Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
FASEB J ; 38(13): e23790, 2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38982638

RESUMEN

Integrase strand transfer inhibitors (INSTIs) based antiretroviral therapy (ART) is currently used as first-line regimen to treat HIV infection. Despite its high efficacy and barrier to resistance, ART-associated neuropsychiatric adverse effects remain a major concern. Recent studies have identified a potential interaction between the INSTI, dolutegravir (DTG), and folate transport pathways at the placental barrier. We hypothesized that such interactions could also occur at the two major blood-brain interfaces: blood-cerebrospinal fluid barrier (BCSFB) and blood-brain barrier (BBB). To address this question, we evaluated the effect of two INSTIs, DTG and bictegravir (BTG), on folate transporters and receptor expression at the mouse BCSFB and the BBB in vitro, ex vivo and in vivo. We demonstrated that DTG but not BTG significantly downregulated the mRNA and/or protein expression of folate transporters (RFC/SLC19A1, PCFT/SLC46A1) in human and mouse BBB models in vitro, and mouse brain capillaries ex vivo. Our in vivo study further revealed a significant downregulation in Slc19a1 and Slc46a1 mRNA expression at the BCSFB and the BBB following a 14-day DTG oral treatment in C57BL/6 mice. However, despite the observed downregulatory effect of DTG in folate transporters/receptor at both brain barriers, a 14-day oral treatment of DTG-based ART did not significantly alter the brain folate level in animals. Interestingly, DTG treatment robustly elevated the mRNA and/or protein expression of pro-inflammatory cytokines and chemokines (Cxcl1, Cxcl2, Cxcl3, Il6, Il23, Il12) in primary cultures of mouse brain microvascular endothelial cells (BBB). DTG oral treatment also significantly upregulated proinflammatory cytokines and chemokine (Il6, Il1ß, Tnfα, Ccl2) at the BCSFB in mice. We additionally observed a downregulated mRNA expression of drug efflux transporters (Abcc1, Abcc4, and Abcb1a) and tight junction protein (Cldn3) at the CP isolated from mice treated with DTG. Despite the structural similarities, BTG only elicited minor effects on the markers of interest at both the BBB and BCSFB. In summary, our current data demonstrates that DTG but not BTG strongly induced inflammatory responses in a rodent BBB and BCSFB model. Together, these data provide valuable insights into the mechanism of DTG-induced brain toxicity, which may contribute to the pathogenesis of DTG-associated neuropsychiatric adverse effect.


Asunto(s)
Barrera Hematoencefálica , Compuestos Heterocíclicos con 3 Anillos , Oxazinas , Piperazinas , Piridonas , Animales , Ratones , Piperazinas/farmacología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Oxazinas/farmacología , Inflamación/inducido químicamente , Inflamación/metabolismo , Ratones Endogámicos C57BL , Femenino , Inhibidores de Integrasa VIH/farmacología , Inhibidores de Integrasa VIH/efectos adversos , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/metabolismo , Masculino , Antirretrovirales/efectos adversos , Encéfalo/metabolismo , Encéfalo/efectos de los fármacos
2.
J Ren Nutr ; 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38490516

RESUMEN

OBJECTIVES: Low plasma folate levels have been reported in patients undergoing hemodialysis and peritoneal dialysis (PD) in clinical studies. However, folate transport has never been mentioned as a factor contributing to low plasma folate levels in patients undergoing PD. The peritoneal equilibrium test (PET) assesses the plasma creatinine level and glucose transport abilities. This study aimed to evaluate the association between plasma folate levels and folate transport during PD based on PET grades. METHODS: This study recruited 50 patients who underwent PD for ≥3 months and were categorized according to PET grades. Data regarding plasma folate levels and dialysate folate were collected. The primary outcomes were the relationship between the PET grade and plasma folate level and between the PET grade and dialysate-to-plasma folate concentration ratio (D/P folate). Furthermore, the difference in the plasma folate level and D/P folate between men and women was assessed. RESULTS: The plasma folate level and the D/P folate significantly differed among the 4 PET groups (both P < .001). PET grade was significantly negatively correlated with plasma folate levels (r = -0.56, P < .001) and positively correlated with D/P folate (r = 0.686, P < .001). In subgroup analysis, neither the plasma folate level nor the D/P folate significantly differed between men and women. CONCLUSIONS: Our study provides clinical evidence that the PET grade is associated with the plasma folate level and D/P folate, regardless of sex. Larger cohort studies are warranted to assess the importance of folate supplementation during PD based on PET grades.

3.
Proc Natl Acad Sci U S A ; 116(35): 17531-17540, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31405972

RESUMEN

Folates are critical for central nervous system function. Folate transport is mediated by 3 major pathways, reduced folate carrier (RFC), proton-coupled folate transporter (PCFT), and folate receptor alpha (FRα/Folr1), known to be regulated by ligand-activated nuclear receptors. Cerebral folate delivery primarily occurs at the choroid plexus through FRα and PCFT; inactivation of these transport systems can result in very low folate levels in the cerebrospinal fluid causing childhood neurodegenerative disorders. These disorders have devastating effects in young children, and current therapeutic approaches are not sufficiently effective. Our group has previously reported in vitro that functional expression of RFC at the blood-brain barrier (BBB) and its upregulation by the vitamin D nuclear receptor (VDR) could provide an alternative route for brain folate uptake. In this study, we further demonstrated in vivo, using Folr1 knockout (KO) mice, that loss of FRα led to a substantial decrease of folate delivery to the brain and that pretreatment of Folr1 KO mice with the VDR activating ligand, calcitriol (1,25-dihydroxyvitamin D3), resulted in over a 6-fold increase in [13C5]-5-formyltetrahydrofolate ([13C5]-5-formylTHF) concentration in brain tissues, with levels comparable to wild-type animals. Brain-to-plasma concentration ratio of [13C5]-5-formylTHF was also significantly higher in calcitriol-treated Folr1 KO mice (15-fold), indicating a remarkable enhancement in brain folate delivery. These findings demonstrate that augmenting RFC functional expression at the BBB could effectively compensate for the loss of Folr1-mediated folate uptake at the choroid plexus, providing a therapeutic approach for neurometabolic disorders caused by defective brain folate transport.


Asunto(s)
Encéfalo/metabolismo , Receptor 1 de Folato/metabolismo , Ácido Fólico/metabolismo , Proteína Portadora de Folato Reducido/metabolismo , Vitamina D/metabolismo , Animales , Transporte Biológico , Biomarcadores , Barrera Hematoencefálica/metabolismo , Cromatografía Liquida , Femenino , Receptor 1 de Folato/genética , Expresión Génica , Inmunohistoquímica , Ratones , Ratones Noqueados , Espectrometría de Masas en Tándem
4.
FASEB J ; 34(8): 10516-10530, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32543769

RESUMEN

Folates are important for neurodevelopment and cognitive function. Folate transport across biological membranes is mediated by three major pathways: folate receptor alpha (FRα), proton-coupled folate transporter (PCFT), and reduced folate carrier (RFC). Brain folate transport primarily occurs at the choroid plexus through FRα and PCFT; inactivation of these transport systems results in suboptimal folate levels in the cerebrospinal fluid (CSF) causing childhood neurological disorders. Our group has reported that upregulation of RFC at the blood-brain barrier (BBB) through interactions with specific transcription factors, that is, vitamin D receptor (VDR) could increase brain folate delivery. This study investigates the role of nuclear respiratory factor 1 (NRF-1) in the regulation of RFC at the BBB. Activation of NRF-1/PGC-1α signaling through treatment with its specific ligand, pyrroloquinoline quinone (PQQ), significantly induced RFC expression and transport activity in hCMEC/D3 cells. In contrast, transfection with NRF-1 or PGC-1α targeting siRNA downregulated RFC functional expression in the same cell system. Applying chromatin immunoprecipitation (ChIP) assay, we further demonstrated that PQQ treatment increased NRF-1 binding to putative NRF-1 binding sites within the SLC19A1 promoter, which encodes for RFC. Additionally, in vivo treatment of wild type mice with PQQ-induced RFC expression in isolated mouse brain capillaries. Together, these findings demonstrate that NRF-1/PGC-1α activation by PQQ upregulates RFC functional expression at the BBB and could potentially enhance brain folate uptake.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Factor Nuclear 1 de Respiración/metabolismo , Proteína Portadora de Folato Reducido/metabolismo , Regulación hacia Arriba/fisiología , Animales , Sitios de Unión/efectos de los fármacos , Sitios de Unión/fisiología , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Receptor 1 de Folato/metabolismo , Ácido Fólico/metabolismo , Humanos , Masculino , Ratones , Cofactor PQQ/farmacología , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/fisiología , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Receptores de Calcitriol/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Regulación hacia Arriba/efectos de los fármacos
5.
Am J Med Genet A ; 185(8): 2526-2531, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34008900

RESUMEN

Cerebral folate transporter deficiency syndrome, caused by FOLR-1 mutations is characterized by late infantile onset, severe developmental regression, epilepsy, and leukodystrophy. An extremely low concentration of 5-methyltetrahydrofolate in the cerebrospinal fluid provides a crucial clue to its diagnosis and is a treatment target. Oral or intravenous folinic acid (5-formyltetrahydrofolate) administration improves clinical symptoms and brain magnetic resonance imaging (MRI) findings. We describe three siblings carrying a novel homozygous FOLR1 nonsense mutation, that were referred due to intractable epilepsy and progressive neurological decline. Brain MRI showed hypomyelination and cerebellar atrophy. Folinic acid (oral and intravenous) supplementation, initiated after over 15 years illness, has failed to result in any sizeable clinical or neurophysiological improvement. Cerebral folate transport deficiency bears overlapping clinical features with many severe developmental encephalopathies. It is crucial to recognize FOLR1 signs and establish an early clinical and molecular diagnosis in order to provide timely folinic acid treatment and improve outcome.


Asunto(s)
Receptor 1 de Folato/deficiencia , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Distrofias Neuroaxonales/diagnóstico , Distrofias Neuroaxonales/genética , Hermanos , Adolescente , Alelos , Encéfalo/diagnóstico por imagen , Encéfalo/efectos de los fármacos , Encéfalo/patología , Consanguinidad , Discapacidades del Desarrollo/diagnóstico , Discapacidades del Desarrollo/genética , Manejo de la Enfermedad , Epilepsia/diagnóstico , Epilepsia/genética , Femenino , Receptor 1 de Folato/genética , Ácido Fólico/administración & dosificación , Pruebas Genéticas , Genotipo , Humanos , Imagen por Resonancia Magnética , Masculino , Mutación , Distrofias Neuroaxonales/terapia , Fenotipo , Síndrome , Resultado del Tratamiento
6.
FASEB J ; 31(8): 3622-3635, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28432198

RESUMEN

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.


Asunto(s)
Cilios/fisiología , Embrión de Mamíferos/metabolismo , Embrión no Mamífero/metabolismo , Ácido Fólico/metabolismo , Tubo Neural/fisiología , Septinas/metabolismo , Animales , Dactinomicina/análogos & derivados , Desarrollo Embrionario/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Células HEK293 , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Metilación , Ratones , Defectos del Tubo Neural/etiología , Plásmidos , Transducción de Señal , Xenopus/embriología
7.
Mol Pharm ; 14(11): 3848-3858, 2017 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-28885847

RESUMEN

Folates are essential for brain development and function. Folate transport in mammalian tissues is mediated by three major folate transport systems, i.e., reduced folate carrier (RFC), proton-coupled folate transporter (PCFT), and folate receptor alpha (FRα), known to be regulated by ligand-activated nuclear receptors, such as vitamin D receptor (VDR). Folate uptake at the choroid plexus, which requires the actions of both FRα and PCFT, is critical to cerebral folate delivery. Inactivating FRα or PCFT mutations cause severe cerebral folate deficiency resulting in early childhood neurodegeneration. The objective of this study was to investigate the role of RFC in folate uptake at the level of the blood-brain barrier (BBB) and its potential regulation by VDR. We detected robust expression of RFC in different in vitro BBB model systems, particularly in immortalized cultures of human cerebral microvascular endothelial cells (hCMEC/D3) and isolated mouse brain capillaries. [3H]-methotrexate uptake by hCMEC/D3 cells at pH 7.4 was inhibited by PT523 and pemetrexed, antifolates with high affinity for RFC. We also showed that activation of VDR through calcitriol (1,25-dihydroxyvitamin D3) exposure up-regulates RFC mRNA and protein expression as well as function in hCMEC/D3 cells and isolated mouse brain capillaries. We further demonstrated that RFC expression could be down-regulated by VDR-targeting siRNA, further confirming the role of VDR in the direct regulation of this folate transporter. Together, these data suggest that augmenting RFC functional expression could constitute a novel strategy for enhancing brain folate delivery for the treatment of neurometabolic disorders caused by loss of FRα or PCFT function.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Receptores de Calcitriol/metabolismo , Proteína Portadora de Folato Reducido/metabolismo , Animales , Transporte Biológico , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Calcitriol/farmacología , Células Cultivadas , Ácido Fólico/metabolismo , Humanos , Masculino , Metotrexato/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Wistar , Receptores de Calcitriol/genética , Proteína Portadora de Folato Reducido/genética
8.
Am J Med Genet A ; 170A(4): 1007-16, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26789141

RESUMEN

Many folate-related genes have been investigated for possible causal roles in neural tube defects (NTDs) and oral clefts. However, no previous reports have examined the major gene responsible for folate uptake, the proton-coupled folate transporter (SLC46A1). We tested for association between these birth defects and single nucleotide polymorphisms in the SLC46A1 gene. The NTD study population included 549 complete and incomplete case-family triads, and 999 controls from Ireland. The oral clefts study population comprised a sample from Utah (495 complete and incomplete case-family triads and 551 controls) and 221 Filipino multiplex cleft families. There was suggestive evidence of increased NTD case risk with the rs17719944 minor allele (odds ratio (OR): 1.29; 95% confidence intervals (CI): [1.00-1.67]), and decreased maternal risk of an NTD pregnancy with the rs4795436 minor allele (OR: 0.62; [0.39-0.99]). In the Utah sample, the rs739439 minor allele was associated with decreased case risk for cleft lip with cleft palate (genotype relative risk (GRR): 0.56 [0.32-0.98]). Additionally, the rs2239907 minor allele was associated with decreased case risk for cleft lip with cleft palate in several models, and with cleft palate only in a recessive model (OR: 0.41; [0.20-0.85]). These associations did not remain statistically significant after correcting for multiple hypothesis testing. Nominal associations between SLC46A1 polymorphisms and both Irish NTDs and oral clefts in the Utah population suggest some role in the etiology of these birth defects, but further investigation in other populations is needed.


Asunto(s)
Labio Leporino/genética , Defectos del Tubo Neural/genética , Polimorfismo de Nucleótido Simple , Transportador de Folato Acoplado a Protón/genética , Alelos , Estudios de Casos y Controles , Frecuencia de los Genes , Estudios de Asociación Genética , Genotipo , Humanos , Factores de Riesgo
9.
Mol Cell Biochem ; 411(1-2): 151-60, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26433955

RESUMEN

Excessive alcohol consumption and dietary folate inadequacy are the main contributors leading to folate deficiency (FD). The present study was planned to study regulation of folate transport in conditions of FD and ethanol exposure in human embryonic kidney cell line. Also, the reversible nature of effects mediated by ethanol exposure and FD was determined by folate repletion and ethanol removal. For ethanol treatment, HEK293 cells were grown in medium containing 100 mM ethanol, and after treatment, one group of cells was shifted on medium that was free from ethanol. For FD treatment, cells were grown in folate-deficient medium followed by shifting of one group of cells on folate containing medium. FD as well as ethanol exposure resulted in an increase in folate uptake which was due to an increase in expression of folate transporters, i.e., reduced folate carrier, proton-coupled folate transporter, and folate receptor, both at the mRNA and protein level. The effects mediated by ethanol exposure and FD were reversible on removal of treatment. Promoter region methylation of folate transporters remained unaffected after FD and ethanol exposure. As far as transcription rate of folate transporters is concerned, an increase in rate of synthesis was observed in both ethanol exposure and FD conditions. Additionally, mRNA life of folate transporters was observed to be reduced by FD. An increased expression of folate transporters under ethanol exposure and FD conditions can be attributed to enhanced rate of synthesis of folate transporters.


Asunto(s)
Etanol/administración & dosificación , Deficiencia de Ácido Fólico/metabolismo , Transportadores de Ácido Fólico/biosíntesis , Ácido Fólico/metabolismo , Células HEK293 , Humanos
10.
J Biol Chem ; 289(36): 25287-95, 2014 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-25053408

RESUMEN

The proton-coupled folate transporter (PCFT) is a folate-proton symporter highly expressed in solid tumors that can selectively target cytotoxic antifolates to tumors under acidic microenvironment conditions. Predicted topology models for PCFT suggest that the loop domain between transmembrane domains (TMDs) 2 and 3 resides in the cytosol. Mutations involving Asp-109 or Arg-113 in the TMD2-3 loop result in loss of activity. By structural homology to other solute carriers, TMD2 may form part of the PCFT substrate binding domain. In this study we mutated the seven cysteine (Cys) residues of human PCFT to serine, creating Cys-less PCFT. Thirty-three single-Cys mutants spanning TMD2 and the TMD2-3 loop in a Cys-less PCFT background were transfected into PCFT-null HeLa cells. All 33 mutants were detected by Western blotting, and 28 were active for [(3)H]methotrexate uptake at pH 5.5. For the active residues, we performed pulldown assays with membrane-impermeable 2-aminoethyl methanethiosulfonate-biotin and streptavidin beads to determine their aqueous-accessibilities. Multiple residues in TMD2 and the TMD2-3 loop domain reacted with 2-aminoethyl methanethiosulfonate-biotin, establishing aqueous accessibilities. Pemetrexed pretreatment inhibited biotinylation of TMD2 mutants G93C and F94C, and biotinylation of these residues inhibited methotrexate transport activity. Our results suggest that the TMD 2-3 loop domain is aqueous-accessible and forms a novel reentrant loop structure. Residues in TMD2 form an aqueous transmembrane pathway for folate substrates, and Gly-93 and Phe-94 may contribute to a substrate binding domain. Characterization of PCFT structure is essential to understanding the transport mechanism including the critical determinants of substrate binding.


Asunto(s)
Sustitución de Aminoácidos , Cisteína/genética , Mutación , Transportador de Folato Acoplado a Protón/genética , Secuencia de Aminoácidos , Sitios de Unión/genética , Transporte Biológico/efectos de los fármacos , Transporte Biológico/genética , Biotinilación , Western Blotting , Membrana Celular/metabolismo , Cisteína/química , Cisteína/metabolismo , Ácido Fólico/metabolismo , Antagonistas del Ácido Fólico/farmacología , Glutamatos/farmacología , Guanina/análogos & derivados , Guanina/farmacología , Células HeLa , Humanos , Cinética , Metotrexato/metabolismo , Datos de Secuencia Molecular , Pemetrexed , Estructura Secundaria de Proteína , Transportador de Folato Acoplado a Protón/química , Transportador de Folato Acoplado a Protón/metabolismo , Tritio
11.
EBioMedicine ; 75: 103771, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34954655

RESUMEN

BACKGROUND: Due to the critical role of folates in neurodevelopment, it is important to understand potential interactions between anti-HIV drugs used during pregnancy, and folate delivery pathways in the placenta. This study investigates the effect of dolutegravir (DTG) exposure on the functional expression of the reduced folate carrier (RFC), proton-coupled folate transporter (PCFT), and folate receptor-α (FRα) in the placenta. METHODS: Human placental cell lines, human placental explants, and a pregnant mouse model treated with clinically relevant concentrations of DTG were used. Gene and protein expression were assessed by qPCR, immunoblot and immunohistochemical assays. Folate transport function was measured by applying radioisotope-based transport assays. FINDINGS: In placental cells, clinically relevant DTG exposure for 3h or 6h was associated with a modest but significant reduction in the expression of RFC and PCFT both at the mRNA and protein levels, as well as decreased uptake of RFC and PCFT substrates [3H]-methotrexate and [3H]-folic acid, respectively. In pregnant mice, DTG administration was associated with an increase in both placental RFC and PCFT mRNA expression, accompanied by a decrease in placental FRα mRNA under folate-deficient dietary conditions. INTERPRETATION: These findings demonstrate a potential interaction between DTG and folate transport pathways in the placenta, particularly in vivo, under folate deficient conditions, potentially impacting folate delivery to the foetus in the context of DTG-based ART during pregnancy. FUNDING: Funded by Ontario HIV Treatment Network, grant #506657; and Eunice Kennedy Shriver National Institute of Child Health & Human Development of the National Institutes of Health, award #R01HD104553.


Asunto(s)
Placenta , Roedores , Animales , Femenino , Transportadores de Ácido Fólico/metabolismo , Compuestos Heterocíclicos con 3 Anillos/farmacología , Humanos , Ratones , Oxazinas , Piperazinas , Placenta/metabolismo , Embarazo , Piridonas , Estados Unidos
12.
Nutrients ; 14(15)2022 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-35956272

RESUMEN

Cerebral folate deficiency syndrome (CFDS) is defined as any neuropsychiatric or developmental disorder characterized by decreased CSF folate levels in the presence of normal folate status outside the nervous system. The specific clinical profile appears to be largely determined by the presence or absence of intrauterine folate deficiency as well as postnatal age at which cerebral folate deficiency occurs. The primary cause of CFDS is identified as the presence of serum folate receptor-alpha (FRα) autoantibodies impairing folate transport across the choroid plexus to the brain whereas, in a minority of cases, mitochondrial disorders, inborn errors of metabolism and loss of function mutations of the FRα (FOLR1) gene are identified. Early recognition and diagnosis of CFDS and prompt intervention is important to improve prognosis with successful outcomes. In this article we focus on FRα autoimmunity and its different age-dependent clinical syndromes, the diagnostic criteria, and treatments to be considered, including prevention strategies in this at-risk population.


Asunto(s)
Deficiencia de Ácido Fólico , Ácido Fólico , Diagnóstico Precoz , Receptor 1 de Folato/deficiencia , Receptor 1 de Folato/genética , Receptor 1 de Folato/uso terapéutico , Ácido Fólico/uso terapéutico , Deficiencia de Ácido Fólico/metabolismo , Humanos , Distrofias Neuroaxonales , Síndrome
13.
Nutrients ; 14(12)2022 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-35745126

RESUMEN

Food fortification and folic acid supplementation during pregnancy have been implemented as strategies to prevent fetal malformations during pregnancy. However, with the emergence of conditions where folate metabolism and transport are disrupted, such as folate receptor alpha autoantibody (FRαAb)-induced folate deficiency, it is critical to find a folate form that is effective and safe for pharmacologic dosing for prolonged periods. Therefore, in this study, we explored the absorption and tissue distribution of folic acid (PGA), 5-methyl-tetrahydrofolate (MTHF), l-folinic acid (levofolinate), and d,l-folinic acid (Leucovorin) in adult rats. During absorption, all forms are converted to MTHF while some unconverted folate form is transported into the blood, especially PGA. The study confirms the rapid distribution of absorbed folate to the placenta and fetus. FRαAb administered, also accumulates rapidly in the placenta and blocks folate transport to the fetus and high folate concentrations are needed to circumvent or overcome the blocking of FRα. In the presence of FRαAb, both Leucovorin and levofolinate are absorbed and distributed to tissues better than the other forms. However, only 50% of the leucovorin is metabolically active whereas levofolinate is fully active and generates higher tetrahydrofolate (THF). Because levofolinate can readily incorporate into the folate cycle without needing methylenetetrahydrofolate reductase (MTHFR) and methionine synthase (MS) in the first pass and is relatively stable, it should be the folate form of choice during pregnancy, other disorders where large daily doses of folate are needed, and food fortification.


Asunto(s)
Ácido Fólico , Animales , Femenino , Embarazo , Ratas , Suplementos Dietéticos , Leucovorina , Metilenotetrahidrofolato Reductasa (NADPH2)/metabolismo , Tetrahidrofolatos/metabolismo , Distribución Tisular
14.
Cancers (Basel) ; 14(1)2021 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-35008360

RESUMEN

New therapies are urgently needed for epithelial ovarian cancer (EOC), the most lethal gynecologic malignancy. To identify new approaches for targeting EOC, metabolic vulnerabilities must be discovered and strategies for the selective delivery of therapeutic agents must be established. Folate receptor (FR) α and the proton-coupled folate transporter (PCFT) are expressed in the majority of EOCs. FRß is expressed on tumor-associated macrophages, a major infiltrating immune population in EOC. One-carbon (C1) metabolism is partitioned between the cytosol and mitochondria and is important for the synthesis of nucleotides, amino acids, glutathione, and other critical metabolites. Novel inhibitors are being developed with the potential for therapeutic targeting of tumors via FRs and the PCFT, as well as for inhibiting C1 metabolism. In this review, we summarize these exciting new developments in targeted therapies for both tumors and the tumor microenvironment in EOC.

15.
Metabolism ; 110: 154306, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32621820

RESUMEN

BACKGROUND: Iron is finely regulated due to its vital roles in organisms and the peroxidase reactivity if excess. Solute Carrier Family 46 Member 1 (SLC46A1), also named PCFT or HCP1, is the main importer of heme­iron in the intestine, but has a high abundance in the liver. Since the liver has a central role in iron homeostasis, whether SLC46A1 regulates hepatic iron metabolism is of interest to be identified. METHODS: The recombinant adeno-associated virus vectors were used to hepatic-specifically inhibit SLC46A1 expression to observe its effects on hepatic iron metabolism. Then the abilities of SLC46A1 in importing heme and folate, and consequent alterations of iron content in hepatocytes were determined. Furthermore, effects of iron on SLC46A1 expression were investigated both in vitro and in vivo. RESULTS: The hepatocyte-specific inhibition of SLC46A1 decreases iron content in the liver and increases iron content in serum. Expressions of iron-related molecules, transferrin receptor 1, hepcidin and ferroportin, are correspondingly altered. Interestingly, free heme concentration in serum is increased, indicating a decreased import of heme by the liver. In hepatocytes, SLC46A1 is capable of importing hemin, increasing intracellular iron content. The import of hemin by SLC46A1 is unaffected by its other substrate, folate. Instead, hemin treatment decreases SLC46A1 expression, reducing the import of folate. In addition, SLC46A1 itself shows to be iron-responsive both in vivo and in vitro, making it available for regulating iron metabolism. CONCLUSION: The results elucidate that SLC46A1 regulates iron metabolism in the liver through a folate-independent manner of importing heme. The iron-responsive characters of SLC46A1 give us a new clue to link heme or iron overload with folate deficiency diseases.


Asunto(s)
Hemo/metabolismo , Hierro/metabolismo , Hígado/metabolismo , Transportador de Folato Acoplado a Protón/fisiología , Animales , Células Cultivadas , Hemina/metabolismo , Hepatocitos/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Transportador de Folato Acoplado a Protón/antagonistas & inhibidores
16.
J Nutr Biochem ; 53: 121-132, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29220669

RESUMEN

The present study has been designed to determine the effect of folate modulation (deficiency/supplementation) with aging on the promoter methylation of tumor suppressor and proto-oncogenes to understand the underlying mechanism of epigenetic alterations. Folate deficiency was induced for 3 and 5 months in weanling, young and adult groups, and after 3 months of folate deficiency, they were repleted with physiological folate (2 mg/kg diet) and folate oversupplementation (8 mg/kg diet) for another 2 months. The methylation facet in the present study revealed that the combined effect of folate deficiency and aging decreased the methylation index. Folate deficiency with age resulted in the up-regulation of proto-oncogenes (C-MYC and C-JUN) and cell cycle regulator gene Cyclin E as a result of promoter hypomethylation. However, in case of tumor suppressor genes (p53, p15ink4b and p16ink4a), the expression levels were found to be decreased at transcriptional level due to promoter hypermethylation. Upon repletion with physiological folate and folate oversupplementation, we found down-regulation of proto-oncogenes and up-regulation of tumor suppressor genes as a result of promoter hypermethylation and hypomethylation, respectively. Deregulation of these important genes due to folate deficiency may contribute toward the pathogenesis at cellular level.


Asunto(s)
Envejecimiento/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Ácido Fólico/farmacología , Hígado/efectos de los fármacos , Envejecimiento/fisiología , Animales , Ciclinas/genética , ADN (Citosina-5-)-Metiltransferasa 1/genética , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Regulación de la Expresión Génica/efectos de los fármacos , Genes Supresores de Tumor/efectos de los fármacos , Genes myc , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Hígado/fisiología , Masculino , Ratas Wistar , S-Adenosilmetionina/metabolismo , Tetrahidrofolatos/farmacocinética , ADN Metiltransferasa 3B
17.
Mol Nutr Food Res ; 60(6): 1501-13, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26990146

RESUMEN

SCOPE: The present study was designed to identify the molecular mechanism of folate modulation and aging on aberrant liver folate transporter system. METHODS AND RESULTS: An in vivo rat model was used, in which weanling, young and adult rats were given folate deficient diet for 3 and 5 months and after 3 months of folate deficiency, one group received physiological folate repletion (2 mg/kg diet) and another group received over supplemented folate diet (8 mg/kg diet) for another 2 months. In adult group, 3 and 5 months of folate deficiency decreased serum and tissue folate levels with decreased uptake of folate, further associated with decreased expression levels of reduced folate carrier (RFC) and increased expression levels of folate exporter (ABCG2) at both mRNA and protein levels, which in turn regulated by promoter hypermethylation of RFC and promoter hypomethylation of ABCG2 gene. CONCLUSION: Promoter hypermethylation of RFC and promoter hypomethylation of ABCG2 may be attributed to the down regulation of RFC and up regulation of ABCG2 at mRNA and protein levels in conditions of 3 and 5 months of folate deficiency in the adult group.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Envejecimiento/genética , Epigénesis Genética , Hígado/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Antígenos de Histocompatibilidad Menor/metabolismo , Proteína Portadora de Folato Reducido/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Animales , Metilación de ADN , Dieta , Suplementos Dietéticos , Ácido Fólico/administración & dosificación , Ácido Fólico/sangre , Deficiencia de Ácido Fólico/sangre , Deficiencia de Ácido Fólico/tratamiento farmacológico , Proteínas de Transporte de Membrana/genética , Antígenos de Histocompatibilidad Menor/genética , Regiones Promotoras Genéticas , Ratas , Proteína Portadora de Folato Reducido/genética
18.
Mol Nutr Food Res ; 59(9): 1713-24, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26012520

RESUMEN

SCOPE: The study was designed to identify the regulatory mechanisms underlying the effects of ethanol exposure on intestinal folate transport and to investigate the reversibility of such effects. METHODS AND RESULTS: Caco-2 cells were grown in control and ethanol containing medium for 96 h. Thereafter, one subgroup of cells was shifted on ethanol free medium and grown for next 72 h. For in vivo studies, rats were given 1g ethanol/kg body weight/day either for 3 or 5 months and after 3 months of ethanol treatment, one group of rats received no ethanol for 2 months. A significant decrease in folic acid transport as well as expression of folate transporters was observed on ethanol treatment and the effects were reversible upon removal of ethanol. Ethanol exposure had no impact on CpG island methylation of the folate transporters however, an increase in their mRNA half-life was observed that seems to be a homeostatic mechanism. Chromatin immunoprecipitation assay revealed a decrease in binding of SP1 transcription factor to the promoter regions of folate transporters. CONCLUSION: Reduced binding of SP1 to the promoter region of folate transporters may be a part of the regulatory mechanism resulting in decreased expression of folate transporters on ethanol exposure.


Asunto(s)
Etanol/toxicidad , Transportadores de Ácido Fólico/genética , Intestinos/efectos de los fármacos , Factor de Transcripción Sp1/metabolismo , Activación Transcripcional/efectos de los fármacos , Alcoholismo/genética , Alcoholismo/metabolismo , Animales , Peso Corporal , Células CACO-2 , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Ácido Fólico/metabolismo , Transportadores de Ácido Fólico/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Masculino , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Antígenos de Histocompatibilidad Menor , Regiones Promotoras Genéticas , Transportador de Folato Acoplado a Protón/genética , Transportador de Folato Acoplado a Protón/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Proteína Portadora de Folato Reducido/genética , Proteína Portadora de Folato Reducido/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
19.
J Nutr Biochem ; 26(10): 1084-94, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26168702

RESUMEN

Folic acid is an essential micronutrient, deficiency of which can lead to disturbance in various metabolic processes of cell. Folate transport across intestine occurs via the involvement of specialized folate transporters viz. proton coupled folate transporter (PCFT) and reduced folate carrier (RFC), which express at the membrane surfaces. The current study was designed to identify the regulatory mechanisms underlying the effects of folate deficiency (FD) on folate transport in human intestinal cell line as well as in rats and to check the reversibility of such effects. Caco-2 cells were grown for five generations in control and FD medium. Following treatment, one subgroup of cells was shifted on folate sufficient medium and grown for three more generations. Similarly, rats were fed an FD diet for 3 and 5 months, and after 3 months of FD treatment, one group of rats were shifted on normal folate-containing diet. Increase in folate transport and expression of folate transporters were observed on FD treatment. However, when cells and rats were shifted to control conditions after treatment, transport and expression of these genes restored to the control level. FD was found to have no impact on promoter methylation of PCFT and RFC; however, messenger RNA stability of transporters was found to be decreased, suggesting some adaptive response. Overall, increased expression of transporters under FD conditions can be attributed to enhanced rate of transcription of folate transporters and also to the increased binding of specificity protein 1 transcription factor to the RFC promoter only.


Asunto(s)
Deficiencia de Ácido Fólico/fisiopatología , Ácido Fólico/metabolismo , Mucosa Intestinal/metabolismo , Animales , Transporte Biológico/fisiología , Células CACO-2 , Medios de Cultivo , Ácido Fólico/administración & dosificación , Expresión Génica , Humanos , Masculino , Transportador de Folato Acoplado a Protón/genética , Transportador de Folato Acoplado a Protón/fisiología , ARN Mensajero/análisis , Ratas , Ratas Wistar , Proteína Portadora de Folato Reducido/genética , Proteína Portadora de Folato Reducido/fisiología
20.
Expert Rev Neurother ; 15(7): 793-802, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26092490

RESUMEN

Cerebral folate deficiency is defined as any neurological condition associated with low cerebrospinal fluid folate concentrations. It is becoming increasingly associated with several neurological diseases, either genetic or environmental. Treatment of cerebral folate deficiency by folate supplementation is generally effective, improving the neurological outcome of some patients. However, to treat cerebral folate deficiency, the proper choice of one of the available folate forms is essential. The distinct brain folate metabolism features compared with peripheral folate metabolic pathways strongly suggest the investigation of different folate forms, such as the biologically active folinic acid and 5-methyltetrahydrofolate, since they are efficiently transported to the brain. Regarding the oral doses of the different folate forms, despite the fact that there are some recommendations, there is no general consensus. Further investigation and designing clinical trials are advisable to elucidate these aspects.


Asunto(s)
Enfermedades Cerebelosas/etiología , Enfermedades Cerebelosas/terapia , Ácido Fólico/metabolismo , Deficiencia de Vitamina B/complicaciones , Humanos , Deficiencia de Vitamina B/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda