Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 38(1): e23346, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-38095297

RESUMEN

Folate deficiency contribute to neural tube defects (NTDs) which could be rescued by folate supplementation. However, the underlying mechanisms are still not fully understood. Besides, there is considerable controversy concerning the forms of folate used for supplementation. To address this controversy, we prepared culture medium with different forms of folate, folic acid (FA), and 5-methyltetrahydrofolate (5mTHF), at concentrations of 5 µM, 500 nM, 50 nM, and folate free, respectively. Mouse embryonic fibroblasts (MEFs) were treated with different folates continuously for three passages, and cell proliferation and F-actin were monitored. We determined that compared to 5mTHF, FA showed stronger effects on promoting cell proliferation and F-actin formation. We also found that FOLR1 protein level was positively regulated by folate concentration and the non-canonical Wnt/planar cell polarity (PCP) pathway signaling was significantly enriched among different folate conditions in RNA-sequencing analyses. We demonstrated for the first time that FOLR1 could promote the transcription of Vangl2, one of PCP core genes. The transcription of Vangl2 was down-regulated under folate-deficient condition, which resulted in a decrease in PCP activity and F-actin formation. In summary, we identified a distinct advantage of FA in cell proliferation and F-actin formation over 5mTHF, as well as demonstrating that FOLR1 could promote transcription of Vangl2 and provide a new mechanism by which folate deficiency can contribute to the etiology of NTDs.


Asunto(s)
Deficiencia de Ácido Fólico , Defectos del Tubo Neural , Animales , Ratones , Ácido Fólico/metabolismo , Actinas/metabolismo , Receptor 1 de Folato/genética , Receptor 1 de Folato/metabolismo , Polaridad Celular/genética , Fibroblastos/metabolismo , Vía de Señalización Wnt , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Deficiencia de Ácido Fólico/metabolismo
2.
Gynecol Oncol ; 188: 52-57, 2024 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-38941962

RESUMEN

OBJECTIVE: Mirvetuximab soravtansine may be a potentially effective therapeutic option for ovarian low-grade serous carcinoma (LGSC), but the prevalence of folate receptor alpha (FRα) overexpression in this tumor type is unknown. We sought to characterize FRα expression in LGSC and its association with clinical and molecular features. METHODS: FRα immunohistochemistry was performed on a tissue microarray comprised of 89 LGSCs and 42 ovarian serous borderline tumors (SBTs). Clinical tumor-normal panel-based sequencing was performed on 78 LGSCs. Associations between FRα-high status and clinicopathologic characteristics and survival outcomes were examined. RESULTS: Of 89 LGSCs, 36 (40%) were FRα-high (≥75% of viable tumor cells exhibiting moderate-to-strong membranous expression). Of 9 patients with LGSC and samples from different timepoints, 4 (44%) had discordant results, with conversion from FRα-negative to FRα-high in 3 (33%) cases. There was no association between FRα-high status with age, race, or progression-free/overall survival. A MAPK pathway genetic alteration, most commonly involving KRAS (n = 23), was present in 45 (58%) LGSCs. Those lacking MAPK pathway alterations were more likely to be FRα-high compared to MAPK-altered LGSCs (61% vs 20%, p < 0.001). In SBTs, FRα-high expression was associated with high-risk (micropapillary) histology and/or subsequent LGSC recurrence compared to conventional SBTs without malignant recurrence (53% vs 9%, p = 0.008). CONCLUSIONS: Future studies of FRα-directed therapy in patients with LGSC are warranted. Discordant FRα status at recurrence suggests potential benefit for retesting. A biomarker-driven approach to direct treatment selection in LGSC is recommended. As high FRα expression is more common amongst tumors lacking MAPK pathway genetic alterations, FRα testing to determine eligibility for mirvetuximab soravtansine therapy is particularly recommended for this subgroup.

