Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
FASEB J ; 38(11): e23738, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38855924

ABSTRACT

Maternal nutrition contributes to gene-environment interactions that influence susceptibility to common congenital anomalies such as neural tube defects (NTDs). Supplemental myo-inositol (MI) can prevent NTDs in some mouse models and shows potential for prevention of human NTDs. We investigated effects of maternal MI intake on embryonic MI status and metabolism in curly tail mice, which are genetically predisposed to NTDs that are inositol-responsive but folic acid resistant. Dietary MI deficiency caused diminished MI in maternal plasma and embryos, showing that de novo synthesis is insufficient to maintain MI levels in either adult or embryonic mice. Under normal maternal dietary conditions, curly tail embryos that developed cranial NTDs had significantly lower MI content than unaffected embryos, revealing an association between diminished MI status and failure of cranial neurulation. Expression of inositol-3-phosphate synthase 1, required for inositol biosynthesis, was less abundant in the cranial neural tube than at other axial levels. Supplemental MI or d-chiro-inositol (DCI) have previously been found to prevent NTDs in curly tail embryos. Here, we investigated the metabolic effects of MI and DCI treatments by mass spectrometry-based metabolome analysis. Among inositol-responsive metabolites, we noted a disproportionate effect on nucleotides, especially purines. We also found altered proportions of 5-methyltetrahydrolate and tetrahydrofolate in MI-treated embryos suggesting altered folate metabolism. Treatment with nucleotides or the one-carbon donor formate has also been found to prevent NTDs in curly tail embryos. Together, these findings suggest that the protective effect of inositol may be mediated through the enhanced supply of nucleotides during neural tube closure.


Subject(s)
Inositol , Neural Tube Defects , Inositol/metabolism , Inositol/pharmacology , Neural Tube Defects/metabolism , Neural Tube Defects/prevention & control , Animals , Female , Mice , Pregnancy , Embryo, Mammalian/metabolism , Maternal Nutritional Physiological Phenomena , Metabolome , Folic Acid/metabolism
2.
Mol Genet Metab ; 142(3): 108496, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38761651

ABSTRACT

Non-Ketotic Hyperglycinemia (NKH) is a rare inborn error of metabolism caused by impaired function of the glycine cleavage system (GCS) and characterised by accumulation of glycine in body fluids and tissues. NKH is an autosomal recessive condition and the majority of affected individuals carry mutations in GLDC (glycine decarboxylase). Current treatments for NKH have limited effect and are not curative. As a monogenic condition with known genetic causation, NKH is potentially amenable to gene therapy. An AAV9-based expression vector was designed to target sites of GCS activity. Using a ubiquitous promoter to drive expression of a GFP reporter, transduction of liver and brain was confirmed following intra-venous and/or intra-cerebroventricular administration to neonatal mice. Using the same capsid and promoter with transgenes to express mouse or human GLDC, vectors were then tested in GLDC-deficient mice that provide a model of NKH. GLDC-deficient mice exhibited elevated plasma glycine concentration and accumulation of glycine in liver and brain tissues as previously observed. Moreover, the folate profile indicated suppression of folate one­carbon metabolism (FOCM) in brain tissue, as found at embryonic stages, and reduced abundance of FOCM metabolites including betaine and choline. Neonatal administration of vector achieved reinstatement of GLDC mRNA and protein expression in GLDC-deficient mice. Treated GLDC-deficient mice showed significant lowering of plasma glycine, confirming functionality of vector expressed protein. AAV9-GLDC treatment also led to lowering of brain tissue glycine, and normalisation of the folate profile indicating restoration of glycine-derived one­carbon supply. These findings support the hypothesis that AAV-mediated gene therapy may offer potential in treatment of NKH.


Subject(s)
Brain , Dependovirus , Disease Models, Animal , Genetic Therapy , Genetic Vectors , Glycine Dehydrogenase (Decarboxylating) , Glycine , Hyperglycinemia, Nonketotic , Liver , Animals , Hyperglycinemia, Nonketotic/genetics , Hyperglycinemia, Nonketotic/metabolism , Hyperglycinemia, Nonketotic/therapy , Glycine Dehydrogenase (Decarboxylating)/genetics , Glycine Dehydrogenase (Decarboxylating)/metabolism , Dependovirus/genetics , Mice , Humans , Genetic Vectors/genetics , Glycine/metabolism , Liver/metabolism , Brain/metabolism , Biomarkers/metabolism , Folic Acid/metabolism
3.
Mol Cell Proteomics ; 23(3): 100718, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38224738

