1
|
Ledet Iii LF, Plaisance CJ, Daniel CP, Wagner MJ, Alvarez I, Burroughs CR, Rieger R, Siddaiah H, Ahmadzadeh S, Shekoohi S, Kaye AD, Varrassi G. Spina Bifida Prevention: A Narrative Review of Folic Acid Supplements for Childbearing Age Women. Cureus 2024; 16:e53008. [PMID: 38406082 PMCID: PMC10894015 DOI: 10.7759/cureus.53008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 01/26/2024] [Indexed: 02/27/2024] Open
Abstract
Neural tube defects (NTDs) are malformations that occur during embryonic development, and they account for most central nervous system birth anomalies. Genetic and environmental factors have been shown to play a role in the etiology of NTDs. The different types of NTDs are classified according to anatomic location and severity of the defect, with most of the neural axis anomalies occurring in the caudal spinal or cranial areas. Spina bifida is a type of NTD that is characterized by an opening in the vertebral arch, and the level of severity is determined by the extent to which the neural tissue protrudes through the opened arch(es). Prevention of NTDs by administration of folic acid has been studied and described in the literature, yet there are approximately 300,000 cases of NTDs that occur annually, with 88,000 deaths occurring per year worldwide. A daily intake of at least 400 μg of folic acid is recommended especially for women of childbearing age. To provide the benefits of folic acid, prenatal vitamins are recommended in pregnancy, and many countries have been fortifying foods such as cereal grain products with folic acid; however, not all countries have instituted folic acid fortification programs. The present investigation includes a description of the pharmacology of folic acid, neural tube formation, defects such as spina bifida, and the relevance of folic acid to developing spina bifida. Women's knowledge and awareness of folic acid regarding its importance in the prevention of spina bifida is a major factor in reducing incidence worldwide.
Collapse
Affiliation(s)
- Lloyd F Ledet Iii
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Connor J Plaisance
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Charles P Daniel
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Maxwell J Wagner
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Ivan Alvarez
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Caroline R Burroughs
- School of Medicine, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Ross Rieger
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Harish Siddaiah
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Shahab Ahmadzadeh
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Sahar Shekoohi
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | - Alan D Kaye
- Department of Anesthesiology, Louisiana State University Health Sciences Center, Shreveport, USA
| | | |
Collapse
|
2
|
Townsend LN, Clarke H, Maddison D, Jones KM, Amadio L, Jefferson A, Chughtai U, Bis DM, Züchner S, Allen ND, Van der Goes van Naters W, Peters OM, Smith GA. Cdk12 maintains the integrity of adult axons by suppressing actin remodeling. Cell Death Discov 2023; 9:348. [PMID: 37730761 PMCID: PMC10511712 DOI: 10.1038/s41420-023-01642-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/25/2023] [Accepted: 09/07/2023] [Indexed: 09/22/2023] Open
Abstract
The role of cyclin-dependent kinases (CDKs) that are ubiquitously expressed in the adult nervous system remains unclear. Cdk12 is enriched in terminally differentiated neurons where its conical role in the cell cycle progression is redundant. We find that in adult neurons Cdk12 acts a negative regulator of actin formation, mitochondrial dynamics and neuronal physiology. Cdk12 maintains the size of the axon at sites proximal to the cell body through the transcription of homeostatic enzymes in the 1-carbon by folate pathway which utilize the amino acid homocysteine. Loss of Cdk12 leads to elevated homocysteine and in turn leads to uncontrolled F-actin formation and axonal swelling. Actin remodeling further induces Drp1-dependent fission of mitochondria and the breakdown of axon-soma filtration barrier allowing soma restricted cargos to enter the axon. We demonstrate that Cdk12 is also an essential gene for long-term neuronal survival and loss of this gene causes age-dependent neurodegeneration. Hyperhomocysteinemia, actin changes, and mitochondrial fragmentation are associated with several neurodegenerative conditions such as Alzheimer's disease and we provide a candidate molecular pathway to link together such pathological events.
Collapse
Affiliation(s)
- L N Townsend
- School of Biosciences, Cardiff University, Cardiff, CF24 4HQ, UK
| | - H Clarke
- School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
- UK Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - D Maddison
- School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
- UK Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - K M Jones
- School of Biosciences, Cardiff University, Cardiff, CF24 4HQ, UK
| | - L Amadio
- School of Biosciences, Cardiff University, Cardiff, CF24 4HQ, UK
- School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
- UK Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - A Jefferson
- School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
- UK Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - U Chughtai
- School of Biosciences, Cardiff University, Cardiff, CF24 4HQ, UK
- School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK
- UK Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - D M Bis
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, FL, USA
| | - S Züchner
- John P. Hussman Institute for Human Genomics, University of Miami, Miami, FL, USA
- Dr. John T. Macdonald Foundation Department of Human Genetics, University of Miami, Miami, FL, USA
| | - N D Allen
- School of Biosciences, Cardiff University, Cardiff, CF24 4HQ, UK
| | | | - O M Peters
- School of Biosciences, Cardiff University, Cardiff, CF24 4HQ, UK
- UK Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - G A Smith
- School of Medicine, Cardiff University, Cardiff, CF24 4HQ, UK.
- UK Dementia Research Institute, Cardiff University, Cardiff, CF24 4HQ, UK.
| |
Collapse
|
3
|
Guo X, Sha Y, Pu X, Xu Y, Yao L, Liu X, He Y, Hu J, Wang J, Li S, Chen G. Coevolution of Rumen Epithelial circRNAs with Their Microbiota and Metabolites in Response to Cold-Season Nutritional Stress in Tibetan Sheep. Int J Mol Sci 2022; 23:ijms231810488. [PMID: 36142400 PMCID: PMC9499677 DOI: 10.3390/ijms231810488] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
This study explores the effects of the coevolution of the host genome (the first genome) and gut microbiome (the second genome) on nutrition stress in Tibetan sheep during the cold season. The rumen epithelial tissue of six Tibetan sheep (Oula-type) was collected as experimental samples during the cold and warm seasons and the study lasted for half a year. The cDNA library was constructed and subjected to high-throughput sequencing. The circRNAs with significant differential expression were identified through bioinformatics analysis and functional prediction, and verified by real-time quantitative PCR (qRT-PCR). The results showed that a total of 56 differentially expressed (DE) circRNAs of rumen epithelial tissue were identified using RNA-seq technology, among which 29 were significantly upregulated in the cold season. The circRNA-miRNA regulatory network showed that DE circRNAs promoted the adaptation of Tibetan sheep in the cold season by targeting miR-150 and oar-miR-370-3p. The results of correlation analysis among circRNAs, microbiota, and metabolites showed that the circRNA NC_040275.1:28680890|28683112 had a very significant positive correlation with acetate, propionate, butyrate, and total volatile fatty acid (VFA) (p < 0.01), and had a significant positive correlation with Ruminococcus-1 (p < 0.05). In addition, circRNA NC_040256.1:78451819|78454934 and metabolites were enriched in the same KEGG pathway biosynthesis of amino acids (ko01230). In conclusion, the host genome and rumen microbiome of Tibetan sheep co-encoded a certain glycoside hydrolase (β-glucosidase) and coevolved efficient VFA transport functions and amino acid anabolic processes; thus, helping Tibetan sheep adapt to nutrient stress in the cold season in high-altitude areas.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiu Liu
- Correspondence: (X.L.); (G.C.)
| | | | | | | | | | | |
Collapse
|
4
|
Turck D, Bohn T, Castenmiller J, De Henauw S, Hirsch‐Ernst KI, Knutsen HK, Maciuk A, Mangelsdorf I, McArdle HJ, Naska A, Peláez C, Siani A, Thies F, Tsabouri S, Vinceti M, Cubadda F, Abrahantes JC, Dumas C, Ercolano V, Titz A, Pentieva K. Conversion of calcium-l-methylfolate and (6S)-5-methyltetrahydrofolic acid glucosamine salt into dietary folate equivalents. EFSA J 2022; 20:e07452. [PMID: 36034319 PMCID: PMC9399872 DOI: 10.2903/j.efsa.2022.7452] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Following a request from the European Commission, the EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA) was asked to deliver a scientific opinion on the conversion of calcium-l-methylfolate and (6S)-5-methyltetrahydrofolic acid glucosamine salt (collectively called 5-MTHF hereafter) into dietary folate equivalents (DFE). Following a systematic review, the conclusions of the opinion are based on one intervention study in adults for intakes < 400 μg/day and three intervention studies in adults for intakes ≥ 400 μg/day. At intakes below 400 μg/day, folic acid (FA) is assumed to be linearly related to responses of biomarkers of intake and status and is an appropriate comparator for deriving a DFE conversion factor for 5-MTHF. It is proposed to use the same factor as for folic acid for conversion of 5-MTHF into DFE for intakes < 400 μg/day. As such intake levels are unlikely to be exceeded through fortified food consumption, the conversion factor of 1.7 relative to natural food folate (NF) could be applied to 5-MTHF added to foods and to food supplements providing < 400 μg/day. At 400 μg/day, 5-MTHF was found to be more bioavailable than folic acid and a conversion factor of 2 is proposed for this intake level and for higher intakes. The derived DFE equations are DFE = NF + 1.7 × FA + 1.7 × 5-MTHF for fortified foods and food supplements providing intakes < 400 μg/day; and DFE = NF + 1.7 × FA + 2.0 × 5-MTHF for food supplements providing intakes ≥ 400 μg/day. Although this assessment applies to calcium-L-methylfolate and 5-MTHF glucosamine salt, it is considered that the influence of the cation on bioavailability is likely to be within the margin of error of the proposed DFE equations. Therefore, the proposed equations can also be applied to 5-MTHF associated with other cations.
Collapse
|
5
|
Liang S, Liu X, Zhao J, Liu R, Huang X, Liu Y, Yang X, Yang X. Effects of high-dose folic acid on protein metabolism in breast muscle and performance of broilers. Poult Sci 2022; 101:101935. [PMID: 35961252 PMCID: PMC9382563 DOI: 10.1016/j.psj.2022.101935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 04/24/2022] [Accepted: 04/26/2022] [Indexed: 11/19/2022] Open
Abstract
Attaining the optimal feed conversion ratio is the unaltered goal for poultry breeding, meat yield is one of the vital reference indexes for that. Folic acid is involved in protein metabolism by acting as a transmitter of one carbon unit, and the detail mechanism for the high-dose folic acid on growth of broiler skeletal muscle is still unclarified. The present study was conducted to investigate the effect and regulatory mechanism of folic acid on deposition and metabolism of protein in broiler breast muscle. A total of 196 one-day-old AA broilers were randomly assigned to 2 treatment groups. The chicks were fed corn-soybean diet with folic acid levels of 1.3 mg/kg (CON) or 13 mg/kg (FA), respectively. The results showed that high dose of folic acid significantly increased the body weight gain, average daily gain, average daily feed intake, and feed conversion ratio of broilers during 1 to 42 d. Compared with control group, folic acid statistically augmented the breast muscle ratio of broilers at 42 d, abdominal fat percentage was also decreased in FA group. Folic acid significantly increased the gene expression of folate receptor (FR) in duodenum and jejunum at 21 d, and its relative expression in jejunum of broilers at 42 d. Furthermore, relative expression of myogenin in broiler breast muscle was upregulated in folic acid group. Folic acid supplementation significantly enhanced the protein expression of phosphorylated serine/threonine kinase (AKT) and ribosomal protein S6 kinase 1 (S6K1) in the breast muscle of broilers at 21 d and 42 d. In conclusion, the results proved that high-dose folic acid activated the AKT/mammalian target of rapamycin (mTOR) pathway and increased the activity of phosphorylation of S6K1, thereby regulating the protein deposition in breast muscle. Meanwhile, the gene expression of the myogenic determinant factor was upregulated by folic acid and then promoted the growth of breast muscle. Consequently, the growth performance, meat production and feeding efficiency were improved of broilers by adding folic acid at 13 mg/kg.