3.
Int J Mol Sci ; 25(2)2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-38256120

RESUMEN

Folate receptor α (FR) was discovered many decades ago, along with drugs that target intracellular folate metabolism, such as pemetrexed and methotrexate. Folate is taken up by the cell via this receptor, which also targeted by many cancer agents due to the over-expression of the receptor by cancer cells. FR is a membrane-bound glycosyl-phosphatidylinositol (GPI) anchor glycoprotein encoded by the folate receptor 1 (FOLR1) gene. FR plays a significant role in DNA synthesis, cell proliferation, DNA repair, and intracellular signaling, all of which are essential for tumorigenesis. FR is more prevalent in cancer cells compared to normal tissues, which makes it an excellent target for oncologic therapeutics. FRα is found in many cancer types, including ovarian cancer, non-small-cell lung cancer (NSCLC), and colon cancer. FR is widely used in antibody drug conjugates, small-molecule-drug conjugates, and chimeric antigen-receptor T cells. Current oncolytic therapeutics include mirvetuximab soravtansine, and ongoing clinical trials are underway to investigate chimeric antigen receptor T cells (CAR-T cells) and vaccines. Additionally, FRα has been used in a myriad of other applications, including as a tool in the identification of tumor types, and as a prognostic marker, as a surrogate of chemotherapy resistance. As such, FRα identification has become an essential part of precision medicine.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Humanos , Receptor 1 de Folato/genética , Medicina de Precisión , Ácido Fólico , Glicosilfosfatidilinositoles
4.
Neurol Sci ; 44(7): 2527-2540, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-36849695

RESUMEN

Neurodevelopmental disorders (NDDs) have broad heterogeneity both clinically and genetically. Inborn errors of metabolism can be one of the reasons of neurodevelopmental disruption causing specific NDDs. Although there is tremendous advance in molecular identification via next-generation sequencing (NGS), there are still many unsolved patients with NDD. Reanalysis of NGS data with different pipelines can at least partially accomplish this challenge. Herein, we report clinic and genetic components of an adult sib-pair with an undiagnosed NDD condition, which has been solved through reanalysis of whole-exome sequencing (WES). Parallel analysis of SNP-based genotyping and WES was performed to focus on variants only in loci with positive logarithm of the odds scores. WES data was analyzed through three different pipelines with two distinct bed files. Reanalysis of WES data led us to detect a homozygous FOLR1 variant (ENST00000393676.5:c.610C > T, p.(Arg204Ter), rs952165627) in the affected sib-pair. Surprisingly, the variant could not be detected in the first analysis as the variant region is not included in the first bed file which may frequently be used. Biochemical tests of CSF have confirmed the genetic analysis, CSF folic acid levels were detected low in sib-pair, and intravenous folinic acid treatment improved the disease course for the first 6 months of follow-up even at late diagnosis age. Although combined analysis of SNP-based genotyping and WES is a powerful tool to reveal the genetic components of heterogeneous diseases, reanalysis of genome data still should be considered in unsolved patients. Also, biochemical screening helps us to decipher undiagnosed NDD that may be a treatable neurometabolic condition.


Asunto(s)
Trastornos del Neurodesarrollo , Hermanos , Adulto , Humanos , Secuenciación del Exoma , Exoma/genética , Trastornos del Neurodesarrollo/diagnóstico , Trastornos del Neurodesarrollo/genética , Homocigoto , Receptor 1 de Folato/genética
5.
Int J Mol Sci ; 24(2)2023 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-36675153

RESUMEN

Folate deficiencies, folate imbalance and associated abnormal methylation are associated with birth defects, developmental delays, neurological conditions and diseases. In the hydrocephalic Texas (H-Tx) rat, 10-formyl tetrahydrofolate dehydrogenase (FDH) is reduced or absent from the CSF and the nuclei of cells in the brain and liver and this is correlated with decreased DNA methylation. In the present study, we tested whether impaired folate metabolism or methylation exists in sexually mature, unaffected H-Tx rats, which may explain the propagation of hydrocephalus in their offspring. We compared normal Sprague Dawley (SD, n = 6) rats with untreated H-Tx (uH-Tx, n = 6 and folate-treated H-Tx (TrH-Tx, n = 4). Structural abnormalities were observed in the testis of uH-Tx rats, with decreased methylation, increased demethylation, and cell death, particularly of sperm. FDH and FRα protein expression was increased in uH-Tx males but not in folate-treated males but tissue folate levels were unchanged. 5-Methylcytosine was significantly reduced in untreated and partially restored in treated individuals, while 5-hydroxymethylcytosine was not significantly changed. Similarly, a decrease in DNA-methyltransferase-1 expression in uH-Tx rats was partially reversed with treatment. The data expose a significant germline methylation error in unaffected adult male H-Tx rats from which hydrocephalic offspring are obtained. Reduced methylation in the testis and sperm was partially recovered by treatment with folate supplements leading us to conclude that this neurological disorder may not be completely eradicated by maternal supplementation alone.


Asunto(s)
Ácido Fólico , Hidrocefalia , Animales , Masculino , Ratas , Metilación de ADN , Ácido Fólico/metabolismo , Ácido Fólico/farmacología , Ácido Fólico/uso terapéutico , Ratas Sprague-Dawley , Semen/metabolismo , Hidrocefalia/congénito , Hidrocefalia/tratamiento farmacológico , Hidrocefalia/genética , Hidrocefalia/metabolismo , Modelos Animales de Enfermedad , Receptor 1 de Folato/genética , Receptor 1 de Folato/metabolismo
6.
Hum Mutat ; 43(12): 2021-2032, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36054333

RESUMEN

Neural tube defects (NTDs) are congenital malformations resulting from abnormal embryonic development of the brain, spine, or spinal column. The genetic etiology of human NTDs remains poorly understood despite intensive investigation. CIC, homolog of the Capicua transcription repressor, has been reported to interact with ataxin-1 (ATXN1) and participate in the pathogenesis of spinocerebellar ataxia type 1. Our previous study demonstrated that CIC loss of function (LoF) variants contributed to the cerebral folate deficiency syndrome by downregulating folate receptor 1 (FOLR1) expression. Given the importance of folate transport in neural tube formation, we hypothesized that CIC variants could contribute to increased risk for NTDs by depressing embryonic folate concentrations. In this study, we examined CIC variants from whole-genome sequencing (WGS) data of 140 isolated spina bifida cases and identified eight missense variants of CIC gene. We tested the pathogenicity of the observed variants through multiple in vitro experiments. We determined that CIC variants decreased the FOLR1 protein level and planar cell polarity (PCP) pathway signaling in a human cell line (HeLa). In a murine cell line (NIH3T3), CIC loss of function variants downregulated PCP signaling. Taken together, this study provides evidence supporting CIC as a risk gene for human NTD.


Asunto(s)
Defectos del Tubo Neural , Proteínas Represoras , Disrafia Espinal , Animales , Femenino , Humanos , Ratones , Embarazo , Receptor 1 de Folato/genética , Ácido Fólico , Mutación Missense , Defectos del Tubo Neural/genética , Células 3T3 NIH , Disrafia Espinal/genética , Células HeLa , Proteínas Represoras/genética
7.
Differentiation ; 117: 1-15, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33302058

RESUMEN

Folic acid is a nutrient essential for embryonic development. Folate deficiency can cause embryonic lethality or neural tube defects and orofacial anomalies. Folate receptor 1 (Folr1) is a folate binding protein that facilitates the cellular uptake of dietary folate. To better understand the biological processes affected by folate deficiency, gene expression profiles of gestational day 9.5 (gd9.5) Folr1-/- embryos were compared to those of gd9.5 Folr1+/+ embryos. The expression of 837 genes/ESTs was found to be differentially altered in Folr1-/- embryos, relative to those observed in wild-type embryos. The 837 differentially expressed genes were subjected to Ingenuity Pathway Analysis. Among the major biological functions affected in Folr1-/- mice were those related to 'digestive system development/function', 'cardiovascular system development/function', 'tissue development', 'cellular development', and 'cell growth and differentiation', while the major canonical pathways affected were those associated with blood coagulation, embryonic stem cell transcription and cardiomyocyte differentiation (via BMP receptors). Cellular proliferation, apoptosis and migration were all significantly affected in the Folr1-/- embryos. Cranial neural crest cells (NCCs) and neural tube explants, grown under folate-deficient conditions, exhibited marked reduction in directed migration that can be attributed, in part, to an altered cytoskeleton caused by perturbations in F-actin formation and/or assembly. The present study revealed that several developmentally relevant biological processes were compromised in Folr1-/- embryos.


Asunto(s)
Diferenciación Celular , Embrión de Mamíferos/metabolismo , Receptor 1 de Folato/fisiología , Ácido Fólico/metabolismo , Regulación del Desarrollo de la Expresión Génica , Cresta Neural/metabolismo , Defectos del Tubo Neural/patología , Animales , Embrión de Mamíferos/citología , Femenino , Perfilación de la Expresión Génica , Edad Gestacional , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Cresta Neural/patología , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo
8.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 47(1): 52-62, 2022 Jan 28.
Artículo en Inglés, Zh | MEDLINE | ID: mdl-35545363

RESUMEN

OBJECTIVES: Maternal periconceptional folic acid supplement is by far the most effective primary prevention strategy to reduce the incidence of congenital heart disease (CHD) in offspring. It was revealed that the underlying mechanisms are complex, including a combination of genetic and environmental factors. The purpose of this study is to investigate the association between periconceptional folic acid supplement, the genetic polymorphisms of maternal folic acid receptor 1 gene (FOLR1) and folic acid receptor 2 gene (FOLR2) and the impact of their interaction on the risk of CHD in offspring, and to provide epidemiological evidence for individualized folic acid dosing in hygienic counseling. METHODS: A case-control study on 569 mothers of CHD infants and 652 mothers of health controls was performed. The interesting points were periconceptional folate supplements, single nucleotide polymorphisms (SNPs) of maternal FOLR1 gene and FOLR2 gene. RESULTS: Mothers who took folate in the periconceptional period were observed a decreased risk of CHD [adjusted odds ratio (aOR)=0.58, 95% CI 0.35 to 0.95]. Our study also found that polymorphisms of maternal FOLR1 gene at rs2071010 (G/A vs G/G: aOR=0.67, 95% CI 0.47 to 0.96) and FOLR2 gene at rs514933 (T/C vs T/T: aOR=0.60, 95% CI 0.43 to 0.84; C/C vs T/T: aOR=0.55, 95% CI 0.33 to 0.90; the dominant model: T/C+ C/C vs T/T: aOR=0.59, 95% CI 0.43 to 0.81; and the addictive model: C/C vs T/C vs T/T: aOR=0.70, 95% CI 0.56 to 0.88) were significantly associated with lower risk of CHD [all P<0.05, false discovery rate P value (FDR_P)<0.1]. Besides, significant interaction between periconceptional folate supplements and rs2071010 G→A (aOR=0.59, 95% CI 0.41-0.86) and rs514933 T→C (aOR=0.52, 95% CI 0.37 to 0.74) on CHD risk were observed (all P<0.05, FDR_P<0.1). CONCLUSIONS: Periconceptional folate supplements, polymorphisms of FOLR1 gene and FOLR2 gene and their interactions are significantly associated with risk of CHD. However, more studies in different ethnic populations with a larger sample and prospective designs are required to confirm our findings.


Asunto(s)
Receptor 1 de Folato , Receptor 2 de Folato , Ácido Fólico , Cardiopatías Congénitas , Estudios de Casos y Controles , Suplementos Dietéticos , Femenino , Receptor 1 de Folato/genética , Receptor 2 de Folato/genética , Ácido Fólico/administración & dosificación , Cardiopatías Congénitas/genética , Hospitales , Humanos , Lactante , Polimorfismo de Nucleótido Simple , Estudios Prospectivos , Factores de Riesgo
9.
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
10.
BMC Med Genet ; 21(1): 235, 2020 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-33243190

RESUMEN

BACKGROUND: Cerebral folate deficiency (CFD) is a neurological disease, hallmarked by remarkable low concentrations of 5-methyltetrahydrofolic acid (5-MTHF) in cerebrospinal fluid (CSF). The primary causes of CFD include the presence of folate receptor (FR) autoantibodies, defects of FR encoding gene FOLR1, mitochondrial diseases and congenital abnormalities in folate metabolism. CASE PRESENTATION: Here we first present a Chinese male CFD patient whose seizure onset at 2 years old with convulsive status epilepticus. Magnetic Resonance Imaging (MRI) revealed the development of encephalomalacia, laminar necrosis in multiple lobes of the brain and cerebellar atrophy. Whole Exome Sequencing (WES) uncovered a homozygous missense variant of c.524G > T (p.C175F) in FOLR1 gene. Further laboratory tests demonstrated the extremely low level of 5-MTHF in the CSF from this patient, which was attributed to cerebral folate transport deficiency. Following the intravenous and oral treatment of calcium folinate, the concentrations of 5-MTHF in CSF were recovered to the normal range and seizure symptoms were relieved as well. CONCLUSIONS: One novel variation of FOLR1 was firstly identified from a Chinese male patient with tonic-clonic seizures, developmental delay, and ataxia. The WES and laboratory results elucidated the etiology of the symptoms. Clinical outcomes were improved by early diagnosis and proper treatment.


Asunto(s)
Encefalomalacia/genética , Receptor 1 de Folato/genética , Deficiencia de Ácido Fólico/genética , Convulsiones/genética , Estado Epiléptico/genética , Edad de Inicio , Corteza Cerebral/diagnóstico por imagen , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Niño , Encefalomalacia/líquido cefalorraquídeo , Encefalomalacia/diagnóstico por imagen , Encefalomalacia/tratamiento farmacológico , Receptor 1 de Folato/deficiencia , Deficiencia de Ácido Fólico/líquido cefalorraquídeo , Deficiencia de Ácido Fólico/diagnóstico por imagen , Deficiencia de Ácido Fólico/tratamiento farmacológico , Homocigoto , Humanos , Leucovorina/uso terapéutico , Imagen por Resonancia Magnética , Masculino , Convulsiones/líquido cefalorraquídeo , Convulsiones/diagnóstico por imagen , Convulsiones/tratamiento farmacológico , Estado Epiléptico/líquido cefalorraquídeo , Estado Epiléptico/diagnóstico por imagen , Estado Epiléptico/tratamiento farmacológico , Tetrahidrofolatos/líquido cefalorraquídeo , Secuenciación del Exoma
11.
Biomarkers ; 25(5): 367-374, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32421366

RESUMEN

Folate receptor alpha (FOLR1), a glycosylphosphatidylinositol-linked protein, is a well characterized folate transporter. However, the prognostic power of FOLR1 in cancer remains controversial. We conducted a meta-analysis to assess the prognostic roles of FOLR1 on different cancers. Twelve studies involving 4471 patients were included in this meta-analysis. The pooled analysis indicated that high FOLR1 significantly predicted poor overall survival (OS) (pooled hazard ratio (HR) = 0.78, 95% confidence interval (CI) = 0.64-0.94, p = 0.009) and the disease-free survival (DFS) (HR = 1.25, 95% CI = 1.07-1.47, p = 0.005). Subgroup analyses based on tumour type found that high FOLR1 level was associated with poor OS in breast cancer (HR = 2.66, 95% CI = 1.54-4.59, p = 0.0005) and endometrial carcinoma (HR = 1.30, 95% CI = 1.05-1.61, p = 0.02). However, FOLR1 has relatively weakly correlation with gender, tumour size and chemotherapy. Additionally, overexpression of FOLR1 was correlated with grade, FIGO stage, vital status and nodule status. The present meta-analysis indicated that the high expression of FOLR1 is associated with the poor survival of cancer patients, which is helpful for the clinical decision-making process.


Asunto(s)
Biomarcadores de Tumor/genética , Receptor 1 de Folato/genética , Neoplasias/genética , Pronóstico , Supervivencia sin Enfermedad , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias/epidemiología , Neoplasias/patología , Modelos de Riesgos Proporcionales
12.
Dev Dyn ; 248(10): 900-917, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31361376

RESUMEN

BACKGROUND: Abnormalities in maternal folate and carbohydrate metabolism have both been shown to induce neural tube defects (NTDs) in humans and animal models. Nevertheless, how these two factors might interact in the development of NTDs remains unclear. RESULTS: In specific mouse models and embryo culture systems, we assessed the effects of combining maternal diabetes with mutations in genes involved in folate transport and metabolism (methylenetetrahydrofolate reductase [Mthfr] and folic acid receptor 1 [Folr1]). When maternal hyperglycemia is combined with alterations in folic acid metabolism, there appears to be an increase in the incidence of congenital malformations in the offspring, with NTDs representing the majority of the malformations detected. CONCLUSIONS: The teratogenic effects of diabetes during pregnancy are exacerbated when combined with altered embryonic folate metabolism.


Asunto(s)
Diabetes Mellitus/genética , Ácido Fólico/metabolismo , Mutación , Defectos del Tubo Neural/etiología , Animales , Modelos Animales de Enfermedad , Técnicas de Cultivo de Embriones , Femenino , Receptor 1 de Folato/genética , Ácido Fólico/genética , Humanos , Metilenotetrahidrofolato Reductasa (NADPH2)/genética , Ratones , Embarazo , Teratogénesis
13.
J Inherit Metab Dis ; 42(4): 655-672, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30916789

RESUMEN

Cerebral folate deficiency is typically defined as a deficiency of the major folate species 5-methyltetrahydrofolate in the cerebrospinal fluid (CSF) in the presence of normal peripheral total folate levels. However, it should be noted that cerebral folate deficiency is also often used to describe conditions where CSF 5-MTHF is low, in the presence of low or undefined peripheral folate levels. Known defects of folate transport are deficiency of the proton coupled folate transporter, associated with systemic as well as cerebral folate deficiency, and deficiency of the folate receptor alpha, leading to an isolated cerebral folate deficiency associated with intractable seizures, developmental delay and/or regression, progressive ataxia and choreoathetoid movement disorders. Inborn errors of folate metabolism include deficiencies of the enzymes methylenetetrahydrofolate reductase, dihydrofolate reductase and 5,10-methenyltetrahydrofolate synthetase. Cerebral folate deficiency is potentially a treatable condition and so prompt recognition of these inborn errors and initiation of appropriate therapy is of paramount importance. Secondary cerebral folate deficiency may be observed in other inherited metabolic diseases, including disorders of the mitochondrial oxidative phosphorylation system, serine deficiency, and pyridoxine dependent epilepsy. Other secondary causes of cerebral folate deficiency include the effects of drugs, immune response activation, toxic insults and oxidative stress. This review describes the absorption, transport and metabolism of folate within the body; analytical methods to measure folate species in blood, plasma and CSF; inherited and acquired causes of cerebral folate deficiency; and possible treatment options in those patients found to have cerebral folate deficiency.


Asunto(s)
Encefalopatías Metabólicas Innatas/diagnóstico , Epilepsia/diagnóstico , Deficiencia de Ácido Fólico/diagnóstico , Ácido Fólico/uso terapéutico , Tetrahidrofolatos/deficiencia , Encéfalo/patología , Encefalopatías Metabólicas Innatas/líquido cefalorraquídeo , Encefalopatías Metabólicas Innatas/tratamiento farmacológico , Encefalopatías Metabólicas Innatas/genética , Diagnóstico Diferencial , Epilepsia/líquido cefalorraquídeo , Epilepsia/tratamiento farmacológico , Epilepsia/genética , Receptor 1 de Folato/genética , Deficiencia de Ácido Fólico/líquido cefalorraquídeo , Deficiencia de Ácido Fólico/tratamiento farmacológico , Deficiencia de Ácido Fólico/genética , Humanos , Tetrahidrofolatos/líquido cefalorraquídeo
14.
Arch Toxicol ; 93(12): 3535-3547, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31641807

RESUMEN

Welding fumes were recently classified as carcinogenic to humans and worldwide millions work as welders or perform welding operations. The purpose of this study was to identify new biomarkers of welding-induced carcinogenesis. We evaluated a panel of 91 putative cancer-related proteins in serum in a cohort of welders working with mild steel (n = 77) and controls (n = 94) from southern Sweden sampled on two occasions 6-year apart using a longitudinal analysis (linear mixed models). The significant results from the longitudinal analysis were tested for reproducibility in welders (n = 88) and controls (n = 69) sampled once during the same sampling period as timepoint 1 or timepoint 2 (linear regression models), i.e., in a cross-sectional setting. The models were adjusted for age, body-mass index, and use of snus. All study participants were non-smokers at recruitment. Exposure to welding fumes was assessed using questionnaires and respirable dust measurement in the breathing zone that was adjusted for personal respiratory protection equipment. The median respirable dust in welders was 0.7 (0.2-4.2) and 0.5 (0.1-1.9) mg/m3 at the first and second timepoints, respectively. We identified 14 cancer-related proteins that were differentially expressed in welders versus controls in the longitudinal analysis, out of which three were also differentially expressed in the cross-sectional analysis (cross-sectional group). Namely, syndecan 1 (SDC1), folate receptor 1 (FOLR1), and secreted protein acidic and cysteine rich (SPARC) were downregulated, in welders compared with controls. In addition, FOLR1 was negatively associated with years welding. Disease and function analysis indicated that the top proteins are related to lung cancer as well as cell invasion and migration. Our study indicates that moderate exposure to welding fumes is associated with changes in circulating levels of putative cancer-related proteins, out of which FOLR1 showed a clear dose-response relationship. It is, however, unclear to which extent these changes are adaptive or potential early biomarkers of cancer.


Asunto(s)
Biomarcadores de Tumor/sangre , Proteínas de Neoplasias/sangre , Neoplasias/sangre , Exposición Profesional/efectos adversos , Soldadura , Adulto , Estudios de Casos y Controles , Estudios Transversales , Biomarcadores Ambientales , Receptor 1 de Folato/sangre , Humanos , Masculino , Persona de Mediana Edad , Osteonectina/sangre , Análisis de Componente Principal , Acero , Suecia , Sindecano-1/sangre
15.
BMC Cancer ; 16: 589, 2016 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-27485273

RESUMEN

BACKGROUND: In this retrospective study we evaluated the respective correlations and clinical relevance of FOLR1 mRNA expression, FOLR1 promoter specific methylation and global DNA hypomethylation in type I and type II ovarian cancer. METHODS: Two hundred fifty four ovarian cancers, 13 borderline tumours and 60 samples of healthy fallopian epithelium and normal ovarian epithelium were retrospectively analysed for FOLR1 expression with RT-PCR. FOLR1 DNA promoter methylation and global DNA hypomethylation (measured by means of LINE1 DNA hypomethylation) were evaluated with MethyLight technique. RESULTS: No correlation between FOLR1 mRNA expression and its specific promoter DNA methylation was found neither in type I nor in type II cancers, however, high FOLR1 mRNA expression was found to be correlated with global DNA hypomethylation in type II cancers (p = 0.033). Strong FOLR1 mRNA expression was revealed for Grades 2-3, FIGO stages III-IV, residual disease > 0, and serous histotype. High FOLR1 expression was found to predict increased platinum sensitivity in type I cancers (odds ratio = 3.288; 1.256-10.75; p = 0.020). One-year survival analysis showed in type I cancers an independent better outcome for strong expression of FOLR1 in FIGO stage III and IV. For the entire follow up period no significant independent outcome for FOLR1 expression was revealed. In type I cancers LINE 1 DNA hypomethylation was found to exhibit a worse PFS and OS which were confirmed to be independent in multivariate COX regression model for both PFS (p = 0.026) and OS (p = 0.012). CONCLUSION: No correlations were found between FOLR1 expression and its specific promoter methylation, however, high FOLR1 mRNA expression was associated with DNA hypomethylation in type II cancers. FOLR1 mRNA expression did not prove to predict clinical outcome in type II cancers, although strong FOLR1 expression generally denotes ovarian cancers with highly aggressive phenotype. In type I cancers, however, strong FOLR1 expression has been found to be a reliable indicator of improved platinum responsiveness reflecting a transient better one-year follow up outcome in highly FOLR1 expressing type I cancers. An independent prognostic role of global DNA hypomethylation was demonstrated in type I tumours.


Asunto(s)
Metilación de ADN , Receptor 1 de Folato/genética , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Regulación hacia Arriba , Anciano , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Elementos de Nucleótido Esparcido Largo , Persona de Mediana Edad , Neoplasias Ováricas/genética , Platino (Metal)/uso terapéutico , Regiones Promotoras Genéticas , Estudios Retrospectivos , Análisis de Supervivencia
16.
Genomics ; 103(1): 40-7, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24184359

RESUMEN

We characterized the gene conversions between the human folate receptor (FOLR) genes and those of five other primate species. We found 26 gene conversions having an average length of 534 nucleotides. The length of these conversions is correlated with sequence similarity, converted regions have a higher GC-content and the average size of converted regions from a functional donor to another functional donor is significantly smaller than the average size from a functional donor to a pseudogene. Furthermore, the few conversions observed in the FOLR1 and FOLR2 genes did not change any amino acids in their coding regions and did not affect their promoter regions. In contrast, the promoter and coding regions of the FOLR3 gene are frequently converted and these conversions changed many amino acids in marmoset. These results suggest that purifying selection is limiting the functional impact that frequent gene conversions have on functional folate receptor genes.


Asunto(s)
Receptor 1 de Folato/genética , Receptor 2 de Folato/genética , Conversión Génica , Primates/genética , Secuencia de Aminoácidos , Animales , Receptor 1 de Folato/metabolismo , Receptor 2 de Folato/metabolismo , Humanos , Datos de Secuencia Molecular , Filogenia , Regiones Promotoras Genéticas
17.
Epileptic Disord ; 16(1): 88-92, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24556562

RESUMEN

Loss-of-function mutations in the FOLR1 gene (MIM *136430), encoding the folate receptor alpha, impair cerebral folate transport and lead to a progressive neurometabolic disorder. We report on a 5-year-old boy with progressive ataxia, from the age of 2 years and 6 months, with myoclonic jerks, regression, and impressive myoclonic tonic spasms with drop attacks, which were partially provoked by touching his face or washing his hands. Delayed myelination and cerebellar atrophy on cranial MRI were important clues to the diagnosis of cerebral folate transport deficiency, which was confirmed by homozygosity for the known nonsense mutation p.R204X in the FOLR1 gene. Computed tomography taken after head injury revealed bilateral calcifications in the basal ganglia as a novel finding in a patient with FOLR1 mutation.


Asunto(s)
Ganglios Basales/patología , Calcinosis/etiología , Receptor 1 de Folato/deficiencia , Síncope/genética , Ganglios Basales/fisiopatología , Preescolar , Electroencefalografía , Predisposición Genética a la Enfermedad , Humanos , Imagen por Resonancia Magnética/métodos , Masculino , Mutación/genética , Síncope/fisiopatología , Grabación en Video/métodos
18.
Cureus ; 16(5): e61032, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38915965

RESUMEN

INTRODUCTION: Gastric cancer (GC) remains a leading cause of cancer-related mortality worldwide, owing to its aggressive nature and poor prognosis. The role of folate receptors, particularly folate receptor 1 (FOLR1) and folate receptor 2 (FOLR2), in cancer has been increasingly recognized due to their overexpression in various malignancies including gastric cancer, and its potential implications in cancer progression, treatment resistance and as therapeutic targets. OBJECTIVE: To evaluate the expression patterns of FOLR1 and FOLR2 in GC patients' tissue and blood specimens and to correlate these patterns with clinicopathological variables. METHODS: A total of 58 gastric cancer patients were enrolled at the Regional Cancer Centre (RCC) from March 2017 to March 2020. Immunohistochemical analysis was performed to examine the expression of FOLR1 and FOLR2 in formalin-fixed paraffin-embedded (FFPE) tissue samples. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) was performed to analyze FOLR1 and FOLR2 expression in blood samples. Statistical analyses were conducted using chi-square tests, independent T-tests, and Kaplan-Meier survival analysis. RESULTS: FOLR1 and FOLR2 were overexpressed in 82.76% and 70.69% of gastric cancer tissues, respectively. High expression levels of FOLR1 were significantly associated with the diffuse type of gastric cancer (p<0.005). qRT-PCR showed significant overexpression of FOLR1 in gastric cancer blood samples compared to control samples, with a median fold change of approximately 14.18 times. Conversely, FOLR2 was significantly underexpressed in gastric cancer samples, with a fold change of 0.30. However, no significant correlation was found between FOLR2 expression and the clinicopathological features. The overall survival analysis did not show a significant difference in survival rates based on the expression levels of FOLR1 and FOLR2. CONCLUSIONS: This study highlights the differential expression patterns of FOLR1 and FOLR2 in gastric cancer and underscores the complexity of their roles in cancer biology. While FOLR1 shows potential as a biomarker for gastric cancer due to its overexpression, further studies are needed to fully elucidate the therapeutic and prognostic implications of folate receptors in gastric cancer.

19.
Cancers (Basel) ; 16(2)2024 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-38254822

RESUMEN

Treatment options for ovarian cancer patients are limited, and a high unmet clinical need remains for targeted and long-lasting, efficient drugs. Genetically modified T cells expressing chimeric antigen receptors (CAR), are promising new drugs that can be directed towards a defined target and have shown efficient, as well as persisting, anti-tumor responses in many patients. We sought to develop novel CAR T cells targeting ovarian cancer and to assess these candidates preclinically. First, we identified potential CAR targets on ovarian cancer samples. We confirmed high and consistent expressions of the tumor-associated antigen FOLR1 on primary ovarian cancer samples. Subsequently, we designed a series of CAR T cell candidates against the identified target and demonstrated their functionality against ovarian cancer cell lines in vitro and in an in vivo xenograft model. Finally, we performed additional in vitro assays recapitulating immune suppressive mechanisms present in solid tumors and developed a process for the automated manufacturing of our CAR T cell candidate. These findings demonstrate the feasibility of anti-FOLR1 CAR T cells for ovarian cancer and potentially other FOLR1-expressing tumors.

20.
Front Immunol ; 15: 1403324, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38694498

RESUMEN

[This corrects the article DOI: 10.3389/fimmu.2023.1254532.].

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA