Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 464
Filter
1.
Fluids Barriers CNS ; 21(1): 67, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39192328

ABSTRACT

BACKGROUND: Folates are a family of B9 vitamins essential for normal growth and development in the central nervous system (CNS). Transport of folates is mediated by three major transport proteins: folate receptor alpha (FRα), proton-coupled folate transporter (PCFT), and reduced folate carrier (RFC). Brain folate uptake occurs at the choroid plexus (CP) epithelium through coordinated actions of FRα and PCFT, or directly into brain parenchyma at the vascular blood-brain barrier (BBB), mediated by RFC. Impaired folate transport can occur due to loss of function mutations in FRα or PCFT, resulting in suboptimal CSF folate levels. Our previous reports have demonstrated RFC upregulation by nuclear respiratory factor-1 (NRF-1) once activated by the natural compound pyrroloquinoline quinone (PQQ). More recently, we have identified folate transporter localization at the arachnoid barrier (AB). The purpose of the present study was to further characterize folate transporters localization and function in AB cells, as well as their regulation by NRF-1/PGC-1α signaling and folate deficiency. METHODS: In immortalized mouse AB cells, polarized localization of RFC and PCFT was assessed by immunocytochemical analysis, with RFC and PCFT functionality examined with transport assays. The effects of PQQ treatment on changes in RFC functional expression were also investigated. Mouse AB cells grown in folate-deficient conditions were assessed for changes in gene expression of the folate transporters, and other key transporters and tight junction proteins. RESULTS: Immunocytochemical analysis revealed apical localization of RFC at the mouse AB epithelium, with PCFT localized on the basolateral side and within intracellular compartments. PQQ led to significant increases in RFC functional expression, mediated by activation of the NRF-1/PGC-1α signalling cascade. Folate deficiency led to significant increases in expression of RFC, MRP3, P-gp, GLUT1 and the tight junction protein claudin-5. CONCLUSION: These results uncover the polarized expression of RFC and PCFT at the AB, with induction of RFC functional expression by activation of the NRF-1/PGC-1α signalling pathway and folate deficiency. These results suggest that the AB may contribute to the flow of folates into the CSF, representing an additional pathway when folate transport at the CP is impaired.


Subject(s)
Folic Acid , Proton-Coupled Folate Transporter , Animals , Folic Acid/metabolism , Mice , Proton-Coupled Folate Transporter/metabolism , Proton-Coupled Folate Transporter/genetics , Folate Receptor 1/metabolism , Folate Receptor 1/genetics , Biological Transport/physiology , Reduced Folate Carrier Protein/metabolism , Reduced Folate Carrier Protein/genetics , Folic Acid Deficiency/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Choroid Plexus/metabolism
2.
J Pharm Biomed Anal ; 247: 116268, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38823222

ABSTRACT

Methotrexate (MTX) is commonly prescribed as the initial treatment for gestational trophoblastic neoplasia (GTN), but MTX monotherapy may not be effective for high-risk GTN and choriocarcinoma. The cellular uptake of MTX is essential for its pharmacological activity. Thus, our study aimed to investigate the cellular pharmacokinetics and transport mechanisms of MTX in choriocarcinoma cells. For the quantification of MTX concentrations in cellular matrix, a liquid chromatography-tandem mass spectrometry method was created and confirmed initially. MTX accumulation in BeWo, JEG-3, and JAR cells was minimal. Additionally, the mRNA levels of folate receptor α (FRα) and breast cancer resistance protein (BCRP) were relatively high in the three choriocarcinoma cell lines, whereas proton-coupled folate transporter (PCFT), reduced folate carrier (RFC), and organic anion transporter (OAT) 4 were low. Furthermore, the expression of other transporters was either very low or undetectable. Notably, the application of inhibitors and small interfering RNAs (siRNAs) targeting FRα, RFC, and PCFT led to a notable decrease in the accumulation of MTX in BeWo cells. Conversely, the co-administration of multidrug resistance protein 1 (MDR1) and BCRP inhibitors increased MTX accumulation. In addition, inhibitors of OATs and organic-anion transporting polypeptides (OATPs) reduced MTX accumulation, while peptide transporter inhibitors had no effect. Results from siRNA knockdown experiments and transporter overexpression cell models indicated that MTX was not a substrate of nucleoside transporters. In conclusion, the results indicate that FRα and multiple transporters such as PCFT, RFC, OAT4, and OATPs are likely involved in the uptake of MTX, whereas MDR1 and BCRP are implicated in the efflux of MTX from choriocarcinoma cells. These results have implications for predicting transporter-mediated drug interactions and offer potential directions for further research on enhancing MTX sensitivity.


