume (PV) measurements. Red blood cell (RBC) and plasma samples were collected at the end of the study and were assessed for folate and MTX content. Plasma samples were analyzed by semitargeted global metabolomic profiling and analyzed by univariate and multivariate analysis. Treatment with MTX was associated with significant reductions in disease activity based on both DAS (p = 0.0006) and PV (p = 0.0006). MTX therapy resulted in significant reductions in 5-methyltetrahydrofolate (5mTHF) levels in plasma (p = 0.02) and RBCs (p = 0.001). Reductions in both RBC and plasma 5mTHF were associated with lower DAS (p = 0.0007, p = 0.01, respectively) and PV (p = 0.001, p = 0.005, respectively). Increases in RBC MTX were associated with lower DAS (p = 0.003) but not PV (p = 0.23). Metabolomic analysis identified N-methylisoleucine (NMI) and quinolone as metabolites significantly altered in disease mice, which were corrected towards healthy control levels in mice treated with MTX. Reductions in plasma NMI were associated with lower DAS (p = 0.0002) and PV (p = 9.5 × 10-6). Increases in plasma quinolone were associated with lower DAS (p = 0.02) and PV (p = 0.01). Receiver-operating characteristic curve analysis identified plasma NMI (AUC = 1.00, p = 2.4 × 10-8), RBC 5mTHF (AUC = 0.99, p = 2.4 × 10-5), and plasma quinolone (AUC = 0.89, p = 0.01) as top discriminating metabolites of MTX treatment. Our data support a relationship between MTX efficacy and its effect on circulating folates and identified 5mTHF, NMI, and quinolone as potential therapeutic biomarkers of disease activity and MTX response in the CIA mouse model of autoimmune arthritis.
Collapse
Affiliation(s)
- Yezan M. Salamoun
- Department of Pharmacy Practice, University of Kansas Medical Center, Kansas City, KS 66160, USA; (K.P.); (Y.K.C.); (M.R.M.)
- Correspondence: (Y.M.S.); (R.S.F.); Tel.: +1-913-945-6904 (Y.M.S. & R.S.F.)
| | - Kishore Polireddy
- Department of Pharmacy Practice, University of Kansas Medical Center, Kansas City, KS 66160, USA; (K.P.); (Y.K.C.); (M.R.M.)
| | - Yu Kyoung Cho
- Department of Pharmacy Practice, University of Kansas Medical Center, Kansas City, KS 66160, USA; (K.P.); (Y.K.C.); (M.R.M.)
| | - Matthew R. Medcalf
- Department of Pharmacy Practice, University of Kansas Medical Center, Kansas City, KS 66160, USA; (K.P.); (Y.K.C.); (M.R.M.)
| | - Ryan S. Funk
- Department of Pharmacy Practice, University of Kansas Medical Center, Kansas City, KS 66160, USA; (K.P.); (Y.K.C.); (M.R.M.)
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Correspondence: (Y.M.S.); (R.S.F.); Tel.: +1-913-945-6904 (Y.M.S. & R.S.F.)
| |
Collapse
|
45
|
Yan L. Folic acid-induced animal model of kidney disease. Animal Model Exp Med 2021; 4:329-342. [PMID: 34977484 PMCID: PMC8690981 DOI: 10.1002/ame2.12194] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/06/2023] Open
Abstract
The kidneys are a vital organ that is vulnerable to both acute kidney injury (AKI) and chronic kidney disease (CKD) which can be caused by numerous risk factors such as ischemia, sepsis, drug toxicity and drug overdose, exposure to heavy metals, and diabetes. In spite of the advances in our understanding of the pathogenesis of AKI and CKD as well AKI transition to CKD, there is still no available therapeutics that can be used to combat kidney disease effectively, highlighting an urgent need to further study the pathological mechanisms underlying AKI, CKD, and AKI progression to CKD. In this regard, animal models of kidney disease are indispensable. This article reviews a widely used animal model of kidney disease, which is induced by folic acid (FA). While a low dose of FA is nutritionally beneficial, a high dose of FA is very toxic to the kidneys. Following a brief description of the procedure for disease induction by FA, major mechanisms of FA-induced kidney injury are then reviewed, including oxidative stress, mitochondrial abnormalities such as impaired bioenergetics and mitophagy, ferroptosis, pyroptosis, and increased expression of fibroblast growth factor 23 (FGF23). Finally, application of this FA-induced kidney disease model as a platform for testing the efficacy of a variety of therapeutic approaches is also discussed. Given that this animal model is simple to create and is reproducible, it should remain useful for both studying the pathological mechanisms of kidney disease and identifying therapeutic targets to fight kidney disease.
Collapse
Affiliation(s)
- Liang‐Jun Yan
- Department of Pharmaceutical SciencesCollege of PharmacyUniversity of North Texas Health Science CenterFort WorthTexasUSA
| |
Collapse
|
46
|
Coulibaly SM, Mesfioui A, Berkiks I, Ennaciri A, Chahirou Y, Diagana Y, Ouichou A, El Midaoui A, El Hessni A. Effects of the Methyl Donors Supplementation on Hippocampal Oxidative Stress, Depression and Anxiety in Chronically High Fructose-treated Rats. Neuroscience 2021; 476:1-11. [PMID: 34543673 DOI: 10.1016/j.neuroscience.2021.09.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 09/05/2021] [Accepted: 09/09/2021] [Indexed: 12/18/2022]
Abstract
Evidence suggests that oxidative stress plays an important role in the development of anxiety and depression. The aim of the present study was to investigate whether methyl donors supplementation could exert beneficial effects on hippocampal oxidative stress, anxiety and depression in chronically high fructose-treated rats, a new animal model of anxiety and mood disorders. Rats were divided into two groups and treated for 10 weeks as follows: Group 1 represents the control group and Group 2 was treated with 23% fructose. After 10 weeks, the fructose-fed animals were divided into two groups and treated for 8 weeks as follows: Group 2 continued to receive fructose while Group 3 was treated with methyl donors and fructose. High fructose-fed rats showed increases in glucose, triglycerides, total cholesterol as well as in the final body weight and the adipose tissue weight. High fructose induced anxiety- and depression-like behaviors. High fructose caused an increase of the nitrite content and the Malondialdehyde (MDA) levels in the hippocampus tissue in association with an induction of damage in the dorsal hippocampus neurons. The 8-weeks dietary supplementation with methyl donors normalized the depression-like behavior, oxidative stress in the hippocampus, reversed the damage observed in the hippocampal neurons. These findings demonstrate that high fructose induced depression in association with the induction of a hippocampal oxidative stress. The anti-depressive action of methyl donors appears to be associated to their anti-oxidative properties since they normalized the nitrite content and the MDA levels at the hippocampus in the high fructose-fed female rats.
Collapse
Affiliation(s)
- Sidi Mohamed Coulibaly
- Laboratory of Genetics, Neuro-endocrinology and Biotechnology, Department of Life Sciences, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| | - Abdelhalem Mesfioui
- Laboratory of Genetics, Neuro-endocrinology and Biotechnology, Department of Life Sciences, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| | - Inssaf Berkiks
- Laboratoire Biologie et Santé, FSK Kénitra and Division of Immunology, University of Cape Town Medical School, South Africa
| | - Abdeljabbar Ennaciri
- Laboratory of Genetics, Neuro-endocrinology and Biotechnology, Department of Life Sciences, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| | - Yassine Chahirou
- Laboratory of Genetics, Neuro-endocrinology and Biotechnology, Department of Life Sciences, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| | - Yacouba Diagana
- Faculté des sciences, Université Noukchott Al Aasriya, Mauritania
| | - Ali Ouichou
- Laboratory of Genetics, Neuro-endocrinology and Biotechnology, Department of Life Sciences, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| | - Adil El Midaoui
- Research Team "Biology, Environment and Health", Department of Biology, FST Errachidia, Moulay Ismail University of Meknes, Morocco.
| | - Aboubaker El Hessni
- Laboratory of Genetics, Neuro-endocrinology and Biotechnology, Department of Life Sciences, Faculty of Sciences, Ibn Tofail University, Kenitra, Morocco
| |
Collapse
|
47
|
Ghergurovich JM, Xu X, Wang JZ, Yang L, Ryseck RP, Wang L, Rabinowitz JD. Methionine synthase supports tumour tetrahydrofolate pools. Nat Metab 2021; 3:1512-1520. [PMID: 34799699 PMCID: PMC9284419 DOI: 10.1038/s42255-021-00465-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 09/01/2021] [Indexed: 01/02/2023]
Abstract
Mammalian cells require activated folates to generate nucleotides for growth and division. The most abundant circulating folate species is 5-methyl tetrahydrofolate (5-methyl-THF), which is used to synthesize methionine from homocysteine via the cobalamin-dependent enzyme methionine synthase (MTR). Cobalamin deficiency traps folates as 5-methyl-THF. Here, we show using isotope tracing that MTR is only a minor source of methionine in cell culture, tissues or xenografted tumours. Instead, MTR is required for cells to avoid folate trapping and assimilate 5-methyl-THF into other folate species. Under conditions of physiological extracellular folates, genetic MTR knockout in tumour cells leads to folate trapping, purine synthesis stalling, nucleotide depletion and impaired growth in cell culture and as xenografts. These defects are rescued by free folate but not one-carbon unit supplementation. Thus, MTR plays a crucial role in liberating THF for use in one-carbon metabolism.
Collapse
Affiliation(s)
- Jonathan M Ghergurovich
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Xincheng Xu
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Joshua Z Wang
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Lifeng Yang
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Rolf-Peter Ryseck
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Lin Wang
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
- Department of Chemistry, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
48
|
Du Y, Zhang Y, Huang M, Wang S, Wang J, Liao K, Wu X, Zhou Q, Zhang X, Wu YD, Peng T. Systematic investigation of the aza-Cope reaction for fluorescence imaging of formaldehyde in vitro and in vivo. Chem Sci 2021; 12:13857-13869. [PMID: 34760171 PMCID: PMC8549814 DOI: 10.1039/d1sc04387k] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/22/2021] [Indexed: 01/02/2023] Open
Abstract
Increasing evidence has highlighted the endogenous production of formaldehyde (FA) in a variety of fundamental biological processes and its involvement in many disease conditions ranging from cancer to neurodegeneration. To examine the physiological and pathological relevance and functions of FA, fluorescent probes for FA imaging in live biological samples are of great significance. Herein we report a systematic investigation of 2-aza-Cope reactions between homoallylamines and FA for identification of a highly efficient 2-aza-Cope reaction moiety and development of fluorescent probes for imaging FA in living systems. By screening a set of N-substituted homoallylamines and comparing them to previously reported homoallylamine structures for reaction with FA, we found that N-p-methoxybenzyl homoallylamine exhibited an optimal 2-aza-Cope reactivity to FA. Theoretical calculations were then performed to demonstrate that the N-substituent on homoallylamine greatly affects the condensation with FA, which is more likely the rate-determining step. Moreover, the newly identified optimal N-p-methoxybenzyl homoallylamine moiety with a self-immolative β-elimination linker was generally utilized to construct a series of fluorescent probes with varying excitation/emission wavelengths for sensitive and selective detection of FA in aqueous solutions and live cells. Among these probes, the near-infrared probe FFP706 has been well demonstrated to enable direct fluorescence visualization of steady-state endogenous FA in live mouse brain tissues and elevated FA levels in a mouse model of breast cancer. This study provides the optimal aza-Cope reaction moiety for FA probe development and new chemical tools for fluorescence imaging and biological investigation of FA in living systems.
Collapse
Affiliation(s)
- Yimeng Du
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Shenzhen 518055 China
| | - Yuqing Zhang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Shenzhen 518055 China
| | - Meirong Huang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Shenzhen 518055 China
| | - Shushu Wang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Shenzhen 518055 China
| | - Jianzheng Wang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Shenzhen 518055 China
| | - Kongke Liao
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Shenzhen 518055 China
| | - Xiaojun Wu
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Shenzhen 518055 China
| | - Qiang Zhou
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Shenzhen 518055 China
| | - Xinhao Zhang
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Shenzhen 518055 China
- Shenzhen Bay Laboratory Shenzhen 518132 China
| | - Yun-Dong Wu
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Shenzhen 518055 China
- Shenzhen Bay Laboratory Shenzhen 518132 China
| | - Tao Peng
- State Key Laboratory of Chemical Oncogenomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Shenzhen 518055 China
| |
Collapse
|
49
|
Bae S, Kamynina E, Guetterman HM, Farinola AF, Caudill MA, Berry RJ, Cassano PA, Stover PJ. Provision of folic acid for reducing arsenic toxicity in arsenic-exposed children and adults. Cochrane Database Syst Rev 2021; 10:CD012649. [PMID: 34661903 PMCID: PMC8522704 DOI: 10.1002/14651858.cd012649.pub2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Arsenic is a common environmental toxin. Exposure to arsenic (particularly its inorganic form) through contaminated food and drinking water is an important public health burden worldwide, and is associated with increased risk of neurotoxicity, congenital anomalies, cancer, and adverse neurodevelopment in children. Arsenic is excreted following methylation reactions, which are mediated by folate. Provision of folate through folic acid supplements could facilitate arsenic methylation and excretion, thereby reducing arsenic toxicity. OBJECTIVES To assess the effects of provision of folic acid (through fortified foods or supplements), alone or in combination with other nutrients, in lessening the burden of arsenic-related health outcomes and reducing arsenic toxicity in arsenic-exposed populations. SEARCH METHODS In September 2020, we searched CENTRAL, MEDLINE, Embase, 10 other international databases, nine regional databases, and two trials registers. SELECTION CRITERIA Randomised controlled trials (RCTs) and quasi-RCTs comparing the provision of folic acid (at any dose or duration), alone or in combination with other nutrients or nutrient supplements, with no intervention, placebo, unfortified food, or the same nutrient or supplements without folic acid, in arsenic-exposed populations of all ages and genders. DATA COLLECTION AND ANALYSIS We used standard methodological procedures expected by Cochrane. MAIN RESULTS We included two RCTs with 822 adults exposed to arsenic-contaminated drinking water in Bangladesh. The RCTs compared 400 µg/d (FA400) or 800 µg/d (FA800) folic acid supplements, given for 12 or 24 weeks, with placebo. One RCT, a multi-armed trial, compared FA400 plus creatine (3 g/d) to creatine alone. We judged both RCTs at low risk of bias in all domains. Due to differences in co-intervention, arsenic exposure, and participants' nutritional status, we could not conduct meta-analyses, and therefore, provide a narrative description of the data. Neither RCT reported on cancer, all-cause mortality, neurocognitive function, or congenital anomalies. Folic acid supplements alone versus placebo Blood arsenic. In arsenic-exposed individuals, FA likely reduces blood arsenic concentrations compared to placebo (2 studies, 536 participants; moderate-certainty evidence). For folate-deficient and folate-replete participants who received arsenic-removal water filters as a co-intervention, FA800 reduced blood arsenic levels more than placebo (percentage change (%change) in geometric mean (GM) FA800 -17.8%, 95% confidence intervals (CI) -25.0 to -9.8; placebo GM -9.5%, 95% CI -16.5 to -1.8; 1 study, 406 participants). In one study with 130 participants with low baseline plasma folate, FA400 reduced total blood arsenic (%change FA400 mean (M) -13.62%, standard error (SE) ± 2.87; placebo M -2.49%, SE ± 3.25), and monomethylarsonic acid (MMA) concentrations (%change FA400 M -22.24%, SE ± 2.86; placebo M -1.24%, SE ± 3.59) more than placebo. Inorganic arsenic (InAs) concentrations reduced in both groups (%change FA400 M -18.54%, SE ± 3.60; placebo M -10.61%, SE ± 3.38). There was little to no change in dimethylarsinic acid (DMA) in either group. Urinary arsenic. In arsenic-exposed individuals, FA likely reduces the proportion of total urinary arsenic excreted as InAs (%InAs) and MMA (%MMA) and increases the proportion excreted as DMA (%DMA) to a greater extent than placebo (2 studies, 546 participants; moderate-certainty evidence), suggesting that FA enhances arsenic methylation. In a mixed folate-deficient and folate-replete population (1 study, 352 participants) receiving arsenic-removal water filters as a co-intervention, groups receiving FA had a greater decrease in %InAs (within-person change FA400 M -0.09%, 95% CI -0.17 to -0.01; FA800 M -0.14%, 95% CI -0.21 to -0.06; placebo M 0.05%, 95% CI 0.00 to 0.10), a greater decrease in %MMA (within-person change FA400 M -1.80%, 95% CI -2.53 to -1.07; FA800 M -2.60%, 95% CI -3.35 to -1.85; placebo M 0.15%, 95% CI -0.37 to 0.68), and a greater increase in %DMA (within-person change FA400 M 3.25%, 95% CI 1.81 to 4.68; FA800 M 4.57%, 95% CI 3.20 to 5.95; placebo M -1.17%, 95% CI -2.18 to -0.17), compared to placebo. In 194 participants with low baseline plasma folate, FA reduced %InAs (%change FA400 M -0.31%, SE ± 0.04; placebo M -0.13%, SE ± 0.04) and %MMA (%change FA400 M -2.6%, SE ± 0.37; placebo M -0.71%, SE ± 0.43), and increased %DMA (%change FA400 M 5.9%, SE ± 0.82; placebo M 2.14%, SE ± 0.71), more than placebo. Plasma homocysteine: In arsenic-exposed individuals, FA400 likely reduces homocysteine concentrations to a greater extent than placebo (2 studies, 448 participants; moderate-certainty evidence), in the mixed folate-deficient and folate-replete population receiving arsenic-removal water filters as a co-intervention (%change in GM FA400 -23.4%, 95% CI -27.1 to -19.5; placebo -1.3%, 95% CI -5.3 to 3.1; 1 study, 254 participants), and participants with low baseline plasma folate (within-person change FA400 M -3.06 µmol/L, SE ± 3.51; placebo M -0.05 µmol/L, SE ± 4.31; 1 study, 194 participants). FA supplements plus other nutrient supplements versus nutrient supplements alone In arsenic-exposed individuals who received arsenic-removal water filters as a co-intervention, FA400 plus creatine may reduce blood arsenic concentrations more than creatine alone (%change in GM FA400 + creatine -14%, 95% CI -22.2 to -5.0; creatine -7.0%, 95% CI -14.8 to 1.5; 1 study, 204 participants; low-certainty evidence); may not change urinary arsenic methylation indices (FA400 + creatine: %InAs M 13.2%, SE ± 7.0; %MMA M 10.8, SE ± 4.1; %DMA M 76, SE ± 7.8; creatine: %InAs M 14.8, SE ± 5.5; %MMA M 12.8, SE ± 4.0; %DMA M 72.4, SE ±7.6; 1 study, 190 participants; low-certainty evidence); and may reduce homocysteine concentrations to a greater extent (%change in GM FA400 + creatinine -21%, 95% CI -25.2 to -16.4; creatine -4.3%, 95% CI -9.0 to 0.7; 1 study, 204 participants; low-certainty evidence) than creatine alone. AUTHORS' CONCLUSIONS There is moderate-certainty evidence that FA supplements may benefit blood arsenic concentration, urinary arsenic methylation profiles, and plasma homocysteine concentration versus placebo. There is low-certainty evidence that FA supplements plus other nutrients may benefit blood arsenic and plasma homocysteine concentrations versus nutrients alone. No studies reported on cancer, all-cause mortality, neurocognitive function, or congenital anomalies. Given the limited number of RCTs, more studies conducted in diverse settings are needed to assess the effects of FA on arsenic-related health outcomes and arsenic toxicity in arsenic-exposed adults and children.
Collapse
Affiliation(s)
- Sajin Bae
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Elena Kamynina
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | | | - Adetutu F Farinola
- Faculty of Public Health, Department of Human Nutrition and Dietetics, University of Ibadan, Ibadan, Nigeria
| | - Marie A Caudill
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Robert J Berry
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | | |
Collapse
|
50
|
Fiddler JL, Xiu Y, Blum JE, Lamarre SG, Phinney WN, Stabler SP, Brosnan ME, Brosnan JT, Thalacker-Mercer AE, Field MS. Reduced Shmt2 Expression Impairs Mitochondrial Folate Accumulation and Respiration, and Leads to Uracil Accumulation in Mouse Mitochondrial DNA. J Nutr 2021; 151:2882-2893. [PMID: 34383924 DOI: 10.1093/jn/nxab211] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/24/2021] [Accepted: 06/08/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Adequate cellular thymidylate (dTMP) pools are essential for preservation of nuclear and mitochondrial genome stability. Previous studies have indicated that disruption in nuclear dTMP synthesis leads to increased uracil misincorporation into DNA, affecting genome stability. To date, the effects of impaired mitochondrial dTMP synthesis in nontransformed tissues have been understudied. OBJECTIVES This study aimed to determine the effects of decreased serine hydroxymethyltransferase 2 (Shmt2) expression and dietary folate deficiency on mitochondrial DNA (mtDNA) integrity and mitochondrial function in mouse tissues. METHODS Liver mtDNA content, and uracil content in liver mtDNA, were measured in Shmt2+/- and Shmt2+/+ mice weaned onto either a folate-sufficient control diet (2 mg/kg folic acid; C) or a modified diet lacking folic acid (0 mg/kg folic acid) for 7 wk. Shmt2+/- and Shmt2+/+ mouse embryonic fibroblast (MEF) cells were cultured in defined culture medium containing either 0 or 25 nM folate (6S-5-formyl-tetrahydrofolate, folinate) to assess proliferative capacity and mitochondrial function. Chi-square tests, linear mixed models, and 2-factor ANOVA with Tukey post hoc analyses were used to analyze data. RESULTS Shmt2 +/- mice exhibited a 48%-67% reduction in SHMT2 protein concentrations in tissues. Interestingly, Shmt2+/- mice consuming the folate-sufficient C diet exhibited a 25% reduction in total folate in liver mitochondria. There was also a >20-fold increase in uracil in liver mtDNA in Shmt2+/- mice consuming the C diet, and dietary folate deficiency also increased uracil content in mouse liver mtDNA from both Shmt2+/+ and Shmt2+/- mice. Furthermore, decreased Shmt2 expression in MEF cells reduced cell proliferation, mitochondrial membrane potential, and oxygen consumption rate. CONCLUSIONS This study demonstrates that Shmt2 heterozygosity and dietary folate deficiency impair mitochondrial dTMP synthesis in mice, as evidenced by the increased uracil in mtDNA. In addition, Shmt2 heterozygosity impairs mitochondrial function in MEF cells. These findings suggest that elevated uracil in mtDNA may impair mitochondrial function.
Collapse
Affiliation(s)
- Joanna L Fiddler
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Yuwen Xiu
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Jamie E Blum
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Simon G Lamarre
- Department of Biology, University of Moncton, Moncton, New Brunswick, Canada
| | | | - Sally P Stabler
- Department of Medicine, University of Colorado, Aurora, CO, USA
| | - Margaret E Brosnan
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - John T Brosnan
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Anna E Thalacker-Mercer
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Martha S Field
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| |
Collapse
|