ABSTRACT

A functional role has been ascribed to the human dihydrofolate reductase 2 (DHFR2) gene based on the enzymatic activity of recombinant versions of the predicted translated protein. However, the in vivo function is still unclear. The high amino acid sequence identity (92%) between DHFR2 and its parental homolog, DHFR, makes analysis of the endogenous protein challenging. This paper describes a targeted mass spectrometry proteomics approach in several human cell lines and tissue types to identify DHFR2-specific peptides as evidence of its translation. We show definitive evidence that the DHFR2 activity in the mitochondria is in fact mediated by DHFR, and not DHFR2. Analysis of Ribo-seq data and an experimental assessment of ribosome association using a sucrose cushion showed that the two main Ensembl annotated mRNA isoforms of DHFR2, 201 and 202, are differentially associated with the ribosome. This indicates a functional role at both the RNA and protein level. However, we were unable to detect DHFR2 protein at a detectable level in most cell types examined despite various RNA isoforms of DHFR2 being relatively abundant. We did detect a DHFR2-specific peptide in embryonic heart, indicating that the protein may have a specific role during embryogenesis. We propose that the main functionality of the DHFR2 gene in adult cells is likely to arise at the RNA level.


Subject(s)
RNA , Tetrahydrofolate Dehydrogenase , Humans , Cell Line , Peptides/metabolism , Protein Biosynthesis , Ribosomes/metabolism , RNA/metabolism , RNA, Messenger/metabolism , Tetrahydrofolate Dehydrogenase/genetics , Tetrahydrofolate Dehydrogenase/metabolism
4.
Hum Mol Genet ; 32(17): 2681-2692, 2023 08 26.
Article in English | MEDLINE | ID: mdl-37364051

ABSTRACT

Orofacial clefts, including cleft lip and palate (CL/P) and neural tube defects (NTDs) are among the most common congenital anomalies, but knowledge of the genetic basis of these conditions remains incomplete. The extent to which genetic risk factors are shared between CL/P, NTDs and related anomalies is also unclear. While identification of causative genes has largely focused on coding and loss of function mutations, it is hypothesized that regulatory mutations account for a portion of the unidentified heritability. We found that excess expression of Grainyhead-like 2 (Grhl2) causes not only spinal NTDs in Axial defects (Axd) mice but also multiple additional defects affecting the cranial region. These include orofacial clefts comprising midline cleft lip and palate and abnormalities of the craniofacial bones and frontal and/or basal encephalocele, in which brain tissue herniates through the cranium or into the nasal cavity. To investigate the causative mutation in the Grhl2Axd strain, whole genome sequencing identified an approximately 4 kb LTR retrotransposon insertion that disrupts the non-coding regulatory region, lying approximately 300 base pairs upstream of the 5' UTR. This insertion also lies within a predicted long non-coding RNA, oriented on the reverse strand, which like Grhl2 is over-expressed in Axd (Grhl2Axd) homozygous mutant embryos. Initial analysis of the GRHL2 upstream region in individuals with NTDs or cleft palate revealed rare or novel variants in a small number of cases. We hypothesize that mutations affecting the regulation of GRHL2 may contribute to craniofacial anomalies and NTDs in humans.


Subject(s)
Abnormalities, Multiple , Cleft Lip , Cleft Palate , Neural Tube Defects , Spinal Dysraphism , Animals , Humans , Mice , Abnormalities, Multiple/genetics , Cleft Lip/genetics , Cleft Palate/genetics , Encephalocele/genetics , Mutation , Neural Tube Defects/genetics , Spinal Dysraphism/genetics
5.
AANA J ; 89(3): 200-203, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34042570

ABSTRACT

A survey was conducted of nurse anesthetists about the practice of conscious sedation for head surgery in a private clinical center. Seventy-eight patients were included from June 2017 to June 2018. Age, sex, medical history, ASA classification, previous sedation experience, level of anxiety, premedication use, amount and type of sedative used, surgical duration, sedation level, incidents, and recovery time were evaluated for all patients. The most significant characteristics were found when evaluating preoperative anxiety, preoperative information, and intraoperative respiratory rate. After data collection, 2 groups were distinguished: group 1: procedure completed without incident (n=57) and group 2: procedure completed with incidents (n=21). The patients' average age was 69.5 years. More than 40% of patients were classified as ASA 2. A total of 49 patients (62.3%) felt stressed or anxious preoperatively. More than 65% of all patients (n=51) did not know what conscious sedation was. Intraoperatively, the average sedation level was the same for all patients. Respiratory rates were not recorded for 12 patients (16%). Most of the complications were experienced by patients with a high level of preoperative anxiety and with a misunderstanding of the anesthetic technique.


Subject(s)
Anesthesia , Anesthetics , Conscious Sedation , Aged , Humans , Hypnotics and Sedatives
6.
Front Genet ; 12: 625120, 2021.
Article in English | MEDLINE | ID: mdl-33569080

ABSTRACT

Glycine cleavage system H protein (GCSH) is a component of the glycine cleavage system (GCS), a conserved protein complex that acts to decarboxylate glycine. Mutation of AMT or GLDC, encoding the GCS components aminomethyltransferase and glycine decarboxylase, can cause malformations of the developing CNS (neural tube defects (NTDs) and ventriculomegaly) as well as a post-natal life-limiting neurometabolic disorder, Non-Ketotic Hyperglycinemia. In contrast, it is unclear whether mutation of GCSH contributes to these conditions and we therefore investigated GCSH loss of function in mice. Mice that were heterozygous for a Gcsh null allele were viable and did not exhibit elevated plasma glycine. Moreover, heterozygous mutation of Gcsh did not increase the frequency of NTDs in Gldc mutant embryos. Homozygous Gcsh null mice were not recovered at post-natal stages. Analysis of litters at E8.5-10.5, revealed the presence of homozygous null embryos which were much smaller than littermates and had failed to develop beyond early post-implantation stages with no visible somites or head-folds. Hence, unlike null mutations of Gldc or Amt, which are compatible with embryonic survival despite the presence of NTDs, loss of Gcsh causes embryonic death prior to mid-gestation. Maternal supplementation with formate did not restore embryonic development beyond E7.5, suggesting that the primary cause of lethality was not loss of glycine cleavage activity or suppression of folate one-carbon metabolism. These findings suggest that GCSH has additional roles beyond function in the glycine cleavage system. We hypothesize that GCSH potentially acts in lipoylation of 2-oxoacid dehydrogenase proteins, as reported in bacteria.

7.
J Inherit Metab Dis ; 43(6): 1186-1198, 2020 11.
Article in English | MEDLINE | ID: mdl-32743799

ABSTRACT

Glycine abundance is modulated in a tissue-specific manner by use in biosynthetic reactions, catabolism by the glycine cleavage system (GCS), and excretion via glycine conjugation. Dysregulation of glycine metabolism is associated with multiple disorders including epilepsy, developmental delay, and birth defects. Mutation of the GCS component glycine decarboxylase (GLDC) in non-ketotic hyperglycinemia (NKH) causes accumulation of glycine in body fluids, but there is a gap in our knowledge regarding the effects on glycine metabolism in tissues. Here, we analysed mice carrying mutations in Gldc that result in severe or mild elevations of plasma glycine and model NKH. Liver of Gldc-deficient mice accumulated glycine and numerous glycine derivatives, including multiple acylglycines, indicating increased flux through reactions mediated by enzymes including glycine-N-acyltransferase and arginine: glycine amidinotransferase. Levels of dysregulated metabolites increased with age and were normalised by liver-specific rescue of Gldc expression. Brain tissue exhibited increased abundance of glycine, as well as derivatives including guanidinoacetate, which may itself be epileptogenic. Elevation of brain tissue glycine occurred even in the presence of only mildly elevated plasma glycine in mice carrying a missense allele of Gldc. Treatment with benzoate enhanced hepatic glycine conjugation thereby lowering plasma and tissue glycine. Moreover, administration of a glycine conjugation pathway intermediate, cinnamate, similarly achieved normalisation of liver glycine derivatives and circulating glycine. Although exogenous benzoate and cinnamate impact glycine levels via activity of glycine-N-acyltransferase, that is not expressed in brain, they are sufficient to lower levels of glycine and derivatives in brain tissue of treated Gldc-deficient mice.


Subject(s)
Brain/metabolism , Glycine Dehydrogenase (Decarboxylating)/genetics , Glycine/metabolism , Hyperglycinemia, Nonketotic/enzymology , Alleles , Animals , Brain/pathology , Hyperglycinemia, Nonketotic/pathology , Mice , Mutation, Missense
8.
Dev Cell ; 52(3): 321-334.e6, 2020 02 10.
Article in English | MEDLINE | ID: mdl-32049039

ABSTRACT

Epithelial fusion is a key process of morphogenesis by which tissue connectivity is established between adjacent epithelial sheets. A striking and poorly understood feature of this process is "zippering," whereby a fusion point moves directionally along an organ rudiment. Here, we uncover the molecular mechanism underlying zippering during mouse spinal neural tube closure. Fusion is initiated via local activation of integrin ß1 and focal anchorage of surface ectoderm cells to a shared point of fibronectin-rich basement membrane, where the neural folds first contact each other. Surface ectoderm cells undergo proximal junction shortening, establishing a transitory semi-rosette-like structure at the zippering point that promotes juxtaposition of cells across the midline enabling fusion propagation. Tissue-specific ablation of integrin ß1 abolishes the semi-rosette formation, preventing zippering and causing spina bifida. We propose integrin-mediated anchorage as an evolutionarily conserved mechanism of general relevance for zippering closure of epithelial gaps whose disturbance can produce clinically important birth defects.


Subject(s)
Embryo, Mammalian/physiology , Epithelial Cells/physiology , Focal Adhesions , Integrin beta1/physiology , Neural Crest/embryology , Neural Tube/embryology , Neurulation , Actomyosin/metabolism , Animals , Cell Fusion , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Neural Crest/metabolism , Neural Crest/physiology , Neural Tube/metabolism , Neural Tube/physiology
9.
Birth Defects Res ; 112(2): 205-211, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31758757

ABSTRACT

BACKGROUND: Neural tube defects (NTDs) result from failure of neural tube closure during embryogenesis. These severe birth defects of the central nervous system include anencephaly and spina bifida, and affect 0.5-2 per 1,000 pregnancies worldwide in humans. It has been demonstrated that acetylation plays a pivotal role during neural tube closure, as animal models for defective histone acetyltransferase proteins display NTDs. Acetylation represents an important component of the complex network of posttranslational regulatory interactions, suggesting a possible fundamental role during primary neurulation events. This study aimed to assess protein acetylation contribution to early patterning of the central nervous system both in human and murine specimens. METHODS: We used both human and mouse (Cited2 -/- ) samples to analyze the dynamic acetylation of proteins during embryo development through immunohistochemistry, western blot analysis and quantitative polymerase chain reaction. RESULTS: We report the dynamic profile of histone and protein acetylation status during neural tube closure. We also report a rescue effect in an animal model by chemical p53 inhibition. CONCLUSIONS: Our data suggest that the p53-acetylation equilibrium may play a role in primary neurulation in mammals.


Subject(s)
Neural Tube Defects/embryology , Neurulation/genetics , Acetylation , Anencephaly/etiology , Anencephaly/physiopathology , Animals , Disease Models, Animal , Embryonic Development/genetics , Embryonic Development/physiology , Histone Acetyltransferases/metabolism , Humans , Mammals , Mice/embryology , Neurulation/physiology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Spinal Dysraphism/etiology , Spinal Dysraphism/physiopathology , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
10.
Dis Model Mech ; 12(11)2019 11 22.
Article in English | MEDLINE | ID: mdl-31636139

ABSTRACT

Neural tube defects (NTDs), including spina bifida and anencephaly, are among the most common birth defects worldwide, but their underlying genetic and cellular causes are not well understood. Some NTDs are preventable by supplemental folic acid. However, despite widespread use of folic acid supplements and implementation of food fortification in many countries, the protective mechanism is unclear. Pax3 mutant (splotch; Sp2H ) mice provide a model in which NTDs are preventable by folic acid and exacerbated by maternal folate deficiency. Here, we found that cell proliferation was diminished in the dorsal neuroepithelium of mutant embryos, corresponding to the region of abolished Pax3 function. This was accompanied by premature neuronal differentiation in the prospective midbrain. Contrary to previous reports, we did not find evidence that increased apoptosis could underlie failed neural tube closure in Pax3 mutant embryos, nor that inhibition of apoptosis could prevent NTDs. These findings suggest that Pax3 functions to maintain the neuroepithelium in a proliferative, undifferentiated state, allowing neurulation to proceed. NTDs in Pax3 mutants were not associated with abnormal abundance of specific folates and were not prevented by formate, a one-carbon donor to folate metabolism. Supplemental folic acid restored proliferation in the cranial neuroepithelium. This effect was mediated by enhanced progression of the cell cycle from S to G2 phase, specifically in the Pax3 mutant dorsal neuroepithelium. We propose that the cell-cycle-promoting effect of folic acid compensates for the loss of Pax3 and thereby prevents cranial NTDs.


Subject(s)
Folic Acid/administration & dosage , Mutation , Neural Tube Defects/etiology , PAX3 Transcription Factor/genetics , Animals , Apoptosis , Cell Cycle/drug effects , Dietary Supplements , Disease Models, Animal , Mice , Mice, Inbred C3H , Mice, Inbred CBA , Neural Tube Defects/prevention & control , PAX3 Transcription Factor/physiology
11.
Nat Commun ; 10(1): 2487, 2019 06 06.
Article in English | MEDLINE | ID: mdl-31171776

ABSTRACT

Lack or excess expression of the surface ectoderm-expressed transcription factor Grainyhead-like2 (Grhl2), each prevent spinal neural tube closure. Here we investigate the causative mechanisms and find reciprocal dysregulation of epithelial genes, cell junction components and actomyosin properties in Grhl2 null and over-expressing embryos. Grhl2 null surface ectoderm shows a shift from epithelial to neuroepithelial identity (with ectopic expression of N-cadherin and Sox2), actomyosin disorganisation, cell shape changes and diminished resistance to neural fold recoil upon ablation of the closure point. In contrast, excessive abundance of Grhl2 generates a super-epithelial surface ectoderm, in which up-regulation of cell-cell junction proteins is associated with an actomyosin-dependent increase in local mechanical stress. This is compatible with apposition of the neural folds but not with progression of closure, unless myosin activity is inhibited. Overall, our findings suggest that Grhl2 plays a crucial role in regulating biomechanical properties of the surface ectoderm that are essential for spinal neurulation.


Subject(s)
Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Neural Tube/embryology , Neuroepithelial Cells/metabolism , Neurulation/genetics , Transcription Factors/genetics , Actomyosin/genetics , Actomyosin/metabolism , Animals , Biomechanical Phenomena , Cadherins/metabolism , Ectoderm/cytology , Ectoderm/embryology , Ectoderm/metabolism , Epithelial Cells/metabolism , Intercellular Junctions/genetics , Intercellular Junctions/metabolism , Mice , Neural Tube/metabolism , SOXB1 Transcription Factors/metabolism , Stress, Mechanical , Transcription Factors/metabolism
12.
Hum Mol Genet ; 27(24): 4218-4230, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30189017

ABSTRACT

The genetic basis of human neural tube defects (NTDs), such as anencephaly and spina bifida (SB), is complex and heterogeneous. Grainyhead-like genes represent candidates for involvement in NTDs based on the presence of SB and exencephaly in mice carrying loss-of-function alleles of Grhl2 or Grhl3. We found that reinstatement of Grhl3 expression, by bacterial artificial chromosome (BAC)-mediated transgenesis, prevents SB in Grhl3-null embryos, as in the Grhl3 hypomorphic curly tail strain. Notably, however, further increase in expression of Grhl3 causes highly penetrant SB. Grhl3 overexpression recapitulates the spinal NTD phenotype of loss-of-function embryos, although the underlying mechanism differs. However, it does not phenocopy other defects of Grhl3-null embryos such as abnormal axial curvature, cranial NTDs (exencephaly) or skin barrier defects, the latter being rescued by the Grhl3-transgene. Grhl2 and Grhl3 can form homodimers and heterodimers, suggesting a possible model in which defects arising from overexpression of Grhl3 result from sequestration of Grhl2 in heterodimers, mimicking Grhl2 loss of function. This hypothesis predicts that increased abundance of Grhl2 would have an ameliorating effect in Grhl3 overexpressing embryo. Instead, we observed a striking additive genetic interaction between Grhl2 and Grhl3 gain-of-function alleles. Severe SB arose in embryos in which both genes were expressed at moderately elevated levels that individually do not cause NTDs. Furthermore, moderate Grhl3 overexpression also interacted with the Vangl2Lp allele to cause SB, demonstrating genetic interaction with the planar cell polarity signalling pathway that is implicated in mouse and human NTDs.


Subject(s)
DNA-Binding Proteins/genetics , Nerve Tissue Proteins/genetics , Neural Tube Defects/genetics , Spinal Dysraphism/genetics , Transcription Factors/genetics , Alleles , Animals , Animals, Genetically Modified/genetics , Disease Models, Animal , Embryonic Development/genetics , Gene Expression Regulation, Developmental/genetics , Humans , Loss of Function Mutation , Mice , Neural Tube Defects/pathology , Protein Multimerization/genetics , Spinal Dysraphism/pathology
13.
PLoS Genet ; 14(8): e1007573, 2018 08.
Article in English | MEDLINE | ID: mdl-30067846

ABSTRACT

Drosophila body pigmentation has emerged as a major Evo-Devo model. Using two Drosophila melanogaster lines, Dark and Pale, selected from a natural population, we analyse here the interaction between genetic variation and environmental factors to produce this complex trait. Indeed, pigmentation varies with genotype in natural populations and is sensitive to temperature during development. We demonstrate that the bric à brac (bab) genes, that are differentially expressed between the two lines and whose expression levels vary with temperature, participate in the pigmentation difference between the Dark and Pale lines. The two lines differ in a bab regulatory sequence, the dimorphic element (called here bDE). Both bDE alleles are temperature-sensitive, but the activity of the bDE allele from the Dark line is lower than that of the bDE allele from the Pale line. Our results suggest that this difference could partly be due to differential regulation by AbdB. bab has been previously reported to be a repressor of abdominal pigmentation. We show here that one of its targets in this process is the pigmentation gene tan (t), regulated via the tan abdominal enhancer (t_MSE). Furthermore, t expression is strongly modulated by temperature in the two lines. Thus, temperature sensitivity of t expression is at least partly a consequence of bab thermal transcriptional plasticity. We therefore propose that a gene regulatory network integrating both genetic variation and temperature sensitivity modulates female abdominal pigmentation. Interestingly, both bDE and t_MSE were previously shown to have been recurrently involved in abdominal pigmentation evolution in drosophilids. We propose that the environmental sensitivity of these enhancers has turned them into evolutionary hotspots.


Subject(s)
DNA-Binding Proteins/physiology , Drosophila Proteins/physiology , Drosophila melanogaster/genetics , Gene Regulatory Networks , Pigmentation/genetics , Transcription Factors/physiology , Alleles , Animals , Base Sequence , Binding Sites , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/physiology , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Evolution, Molecular , Female , Gene Expression Regulation , Genetic Variation , Genotyping Techniques , Sequence Analysis, DNA , Temperature , Transcription Factors/genetics
14.
Inflamm Res ; 67(7): 597-608, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29687146

ABSTRACT

OBJECTIVE AND DESIGN: The present work investigates the modulation of experimental autoimmune encephalomyelitis (EAE) using genistein before the EAE induction. MATERIAL: Female C57BL/6 mice (n = 96 mice/experiment), 4-6 weeks old, were used to induce the EAE. The mice were divided into three experimental groups: non-immunized group, immunized group (EAE), and immunized and treated with genistein group (Genistein). TREATMENT: Genistein was used at a dose of 200 mg/kg s.c. and were initiated 2 days before the immunization and continued daily until day 6 postimmunization. METHODS: Animals were monitored daily for clinical signs of EAE up to day 21. Inflammatory infiltration, demyelination, Toll-like receptor (TLR) expression, cytokines and transcription factors were analyzed in spinal cords. RESULTS: The present study demonstrates, for the first time, the genistein ability to modulate the factors involved in the innate immune response in the early stages of EAE. The genistein therapy delayed the onset of the disease, with reduced inflammatory infiltration and demyelination. In addition, the expression of TLR3, TLR9 and IFN-ß were increased in genistein group, with reduction in the factors of TH1 and Th17 cells. CONCLUSION: These findings shed light on the potential of genistein as a prophylactic strategy for multiple sclerosis (MS) prevention.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Genistein/pharmacology , Genistein/therapeutic use , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Toll-Like Receptors/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Female , Macrophages/drug effects , Macrophages/immunology , Mice, Inbred C57BL , Multiple Sclerosis/prevention & control , Myelin Sheath/drug effects , Spinal Cord/drug effects , Spinal Cord/immunology , Spinal Cord/pathology
15.
Dev Biol ; 435(2): 130-137, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29397878

ABSTRACT

Failure of neural tube closure leads to neural tube defects (NTDs), common congenital abnormalities in humans. Among the genes whose loss of function causes NTDs in mice, Grainyhead-like3 (Grhl3) is essential for spinal neural tube closure, with null mutants exhibiting fully penetrant spina bifida. During spinal neurulation Grhl3 is initially expressed in the surface (non-neural) ectoderm, subsequently in the neuroepithelial component of the neural folds and at the node-streak border, and finally in the hindgut endoderm. Here, we show that endoderm-specific knockout of Grhl3 causes late-arising spinal NTDs, preceded by increased ventral curvature of the caudal region which was shown previously to suppress closure of the spinal neural folds. This finding supports the hypothesis that diminished Grhl3 expression in the hindgut is the cause of spinal NTDs in the curly tail, carrying a hypomorphic Grhl3 allele. Complete loss of Grhl3 function produces a more severe phenotype in which closure fails earlier in neurulation, before the stage of onset of expression in the hindgut of wild-type embryos. This implicates additional tissues and NTD mechanisms in Grhl3 null embryos. Conditional knockout of Grhl3 in the neural plate and node-streak border has minimal effect on closure, suggesting that abnormal function of surface ectoderm, where Grhl3 transcripts are first detected, is primarily responsible for early failure of spinal neurulation in Grhl3 null embryos.


Subject(s)
DNA-Binding Proteins/physiology , Neural Tube Defects/genetics , Neural Tube/physiology , Neurulation/genetics , Transcription Factors/physiology , Animals , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Embryonic Development , Gene Expression Regulation, Developmental , Genes, Reporter , Germ Layers/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Neural Plate/metabolism , Neural Tube Defects/embryology , Neural Tube Defects/pathology , Organ Specificity , RNA, Messenger/biosynthesis , Spinal Dysraphism/embryology , Spinal Dysraphism/genetics , Transcription Factors/deficiency , Transcription Factors/genetics
16.
PLoS Genet ; 12(8): e1006218, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27508387

ABSTRACT

Phenotypic plasticity is the ability of a given genotype to produce different phenotypes in response to distinct environmental conditions. Phenotypic plasticity can be adaptive. Furthermore, it is thought to facilitate evolution. Although phenotypic plasticity is a widespread phenomenon, its molecular mechanisms are only beginning to be unravelled. Environmental conditions can affect gene expression through modification of chromatin structure, mainly via histone modifications, nucleosome remodelling or DNA methylation, suggesting that phenotypic plasticity might partly be due to chromatin plasticity. As a model of phenotypic plasticity, we study abdominal pigmentation of Drosophila melanogaster females, which is temperature sensitive. Abdominal pigmentation is indeed darker in females grown at 18°C than at 29°C. This phenomenon is thought to be adaptive as the dark pigmentation produced at lower temperature increases body temperature. We show here that temperature modulates the expression of tan (t), a pigmentation gene involved in melanin production. t is expressed 7 times more at 18°C than at 29°C in female abdominal epidermis. Genetic experiments show that modulation of t expression by temperature is essential for female abdominal pigmentation plasticity. Temperature modulates the activity of an enhancer of t without modifying compaction of its chromatin or level of the active histone mark H3K27ac. By contrast, the active mark H3K4me3 on the t promoter is strongly modulated by temperature. The H3K4 methyl-transferase involved in this process is likely Trithorax, as we show that it regulates t expression and the H3K4me3 level on the t promoter and also participates in female pigmentation and its plasticity. Interestingly, t was previously shown to be involved in inter-individual variation of female abdominal pigmentation in Drosophila melanogaster, and in abdominal pigmentation divergence between Drosophila species. Sensitivity of t expression to environmental conditions might therefore give more substrate for selection, explaining why this gene has frequently been involved in evolution of pigmentation.


Subject(s)
Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Gene-Environment Interaction , Selection, Genetic/genetics , Animals , Chromatin/genetics , Drosophila Proteins/biosynthesis , Drosophila melanogaster/physiology , Female , Gene Expression Regulation , Genotype , Histone-Lysine N-Methyltransferase/genetics , Melanins/biosynthesis , Phenotype , Pigmentation/genetics , Promoter Regions, Genetic , Temperature
17.
Elife ; 5: e13273, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27114066

ABSTRACT

Epithelial fusion is a crucial process in embryonic development, and its failure underlies several clinically important birth defects. For example, failure of neural fold fusion during neurulation leads to open neural tube defects including spina bifida. Using mouse embryos, we show that cell protrusions emanating from the apposed neural fold tips, at the interface between the neuroepithelium and the surface ectoderm, are required for completion of neural tube closure. By genetically ablating the cytoskeletal regulators Rac1 or Cdc42 in the dorsal neuroepithelium, or in the surface ectoderm, we show that these protrusions originate from surface ectodermal cells and that Rac1 is necessary for the formation of membrane ruffles which typify late closure stages, whereas Cdc42 is required for the predominance of filopodia in early neurulation. This study provides evidence for the essential role and molecular regulation of membrane protrusions prior to fusion of a key organ primordium in mammalian development.


Subject(s)
Cell Surface Extensions/metabolism , Ectoderm/cytology , Ectoderm/enzymology , Neural Crest/embryology , Neural Tube/embryology , Neuropeptides/metabolism , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Mice , Neurulation
18.
Biochimie ; 126: 63-70, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26924399

ABSTRACT

The curly tail mouse provides a model for neural tube defects (spina bifida and exencephaly) that are resistant to prevention by folic acid. The major ct gene, responsible for spina bifida, corresponds to a hypomorphic allele of grainyhead-like 3 (Grhl3) but the frequency of NTDs is strongly influenced by modifiers in the genetic background. Moreover, exencephaly in the curly tail strain is not prevented by reinstatement of Grhl3 expression. In the current study we found that expression of Mthfd1L, encoding a key component of mitochondrial folate one-carbon metabolism (FOCM), is significantly reduced in ct/ct embryos compared to a partially congenic wild-type strain. This expression change is not attributable to regulation by Grhl3 or the genetic background at the Mthfd1L locus. Mitochondrial FOCM provides one-carbon units as formate for FOCM reactions in the cytosol. We found that maternal supplementation with formate prevented NTDs in curly tail embryos and also resulted in increased litter size. Analysis of the folate profile of neurulation-stage embryos showed that formate supplementation resulted in an increased proportion of formyl-THF and THF but a reduction in proportion of 5-methyl THF. In contrast, THF decreased and 5-methyl THF was relatively more abundant in the liver of supplemented dams than in controls. In embryos cultured through the period of spinal neurulation, incorporation of labelled thymidine and adenine into genomic DNA was suppressed by supplemental formate, suggesting that de novo folate-dependent biosynthesis of nucleotides (thymidylate and purines) was enhanced. We hypothesise that reduced Mthfd1L expression may contribute to susceptibility to NTDs in the curly tail strain and that formate acts as a one-carbon donor to prevent NTDs.


Subject(s)
Folic Acid/metabolism , Formates/pharmacology , Nucleotides/biosynthesis , Spinal Dysraphism , Animals , Disease Models, Animal , Mice , Spinal Dysraphism/metabolism , Spinal Dysraphism/prevention & control
19.
J Immunol Res ; 2015: 931574, 2015.
Article in English | MEDLINE | ID: mdl-25759850

ABSTRACT

Many immune-based intestinal disorders, such as ulcerative colitis and Crohn's disease, as well as other illnesses, may have the intestines as an initial cause or aggravator in the development of diseases, even apparently not correlating directly to the intestine. Diabetes, obesity, multiple sclerosis, depression, and anxiety are examples of other illnesses discussed in the literature. In parallel, importance of the gut microbiota in intestinal homeostasis and immunologic conflict between tolerance towards commensal microorganisms and combat of pathogens is well known. Recent researches show that the immune system, when altered by the gut microbiota, influences the state in which these diseases are presented in the patient directly and indirectly. At the present moment, a considerable number of investigations about this subject have been performed and published. However, due to difficulties on correlating information, several speculations and hypotheses are generated. Thus, the present review aims at bringing together how these interactions work-gut microbiota, immune system, and their influence in the neuroimmune system.


Subject(s)
Gastrointestinal Microbiome/immunology , Immune System , Nervous System , Neuroimmunomodulation , Animals , Autoimmune Diseases/etiology , Autoimmune Diseases/metabolism , Disease Models, Animal , Gastrointestinal Tract/immunology , Gastrointestinal Tract/microbiology , Homeostasis/physiology , Humans , Signal Transduction
20.
Cell Immunol ; 293(2): 87-94, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25585346

ABSTRACT

Multiple sclerosis (MS) shows distinct clinical courses. Experimental autoimmune encephalomyelitis (EAE), a model to study multiple sclerosis, can be induced by different protocols, which show distinct cytokine and antibody production. The factors involved in this heterogeneity remain unclear. The relevance of MOG concentration in triggering a regulatory response in the chronic model of EAE is imprecise. The aim of this study was investigate if 100 or 300 µg of MOG(35-55) could induce different EAE profiles. Modifications in the concentration of MOG were able to change the patterns of chemokines, cytokines, percentage of cells, inflammatory infiltrate and the development of a regulatory response. However, these changes were unable to modify the intensity of response, which explains the chronic progression of the disease in both concentrations. The results presented in this study contribute to understanding the intricate mechanisms that trigger EAE and provide insights into the pathogenesis of various forms of MS.


Subject(s)
Brain/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Inflammation/immunology , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Spinal Cord/immunology , Animals , Brain/pathology , Chemokines/analysis , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Flow Cytometry , Mice , Mice, Inbred C57BL , Spinal Cord/pathology , Statistics, Nonparametric
SELECTION OF CITATIONS
SEARCH DETAIL
...