Collapse
Affiliation(s)
- Saisai Liang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xinshuai Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Jianfei Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Rui Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xinhuo Huang
- Nano Vitamin Engineering Research Center of Shaanxi Province, Xi'an, China
| | - Yanli Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xiaojun Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Xin Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China.
| |
Collapse
|
6
|
Quevedo-Ocampo J, Escobedo-Calvario A, Souza-Arroyo V, Miranda-Labra RU, Bucio-Ortiz L, Gutiérrez-Ruiz MC, Chávez-Rodríguez L, Gomez-Quiroz LE. Folate Metabolism in Hepatocellular Carcinoma. What Do We Know So Far? Technol Cancer Res Treat 2022; 21:15330338221144446. [PMID: 36503290 DOI: 10.1177/15330338221144446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cancer cells are characterized by accelerated proliferation and an outstanding adaptation of their metabolic pathways to meet energy demands. The folate cycle, also known as folate metabolism or one-carbon metabolism, through enzymatic interconversions, provides metabolites necessary for nucleotide synthesis, methylation, and reduction power, helping to maintain the high rate of proliferation; therefore, the study of this metabolic pathway is of great importance in the study of cancer. Moreover, multiple enzymes involved in this cycle have been implicated in different types of cancer, corroborating the cell's adaptations under this pathology. During the last decade, nonalcoholic fatty liver disease has emerged as the leading etiology related to the rise in the incidence and deaths of hepatocellular carcinoma. Specifically, cholesterol accumulation has been a determinant promoter of tumor formation, with solid evidence that an enriched-cholesterol diet plays a crucial role in accelerating the development of an aggressive subtype of hepatocellular carcinoma compared to other models. In this review, we will discuss the most recent findings to understand the contribution of folate metabolism to cancer cells and tumor microenvironment while creating a link between the dynamics given by cholesterol and methylenetetrahydrofolate dehydrogenase 1-like, a key enzyme of the cycle located in the mitochondrial compartment.
Collapse
Affiliation(s)
- Jaqueline Quevedo-Ocampo
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Posgrado en Biología Experimental, DCBS, Universidad Autónoma Metrolitana-Iztapalapa, Mexico City, Mexico
| | - Alejandro Escobedo-Calvario
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Posgrado en Biología Experimental, DCBS, Universidad Autónoma Metrolitana-Iztapalapa, Mexico City, Mexico
| | - Verónica Souza-Arroyo
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional IIB/UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Roxana U Miranda-Labra
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional IIB/UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Leticia Bucio-Ortiz
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional IIB/UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - María C Gutiérrez-Ruiz
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional IIB/UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Lisette Chávez-Rodríguez
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Posgrado en Biología Experimental, DCBS, Universidad Autónoma Metrolitana-Iztapalapa, Mexico City, Mexico
| | - Luis E Gomez-Quiroz
- Área de Medicina Experimental y Traslacional, Departamento de Ciencias de la Salud, 27786Universidad Autónoma Metropolitana-Iztapalapa, Mexico City, Mexico.,Laboratorio de Medicina Experimental, Unidad de Medicina Traslacional IIB/UNAM, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| |
Collapse
|
7
|
Narawa T, Narita Y, Hosokawa S, Itoh T. Functional role of serine 318 of the proton-coupled folate transporter in methotrexate transport. Drug Metab Pharmacokinet 2021; 41:100421. [PMID: 34619546 DOI: 10.1016/j.dmpk.2021.100421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 08/08/2021] [Accepted: 09/16/2021] [Indexed: 10/20/2022]
Abstract
This study revealed the importance of serine 318 (S318) residue for proton-coupled folate transporter (PCFT, SLC46A1) functioning. Substitution of S318 with arginine or lysine impaired transport of methotrexate (MTX), but substitution with alanine (has a simple side chain structure), or cysteine (structurally similar to serine), had no significant effect on MTX transport. The initial uptake rate of MTX by S318A and S318C mutant at pH 5.0, followed by Michaelis-Menten kinetics with a Km value of approximately 2.3 μM (for S318A) and 2.9 μM (for S318C), was similar to that of the wild-type. The normalized Vmax value of the S318A mutant, calculated by dividing the Vmax value by the Western blot protein band's relative intensity, was approximately 2-fold greater than that of the wild-type. The normalized Vmax value of the S318C mutant was approximately 0.8-fold smaller than the wild-type. Results obtained showed that the substitution of S318 with basic amino acid residues results in the loss of transport activity, even though PCFT mutants are expressed at the cell membrane. Alternatively, the substitution of S318 with neutral amino acids did not significantly affect the transport function of PCFT.
Collapse
Affiliation(s)
- Tomoya Narawa
- Laboratory of Pharmaceutics, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Tokyo, 108-8641, Japan.
| | - Yuuki Narita
- Laboratory of Pharmaceutics, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Tokyo, 108-8641, Japan
| | - Sayuri Hosokawa
- Laboratory of Pharmaceutics, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Tokyo, 108-8641, Japan
| | - Tomoo Itoh
- Laboratory of Pharmaceutics, School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Tokyo, 108-8641, Japan
| |
Collapse
|
8
|
Shulpekova Y, Nechaev V, Kardasheva S, Sedova A, Kurbatova A, Bueverova E, Kopylov A, Malsagova K, Dlamini JC, Ivashkin V. The Concept of Folic Acid in Health and Disease. Molecules 2021; 26:molecules26123731. [PMID: 34207319 PMCID: PMC8235569 DOI: 10.3390/molecules26123731] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/12/2021] [Accepted: 06/17/2021] [Indexed: 12/18/2022] Open
Abstract
Folates have a pterine core structure and high metabolic activity due to their ability to accept electrons and react with O-, S-, N-, C-bounds. Folates play a role as cofactors in essential one-carbon pathways donating methyl-groups to choline phospholipids, creatine, epinephrine, DNA. Compounds similar to folates are ubiquitous and have been found in different animals, plants, and microorganisms. Folates enter the body from the diet and are also synthesized by intestinal bacteria with consequent adsorption from the colon. Three types of folate and antifolate cellular transporters have been found, differing in tissue localization, substrate affinity, type of transferring, and optimal pH for function. Laboratory criteria of folate deficiency are accepted by WHO. Severe folate deficiencies, manifesting in early life, are seen in hereditary folate malabsorption and cerebral folate deficiency. Acquired folate deficiency is quite common and is associated with poor diet and malabsorption, alcohol consumption, obesity, and kidney failure. Given the observational data that folates have a protective effect against neural tube defects, ischemic events, and cancer, food folic acid fortification was introduced in many countries. However, high physiological folate concentrations and folate overload may increase the risk of impaired brain development in embryogenesis and possess a growth advantage for precancerous altered cells.
Collapse
Affiliation(s)
- Yulia Shulpekova
- Department of Internal Diseases Propedeutics, Sechenov University, 119121 Moscow, Russia; (Y.S.); (V.N.); (S.K.); (A.S.); (A.K.); (E.B.); (V.I.)
| | - Vladimir Nechaev
- Department of Internal Diseases Propedeutics, Sechenov University, 119121 Moscow, Russia; (Y.S.); (V.N.); (S.K.); (A.S.); (A.K.); (E.B.); (V.I.)
| | - Svetlana Kardasheva
- Department of Internal Diseases Propedeutics, Sechenov University, 119121 Moscow, Russia; (Y.S.); (V.N.); (S.K.); (A.S.); (A.K.); (E.B.); (V.I.)
| | - Alla Sedova
- Department of Internal Diseases Propedeutics, Sechenov University, 119121 Moscow, Russia; (Y.S.); (V.N.); (S.K.); (A.S.); (A.K.); (E.B.); (V.I.)
| | - Anastasia Kurbatova
- Department of Internal Diseases Propedeutics, Sechenov University, 119121 Moscow, Russia; (Y.S.); (V.N.); (S.K.); (A.S.); (A.K.); (E.B.); (V.I.)
| | - Elena Bueverova
- Department of Internal Diseases Propedeutics, Sechenov University, 119121 Moscow, Russia; (Y.S.); (V.N.); (S.K.); (A.S.); (A.K.); (E.B.); (V.I.)
| | - Arthur Kopylov
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 119121 Moscow, Russia;
| | - Kristina Malsagova
- Biobanking Group, Branch of Institute of Biomedical Chemistry “Scientific and Education Center”, 119121 Moscow, Russia;
- Correspondence: ; Tel.: +7-499-764-9878
| | | | - Vladimir Ivashkin
- Department of Internal Diseases Propedeutics, Sechenov University, 119121 Moscow, Russia; (Y.S.); (V.N.); (S.K.); (A.S.); (A.K.); (E.B.); (V.I.)
| |
Collapse
|
9
|
Boss SD, Ametamey SM. Development of Folate Receptor-Targeted PET Radiopharmaceuticals for Tumor Imaging-A Bench-to-Bedside Journey. Cancers (Basel) 2020; 12:cancers12061508. [PMID: 32527010 PMCID: PMC7352234 DOI: 10.3390/cancers12061508] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/02/2020] [Accepted: 06/05/2020] [Indexed: 01/05/2023] Open
Abstract
The folate receptor-α (FR-α) is overexpressed in many epithelial cancers, including ovary, uterus, kidneys, breast, lung, colon and prostate carcinomas, but shows limited expression in normal tissues such as kidneys, salivary glands, choroid plexus and placenta. FR-α has therefore emerged as a promising target for the delivery of therapeutic and imaging agents to FR-positive tumors. A series of folate-based PET (positron emission tomography) radiopharmaceuticals have been developed for the selective targeting of FR-positive malignancies. This review provides an overview on the research progress made so far regarding the design, radiosynthesis and the utility of the folate-derived PET radioconjugates for targeting FR-positive tumors. For the most part, results from folate radioconjugates labeled with fluorine-18 (t1/2 = 109.8 min) and gallium-68 (t1/2 = 67.7 min) have been presented but folates labeled with "exotic" and new PET radionuclides such as copper-64 (t1/2 = 12.7 h), terbium-152 (t1/2 = 17.5 h), scandium-44 (t1/2 = 3.97 h), cobalt-55 (t1/2 = 17.5 h) and zirconium-89 (t1/2 = 78.4 h) are also discussed. For tumor imaging, none of the reported PET radiolabeled folates reported to date has made the complete bench-to-bedside journey except [18F]AzaFol, which made it to patients with metastatic ovarian and lung cancers in a multicenter first-in-human trial. In the near future, however, we expect more clinical trials with folate-based PET radiopharmaceuticals given the increasing clinical interest in imaging and the treatment of FR-related malignancies.
Collapse
Affiliation(s)
- Silvan D. Boss
- SWAN Isotopen AG, University Hospital Bern, 3010 Bern, Switzerland;
| | - Simon Mensah Ametamey
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, 8093 Zurich, Switzerland
- Correspondence:
| |
Collapse
|
10
|
Deng J, Chen L, Xue H, Zeng FX, Niu PG, Shi DH. Contribution of genetic polymorphism of methylene tetrahydrofolate reductase on the effect of methotrexate in ectopic pregnancy patients. J Clin Lab Anal 2019; 34:e23030. [PMID: 31502727 PMCID: PMC6977338 DOI: 10.1002/jcla.23030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/13/2019] [Accepted: 08/13/2019] [Indexed: 01/22/2023] Open
Abstract
Background Methotrexate (MTX) is the prior drug in ectopic pregnancy (EP). However, approximately 10% of patients suffer from failure by MTX therapy. Reduced folate carrier 1 (RFC1), methylene tetrahydrofolate reductase (MTHFR), and dihydrofolate reductase (DHFR) are involved in the transport and effects of MTX in vivo. In the present study, we aim to investigate the relationship between the genetic polymorphisms of RFC1, MTHFR, and DHFR and the clinical efficacy of MTX in tubal pregnancies. Methods 100 patients of EP were enrolled in this study. Polymorphisms of RFC1 G80A, MTHFR C677T, and DHFR A‐317G were genotyped. β‐hCG level was detected in day 0, 4, and 7 after MTX injection. Association of MTX efficacy and genetic polymorphisms was analyzed. Results Methylene tetrahydrofolate reductase C677T was associated with MTX treatment (P = .017). The success rate of first MTX injection was superior in patients with harboring mutation allele of MTHFR gene than that in patients with wild‐type gene (P = .001). However, there was no significant association between the polymorphisms of RFC1 G80A, DHFR A‐317G, and surgical treatment (P = .709 and .476, respectively). In addition, β‐hCG level decrement was not significantly changed by MTX injection with different polymorphisms of RFC1, MTHFR, and DHFR on either day 4 (P = .214, 0.197 and 0.270, respectively) or day 7 (P = .172, .554, and .726, respectively). Conclusion Our results suggested that the reliable indicator was polymorphism of MTHFR C677T in failure by MTX injection.
Collapse
Affiliation(s)
- Jie Deng
- Department of Pharmacy, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Li Chen
- Department of Gynecology, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Heng Xue
- Laboratory Medicine, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Fan-Xiang Zeng
- Department of Pharmacy, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Pei-Guang Niu
- Department of Pharmacy, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Dao-Hua Shi
- Department of Pharmacy, Fujian Provincial Maternity and Children's Hospital, Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
11
|
A pilot trial examining the absorption of oral forms of folate supplementation in a healthy population: A randomised control trial. ADVANCES IN INTEGRATIVE MEDICINE 2019. [DOI: 10.1016/j.aimed.2018.09.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
12
|
Diao W, Cai H, Chen L, Jin X, Liao X, Jia Z. Recent Advances in Prostate-Specific Membrane Antigen-Based Radiopharmaceuticals. Curr Top Med Chem 2019; 19:33-56. [PMID: 30706785 DOI: 10.2174/1568026619666190201100739] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 12/13/2018] [Accepted: 12/15/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND Prostate cancer (PCa) is the most common sex-related malignancy with high mortality in men worldwide. Prostate-specific membrane antigen (PSMA) is overexpressed on the surface of most prostate tumor cells and considered a valuable target for both diagnosis and therapy of prostate cancer. A series of radiolabeled agents have been developed based on the featured PSMA ligands in the previous decade and have demonstrated promising outcomes in clinical research of primary and recurrent PCa. Furthermore, the inspiring response and safety of lutetium-177-PSMA-617 (177Lu-PSMA-617) radiotherapy represent the potential for expanded therapeutic options for metastatic castration-resistant PCa. Retrospective cohort studies have revealed that radiolabeled PSMA agents are the mainstays of the current success, especially in detecting prostate cancer with metastasis and biochemical recurrence. OBJECTIVE This review is intended to present a comprehensive overview of the current literature on PSMA ligand-based agents for both radionuclide imaging and therapeutic approaches, with a focus on those that have been clinically adopted. CONCLUSION PSMA-based diagnosis and therapy hold great promise for improving the clinical management of prostate cancer.
Collapse
Affiliation(s)
- Wei Diao
- Department of Nuclear Medicine, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Huawei Cai
- Department of Nuclear Medicine, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Lihong Chen
- Department of Biochemistry & Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xi Jin
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Xinyang Liao
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Zhiyun Jia
- Department of Nuclear Medicine, West China Hospital, Sichuan University, 610041, Chengdu, China
| |
Collapse
|
13
|
Folic acid deficiency and vision: a review. Graefes Arch Clin Exp Ophthalmol 2019; 257:1573-1580. [PMID: 30919078 DOI: 10.1007/s00417-019-04304-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/10/2019] [Accepted: 03/20/2019] [Indexed: 02/03/2023] Open
Abstract
Folic acid (FA), also termed folate, is an essential vitamin for health at all ages since it participates in the biosynthesis of nucleotides, amino acids, neurotransmitters, and certain vitamins. It is therefore crucial for rapidly growing tissues such as those of the fetus. It is becoming clear that FA deficiency and impaired folate pathways are implicated in many diseases of both early life and old age. FA can be transported into the cell by the folate receptor, the reduced folate transporter, and proton-coupled folate transporter. Folate transport proteins are present in certain eye tissues, which explains why FA plays an important role in eye development. The purpose of this literature review is to investigate the evidence relating FA deficiency to eye diseases.
Collapse
|
14
|
Zheng Y, Cantley LC. Toward a better understanding of folate metabolism in health and disease. J Exp Med 2019; 216:253-266. [PMID: 30587505 PMCID: PMC6363433 DOI: 10.1084/jem.20181965] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/18/2018] [Accepted: 12/03/2018] [Indexed: 12/15/2022] Open
Abstract
Folate metabolism is crucial for many biochemical processes, including purine and thymidine monophosphate (dTMP) biosynthesis, mitochondrial protein translation, and methionine regeneration. These biochemical processes in turn support critical cellular functions such as cell proliferation, mitochondrial respiration, and epigenetic regulation. Not surprisingly, abnormal folate metabolism has been causally linked with a myriad of diseases. In this review, we provide a historical perspective, delve into folate chemistry that is often overlooked, and point out various missing links and underdeveloped areas in folate metabolism for future exploration.
Collapse
Affiliation(s)
- Yuxiang Zheng
- Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine, New York, NY
| | - Lewis C Cantley
- Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine, New York, NY
| |
Collapse
|
15
|
Sijilmassi O, López-Alonso JM, Del Río Sevilla A, Murillo González J, Barrio Asensio MDC. Biometric Alterations of Mouse Embryonic Eye Structures Due to Short-Term Folic Acid Deficiency. Curr Eye Res 2018; 44:428-435. [PMID: 30403890 DOI: 10.1080/02713683.2018.1545911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Folic acid (FA) is an essential nutrient for normal embryonic development. FA deficiency (FAD) in maternal diet increases the risk of several defects among the progeny, especially, neural tube defects. The eye begins its development from the neural tube; however, the relationship between FAD and ocular development in the offspring has been little explored and it isn't known how the FAD affects the formation of the eye. Our objective was to analyze the effect of maternal FAD on mouse embryos ocular biometry. METHODS Female mice C57/BL/6J were distributed into three different groups, according to the assigned diet: control group fed a standard FA diet (2 mg FA/kg), FAD group for short term fed (0 mg FA/kg + 1% succinylsulfathiazole) from the day after mating until day 14.5 of gestation, and FAD group for long term fed the same FA-deficient diet for 6 weeks prior mating and continued with this diet during gestation. A total of 57 embryos (19 embryos of each dietary group) at 14.5 gestational days were evaluated. As indicators of changes in ocular biometry, we analyze two parameters: area and circularity of the lens and whole eye, and the area of the retina. The program used in the treatment and selection of the areas of interest was ImageJ. The statistical analysis was performed by IBM SPSS Statistics 19. RESULTS Regarding the measures of the area, FA-deficient lenses and eyes were smaller than that of controls. We have also observed increase in the size of the neural retina, spatially, in embryos from females fed FAD diet during long term. On the other hand, as regard to circularity measures, we have seen that eyes and lenses were more circular than control. CONCLUSION Maternal FAD diet for a very short term generates morphological changes in ocular structures to the offspring.
Collapse
Affiliation(s)
- Ouafa Sijilmassi
- a Faculty of Optics and Optometry, Anatomy and Human Embryology Department , Universidad Complutense De Madrid , Madrid , Spain.,b Faculty of Optics and Optometry, Optics Department , Universidad Complutense De Madrid , Madrid , Spain
| | - José Manuel López-Alonso
- b Faculty of Optics and Optometry, Optics Department , Universidad Complutense De Madrid , Madrid , Spain
| | - Aurora Del Río Sevilla
- a Faculty of Optics and Optometry, Anatomy and Human Embryology Department , Universidad Complutense De Madrid , Madrid , Spain
| | - Jorge Murillo González
- c Faculty of medicine, Anatomy and Human Embryology Department , Universidad Complutense De Madrid , Madrid , Spain
| | - María Del Carmen Barrio Asensio
- a Faculty of Optics and Optometry, Anatomy and Human Embryology Department , Universidad Complutense De Madrid , Madrid , Spain
| |
Collapse
|
16
|
Wu YJ, Wang C, Wei W. The effects of DMARDs on the expression and function of P-gp, MRPs, BCRP in the treatment of autoimmune diseases. Biomed Pharmacother 2018; 105:870-878. [DOI: 10.1016/j.biopha.2018.06.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/24/2018] [Accepted: 06/04/2018] [Indexed: 12/17/2022] Open
|
17
|
Abbasi IHR, Abbasi F, Wang L, Abd El Hack ME, Swelum AA, Hao R, Yao J, Cao Y. Folate promotes S-adenosyl methionine reactions and the microbial methylation cycle and boosts ruminants production and reproduction. AMB Express 2018; 8:65. [PMID: 29687201 PMCID: PMC5913057 DOI: 10.1186/s13568-018-0592-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 04/13/2018] [Indexed: 12/14/2022] Open
Abstract
Folate has gained significant attention due to its vital role in biological methylation and epigenetic machinery. Folate, or vitamin (B9), is only produced through a de novo mechanism by plants and micro-organisms in the rumen of mature animals. Although limited research has been conducted on folate in ruminants, it has been noted that ruminal synthesis could not maintain folate levels in high yielding dairy animals. Folate has an essential role in one-carbon metabolism and is a strong antiproliferative agent. Folate increases DNA stability, being crucial for DNA synthesis and repair, the methylation cycle, and preventing oxidation of DNA by free radicals. Folate is also critical for cell division, metabolism of proteins, synthesis of purine and pyrimidine, and increasing the de novo delivery of methyl groups and S-adenosylmethionine. However, in ruminants, metabolism of B12 and B9 vitamins are closely connected and utilization of folate by cells is significantly affected by B12 vitamin concentration. Supplementation of folate through diet, particularly in early lactation, enhanced metabolic efficiency, lactational performance, and nutritional quality of milk. Impaired absorption, oxidative degradation, or deficient supply of folate in ruminants affects DNA stability, cell division, homocysteine remethylation to methionine, de novo synthesis of S-adenosylmethionine, and increases DNA hypomethylation, uracil misincorporation into DNA, chromosomal damage, abnormal cell growth, oxidative species, premature birth, low calf weight, placental tube defects, and decreases production and reproduction of ruminant animals. However, more studies are needed to overcome these problems and reduce enormous dietary supplement waste and impaired absorption of folate in ruminants. This review was aimed to highlight the vital role of folic acid in ruminants performance.
Collapse
|
18
|
Balashova OA, Visina O, Borodinsky LN. Folate action in nervous system development and disease. Dev Neurobiol 2018; 78:391-402. [PMID: 29380544 DOI: 10.1002/dneu.22579] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 01/24/2018] [Accepted: 01/24/2018] [Indexed: 01/04/2023]
Abstract
The vitamin folic acid has been recognized as a crucial environmental factor for nervous system development. From the early fetal stages of the formation of the presumptive spinal cord and brain to the maturation and maintenance of the nervous system during infancy and childhood, folate levels and its supplementation have been considered influential in the clinical outcome of infants and children affected by neurological diseases. Despite the vast epidemiological information recorded on folate function and neural tube defects, neural development and neurodegenerative diseases, the mechanisms of folate action in the developing neural tissue have remained elusive. Here we compiled studies that argue for a unique role for folate in nervous system development and function and its consequences to neural disease and repair. © 2018 Wiley Periodicals, Inc. Develop Neurobiol 78: 391-402, 2018.
Collapse
Affiliation(s)
- Olga A Balashova
- Department of Physiology & Membrane Biology and Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children, University of California Davis School of Medicine, Sacramento, California
| | - Olesya Visina
- Department of Physiology & Membrane Biology and Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children, University of California Davis School of Medicine, Sacramento, California
| | - Laura N Borodinsky
- Department of Physiology & Membrane Biology and Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children, University of California Davis School of Medicine, Sacramento, California
| |
Collapse
|
19
|
Abstract
Abstract
Clinical practice and experimental studies have shown the necessity of sufficient quantities of folic acid intake for normal embryogenesis and fetal development in the prevention of neural tube defects (NTDs) and neurological malformations. So, women of childbearing age must be sure to have an adequate folate intake periconceptionally, prior to and during pregnancy. Folic acid fortification of all enriched cereal grain product flour has been implemented in many countries. Thus, hundreds of thousands of people have been exposed to an increased intake of folic acid. Folate plays an essential role in the biosynthesis of methionine. Methionine is the principal aminopropyl donor required for polyamine biosynthesis, which is up-regulated in actively growing cells, including cancer cells. Folates are important in RNA and DNA synthesis, DNA stability and integrity. Clinical and epidemiological evidence links folate deficiency to DNA damage and cancer. On the other hand, long-term folate oversupplementation leads to adverse toxic effects, resulting in the appearance of malignancy. Considering the relationship of polyamines and rapidly proliferating tissues (especially cancers), there is a need for better investigation of the relationship between the ingestion of high amounts of folic acid in food supplementation and polyamine metabolism, related to malignant processes in the human body.
Collapse
|
20
|
Ceborska M. Folate appended cyclodextrins for drug, DNA, and siRNA delivery. Eur J Pharm Biopharm 2017; 120:133-145. [DOI: 10.1016/j.ejpb.2017.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 09/01/2017] [Accepted: 09/08/2017] [Indexed: 12/11/2022]
|
21
|
O’Connor OA, Amengual J, Colbourn D, Deng C, Sawas A. Pralatrexate: a comprehensive update on pharmacology, clinical activity and strategies to optimize use. Leuk Lymphoma 2017; 58:2548-2557. [DOI: 10.1080/10428194.2017.1306642] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Owen A. O’Connor
- Department of Medicine, College of Physicians and Surgeons, Center for Lymphoid Malignancies, Columbia University Medical Center, New York, NY, USA
| | - Jennifer Amengual
- Department of Medicine, College of Physicians and Surgeons, Center for Lymphoid Malignancies, Columbia University Medical Center, New York, NY, USA
| | - Donald Colbourn
- Department of Medicine, College of Physicians and Surgeons, Center for Lymphoid Malignancies, Columbia University Medical Center, New York, NY, USA
| | - Changchun Deng
- Department of Medicine, College of Physicians and Surgeons, Center for Lymphoid Malignancies, Columbia University Medical Center, New York, NY, USA
| | - Ahmed Sawas
- Department of Medicine, College of Physicians and Surgeons, Center for Lymphoid Malignancies, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
22
|
Watanabe S, Ohtani Y, Tatsukami Y, Aoki W, Amemiya T, Sukekiyo Y, Kubokawa S, Ueda M. Folate Biofortification in Hydroponically Cultivated Spinach by the Addition of Phenylalanine. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:4605-4610. [PMID: 28548831 DOI: 10.1021/acs.jafc.7b01375] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Folate is an important vitamin mainly ingested from vegetables, and folate deficiency causes various health problems. Recently, several studies demonstrated folate biofortification in plants or food crops by metabolic engineering through genetic modifications. However, the production and sales of genetically modified foods are under strict regulation. Here, we developed a new approach to achieve folate biofortification in spinach (Spinacia oleracea) without genetic modification. We hydroponically cultivated spinach with the addition of three candidate compounds expected to fortify folate. As a result of liquid chromatography tandem mass spectrometry analysis, we found that the addition of phenylalanine increased the folate content up to 2.0-fold (306 μg in 100 g of fresh spinach), representing 76.5% of the recommended daily allowance for adults. By measuring the intermediates of folate biosynthesis, we revealed that phenylalanine activated folate biosynthesis in spinach by increasing the levels of pteridine and p-aminobenzoic acid. Our approach is a promising and practical approach to cultivate nutrient-enriched vegetables.
Collapse
Affiliation(s)
- Sho Watanabe
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University , Sakyo-ku, Kyoto 606-8502, Japan
| | - Yuta Ohtani
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University , Sakyo-ku, Kyoto 606-8502, Japan
| | - Yohei Tatsukami
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University , Sakyo-ku, Kyoto 606-8502, Japan
- Japan Society for the Promotion of Science , Sakyo-ku, Kyoto 606-8502, Japan
| | - Wataru Aoki
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University , Sakyo-ku, Kyoto 606-8502, Japan
- Kyoto Integrated Science & Technology Bio-Analysis Center , Shimogyo-ku, Kyoto 600-8813, Japan
| | - Takashi Amemiya
- Mitsubishi Plastic, Inc. , Chiyoda-ku, Tokyo 100-8252, Japan
| | | | | | - Mitsuyoshi Ueda
- Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University , Sakyo-ku, Kyoto 606-8502, Japan
- Kyoto Integrated Science & Technology Bio-Analysis Center , Shimogyo-ku, Kyoto 600-8813, Japan
| |
Collapse
|
23
|
Silva E, Rosario FJ, Powell TL, Jansson T. Mechanistic Target of Rapamycin Is a Novel Molecular Mechanism Linking Folate Availability and Cell Function. J Nutr 2017; 147:1237-1242. [PMID: 28592519 DOI: 10.3945/jn.117.248823] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/16/2017] [Accepted: 05/11/2017] [Indexed: 12/15/2022] Open
Abstract
Folate deficiency has been linked to a wide range of disorders, including cancer, neural tube defects, and fetal growth restriction. Folate regulates cellular function mediated by its involvement in the synthesis of nucleotides, which are needed for DNA synthesis, and its function as a methyl donor, which is critical for DNA methylation. Here we review current data showing that folate sensing by mechanistic target of rapamycin (mTOR) constitutes a novel and distinct pathway by which folate modulates cell functions such as nutrient transport, protein synthesis, and mitochondrial respiration. The mTOR signaling pathway responds to growth factors and changes in nutrient availability to control cell growth, proliferation, and metabolism. mTOR exists in 2 complexes, mTOR complex (mTORC) 1 and mTORC2, which have distinct upstream regulators and downstream targets. Folate deficiency in pregnant mice caused a marked inhibition of mTORC1 and mTORC2 signaling in multiple maternal and fetal tissues, downregulation of placental amino acid transporters, and fetal growth restriction. In addition, folate deficiency in primary human trophoblast (PHT) cells resulted in inhibition of mTORC1 and mTORC2 signaling and decreased the activity of key amino acid transporters. Folate sensing by mTOR in PHT cells is independent of the accumulation of homocysteine and requires the proton-coupled folate transporter (PCFT; solute carrier 46A1). Furthermore, mTORC1 and mTORC2 regulate trophoblast folate uptake by modulating the cell surface expression of folate receptor α and the reduced folate carrier. These findings, which provide a novel link between folate availability and cell function, growth, and proliferation, may have broad biological significance given the critical role of folate in normal cell function and the multiple diseases that have been associated with decreased or excessive folate availability. Low maternal folate concentrations are linked to restricted fetal growth, and we propose that the underlying mechanisms involve trophoblast mTOR folate sensing resulting in inhibition of mTORC1 and mTORC2 and downregulation of placental amino acid transporters.
Collapse
Affiliation(s)
- Elena Silva
- Departments of Obstetrics and Gynecology and
| | | | - Theresa L Powell
- Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, CO
| | | |
Collapse
|
24
|
Toriyama M, Toriyama M, Wallingford JB, Finnell RH. Folate-dependent methylation of septins governs ciliogenesis during neural tube closure. FASEB J 2017; 31:3622-3635. [PMID: 28432198 DOI: 10.1096/fj.201700092r] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/11/2017] [Indexed: 12/15/2022]
Abstract
Periconception maternal folic acid (vitamin B9) supplementation can reduce the prevalence of neural tube defects (NTDs), although just how folates benefit the developing embryo and promote closing of the neural tube and other morphologic processes during development remains unknown. Folate contributes to a 1-carbon metabolism, which is essential for purine biosynthesis and methionine recycling and affects methylation of DNA, histones, and nonhistone proteins. Herein, we used animal models and cultured mammalian cells to demonstrate that disruption of the methylation pathway mediated by folate compromises normal neural tube closure (NTC) and ciliogenesis. We demonstrate that the embryos with NTD failed to adequately methylate septin2, a key regulator of cilium structure and function. We report that methylation of septin2 affected its GTP binding activity and formation of the septin2-6-7 complex. We propose that folic acid promotes normal NTC in some embryos by regulating the methylation of septin2, which is critical for normal cilium formation during early embryonic development.-Toriyama, M., Toriyama, M., Wallingford, J. B., Finnell, R. H. Folate-dependent methylation of septins governs ciliogenesis during neural tube closure.
Collapse
Affiliation(s)
- Manami Toriyama
- Department of Pediatrics, Dell Pediatric Research Institute, The University of Texas at Austin Dell Medical School, Austin, Texas, USA
| | - Michinori Toriyama
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - John B Wallingford
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - Richard H Finnell
- Department of Pediatrics, Dell Pediatric Research Institute, The University of Texas at Austin Dell Medical School, Austin, Texas, USA;
| |
Collapse
|
25
|
Xu J, Sinclair KD. One-carbon metabolism and epigenetic regulation of embryo development. Reprod Fertil Dev 2017; 27:667-76. [PMID: 25710200 DOI: 10.1071/rd14377] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 01/08/2015] [Indexed: 12/15/2022] Open
Abstract
One-carbon (1C) metabolism consists of an integrated series of metabolic pathways that include the folate cycle and methionine remethylation and trans-sulfuration pathways. Most, but not all, 1C metabolic enzymes are expressed in somatic cells of the ovary, mammalian oocytes and in preimplantation embryos. The metabolic implications of this, with regard to the provision of methyl donors (e.g. betaine) and 1C cofactors (e.g. vitamin B12), together with consequences of polymorphic variances in genes encoding 1C enzymes, are not fully understood but are the subject of ongoing investigations at the authors' laboratory. However, deficiencies in 1C-related substrates and/or cofactors during the periconception period are known to lead to epigenetic alterations in DNA and histone methylation in genes that regulate key developmental processes in the embryo. Such epigenetic modifications have been demonstrated to negatively impact on the subsequent health and metabolism of offspring. For this reason, parental nutrition around the time of conception has become a focal point of investigation in many laboratories with the aim of providing improved nutritional advice to couples. These issues are considered in detail in this article, which offers a contemporary overview of the effects of 1C metabolism on epigenetic programming in mammalian gametes and the early embryo.
Collapse
Affiliation(s)
- Juan Xu
- School of Bioscience, University of Nottingham, Sutton Bonington, Leicestershire LE12 5RD, UK
| | - Kevin D Sinclair
- School of Bioscience, University of Nottingham, Sutton Bonington, Leicestershire LE12 5RD, UK
| |
Collapse
|
26
|
Wang D, Wang F, Shi KH, Tao H, Li Y, Zhao R, Lu H, Duan W, Qiao B, Zhao SM, Wang H, Zhao JY. Lower Circulating Folate Induced by a Fidgetin Intronic Variant Is Associated With Reduced Congenital Heart Disease Susceptibility. Circulation 2017; 135:1733-1748. [PMID: 28302752 DOI: 10.1161/circulationaha.116.025164] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 03/07/2017] [Indexed: 02/05/2023]
Abstract
BACKGROUND Folate deficiency is an independent risk factor for congenital heart disease (CHD); however, the maternal plasma folate level is paradoxically not a good diagnostic marker. Genome-wide surveys have identified variants of nonfolate metabolic genes associated with the plasma folate level, suggesting that these genetic polymorphisms are potential risk factors for CHD. METHODS To examine the effects of folate concentration-related variations on CHD risk in the Han Chinese population, we performed 3 independent case-control studies including a total of 1489 patients with CHD and 1745 control subjects. The expression of the Fidgetin (FIGN) was detected in human cardiovascular and decidua tissue specimens with quantitative real-time polymerase chain reaction and Western blotting. The molecular mechanisms were investigated by luciferase reporter assays, surface plasmon resonance, and chromatin immunoprecipitation. FIGN-interacting proteins were confirmed by tandem affinity purification and coimmunoprecipitation. Proteasome activity and metabolite concentrations in the folate pathway were quantified with a commercial proteasome activity assay and immunoassays, respectively. RESULTS The +94762G>C (rs2119289) variant in intron 4 of the FIGN gene was associated with significant reduction in CHD susceptibility (P=5.1×10-14 for the allele, P=8.5×10--13 for the genotype). Analysis of combined samples indicated that CHD risks in individuals carrying heterozygous (GC) or homozygous (CC) genotypes were reduced by 44% (odds ratio [OR]=0.56; 95% confidence interval [CI]=0.47-0.67) and 66% (OR=0.34; 95% CI=0.23-0.50), respectively, compared with those with the major GG genotype. Minor C allele carriers who had decreased plasma folate levels exhibited significantly increased FIGN expression because the transcription suppressor CREB1 did not bind the alternative promoter of FIGN isoform X3. Mechanistically, increased FIGN expression led to the accumulation of both reduced folate carrier 1 and dihydrofolate reductase via inhibition of their proteasomal degradation, which promoted folate absorption and metabolism. CONCLUSIONS We report a previously undocumented finding that decreased circulating folate levels induced by increased folate transmembrane transport and utilization, as determined by the FIGN intronic variant, serves as a protective mechanism against CHD. Our results may explain why circulating folate levels do not have a good diagnostic value.
Collapse
Affiliation(s)
- Dan Wang
- From Obstetrics and Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai, China (D.W., Y.L., R.Z., H.L., S.-M.Z., H.W., J.-Y.Z.); Key Laboratory of Reproduction Regulation of NPFPC, Institute of Reproduction and Development and Children's Hospital of Fudan University, Fudan University, Shanghai, China (D.W., F.W., Y.L., R.Z., S.-M.Z., H.W., J.-Y.Z.); MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China (S.-M.Z., H.W., J.-Y.Z.); Department of Cardiothoracic Surgery, Second Hospital of Anhui Medical University, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Cardiovascular Research Center, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China (W.D., B.Q.); Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China (S.M.-Z., J.-Y.Z.); and Department of Neonatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (D.W.)
| | - Feng Wang
- From Obstetrics and Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai, China (D.W., Y.L., R.Z., H.L., S.-M.Z., H.W., J.-Y.Z.); Key Laboratory of Reproduction Regulation of NPFPC, Institute of Reproduction and Development and Children's Hospital of Fudan University, Fudan University, Shanghai, China (D.W., F.W., Y.L., R.Z., S.-M.Z., H.W., J.-Y.Z.); MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China (S.-M.Z., H.W., J.-Y.Z.); Department of Cardiothoracic Surgery, Second Hospital of Anhui Medical University, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Cardiovascular Research Center, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China (W.D., B.Q.); Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China (S.M.-Z., J.-Y.Z.); and Department of Neonatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (D.W.)
| | - Kai-Hu Shi
- From Obstetrics and Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai, China (D.W., Y.L., R.Z., H.L., S.-M.Z., H.W., J.-Y.Z.); Key Laboratory of Reproduction Regulation of NPFPC, Institute of Reproduction and Development and Children's Hospital of Fudan University, Fudan University, Shanghai, China (D.W., F.W., Y.L., R.Z., S.-M.Z., H.W., J.-Y.Z.); MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China (S.-M.Z., H.W., J.-Y.Z.); Department of Cardiothoracic Surgery, Second Hospital of Anhui Medical University, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Cardiovascular Research Center, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China (W.D., B.Q.); Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China (S.M.-Z., J.-Y.Z.); and Department of Neonatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (D.W.)
| | - Hui Tao
- From Obstetrics and Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai, China (D.W., Y.L., R.Z., H.L., S.-M.Z., H.W., J.-Y.Z.); Key Laboratory of Reproduction Regulation of NPFPC, Institute of Reproduction and Development and Children's Hospital of Fudan University, Fudan University, Shanghai, China (D.W., F.W., Y.L., R.Z., S.-M.Z., H.W., J.-Y.Z.); MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China (S.-M.Z., H.W., J.-Y.Z.); Department of Cardiothoracic Surgery, Second Hospital of Anhui Medical University, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Cardiovascular Research Center, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China (W.D., B.Q.); Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China (S.M.-Z., J.-Y.Z.); and Department of Neonatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (D.W.)
| | - Yang Li
- From Obstetrics and Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai, China (D.W., Y.L., R.Z., H.L., S.-M.Z., H.W., J.-Y.Z.); Key Laboratory of Reproduction Regulation of NPFPC, Institute of Reproduction and Development and Children's Hospital of Fudan University, Fudan University, Shanghai, China (D.W., F.W., Y.L., R.Z., S.-M.Z., H.W., J.-Y.Z.); MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China (S.-M.Z., H.W., J.-Y.Z.); Department of Cardiothoracic Surgery, Second Hospital of Anhui Medical University, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Cardiovascular Research Center, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China (W.D., B.Q.); Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China (S.M.-Z., J.-Y.Z.); and Department of Neonatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (D.W.)
| | - Rui Zhao
- From Obstetrics and Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai, China (D.W., Y.L., R.Z., H.L., S.-M.Z., H.W., J.-Y.Z.); Key Laboratory of Reproduction Regulation of NPFPC, Institute of Reproduction and Development and Children's Hospital of Fudan University, Fudan University, Shanghai, China (D.W., F.W., Y.L., R.Z., S.-M.Z., H.W., J.-Y.Z.); MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China (S.-M.Z., H.W., J.-Y.Z.); Department of Cardiothoracic Surgery, Second Hospital of Anhui Medical University, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Cardiovascular Research Center, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China (W.D., B.Q.); Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China (S.M.-Z., J.-Y.Z.); and Department of Neonatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (D.W.)
| | - Han Lu
- From Obstetrics and Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai, China (D.W., Y.L., R.Z., H.L., S.-M.Z., H.W., J.-Y.Z.); Key Laboratory of Reproduction Regulation of NPFPC, Institute of Reproduction and Development and Children's Hospital of Fudan University, Fudan University, Shanghai, China (D.W., F.W., Y.L., R.Z., S.-M.Z., H.W., J.-Y.Z.); MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China (S.-M.Z., H.W., J.-Y.Z.); Department of Cardiothoracic Surgery, Second Hospital of Anhui Medical University, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Cardiovascular Research Center, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China (W.D., B.Q.); Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China (S.M.-Z., J.-Y.Z.); and Department of Neonatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (D.W.)
| | - Wenyuan Duan
- From Obstetrics and Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai, China (D.W., Y.L., R.Z., H.L., S.-M.Z., H.W., J.-Y.Z.); Key Laboratory of Reproduction Regulation of NPFPC, Institute of Reproduction and Development and Children's Hospital of Fudan University, Fudan University, Shanghai, China (D.W., F.W., Y.L., R.Z., S.-M.Z., H.W., J.-Y.Z.); MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China (S.-M.Z., H.W., J.-Y.Z.); Department of Cardiothoracic Surgery, Second Hospital of Anhui Medical University, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Cardiovascular Research Center, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China (W.D., B.Q.); Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China (S.M.-Z., J.-Y.Z.); and Department of Neonatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (D.W.)
| | - Bin Qiao
- From Obstetrics and Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai, China (D.W., Y.L., R.Z., H.L., S.-M.Z., H.W., J.-Y.Z.); Key Laboratory of Reproduction Regulation of NPFPC, Institute of Reproduction and Development and Children's Hospital of Fudan University, Fudan University, Shanghai, China (D.W., F.W., Y.L., R.Z., S.-M.Z., H.W., J.-Y.Z.); MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China (S.-M.Z., H.W., J.-Y.Z.); Department of Cardiothoracic Surgery, Second Hospital of Anhui Medical University, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Cardiovascular Research Center, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China (W.D., B.Q.); Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China (S.M.-Z., J.-Y.Z.); and Department of Neonatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (D.W.)
| | - Shi-Min Zhao
- From Obstetrics and Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai, China (D.W., Y.L., R.Z., H.L., S.-M.Z., H.W., J.-Y.Z.); Key Laboratory of Reproduction Regulation of NPFPC, Institute of Reproduction and Development and Children's Hospital of Fudan University, Fudan University, Shanghai, China (D.W., F.W., Y.L., R.Z., S.-M.Z., H.W., J.-Y.Z.); MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China (S.-M.Z., H.W., J.-Y.Z.); Department of Cardiothoracic Surgery, Second Hospital of Anhui Medical University, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Cardiovascular Research Center, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China (W.D., B.Q.); Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China (S.M.-Z., J.-Y.Z.); and Department of Neonatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (D.W.).
| | - Hongyan Wang
- From Obstetrics and Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai, China (D.W., Y.L., R.Z., H.L., S.-M.Z., H.W., J.-Y.Z.); Key Laboratory of Reproduction Regulation of NPFPC, Institute of Reproduction and Development and Children's Hospital of Fudan University, Fudan University, Shanghai, China (D.W., F.W., Y.L., R.Z., S.-M.Z., H.W., J.-Y.Z.); MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China (S.-M.Z., H.W., J.-Y.Z.); Department of Cardiothoracic Surgery, Second Hospital of Anhui Medical University, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Cardiovascular Research Center, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China (W.D., B.Q.); Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China (S.M.-Z., J.-Y.Z.); and Department of Neonatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (D.W.).
| | - Jian-Yuan Zhao
- From Obstetrics and Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, School of Life Sciences and Institutes of Biomedical Sciences, Fudan University, Shanghai, China (D.W., Y.L., R.Z., H.L., S.-M.Z., H.W., J.-Y.Z.); Key Laboratory of Reproduction Regulation of NPFPC, Institute of Reproduction and Development and Children's Hospital of Fudan University, Fudan University, Shanghai, China (D.W., F.W., Y.L., R.Z., S.-M.Z., H.W., J.-Y.Z.); MOE Key Laboratory of Contemporary Anthropology, Collaborative Innovation Center for Genetics and Development, Fudan University, Shanghai, China (S.-M.Z., H.W., J.-Y.Z.); Department of Cardiothoracic Surgery, Second Hospital of Anhui Medical University, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Cardiovascular Research Center, Anhui Medical University, Hefei, China (K.-H.S., H.T.); Institute of Cardiovascular Disease, General Hospital of Jinan Military Region, Jinan, China (W.D., B.Q.); Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China (S.M.-Z., J.-Y.Z.); and Department of Neonatology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China (D.W.).
| |
Collapse
|
27
|
Balashova OA, Visina O, Borodinsky LN. Folate receptor 1 is necessary for neural plate cell apical constriction during Xenopus neural tube formation. Development 2017; 144:1518-1530. [PMID: 28255006 DOI: 10.1242/dev.137315] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 02/17/2017] [Indexed: 12/26/2022]
Abstract
Folate supplementation prevents up to 70% of neural tube defects (NTDs), which result from a failure of neural tube closure during embryogenesis. The elucidation of the mechanisms underlying folate action has been challenging. This study introduces Xenopus laevis as a model to determine the cellular and molecular mechanisms involved in folate action during neural tube formation. We show that knockdown of folate receptor 1 (Folr1; also known as FRα) impairs neural tube formation and leads to NTDs. Folr1 knockdown in neural plate cells only is necessary and sufficient to induce NTDs. Folr1-deficient neural plate cells fail to constrict, resulting in widening of the neural plate midline and defective neural tube closure. Pharmacological inhibition of folate action by methotrexate during neurulation induces NTDs by inhibiting folate interaction with its uptake systems. Our findings support a model in which the folate receptor interacts with cell adhesion molecules, thus regulating the apical cell membrane remodeling and cytoskeletal dynamics necessary for neural plate folding. Further studies in this organism could unveil novel cellular and molecular events mediated by folate and lead to new ways of preventing NTDs.
Collapse
Affiliation(s)
- Olga A Balashova
- Department of Physiology & Membrane Biology and Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Olesya Visina
- Department of Physiology & Membrane Biology and Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Laura N Borodinsky
- Department of Physiology & Membrane Biology and Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children, University of California Davis School of Medicine, Sacramento, CA 95817, USA
| |
Collapse
|
28
|
Vats K, Subramanian S, Mathur A, Sarma HD, Banerjee S. Radiosynthesis and evaluation of a 99mTc-folic acid radiotracer prepared using [ 99mTcN(PNP)] 2+ metal fragment. Bioorg Med Chem Lett 2017; 27:1329-1332. [PMID: 28174106 DOI: 10.1016/j.bmcl.2016.03.090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 03/10/2016] [Accepted: 03/25/2016] [Indexed: 11/16/2022]
Abstract
Folate receptors (FR) are over-expressed on a wide variety of tumor cells and are a potential molecular target for radiolabeled folates. In this respect, several SPECT and PET based radiofolates have been evaluated in the past albeit with their high renal uptake posing limitation towards their clinical use. To overcome this, a new 99mTc labeled folic acid was synthesized via the use of [99mTcN(PNP)]2+ metal fragment, where the presence of the latter pharmacophore redirects in vivo clearance via the hepatobiliary pathway. In this respect, folic acid was derivatized at the γ-acid group with a cysteine BFCA (bifunctional chelating agent) and subsequently reacted with the preformed [99mTcN]2+ intermediate in presence of PNP2 (bisphosphine) ligand, to yield the final complex. While preliminary, in vivo distribution of the complex exhibited high association of activity with liver and intestines and provided support to the rationality of the present design as clearance of labeled folic acid could be effected via the hepatic route, the in vitro studies of the folic acid-cysteine conjugate carried out in KB-31 cells, did not show much promise with reduction in receptor affinity in comparison with the native folic acid. The route followed herein to prepare a folic-acid based radiotracer constitutes the first report of radiolabeling folic acid using the [99mTcN(PNP)]2+ as a radiosynthon. Modification in the structure of conjugate by linking the BFCA through a long-chain linker can be envisaged to improve the affinity of [99mTcN(PNP)]-folic acid complex towards FRs.
Collapse
Affiliation(s)
- Kusum Vats
- Radiopharmaceuticals Chemistry Section, Radiochemistry & Isotope Group, Bhabha Atomic Research Centre, Mumbai 400085, India; Homi Bhabha National Institute, Anushakti Nagar, Mumbai 400094, India
| | - Suresh Subramanian
- Isotope Production and Applications Division, Bhabha Atomic Research Centre, Mumbai 400085, India
| | - Anupam Mathur
- Radiopharmaceuticals Program, Board of Radiation and Isotope Technology, Navi Mumbai 400703, India
| | - Haladhar Dev Sarma
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai 400085, India
| | - Sharmila Banerjee
- Radiopharmaceuticals Chemistry Section, Radiochemistry & Isotope Group, Bhabha Atomic Research Centre, Mumbai 400085, India; Homi Bhabha National Institute, Anushakti Nagar, Mumbai 400094, India.
| |
Collapse
|
29
|
Brasil FB, Amarante LH, Oliveira MRD. Maternal folic acid consumption during gestation and its long-term effects on offspring's liver: a systematic review. REVISTA BRASILEIRA DE SAÚDE MATERNO INFANTIL 2017. [DOI: 10.1590/1806-93042017000100002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Abstract Objectives: describing the effects of maternal supplementation with folic acid (FA) exclusively during gestation on offspring's liver at later stages in life. Supplementation with FA during gestation has been recommended by the medical society worldwide. The liver has a central role on the substances of metabolism and homeostasis and some studies have shown that a high intake of FA at other periods in life may cause hepatic damage. Methods: a systematic review through which the following databases were consulted: Medline, through platforms of Pubmed, Lilacs and Scielo. The research was performed by keywords such as: "Folic acid", "Gestation", "Rat", "Offspring" and "Liver". Articles which evaluate the effect of FA consumption during both gestation and lactation were excluded. Results: FA consumption avoids disorders on expression of peroxisome proliferator-activated receptor alpha (PPARα) and glucocorticoid receptor (GccR), its lack did not change enzyme activity of the male offspring's liver in adulthood. Supplementation with FA during gestation did not change iron hepatic levels or lipid composition, but had an antioxidant effect on it. Conclusions: supplementation with FA at recommended doses did not cause toxic effects and is very likely to avoid deleterious effects in the liver of the offspring regarding the epigenetic level.
Collapse
|
30
|
Jackson RA, Nguyen ML, Barrett AN, Tan YY, Choolani MA, Chen ES. Synthetic combinations of missense polymorphic genetic changes underlying Down syndrome susceptibility. Cell Mol Life Sci 2016; 73:4001-17. [PMID: 27245382 PMCID: PMC11108497 DOI: 10.1007/s00018-016-2276-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/10/2016] [Accepted: 05/12/2016] [Indexed: 02/08/2023]
Abstract
Single nucleotide polymorphisms (SNPs) are important biomolecular markers in health and disease. Down syndrome, or Trisomy 21, is the most frequently occurring chromosomal abnormality in live-born children. Here, we highlight associations between SNPs in several important enzymes involved in the one-carbon folate metabolic pathway and the elevated maternal risk of having a child with Down syndrome. Our survey highlights that the combination of SNPs may be a more reliable predictor of the Down syndrome phenotype than single SNPs alone. We also describe recent links between SNPs in p53 and its related pathway proteins and Down syndrome, as well as highlight several proteins that help to associate apoptosis and p53 signaling with the Down syndrome phenotype. In addition to a comprehensive review of the literature, we also demonstrate that several SNPs reside within the same regions as these Down syndrome-linked SNPs, and propose that these closely located nucleotide changes may provide new candidates for future exploration.
Collapse
Affiliation(s)
- Rebecca A Jackson
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, #05-05, MD7, 8 Medical Drive, Singapore, 117597, Singapore
| | - Mai Linh Nguyen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, #05-05, MD7, 8 Medical Drive, Singapore, 117597, Singapore
| | - Angela N Barrett
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, #05-05, MD7, 8 Medical Drive, Singapore, 117597, Singapore
| | - Yuan Yee Tan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, #05-05, MD7, 8 Medical Drive, Singapore, 117597, Singapore
| | - Mahesh A Choolani
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, #05-05, MD7, 8 Medical Drive, Singapore, 117597, Singapore.
- National University Health System, Singapore, Singapore.
| | - Ee Sin Chen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, #05-05, MD7, 8 Medical Drive, Singapore, 117597, Singapore.
- National University Health System, Singapore, Singapore.
- NUS Graduate School of Science and Engineering, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
31
|
Dewar S, Sienkiewicz N, Ong HB, Wall RJ, Horn D, Fairlamb AH. The Role of Folate Transport in Antifolate Drug Action in Trypanosoma brucei. J Biol Chem 2016; 291:24768-24778. [PMID: 27703008 PMCID: PMC5114424 DOI: 10.1074/jbc.m116.750422] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 09/14/2016] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to identify and characterize mechanisms of resistance to antifolate drugs in African trypanosomes. Genome-wide RNAi library screens were undertaken in bloodstream form Trypanosoma brucei exposed to the antifolates methotrexate and raltitrexed. In conjunction with drug susceptibility and folate transport studies, RNAi knockdown was used to validate the functions of the putative folate transporters. The transport kinetics of folate and methotrexate were further characterized in whole cells. RNA interference target sequencing experiments identified a tandem array of genes encoding a folate transporter family, TbFT1-3, as major contributors to antifolate drug uptake. RNAi knockdown of TbFT1-3 substantially reduced folate transport into trypanosomes and reduced the parasite's susceptibly to the classical antifolates methotrexate and raltitrexed. In contrast, knockdown of TbFT1-3 increased susceptibly to the non-classical antifolates pyrimethamine and nolatrexed. Both folate and methotrexate transport were inhibited by classical antifolates but not by non-classical antifolates or biopterin. Thus, TbFT1-3 mediates the uptake of folate and classical antifolates in trypanosomes, and TbFT1-3 loss-of-function is a mechanism of antifolate drug resistance.
Collapse
Affiliation(s)
- Simon Dewar
- From the Division of Biological Chemistry and Drug Discovery, Wellcome Trust Building, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | - Natasha Sienkiewicz
- From the Division of Biological Chemistry and Drug Discovery, Wellcome Trust Building, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | - Han B Ong
- From the Division of Biological Chemistry and Drug Discovery, Wellcome Trust Building, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | - Richard J Wall
- From the Division of Biological Chemistry and Drug Discovery, Wellcome Trust Building, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | - David Horn
- From the Division of Biological Chemistry and Drug Discovery, Wellcome Trust Building, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | - Alan H Fairlamb
- From the Division of Biological Chemistry and Drug Discovery, Wellcome Trust Building, College of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom.
| |
Collapse
|
32
|
Zaruma-Torres F, Lares-Asseff I, Lima A, Reyes-Espinoza A, Loera-Castañeda V, Sosa-Macías M, Galaviz-Hernández C, Arias-Peláez MC, Reyes-López MA, Quiñones LA. Genetic Polymorphisms Associated to Folate Transport as Predictors of Increased Risk for Acute Lymphoblastic Leukemia in Mexican Children. Front Pharmacol 2016; 7:238. [PMID: 27547186 PMCID: PMC4974492 DOI: 10.3389/fphar.2016.00238] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Accepted: 07/21/2016] [Indexed: 01/25/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a frequent neoplasia occurring in children. The most commonly used drug for the treatment of ALL is methotrexate (MTX), an anti-folate agent. Previous studies suggest that folate transporters play a role in ALL prognosis and that genetic polymorphism of genes encoding folate transporters may increase the risk of ALL. Therefore, the main goal of this study was to determine the associations among six genetic polymorphisms in four genes related with the folate transporter pathway to determine a relationship with the occurrence of ALL in Mexican children. A case-control study was performed in 73 ALL children and 133 healthy children from Northern and Northwestern Mexico. COL18A1 (rs2274808), SLC19A1 (rs2838956), ABCB1 (rs1045642 and rs1128503), and ABCC5 (rs9838667 and rs3792585). Polymorphisms were assayed through qPCR. Our results showed an increased ALL risk in children carrying CT genotype (OR = 2.55, CI 95% 1.11–5.83, p = 0.0001) and TT genotype (OR = 21.05, CI 95% 5.62–78.87, p < 0.0001) of COL18A1 rs2274808; in SLC19A1 rs2838956 AG carriers (OR = 44.69, CI 95% 10.42–191.63, p = 0.0001); in ABCB1 rs1045642 TT carriers (OR = 13.76, CI 95% 5.94–31.88, p = 0.0001); in ABCC5 rs9838667 AC carriers (OR = 2.61, CI 95% 1.05–6.48, p < 0.05); and in ABCC5 rs3792585 CC carriers (OR = 9.99, CI 95% 3.19–31.28, p = 0.004). Moreover, several combinations of genetic polymorphisms were found to be significantly associated with a risk for ALL. Finally, two combinations of ABCC5 polymorphisms resulted in protection from this neoplasia. In conclusion, certain genetic polymorphisms related to the folate transport pathway, particularly COL18A1 rs2274808, SLC19A1 rs2838956, ABCB1 rs1045642, and ABCC5 rs3792585, were associated with an increased risk for ALL in Mexican children.
Collapse
Affiliation(s)
- Fausto Zaruma-Torres
- Pharmacogenomics Academia, National Polytechnic Institute-CIIDIRDurango, Mexico; School of Biochemistry and Pharmacy, University of CuencaCuenca, Ecuador
| | - Ismael Lares-Asseff
- Pharmacogenomics Academia, National Polytechnic Institute-CIIDIR Durango, Mexico
| | - Aurea Lima
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde Gandra, Portugal
| | | | | | - Martha Sosa-Macías
- Pharmacogenomics Academia, National Polytechnic Institute-CIIDIR Durango, Mexico
| | | | - María C Arias-Peláez
- Institute of Scientific Research of the University Juarez of State of Durango Durango, Mexico
| | - Miguel A Reyes-López
- Center of Biotechnology Genomics, National Polytechnic Institute Reynosa, Mexico
| | - Luis A Quiñones
- Pharmacological and Molecular Program, Laboratory of Chemical Carcinogenesis and Pharmacogenetics (CQF), Faculty of Medicine, ICBM, University of Chile Santiago, Chile
| |
Collapse
|
33
|
Kim YJ. Nutritional concerns in pediatric inflammatory bowel disease. KOREAN JOURNAL OF PEDIATRICS 2016; 59:247-51. [PMID: 27462352 PMCID: PMC4958701 DOI: 10.3345/kjp.2016.59.6.247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 12/15/2015] [Accepted: 12/16/2015] [Indexed: 01/09/2023]
Abstract
The pathophysiology and fundamental etiologic mechanism of inflammatory bowel disease (IBD) is not well understood even though therapeutic regimens and drugs are rapidly evolutionary. IBD has complicated connections with genetic, immunologic, gut microbial, environmental, and nutritional factors. It is not clearly well known to the physicians how to feed, what nutrients are more helpful, and what food to be avoided. This review discusses the issues of growth and important nutritional concerns in the management of IBD in childhood.
Collapse
Affiliation(s)
- Yong Joo Kim
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Korea
| |
Collapse
|
34
|
Assessment of folate receptor-β expression in human neoplastic tissues. Oncotarget 2016; 6:14700-9. [PMID: 25909292 PMCID: PMC4546498 DOI: 10.18632/oncotarget.3739] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 03/03/2015] [Indexed: 11/25/2022] Open
Abstract
Over-expression of folate receptor alpha on cancer cells has been frequently exploited for delivery of folate-targeted imaging and therapeutic agents to tumors. Because limited information exists on expression of the beta isoform of the folate receptor in human cancers (FR-β), we have evaluated the immunohistochemical staining pattern of FR-β in 992 tumor sections from 20 different human cancer types using a new anti-human FR-β monoclonal antibody. FR-β expression was shown to be more pronounced in cells within the stroma, primarily macrophages and macrophage-like cells than cancer cells in every cancer type studied. Moreover, FR-β expression in both cancer and stromal cells was found to be statistically more prominent in females than males. A significant positive correlation was also observed between FR-β expression on stromal cells and both the stage of the cancer and the presence of lymph node metastases. Based on these data we conclude FR-β may constitute a good target for specific delivery of therapeutic agents to activated macrophages and that accumulation of FR-β positive macrophages in the stroma could serve as a useful indicator of a tumor's metastatic potential.
Collapse
|
35
|
Folic acid induces cell type-specific changes in the transcriptome of breast cancer cell lines: a proof-of-concept study. J Nutr Sci 2016; 5:e17. [PMID: 27293554 PMCID: PMC4891697 DOI: 10.1017/jns.2016.8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 01/28/2016] [Accepted: 02/29/2016] [Indexed: 02/08/2023] Open
Abstract
The effect of folic acid (FA) on breast cancer (BC) risk is uncertain. We hypothesised
that this uncertainty may be due, in part, to differential effects of FA between BC cells
with different phenotypes. To test this we investigated the effect of treatment with FA
concentrations within the range of unmetabolised FA reported in humans on the expression
of the transcriptome of non-transformed (MCF10A) and cancerous (MCF7 and Hs578T) BC cells.
The total number of transcripts altered was: MCF10A, seventy-five (seventy up-regulated);
MCF7, twenty-four (fourteen up-regulated); and Hs578T, 328 (156 up-regulated). Only the
cancer-associated gene TAGLN was altered by FA in all three cell lines.
In MCF10A and Hs578T cells, FA treatment decreased pathways associated with apoptosis,
cell death and senescence, but increased those associated with cell proliferation. The
folate transporters SLC19A1, SLC46A1 and FOLR1 were differentially expressed between cell
lines tested. However, the level of expression was not altered by FA treatment. These
findings suggest that physiological concentrations of FA can induce cell type-specific
changes in gene regulation in a manner that is consistent with proliferative phenotype.
This has implications for understanding the role of FA in BC risk. In addition, these
findings support the suggestion that differences in gene expression induced by FA may
involve differential activities of folate transporters. Together these findings indicate
the need for further studies of the effect of FA on BC.
Collapse
|
36
|
Naggar H, Van Ells TK, Ganapathy V, Smith SB. Regulation of Reduced-Folate Transporter-1 in Retinal Pigment Epithelial Cells by Folate. Curr Eye Res 2015. [DOI: 10.1080/02713680490894559a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
37
|
Methotrexate influx via folate transporters into alveolar epithelial cell line A549. Drug Metab Pharmacokinet 2015; 30:276-81. [PMID: 26190800 DOI: 10.1016/j.dmpk.2015.04.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 04/22/2015] [Accepted: 04/29/2015] [Indexed: 12/31/2022]
Abstract
Methotrexate (MTX), a drug used for the treatment of certain cancers as well as rheumatoid arthritis, sometimes induces serious interstitial lung injury. Although lung toxicity of MTX is related to its accumulation, the information concerning MTX transport in the lungs is lacking. In this study, we investigated the mechanisms underlying MTX influx into human alveolar epithelial cell line A549. MTX influx into A549 cells was time-, pH-, and temperature-dependent and showed saturation kinetics. The influx was inhibited by folic acid with IC50 values of 256.1 μM at pH 7.4 and 1.6 μM at pH 5.5, indicating that the mechanisms underlying MTX influx would be different at these pHs. We then examined the role of two folate transporters in MTX influx, reduced folate carrier (RFC) and proton-coupled folate transporter (PCFT). The expression of RFC and PCFT mRNAs in A549 cells was confirmed by reverse transcription polymerase chain reaction. In addition, MTX influx was inhibited by thiamine monophosphate, an RFC inhibitor, at pH 7.4, and by sulfasalazine, a PCFT inhibitor, at pH 5.5. These results indicated that RFC and PCFT are predominantly involved in MTX influx into A549 cells at pH 7.4 and pH 5.5, respectively.
Collapse
|
38
|
Kim MH, Kim WH, Kim CG, Kim DW. Synthesis and Evaluation of (99m)Tc-Labeled Folate-Tripeptide Conjugate as a Folate Receptor-Targeted Imaging Agent in a Tumor-Bearing Mouse Model. Nucl Med Mol Imaging 2015; 49:200-7. [PMID: 26279693 DOI: 10.1007/s13139-015-0336-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 04/08/2015] [Accepted: 04/12/2015] [Indexed: 11/25/2022] Open
Abstract
PURPOSE The folate receptor (FR) is an attractive molecular target since it is overexpressed in a variety of human tumors. The purpose of the present study was to synthesize and evaluate the feasibility of a novel (99m)Tc-ECG-EDA (Glu-Cys-Gly-ethylenediamine)-folate as an FR-positive tumor imaging agent in a mouse tumor model. MATERIALS AND METHODS ECG-EDA-folate was synthesized using solid phase peptide synthesis (SPPS) and radiolabeled with (99m)Tc using tripeptide ECG as a chelator. FR-positive KB cells were inoculated in athymic nude mice. Following injection of (99m)Tc-ECG-EDA-folate, serial scintigraphy and micro-SPECT/CT imaging were performed at various time points with and without pre-administration of excess free folate. Mean count densities (MCD) for regions of interest drawn on KB tumors and major normal organs at each time point were measured, and uptake ratios of tumor to normal organs were calculated. RESULTS ECG-EDA-folate was labeled with (99m)Tc with high radiolabeling efficiency and stability (>96 %). FR-positive tumors were clearly visualized on both scintigraphy and micro-SPECT/CT images and the tumor uptake of (99m)Tc-ECG-EDA-folate was markedly suppressed with faint visualization of tumors by pre-administration of excess free folate on serial planar scintigraphy, indicating FR-specific binding of the agent. Furthermore, semiquantitative analysis of MCD data showed again that both tumor MCD and tumor-to-normal organ ratios decreased considerably by pre-administration of excess free folate, supporting FR-specific tumor uptake. Tumor-to-normal organ ratios approximately increased with time after injection until 4 h. CONCLUSION The present study demonstrated that (99m)Tc-ECG-EDA-folate can bind specifically to FR with clear visualization of FR-positive tumors in a mouse tumor model.
Collapse
Affiliation(s)
- Myoung Hyoun Kim
- Department of Nuclear Medicine, Wonkwang University School of Medicine, Iksan, Korea
| | - Woo Hyoung Kim
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Korea
| | - Chang Guhn Kim
- Department of Nuclear Medicine, Wonkwang University School of Medicine, Iksan, Korea ; Institute of Wonkwang Medical Science, Iksan, Korea
| | - Dae-Weung Kim
- Department of Nuclear Medicine, Wonkwang University School of Medicine, Iksan, Korea ; Institute of Wonkwang Medical Science, Iksan, Korea
| |
Collapse
|
39
|
Narawa T, Yano T, Itoh T. Stereoselective recognition of amethopterin enantiomers by the rat proton-coupled folate transporter. Biol Pharm Bull 2015; 38:545-51. [PMID: 25832635 DOI: 10.1248/bpb.b14-00688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The stereoselective transport of methotrexate (L-amethopterin, L-MTX) and its enantiomer (D-amethopterin, D-MTX) by the rat proton-coupled folate transporter (rPCFT) were examined using rPCFT-expressing HEK293 cells. The initial rate of uptake of [3H]-L-MTX by the rPCFT followed Michaelis-Menten kinetics, with a Km value of 2.1 µM. Dixon plots revealed that the uptake of L-MTX by the rPCFT was inhibited in a competitive manner by unlabeled L-MTX and D-MTX, with Ki values of approximately 1.3 and 150 µM, respectively. The initial rate of uptake of D-MTX by the rPCFT also followed Michaelis-Menten kinetics, with a Km value of 190 µM. The results of the current study demonstrate that the different enantiomers of MTX are transported in a highly stereoselective manner by the rPCFT, with the uptake clearance of L-MTX being approximately 46-fold greater than that of D-MTX. The observed stereoselectivity of the rPCFT was found to be comparable with that of the human PCFT.
Collapse
|
40
|
Hu J, Wang B, Sahyoun NR. Application of the Key Events Dose-response Framework to Folate Metabolism. Crit Rev Food Sci Nutr 2015; 56:1325-33. [DOI: 10.1080/10408398.2013.807221] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
41
|
Ramaekers VT, Thöny B, Sequeira JM, Ansseau M, Philippe P, Boemer F, Bours V, Quadros EV. Folinic acid treatment for schizophrenia associated with folate receptor autoantibodies. Mol Genet Metab 2014; 113:307-14. [PMID: 25456743 DOI: 10.1016/j.ymgme.2014.10.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Revised: 09/30/2014] [Accepted: 10/01/2014] [Indexed: 12/14/2022]
Abstract
BACKGROUND Auto-antibodies against folate receptor alpha (FRα) at the choroid plexus that block N(5)-methyltetrahydrofolate (MTHF) transfer to the brain were identified in catatonic schizophrenia. Acoustic hallucinations disappeared following folinic acid treatment. Folate transport to the CNS prevents homocysteine accumulation and delivers one-carbon units for methyl-transfer reactions and synthesis of purines. The guanosine derivative tetrahydrobiopterin acts as common co-factor for the enzymes producing dopamine, serotonin and nitric oxide. METHODS Our study selected patients with schizophrenia unresponsive to conventional treatment. Serum from these patients with normal plasma homocysteine, folate and vitamin B12 was tested for FR autoantibodies of the blocking type on serial samples each week. Spinal fluid was analyzed for MTHF and the metabolites of pterins, dopamine and serotonin. The clinical response to folinic acid treatment was evaluated. RESULTS Fifteen of 18 patients (83.3%) had positive serum FR auto-antibodies compared to only 1 in 30 controls (3.3%) (χ(2)=21.6; p<0.0001). FRα antibody titers in patients fluctuated over time varying between negative and high titers, modulating folate flux to the CNS, which explained low CSF folate values in 6 and normal values in 7 patients. The mean±SD for CSF MTHF was diminished compared to previously established controls (t-test: 3.90; p=0.0002). A positive linear correlation existed between CSF MTHF and biopterin levels. CSF dopamine and serotonin metabolites were low or in the lower normal range. Administration of folinic acid (0.3-1mg/kg/day) to 7 participating patients during at least six months resulted in clinical improvement. CONCLUSION Assessment of FR auto-antibodies in serum is recommended for schizophrenic patients. Clinical negative or positive symptoms are speculated to be influenced by the level and evolution of FRα antibody titers which determine folate flux to the brain with up- or down-regulation of brain folate intermediates linked to metabolic processes affecting homocysteine levels, synthesis of tetrahydrobiopterin and neurotransmitters. Folinic acid intervention appears to stabilize the disease process.
Collapse
Affiliation(s)
- V T Ramaekers
- Division of Paediatric Neurology, Centre Hospitalier Universitaire de Liège, Liège, Belgium; Centre for Autism Liège, Centre Hospitalier Universitaire de Liège, Liège, Belgium.
| | - B Thöny
- Division of Metabolism, University Children's Hospital Zurich, Switzerland
| | - J M Sequeira
- Department of Medicine, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - M Ansseau
- Department of Psychiatry, Centre Hospitalier Universitaire de Liège, Liège, Belgium
| | - P Philippe
- Centre for Autism Liège, Centre Hospitalier Universitaire de Liège, Liège, Belgium
| | - F Boemer
- Department of Human Genetics and Metabolism, Centre Hospitalier Universitaire de Liège, Liège, Belgium
| | - V Bours
- Department of Human Genetics and Metabolism, Centre Hospitalier Universitaire de Liège, Liège, Belgium
| | - E V Quadros
- Department of Medicine, SUNY Downstate Medical Center, Brooklyn, NY, USA
| |
Collapse
|
42
|
Muthukumar T, Chamundeeswari M, Prabhavathi S, Gurunathan B, Chandhuru J, Sastry TP. Carbon nanoparticle from a natural source fabricated for folate receptor targeting, imaging and drug delivery application in A549 lung cancer cells. Eur J Pharm Biopharm 2014; 88:730-6. [DOI: 10.1016/j.ejpb.2014.09.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 09/26/2014] [Accepted: 09/29/2014] [Indexed: 12/26/2022]
|
43
|
Abstract
Folate is essential for fetal development, and its deficiency during gestation causes behavioural deficits in the offspring. The present study investigated its influence during weaning on brain function in the pups of rats that were put on a folate-deficient (FD) diet on postnatal day (PND) 1. Systemic folate deficiency in pups on the FD diet (n 15) was evident from the dramatically lower hepatic folate concentrations (median 23·7, range 8·1–48·4 ng/mg protein) and higher homocysteine concentrations (median 27·7, range 14·7–45·5 pmol/mg protein), respectively, compared with those of pups on the normal diet (ND; n 9) (median 114·5, range 64·5–158·5 ng/mg protein and median 15·5, range 11·6–18·9 pmol/mg protein) on PND 23. Brain folate concentrations although low were similar in pups on the FD diet (median 10·5, range 5·5–24·5 ng/mg protein) and ND (median 11·1, range 7·1–24·2 ng/mg protein). There was a high accumulation of homocysteine in the brain of FD pups, mostly in the hippocampus (median 58·1, range 40·8–99·7 pmol/mg protein) and cerebellum (median 69·1, range 50·8–126·6 pmol/mg protein), indicating metabolic folate deficiency despite normal brain folate concentrations. Developmental deficits or autistic traits were more frequent in the FD group than in the ND group and repetitive self-grooming occurred, on average, three times (range 1–8) v. once (range 0–3) during 5 min, respectively. Long-term memory or spatial learning and set-shifting deficits affected 12 to 62 % of rats in the FD group compared with none in the ND group. Post-weaning folic acid supplementation did not correct these deficits. These observations indicate that folate deficiency during weaning affects postnatal development even when gestational folate supply is normal.
Collapse
|
44
|
Fujiwara R, Takenaka S, Hashimoto M, Narawa T, Itoh T. Expression of human solute carrier family transporters in skin: possible contributor to drug-induced skin disorders. Sci Rep 2014; 4:5251. [PMID: 24918694 PMCID: PMC4052716 DOI: 10.1038/srep05251] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Accepted: 05/19/2014] [Indexed: 12/15/2022] Open
Abstract
Solute carrier (SLC) transporters play important roles in absorption and disposition of drugs in cells; however, the expression pattern of human SLC transporters in the skin has not been determined. In the present study, the expression patterns of 28 human SLC transporters were determined in the human skin. Most of the SLC transporter family members were either highly or moderately expressed in the liver, while their expression was limited in the skin and small intestine. Treatment of human keratinocytes with a reactive metabolite of ibuprofen significantly reduced cell viability. Expression array analysis revealed that S100 calcium binding protein A7A (S100A7A) was induced nearly 50-fold in dermal cells treated with ibuprofen acyl-glucuronide. Determination of the expression of drug-metabolizing enzymes as well as drug transporters prior to the administration of drugs would make it possible to avoid the development of idiosyncratic skin diseases in individuals.
Collapse
Affiliation(s)
- Ryoichi Fujiwara
- School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, JAPAN
| | - Saya Takenaka
- School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, JAPAN
| | - Mitsuhiro Hashimoto
- School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, JAPAN
| | - Tomoya Narawa
- School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, JAPAN
| | - Tomoo Itoh
- School of Pharmacy, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, JAPAN
| |
Collapse
|
45
|
Kissei M, Itoh T, Narawa T. Effect of epigallocatechin gallate on drug transport mediated by the proton-coupled folate transporter. Drug Metab Pharmacokinet 2014; 29:367-72. [PMID: 24695276 DOI: 10.2133/dmpk.dmpk-14-rg-015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Folic acid (FA) is a water-soluble vitamin, and orally ingested FA is absorbed from the small intestine by the proton-coupled folate transporter (PCFT). In the present study, we investigated whether epigallocatechin gallate (EGCG), one of the tea catechins, affects the transport of FA by PCFT. EGCG inhibited the uptake of FA into Caco-2 cells and human PCFT-expressing HEK293 cells (PCFT-HEK293 cells). The initial rate of uptake of FA into PCFT-HEK293 cells followed Michaelis-Menten kinetics (K(m) = 1.9 µM). Dixon plots revealed that PCFT-mediated FA uptake was competitively inhibited by EGCG (K(i) ≒ 9 µM). The uptake of the PCFT substrate methotrexate (MTX) was competitively inhibited by EGCG as well (K(i) ≒ 15 µM). In conclusion, it is suggested that when FA or MTX is ingested with tea, it is likely that the intestinal absorption of these compounds by PCFT is inhibited, which could result in insufficient efficacy.
Collapse
|
46
|
Scaglione F, Panzavolta G. Folate, folic acid and 5-methyltetrahydrofolate are not the same thing. Xenobiotica 2014; 44:480-8. [PMID: 24494987 DOI: 10.3109/00498254.2013.845705] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
1. Folate, an essential micronutrient, is a critical cofactor in one-carbon metabolism. Mammals cannot synthesize folate and depend on supplementation to maintain normal levels. Low folate status may be caused by low dietary intake, poor absorption of ingested folate and alteration of folate metabolism due to genetic defects or drug interactions. 2. Folate deficiency has been linked with an increased risk of neural tube defects, cardiovascular disease, cancer and cognitive dysfunction. Most countries have established recommended intakes of folate through folic acid supplements or fortified foods. External supplementation of folate may occur as folic acid, folinic acid or 5-methyltetrahydrofolate (5-MTHF). 3. Naturally occurring 5-MTHF has important advantages over synthetic folic acid - it is well absorbed even when gastrointestinal pH is altered and its bioavailability is not affected by metabolic defects. Using 5-MTHF instead of folic acid reduces the potential for masking haematological symptoms of vitamin B12 deficiency, reduces interactions with drugs that inhibit dihydrofolate reductase and overcomes metabolic defects caused by methylenetetrahydrofolate reductase polymorphism. Use of 5-MTHF also prevents the potential negative effects of unconverted folic acid in the peripheral circulation. 4. We review the evidence for the use of 5-MTHF in preventing folate deficiency.
Collapse
Affiliation(s)
- Francesco Scaglione
- Department of Medical Biotechnology and Translational Medicine, University of Milan , Milan , Italy
| | | |
Collapse
|
47
|
López KA, Piña MN, Alemany R, Vögler O, Barceló F, Morey J. Antifolate-modified iron oxide nanoparticles for targeted cancer therapy: inclusion vs. covalent union. RSC Adv 2014. [DOI: 10.1039/c4ra01216j] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In this work four different iron oxide nanoparticles for the delivery of antitumoral drugs into cancer cells were synthesized and characterized.
Collapse
Affiliation(s)
- K. A. López
- Department of Chemistry
- University of the Balearic Islands
- 07122 Palma de Mallorca, Spain
| | - M. N. Piña
- Department of Chemistry
- University of the Balearic Islands
- 07122 Palma de Mallorca, Spain
| | - R. Alemany
- Department of Biology
- University of the Balearic Islands
- 07122 Palma de Mallorca, Spain
| | - O. Vögler
- Department of Biology
- University of the Balearic Islands
- 07122 Palma de Mallorca, Spain
| | - F. Barceló
- Department of Biology
- University of the Balearic Islands
- 07122 Palma de Mallorca, Spain
| | - J. Morey
- Department of Chemistry
- University of the Balearic Islands
- 07122 Palma de Mallorca, Spain
| |
Collapse
|
48
|
Nazki FH, Sameer AS, Ganaie BA. Folate: Metabolism, genes, polymorphisms and the associated diseases. Gene 2014; 533:11-20. [DOI: 10.1016/j.gene.2013.09.063] [Citation(s) in RCA: 206] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Revised: 08/19/2013] [Accepted: 09/17/2013] [Indexed: 12/22/2022]
|
49
|
Korell J, Stamp LK, Barclay ML, Dalrymple JM, Drake J, Zhang M, Duffull SB. A population pharmacokinetic model for low-dose methotrexate and its polyglutamated metabolites in red blood cells. Clin Pharmacokinet 2013; 52:475-85. [PMID: 23483363 DOI: 10.1007/s40262-013-0052-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Measurement of intracellular concentrations of methotrexate (MTX) and its polyglutamated metabolites (MTXGlu(2-5)) in red blood cells (RBCs) has been suggested as a potential means of monitoring low-dose MTX treatment of rheumatoid arthritis (RA). However, a possible correlation between RBC MTX and MTXGlu2-5 concentrations and clinical outcomes of MTX treatment in RA is debated. A better understanding of the dose-concentration-time relationship of MTX and MTXGlu(2-5) in RBCs by population pharmacokinetic modelling is desirable and will facilitate assessing a potential RBC concentration-effect relationship in the future. AIM The purpose of this analysis was to describe the pharmacokinetics of MTX and MTXGlu(2-5) in RBCs. Secondary objectives included investigation of deglutamation reactions and the loss of MTX and MTXGlu(2-5) from the RBC. METHODS A model was developed using NONMEM(®) version 7.2 based on RBC data obtained from 48 patients with RA receiving once-weekly low-dose MTX treatment. This model was linked to a fixed two-compartment model that was used to describe the pharmacokinetics of MTX in the plasma. A series of five compartments were used to describe the intracellular pharmacokinetics of MTX and MTXGlu(2-5) in RBCs. Biologically plausible covariates were tested for a significant effect on MTX plasma clearance and the intracellular volume of distribution of all MTX species in RBCs ([Formula: see text]). The developed model was used to test hypotheses related to the enzymatic deglutamation of MTXGlu(2-5) and potential loss of MTXGlu(2-5) from RBCs. RESULTS The final RBC pharmacokinetic model required the intracellular volumes of distribution for the parent and metabolites to be set to the value estimated for the parent drug MTX alone, and the rate constants describing the polyglutamation steps were fixed at literature values. Significant covariates included effect of body surface area-adjusted estimated glomerular filtration rate on renal plasma clearance and effect of allometrically scaled total body weight with a fixed exponent of 0.75 on non-renal plasma clearance of MTX. The only significant covariate with an effect on [Formula: see text] was mean corpuscular volume (MCV). The model supported single deglutamation steps and a single mechanism of MTX and MTXGlu(2-5) loss from RBCs. CONCLUSIONS The developed model enabled acceptable description of the intracellular kinetics of MTX and MTXGlu(2-5) in RBCs. In the future it can form the basis of a full pharmacokinetic-pharmacodynamic model to assess the time-RBC concentration-effect relationship of low-dose MTX treatment in RA.
Collapse
Affiliation(s)
- Julia Korell
- School of Pharmacy, University of Otago, PO Box 56, 9054 Dunedin, New Zealand.
| | | | | | | | | | | | | |
Collapse
|
50
|
Yan J, Jin G, Du L, Yang Q. Modulation of intestinal folate absorption by erythropoietin in vitro. Mol Pharm 2013; 11:358-66. [PMID: 24294939 DOI: 10.1021/mp400318c] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Besides the direct stimulation of erythropoiesis, erythropoietin (EPO) therapy in renal anemia may also play a regulatory role in maintaining the homeostasis of hematopoietic nutrients. It has been reported that EPO can stimulate intestinal iron absorption. However, the involvement of EPO in intestinal folate absorption remains elusive. The objective of this study was to investigate the effect of EPO on intestinal transport of folate in vitro and to elucidate the possible mechanism(s) involved in this regulation. Transport assays of folic acid were performed in Caco-2 monolayers treated with EPO. The effect of EPO on the expression of transporters involved in the folate absorption was investigated. The possible involvement of three main EPO signaling pathways, the janus protein tyrosine kinase 2 (JAK-2) pathway, extracellular signal regulated kinases (ERK) pathway, and phosphatidylinositol 3 kinase/Akt (PI3K/Akt) pathway, in the transporter regulation was explored. The absorptive flux (apical to basolateral) of folic acid was enhanced by EPO treatment in a dose-dependent manner, which was companied with the significant up-regulation of reduced folate carrier (RFC) and apical proton coupled folate transporter (PCFT). The efflux (basolaterial to apical) of folic acid was enhanced only by the high dose of EPO treatment, which was associated with the significant up-regulation of apical multidrug resistance-associated protein 2 (MRP2). The expression levels of all of these transporters were up-regulated by EPO treatment in a dose- and time-dependent manner. Transporter expression in response to blocking EPO induced activation of JAK-2, ERK, and PI3K/Akt was changed to a different extent. As a conclusion, intestinal folate absorption was enhanced by EPO treatment in vitro. Our findings provided direct evidence to establish the correlation between EPO and folate homeostasis.
Collapse
Affiliation(s)
- Junkai Yan
- State Key Laboratory of Genetic Engineering, Department of Biochemistry, School of Life Sciences, Fudan University , Handan Road 220, Shanghai, China
| | | | | | | |
Collapse
|