Subject(s)
Choriocarcinoma , Methotrexate , Tandem Mass Spectrometry , Methotrexate/pharmacology , Humans , Choriocarcinoma/metabolism , Choriocarcinoma/drug therapy , Tandem Mass Spectrometry/methods , Cell Line, Tumor , Biological Transport , Chromatography, Liquid/methods , Female , Neoplasm Proteins/metabolism , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Uterine Neoplasms/drug therapy , Uterine Neoplasms/metabolism , Pregnancy , Folate Receptor 1/metabolism , Folate Receptor 1/genetics , RNA, Small Interfering , Reduced Folate Carrier Protein/metabolism , Reduced Folate Carrier Protein/genetics , Liquid Chromatography-Mass Spectrometry
3.
Am J Physiol Endocrinol Metab ; 326(4): E482-E492, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38324257

ABSTRACT

Vitamin D (VD) is a fat-soluble sterol that possesses a wide range of physiological functions. The present study aimed to evaluate the effects of VD on folate metabolism in zebrafish and further investigated the underlying mechanism. Wild-type (WT) zebrafish were fed with a diet containing 0 IU/kg VD3 or 800 IU/kg VD3 for 3 wk. Meanwhile, cyp2r1 mutant zebrafish with impaired VD metabolism was used as another model of VD deficiency. Our results showed that VD deficiency in zebrafish suppressed the gene expression of folate transporters, including reduced folate carrier (RFC) and proton-coupled folate transporter (PCFT) in the intestine. Moreover, VD influenced the gene expression of several enzymes related to cellular folate metabolism in the intestine and liver of zebrafish. Importantly, VD-deficient zebrafish contained a remarkably lower level of folate content in the liver. Notably, VD was incapable of altering folate metabolism in zebrafish when gut microbiota was depleted by antibiotic treatment. Further studies proved that gut commensals from VD-deficient fish displayed a lower capacity to produce folate than those from WT fish. Our study revealed the potential correlation between VD and folate metabolism in zebrafish, and gut microbiota played a key role in VD-regulated folate metabolism in zebrafish.NEW & NOTEWORTHY Our study has identified that VD influences intestinal uptake and transport of folate in zebrafish while also altering hepatic folate metabolism and storage. Interestingly, the regulatory effects of VD on folate transport and metabolism diminished after the gut flora was interrupted by antibiotic treatment, suggesting that the regulatory effects of VD on folate metabolism in zebrafish are most likely dependent on the intestinal flora.


Subject(s)
Vitamin D Deficiency , Vitamin D , Animals , Zebrafish , Folic Acid/pharmacology , Folic Acid/metabolism , Vitamins , Reduced Folate Carrier Protein/genetics , Reduced Folate Carrier Protein/metabolism , Anti-Bacterial Agents
4.
J World Fed Orthod ; 13(3): 123-127, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38395727

ABSTRACT

BACKGROUND: The disruption of craniofacial developmental pathways during early embryogenesis can lead to conditions such as nonsyndromic cleft lip with or without cleft palate (NSCL/P). Several lines of evidence indicate that inadequate maternal nutrition causes low folate levels during the periconceptional period, resulting in NSCL/P. Although substantial research has been conducted on the possible link between SLC19A1 genetic variants and NSCL/P, the association between SLC19A1 80G>A (rs1051266) and NSCL/P remains unclear. In the present study, the associations of SLC19A1 80G>A with NSCL/P risk were assessed by calculating the pooled odds ratios (ORs) and 95% confidence intervals (CIs) by meta-analyses. METHODS: Following the PRISMA guidelines, a meta-analysis was conducted on 10 studies assessing the NSCL/P risk associated with SLC19A1 80G>A variant. To ascertain the degree of relationship between the SLC19A1 80G>A genetic variant and the risk of NSCL/P, data were analyzed in allelic, recessive and dominant genetic models. CI of OR for each study and the pooled data were obtained. All statistical analyses were conducted utilizing the MetaGenyo software tool, which integrates the adjustment of P values for multiple testing through the Bonferroni method. RESULTS: The pooled analysis showed that SLC19A1 80G>A variant significantly increased the NSCL/P risk in the allelic model (OR 1.39; 95% CI 1.00-1.92), recessive model (OR 1.37; 95% CI 1.03-1.82) and dominant models (OR 1.7; 95% CI 1.05-2.90). Publication bias was not observed. CONCLUSIONS: This study supports that the SLC19A1 80G>A genetic variant is associated with NSCL/P risk.


Subject(s)
Cleft Lip , Cleft Palate , Genetic Predisposition to Disease , Genetic Variation , Reduced Folate Carrier Protein , Cleft Lip/genetics , Cleft Palate/genetics , Humans , Reduced Folate Carrier Protein/genetics , Alleles , Polymorphism, Single Nucleotide
5.
Fluids Barriers CNS ; 20(1): 47, 2023 Jun 16.
Article in English | MEDLINE | ID: mdl-37328777

ABSTRACT

BACKGROUND: Reduced folate carrier 1 (RFC1; SLC19a1) is the main responsible transporter for the B9 family of vitamins named folates, which are essential for normal tissue growth and development. While folate deficiency resulted in retinal vasculopathy, the expression and the role of RFC1 in blood-retinal barrier (BRB) are not well known. METHODS: We used whole mount retinas and trypsin digested microvessel samples of adult mice. To knockdown RFC1, we delivered RFC1-targeted short interfering RNA (RFC1-siRNA) intravitreally; while, to upregulate RFC1 we delivered lentiviral vector overexpressing RFC1. Retinal ischemia was induced 1-h by applying FeCl3 to central retinal artery. We used RT-qPCR and Western blotting to determine RFC1. Endothelium (CD31), pericytes (PDGFR-beta, CD13, NG2), tight-junctions (Occludin, Claudin-5 and ZO-1), main basal membrane protein (Collagen-4), endogenous IgG and RFC1 were determined immunohistochemically. RESULTS: Our analyses on whole mount retinas and trypsin digested microvessel samples of adult mice revealed the presence of RFC1 in the inner BRB and colocalization with endothelial cells and pericytes. Knocking down RFC1 expression via siRNA delivery resulted in the disintegration of tight junction proteins and collagen-4 in twenty-four hours, which was accompanied by significant endogenous IgG extravasation. This indicated the impairment of BRB integrity after an abrupt RFC1 decrease. Furthermore, lentiviral vector-mediated RFC1 overexpression resulted in increased tight junction proteins and collagen-4, confirming the structural role of RFC1 in the inner BRB. Acute retinal ischemia decreased collagen-4 and occludin levels and led to an increase in RFC1. Besides, the pre-ischemic overexpression of RFC1 partially rescued collagen-4 and occludin levels which would be decreased after ischemia. CONCLUSION: In conclusion, our study clarifies the presence of RFC1 protein in the inner BRB, which has recently been defined as hypoxia-immune-related gene in other tissues and offers a novel perspective of retinal RFC1. Hence, other than being a folate carrier, RFC1 is an acute regulator of the inner BRB in healthy and ischemic retinas.


Subject(s)
Blood-Retinal Barrier , Endothelial Cells , Reduced Folate Carrier Protein , Animals , Mice , Blood-Retinal Barrier/metabolism , Endothelial Cells/metabolism , Folic Acid/metabolism , Immunoglobulin G , Occludin/metabolism , Reduced Folate Carrier Protein/genetics , Reduced Folate Carrier Protein/metabolism , RNA, Small Interfering/metabolism , Trypsin/metabolism
6.
Exp Gerontol ; 178: 112208, 2023 07.
Article in English | MEDLINE | ID: mdl-37201763

ABSTRACT

BACKGROUND: Plasma folate levels are closely related to antioxidant capacity and are regulated by folate pathway gene polymorphism. However, few studies have explored the gender-specific association of folate pathway gene polymorphism with oxidative stress biomarkers. The present study was designed to explore the gender-specific independent and combined impacts of solute carrier family 19 member 1 (SLC19A1) and methylenetetrahydrofolate reductase (MTHFR) genetic polymorphisms on oxidative stress biomarkers in older adults. METHODS: A total of 401 subjects were recruited, including 145 males and 256 females. Demographic characteristics of the participants were collected by using a self-administered questionnaire. Fasting venous blood samples were taken for folate pathway gene genotyping, circulating lipids parameters and erythrocyte oxidative stress biomarkers measurement. The difference of genotype distribution and the Hardy-Weinberg equilibrium was calculated by the Chi-square test. The general linear model was applied to compare the plasma folate levels and erythrocyte oxidative stress biomarkers. Multiple linear regression was used to explore the correlation between genetic risk scores and oxidative stress biomarkers. Logistic regression was used to explore the association of genetic risk scores of folate pathway gene with folate deficiency. RESULTS: The male subjects have lower plasma folate and HDL-C levels than the female ones, and the male carrying MTHFR rs1801133 (CC) or MTHFR rs2274976 (GA) genotypes have higher erythrocyte SOD activity. The plasma folate levels, erythrocyte SOD and GSH-PX activities were negatively correlated with genetic risk scores in the male subjects. A positive correlation between the genetic risk scores and folate deficiency was observed in the male subjects. CONCLUSIONS: There was association between folate pathway gene polymorphism of Solute Carrier Family 19 Member 1 (SLC19A1) and Methylenetetrahydrofolate Reductase (MTHFR) with erythrocyte SOD and GSH-PX activities, and folate levels in male but not in female aging subjects. Genetic variant of genes involved in folate metabolism has strong impact on plasma folate levels in the male aging subjects. Our data demonstrated that there was a potential interaction of gender and its genetic background in affecting the body's antioxidant capacity and the risk of folate deficiency in aging subjects.


Subject(s)
Folic Acid Deficiency , Methylenetetrahydrofolate Reductase (NADPH2) , Aged , Female , Humans , Male , Antioxidants/metabolism , Biomarkers/metabolism , Folic Acid , Folic Acid Deficiency/genetics , Genotype , Homocysteine/metabolism , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/metabolism , Oxidative Stress , Polymorphism, Genetic , Reduced Folate Carrier Protein/genetics , Reduced Folate Carrier Protein/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Sex Characteristics
7.
Pharmacogenomics ; 24(5): 261-268, 2023 04.
Article in English | MEDLINE | ID: mdl-36999405

ABSTRACT

Aims: To analyze the methylation level in the promoter region of SLC19A1 in adult acute lymphoblastic leukemia (ALL) patients, and explore the relationship between methotrexate (MTX) drug metabolism and SLC19A1 methylation. Methods: The methylation levels of the SLC19A1 promoter region in 52 adult ALL patients who received high-dose MTX chemotherapy were retrospectively analyzed in combination with clinical indicators and plasma MTX concentration. Results: Methylation levels of 17 CpG units were differently correlated with clinical parameters of ALL patients including gender, age, immunophenotype and Philadelphia chromosome status. Patients with delayed MTX drug excretion had higher methylation levels in the SLC19A1 promoter region. Conclusion: The methylation may affect the MTX plasma level and adverse reactions, which may predict patients at risk of adverse reactions after high-dose MTX therapy.


Subject(s)
Methotrexate , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Adult , Methotrexate/adverse effects , Antimetabolites, Antineoplastic/adverse effects , Methylation , Retrospective Studies , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Promoter Regions, Genetic/genetics , Reduced Folate Carrier Protein/genetics
9.
Immunopharmacol Immunotoxicol ; 45(1): 16-25, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35850595

ABSTRACT

BACKGROUND: The first-line anti-rheumatic drug methotrexate (MTX) is used in the combination. Because of the unpredictable adverse reactions, optimization of relevant regimens is necessary and meaningful. This study aimed to study the possible interaction between Securidaca inappendiculate Hassk. Derived xanthones and MTX. METHODS: We established adjuvant-induced arthritis (AIA) model, which was treated with MTX and MTX + xanthone-rich fraction (XRF). The clinical efficacy was evaluated by histopathological examination, and LC-MS was used to monitor the blood concentration of MTX. Western blotting and immunohistochemistry were used to detect protein expression. In vitro, we assessed the activity of related transporters by cellular uptake assay based on HEK-293T cells. RESULTS: Compared with MTX-treated rats, inflammation in the immunized rats in the MTX + XRF group was obvious, indicating that XRF antagonized the anti-rheumatic effect of MTX. Meanwhile, XRF reduced liver and kidney injuries caused by MTX in addition to MTX. Results from immunohistochemical and nappendiculat assays suggested that XRF may reduce uptake of MTX by down-regulating reduced folate carrier 1 (RFC1). CONCLUSION: This study indicated that XRF could reduce the plasma concentration of MTX by inhibiting the expression of RFC1, antagonize the therapeutic effect of MTX on AIA rats, and reduce its oral bioavailability. The combination of S. inappendiculate and MTX should be further optimized to achieve the goal of increasing efficiency and reducing toxicity.


Subject(s)
Antirheumatic Agents , Securidaca , Xanthones , Rats , Animals , Methotrexate/pharmacology , Securidaca/metabolism , Reduced Folate Carrier Protein , Antirheumatic Agents/pharmacology , Antirheumatic Agents/therapeutic use , Xanthones/pharmacology
10.
Genes Immun ; 24(1): 12-20, 2023 02.
Article in English | MEDLINE | ID: mdl-36517554

ABSTRACT

Insufficient dietary folate intake, hereditary malabsorption, or defects in folate transport may lead to combined immunodeficiency (CID). Although loss of function mutations in the major intestinal folate transporter PCFT/SLC46A1 was shown to be associated with CID, the evidence for pathogenic variants of RFC/SLC19A1 resulting in immunodeficiency was lacking. We report two cousins carrying a homozygous pathogenic variant c.1042 G > A, resulting in p.G348R substitution who showed symptoms of immunodeficiency associated with defects of folate transport. SLC19A1 expression by peripheral blood mononuclear cells (PBMC) was quantified by real-time qPCR and immunostaining. T cell proliferation, methotrexate resistance, NK cell cytotoxicity, Treg cells and cytokine production by T cells were examined by flow cytometric assays. Patients were treated with and benefited from folinic acid. Studies revealed normal NK cell cytotoxicity, Treg cell counts, and naive-memory T cell percentages. Although SLC19A1 mRNA and protein expression were unaltered, remarkably, mitogen induced-T cell proliferation was significantly reduced at suboptimal folic acid and supraoptimal folinic acid concentrations. In addition, patients' PBMCs were resistant to methotrexate-induced apoptosis supporting a functionally defective SLC19A1. This study presents the second pathogenic SLC19A1 variant in the literature, providing the first experimental evidence that functionally defective variants of SLC19A1 may present with symptoms of immunodeficiency.


Subject(s)
Immunologic Deficiency Syndromes , Leucovorin , Reduced Folate Carrier Protein , Humans , Folic Acid/genetics , Folic Acid/metabolism , Leucovorin/therapeutic use , Leucovorin/metabolism , Leukocytes, Mononuclear/metabolism , Methotrexate/pharmacology , Methotrexate/therapeutic use , Proton-Coupled Folate Transporter/genetics , Proton-Coupled Folate Transporter/metabolism , Reduced Folate Carrier Protein/genetics , Immunologic Deficiency Syndromes/drug therapy , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/metabolism
11.
Nature ; 612(7938): 170-176, 2022 12.
Article in English | MEDLINE | ID: mdl-36265513

ABSTRACT

Cyclic dinucleotides (CDNs) are ubiquitous signalling molecules in all domains of life1,2. Mammalian cells produce one CDN, 2'3'-cGAMP, through cyclic GMP-AMP synthase after detecting cytosolic DNA signals3-7. 2'3'-cGAMP, as well as bacterial and synthetic CDN analogues, can act as second messengers to activate stimulator of interferon genes (STING) and elicit broad downstream responses8-21. Extracellular CDNs must traverse the cell membrane to activate STING, a process that is dependent on the solute carrier SLC19A122,23. Moreover, SLC19A1 represents the major transporter for folate nutrients and antifolate therapeutics24,25, thereby placing SLC19A1 as a key factor in multiple physiological and pathological processes. How SLC19A1 recognizes and transports CDNs, folate and antifolate is unclear. Here we report cryo-electron microscopy structures of human SLC19A1 (hSLC19A1) in a substrate-free state and in complexes with multiple CDNs from different sources, a predominant natural folate and a new-generation antifolate drug. The structural and mutagenesis results demonstrate that hSLC19A1 uses unique yet divergent mechanisms to recognize CDN- and folate-type substrates. Two CDN molecules bind within the hSLC19A1 cavity as a compact dual-molecule unit, whereas folate and antifolate bind as a monomer and occupy a distinct pocket of the cavity. Moreover, the structures enable accurate mapping and potential mechanistic interpretation of hSLC19A1 with loss-of-activity and disease-related mutations. Our research provides a framework for understanding the mechanism of SLC19-family transporters and is a foundation for the development of potential therapeutics.


Subject(s)
Cryoelectron Microscopy , Dinucleoside Phosphates , Folic Acid Antagonists , Folic Acid , Nucleotides, Cyclic , Animals , Humans , Dinucleoside Phosphates/metabolism , Folic Acid/metabolism , Folic Acid Antagonists/pharmacology , Mammals/metabolism , Nucleotides, Cyclic/metabolism , Reduced Folate Carrier Protein/chemistry , Reduced Folate Carrier Protein/genetics , Reduced Folate Carrier Protein/metabolism , Reduced Folate Carrier Protein/ultrastructure
12.
Nature ; 609(7929): 1056-1062, 2022 09.
Article in English | MEDLINE | ID: mdl-36071163

ABSTRACT

Folates are essential nutrients with important roles as cofactors in one-carbon transfer reactions, being heavily utilized in the synthesis of nucleic acids and the metabolism of amino acids during cell division1,2. Mammals lack de novo folate synthesis pathways and thus rely on folate uptake from the extracellular milieu3. The human reduced folate carrier (hRFC, also known as SLC19A1) is the major importer of folates into the cell1,3, as well as chemotherapeutic agents such as methotrexate4-6. As an anion exchanger, RFC couples the import of folates and antifolates to anion export across the cell membrane and it is a major determinant in methotrexate (antifolate) sensitivity, as genetic variants and its depletion result in drug resistance4-8. Despite its importance, the molecular basis of substrate specificity by hRFC remains unclear. Here we present cryo-electron microscopy structures of hRFC in the apo state and captured in complex with methotrexate. Combined with molecular dynamics simulations and functional experiments, our study uncovers key determinants of hRFC transport selectivity among folates and antifolate drugs while shedding light on important features of anion recognition by hRFC.


Subject(s)
Cryoelectron Microscopy , Folic Acid Antagonists , Methotrexate , Reduced Folate Carrier Protein , Anions/metabolism , Apoproteins/genetics , Apoproteins/metabolism , Biological Transport , Carbon/metabolism , Folic Acid/metabolism , Folic Acid Antagonists/chemistry , Folic Acid Antagonists/metabolism , Humans , Methotrexate/chemistry , Methotrexate/metabolism , Molecular Dynamics Simulation , Reduced Folate Carrier Protein/genetics , Reduced Folate Carrier Protein/metabolism , Reduced Folate Carrier Protein/ultrastructure , Substrate Specificity
13.
Front Immunol ; 13: 843369, 2022.
Article in English | MEDLINE | ID: mdl-35958555

ABSTRACT

Background: Multiple myeloma (MM) remains an incurable malignant tumor of plasma cells. Increasing evidence has reported that hypoxia and immune status contribute to the progression of MM. In this research, the prognostic value of the hypoxia-immune-related gene SLC19A1 in MM was evaluated by bioinformatics analysis. Method: RNA-sequencing (RNA-seq) data along with clinical information on MM were downloaded from the Gene Expression Omnibus (GEO) database. Consistent clustering analysis and ESTIMATE algorithms were performed to establish the MM sample subgroups related to hypoxia and immune status, respectively, based on the GSE24080 dataset. The differentially expressed analysis was performed to identify the hypoxia-immune-related genes. Subsequently, a hypoxia-immune-gene risk signature for MM patients was constructed by univariate and multivariate Cox regression analyses, which was also verified in the GSE4581 dataset. Furthermore, the mRNA expression of SLC19A1 was determined using qRT-PCR in 19 MM patients, and the correlations between the genetic expression of SLC19A1 and clinical features were further analyzed. Result: A total of 47 genes were identified as hypoxia-immune-related genes for MM. Among these genes, SLC19A1 was screened to construct a risk score model that had better predictive power for MM. The constructed prognostic signature based on SLC19A1 was verified in the GSE4581 dataset. All independent prognostic factors (age, ß2-microglobulin, LDH, albumin, MRI, and gene risk score) were used to develop a nomogram that showed a better performance for predicting the survival probability of MM patients for 1-5 years. Furthermore, SLC19A1 was highly expressed in newly diagnosed and relapsed MM patients, and high expression of SLC19A1 is correlated with higher bone marrow aspiration plasma cells and ß2-microglobulin levels in MM patients. Conclusion: In conclusion, our results suggest that SLC19A1 is aberrantly expressed in MM and highly expressed SLC19A1 might be a biomarker correlated with inferior prognosis. More importantly, we identified SLC19A1 as a hypoxia-immune-related gene in MM. Future functional and mechanistic studies will further clarify the roles of SLC19A1 in MM.


Subject(s)
Multiple Myeloma , Biomarkers, Tumor/metabolism , Humans , Hypoxia/genetics , Multiple Myeloma/diagnosis , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , Multivariate Analysis , Prognosis , Reduced Folate Carrier Protein
14.
Alcohol Alcohol ; 57(5): 581-588, 2022 Sep 10.
Article in English | MEDLINE | ID: mdl-35952336

ABSTRACT

AIMS: Wernicke-Korsakoff syndrome (WKS) is commonly associated with chronic alcohol misuse, a condition known to have multiple detrimental effects on thiamine metabolism. This study was conducted to identify genetic variants that may contribute to the development of WKS in individuals with alcohol dependence syndrome through alteration of thiamine transport into cells. METHODS: Exome sequencing data from a panel of genes related to alcohol metabolism and thiamine pathways were analysed in a discovery cohort of 29 individuals with WKS to identify possible genetic risk variants associated with its development. Variant frequencies in this discovery cohort were compared with European frequencies in the Genome Aggregation Database browser, and those present at significantly higher frequencies were genotyped in an additional cohort of 87 alcohol-dependent cases with WKS and 197 alcohol-dependent cognitively intact controls. RESULTS: Thirty non-synonymous variants were identified in the discovery cohort and, after filtering, 23 were taken forward and genotyped in the case-control cohort. Of these SLC19A1:rs1051266:G was nominally associated with WKS. SLC19A1 encodes the reduced folate carrier, a major transporter for physiological folate in plasma; rs1051266 is reported to impact folate transport. Thiamine pyrophosphate (TPP) efflux was significantly decreased in HEK293 cells, stably transfected with rs1051266:G, under thiamine deficient conditions when compared with the efflux from cells transfected with rs1051266:A (P = 5.7 × 10-11). CONCLUSION: This study provides evidence for the role of genetic variation in the SLC19A1 gene, which may contribute to the development of WKS in vivo through modulation of TPP transport in cells.


Subject(s)
Alcoholism , Korsakoff Syndrome , Reduced Folate Carrier Protein , Thiamine Deficiency , Alcoholism/complications , Ethanol , Folic Acid , Genetic Variation/genetics , HEK293 Cells , Humans , Korsakoff Syndrome/complications , Reduced Folate Carrier Protein/genetics , Thiamine , Thiamine Deficiency/genetics , Thiamine Pyrophosphate/metabolism
15.
J Clin Pharm Ther ; 47(12): 2049-2067, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35896189

ABSTRACT

WHAT IS KNOWN AND OBJECTIVE: Solute Carrier (SLC) transporters are known mediators of drug disposition that facilitate the influx of substrates and various chemotherapeutic agents into cells. Polymorphisms in the SLC19A1, SLCO1B1, and SLCO1B3 gene influence the prognosis in the cancer patients, but little is known about their role in lung cancer in Asians. So, the current study aims to investigate the polymorphisms in SLC19A1, SLCO1B1, and SLCO1B3 genes in Northern Indian lung cancer patients. METHODS: Patients with lung cancer who had a confirmed histology and cytology diagnosis were enrolled in the study. SLC polymorphisms were assessed by Polymerase Chain Reaction-Restriction Fragment Length Polymorphism (PCR-RFLP) for variations in SLC19A1 (G80 A), SLCO1B1 (A388 G, T521 C), and SLCO1B3 (A1683-5676 G). RESULTS AND DISCUSSION: Our results showed that mutant genotype for SLC19A1 G80 A polymorphism had higher median survival time (MST) compared to wild genotype. ADCC patients with mutant genotype showed better survival compared to wild genotype for SLC19A1 G80 A. SCLC patients G80 A polymorphism showed increased survival in patients with mutant genotype (p = 0.04). In SLCO1B3, A1683-5676 G patients carrying heterozygous alleles and administered with platinum and docetaxel showed inferior survival (p = 0.006). In T521 C variant, patients with carrier genotype had reduced chances of developing anaemia (p = 0.04). Patients with SLC19A1 and SLCO1B3 variants showed lower incidence of thrombocytopenia and nephrotoxicity. WHAT IS NEW AND CONCLUSION: Our findings imply that Solute Carrier gene polymorphisms modulate the overall survival in lung cancer patients undergoing platin-based doublet chemotherapy, also these polymorphisms have a modifying impact on the associated adverse events/toxicity.


Subject(s)
Lung Neoplasms , Polymorphism, Single Nucleotide , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Docetaxel/adverse effects , Genotype , Alleles , Liver-Specific Organic Anion Transporter 1/genetics , Solute Carrier Organic Anion Transporter Family Member 1B3/genetics , Solute Carrier Organic Anion Transporter Family Member 1B3/therapeutic use , Reduced Folate Carrier Protein/genetics
18.
Pharmacogenomics J ; 21(5): 608-621, 2021 10.
Article in English | MEDLINE | ID: mdl-34302046

ABSTRACT

Methotrexate (MTX) is the first-line therapy for rheumatoid arthritis. Nevertheless, MTX resistance is quite a common issue in clinical practice. There are some premises that aryl hydrocarbon receptor (AhR) gene battery may take part in MTX metabolism. In the present retrospective study, we analyzed genes expression of AHR genes battery associated with MTX metabolism in whole blood of RA patients with good and poor response to MTX treatment. Additionally, sequencing, genotyping and bioinformatics analysis of AHR repressor gene (AHRR) c.565C > G (rs2292596) and c.1933G > C (rs34453673) have been performed. Theoretically, both changes may have an impact on H3K36me3 and H3K27me3. Evolutionary analysis revealed that rs2292596 may be possibly damaging. Allele G in rs2292596 and DAS28 seems to be associated with a higher risk of poor response to MTX treatment in RA. RA patients with poor response to MTX treatment revealed upregulated AhR and SLC19A1 mRNA level. Treatment with IL-6 inhibitor may be helpful to overcome the low-dose MTX resistance. Analysis of gene expression revealed possible another cause of poor response to MTX treatment which is different from that observed in the case of acute lymphoblastic leukemia.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Methotrexate/therapeutic use , Receptors, Aryl Hydrocarbon/genetics , Adult , Aged , Aged, 80 and over , Alleles , Arthritis, Rheumatoid/genetics , Drug Resistance/genetics , Female , Genes/genetics , Genotyping Techniques , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Reduced Folate Carrier Protein/genetics , Treatment Outcome , Young Adult
19.
Drug Metab Pharmacokinet ; 40: 100409, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34293696

ABSTRACT

Valproate (VPA), an antiepileptic drug, is known to inhibit histone deacetylases (HDACs). Exposure to VPA during pregnancy increases several fetal risks. The maintenance of folate level during pregnancy is essential for adequate fetal development, and the placenta plays a critical role in supplying nutrients to the fetus. The aim of this study was to elucidate the effects of VPA on the gene expression of folate carriers and metabolizing enzymes in the rat placenta at both mid and late gestation periods. Pregnant rats were orally administered VPA on a single day or 4 days (repeated administration). Gene expression of folate carriers (Folr1, Slc19a1, Slc46a1) and metabolizing enzymes (Cth, Mtr, Mtrr, Mthfr, Dhfr) was assessed in the placenta on gestational day (GD) 13 or GD20. In the control rats, the expression of Folr1, Slc46a1, Cth, and Mthfr tended to be upregulated, whereas that of Mtrr and Dhfr was downregulated during gestation; the expression of Slc19a1 and Mtr did not change. Repeated VPA administration reduced the placental expression of Folr1and Mtr on GD20 and increased the expression of Dhfr on GD13 compared with the control. These findings indicate that administration of VPA alters the placental gene expression of folate carriers and metabolism-related enzymes.


Subject(s)
Placenta , Valproic Acid , Animals , Anticonvulsants/therapeutic use , Female , Folic Acid , Histone Deacetylase Inhibitors/pharmacology , Membrane Transport Proteins , Minor Histocompatibility Antigens , Pregnancy , Proton-Coupled Folate Transporter/genetics , Rats , Reduced Folate Carrier Protein/genetics
20.
Zhongguo Dang Dai Er Ke Za Zhi ; 23(6): 547-554, 2021 Jun.
Article in Chinese | MEDLINE | ID: mdl-34130774

ABSTRACT

OBJECTIVE: To study the association between maternal reduced folate carrier (RFC) gene polymorphisms and congenital heart disease (CHD) in offspring. METHODS: A hospital-based case-control study was conducted. The mothers of 683 infants with CHD who attended the Department of Cardiothoracic Surgery, Hunan Children's Hospital, from November 2017 to March 2020 were enrolled as the case group. The mothers of 740 healthy infants without any deformity who attended the hospital during the same period of time were enrolled as the control group. A questionnaire survey was performed to collect the exposure data of subjects. Venous blood samples of 5 mL were collected from the mothers for genetic polymorphism detection. A multivariate logistic regression analysis was used to evaluate the association of RFC gene polymorphisms and their haplotypes with CHD. A generalized multifactor dimensionality reduction method was used to analyze gene-gene interactions. RESULTS: After control for confounding factors, the multivariate logistic regression analysis showed that maternal RFC gene polymorphisms at rs2236484 (AG vs AA:OR=1.91, 95%CI:1.45-2.51; GG vs AA: OR=1.96, 95%CI:1.40-2.75) and rs2330183 (CT vs CC:OR=1.39, 95%CI:1.06-1.83) were significantly associated with the risk of CHD in offspring. The haplotypes of G-G (OR=1.21, 95%CI:1.03-1.41) and T-G (OR=1.25, 95%CI:1.07-1.46) in mothers significantly increased the risk of CHD in offspring. The interaction analysis showed significant gene-gene interactions between different SNPs of the RFC gene in CHD (P < 0.05). CONCLUSIONS: Maternal RFC gene polymorphisms and interactions between different SNPs are significantly associated with the risk of CHD in offspring.


Subject(s)
Heart Defects, Congenital , Polymorphism, Single Nucleotide , Case-Control Studies , Child , Female , Genetic Predisposition to Disease , Genotype , Heart Defects, Congenital/genetics , Humans , Infant , Reduced Folate Carrier Protein/genetics , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL