Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
FASEB J ; 38(14): e23815, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-38989587

ABSTRACT

To investigate how the fatty acid composition of brain phospholipids influences brain-specific processes, we leveraged the AdipoR2 (adiponectin receptor 2) knockout mouse model in which the brain is enlarged, and cellular membranes are excessively rich in saturated fatty acids. Lipidomics analysis of brains at 2, 7, and 18 months of age showed that phosphatidylcholines, which make up about two-thirds of all cerebrum membrane lipids, contain a gross excess of saturated fatty acids in AdipoR2 knockout mice, and that this is mostly attributed to an excess palmitic acid (C16:0) at the expense of oleic acid (C18:1), consistent with a defect in fatty acid desaturation and elongation in the mutant. Specifically, there was a ~12% increase in the overall saturated fatty acid content within phosphatidylcholines and a ~30% increase in phosphatidylcholines containing two palmitic acids. Phosphatidylethanolamines, sphingomyelins, ceramides, lactosylceramides, and dihydroceramides also showed an excess of saturated fatty acids in the AdipoR2 knockout mice while nervonic acid (C24:1) was enriched at the expense of shorter saturated fatty acids in glyceroceramides. Similar defects were found in the cerebellum and myelin sheaths. Histology showed that cell density is lower in the cerebrum of AdipoR2 knockout mice, but electron microscopy did not detect reproducible defects in the ultrastructure of cerebrum neurons, though proteomics analysis showed an enrichment of electron transport chain proteins in the cerebellum. Behavioral tests showed that older (33 weeks old) AdipoR2 knockout mice are hyperactive and anxious compared to control mice of a similar age. Also, in contrast to control mice, the AdipoR2 knockout mice do not gain weight in old age but do have normal lifespans. We conclude that an excess fatty acid saturation in brain phospholipids is accompanied by hyperactivity but seems otherwise well tolerated.


Subject(s)
Aging , Brain , Fatty Acids , Mice, Knockout , Receptors, Adiponectin , Animals , Mice , Brain/metabolism , Fatty Acids/metabolism , Aging/metabolism , Receptors, Adiponectin/metabolism , Receptors, Adiponectin/genetics , Male , Mice, Inbred C57BL , Phosphatidylcholines/metabolism , Phospholipids/metabolism
2.
Hum Mol Genet ; 28(8): 1286-1297, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30561639

ABSTRACT

Molecular mechanisms governing the development of the human cochlea remain largely unknown. Through genome sequencing, we identified a homozygous FOXF2 variant c.325A>T (p.I109F) in a child with profound sensorineural hearing loss (SNHL) associated with incomplete partition type I anomaly of the cochlea. This variant is not found in public databases or in over 1000 ethnicity-matched control individuals. I109 is a highly conserved residue in the forkhead box (Fox) domain of FOXF2, a member of the Fox protein family of transcription factors that regulate the expression of genes involved in embryogenic development as well as adult life. Our in vitro studies show that the half-life of mutant FOXF2 is reduced compared to that of wild type. Foxf2 is expressed in the cochlea of developing and adult mice. The mouse knockout of Foxf2 shows shortened and malformed cochleae, in addition to altered shape of hair cells with innervation and planar cell polarity defects. Expressions of Eya1 and Pax3, genes essential for cochlear development, are reduced in the cochleae of Foxf2 knockout mice. We conclude that FOXF2 plays a major role in cochlear development and its dysfunction leads to SNHL and developmental anomalies of the cochlea in humans and mice.


Subject(s)
Cochlea/embryology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/physiology , Adult , Animals , Child , Cochlea/metabolism , Cochlea/physiology , Embryonic Development , Female , Hair Cells, Auditory/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Organogenesis , PAX3 Transcription Factor/genetics , PAX3 Transcription Factor/physiology , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/physiology , Signal Transduction/genetics , Whole Genome Sequencing
3.
Biotechniques ; 55(1): 42-4, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23834385

ABSTRACT

Current protocols for separating adult intestinal epithelial cells from the underlying muscular and mesenchymal tissues typically involve extended incubations, harsh mechanical treatment, and exposure to either proteases or chelating agents. The drawbacks of these approaches include fragmentation, contamination with other cell types, reduced viability, and under-representation of crypt cells. Here we describe a gentle procedure that allows harvesting of pure, fully viable sheets of murine intestinal epithelium, with intact crypts and villi, without enzymes or EDTA. The mesenchyme retains intact villus core projections, is virtually free from epithelial cells, and can be cultured in vitro.


Subject(s)
Histological Techniques/instrumentation , Histological Techniques/methods , Intestinal Mucosa/anatomy & histology , Intestinal Mucosa/physiology , Mesoderm/anatomy & histology , Mesoderm/physiology , Animals , Mice , Mice, Transgenic
4.
Biochem Biophys Res Commun ; 437(2): 274-9, 2013 Jul 26.
Article in English | MEDLINE | ID: mdl-23811271

ABSTRACT

The very low density lipoprotein receptor (VLDLr) is highly upregulated during hypoxia in mouse cardiomyocytes and in human and mouse ischemic hearts causing a detrimental lipid accumulation. To know how the gene is regulated is important for future studies. In this study, we have thoroughly mapped the 5'-flanking region of the mouse VLDLr promoter and show that the hypoxia-mediated increase in VLDLr expression is dependent on Hif-1α binding to a hypoxia responsive element (HRE) located at -162 to -158bp 5'of translation start. We show that classical HRE sites and the previously described PPARγ and Sp1 binding are not involved in the hypoxia-induced regulation of the VLDLr promoter. Using a chromatin immunoprecipitation (ChIP) assay, we show that Hif-1α specifically binds and activates the mouse VLDLr promoter at the previously described non-classical HRE in HL-1 cells. We also show that the same HRE is present and active in response to hypoxia in human cardiomyocytes, however at a different location (-812bp from translation start). These results conclude that in the hypoxic hearts of mice and men, the VLDLr gene is regulated by a direct binding of Hif-1α to the VLDLr promoter.


Subject(s)
Hypoxia/physiopathology , Receptors, LDL/physiology , Animals , Binding Sites , Cell Line , DNA Primers , Mice , PPAR gamma/metabolism , Promoter Regions, Genetic , Receptors, LDL/genetics , Sp1 Transcription Factor/metabolism
5.
Am J Med Genet A ; 161A(4): 764-70, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23444129

ABSTRACT

Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is a congenital malformation that leads to severe pulmonary hypertension and respiratory failure. It has been associated with deletion of, or mutation in, FOXF1 on 16q24.1, a gene encoding a forkhead transcription factor expressed in the mesenchyme of the developing lung. Here we report on the identification of a pericentric inversion on chromosome 16 (p11.2q24.1) in a case of lethal ACDMPV with atrioventricular septal defect and duodenal atresia. Array-CGH indicated that the inversion is balanced, and FISH showed that the q-arm breakpoint occurs 134 ± 10 kb upstream (5'; centromeric) of FOXF1. This is suggestive of cis-regulatory elements located more than 130 kb 5' of FOXF1, and analysis of genome-wide data sets of chromatin modifications in two different cell types suggested that the FOXF1 regulatory domain covers more than 300 kb, and perhaps up to 433 kb, upstream of the gene, but only 3 kb downstream. The 588 kb gene-free region between FOXF1 and the next gene in the centromeric direction, IRF8, is highly conserved between species and divided into two distinct regulatory domains by an insulator element. Another putative insulator occurs just downstream of FOXF1. Our results further strengthen the association between FOXF1 and a spectrum of malformations that include ACDMPV, atrioventricular septal defects, and gastrointestinal atresia. Furthermore, the presented analysis aids in defining the critical genomic region for this syndrome.


Subject(s)
Chromosome Inversion , Forkhead Transcription Factors/genetics , Persistent Fetal Circulation Syndrome/genetics , Persistent Fetal Circulation Syndrome/pathology , Autopsy , Chromosomes, Human, Pair 16 , Comparative Genomic Hybridization , Fatal Outcome , Female , Humans , In Situ Hybridization, Fluorescence , Infant, Newborn , Interferon Regulatory Factors/genetics
6.
Gastroenterology ; 144(5): 1001-11, 2013 May.
Article in English | MEDLINE | ID: mdl-23376422

ABSTRACT

BACKGROUND & AIMS: The stem cell niche at the base of the intestinal crypts, as well as stemness and high clonogenicity in colon cancer cells, depend on Wnt signaling to ß-catenin. Fibroblasts modulate the Wnt pathway in normal and neoplastic epithelial cells via unclear mechanisms. We investigated how in intestinal fibroblasts the forkhead transcription factor Foxf2 controls Wnt signaling to affect numbers of stem cells and formation and growth of adenomas in mice. METHODS: We created mice with different copy numbers of Foxf2 by generating Foxf2(-/+) mice and a transgenic strain, Tg(FOXF2). Adenoma formation was investigated in Apc(Min/+) mice, stem cells were counted in mice with the Lgr5-enhanced green fluorescent protein knock-in allele, proliferation was measured by incorporation of bromodeoxyuridine, Foxf2 and Sfrp1 were localized by immunohistochemistry, and signaling pathways were analyzed by quantitative polymerase chain reaction and immunoblot assays. RESULTS: Epithelial ß-catenin was stabilized in Foxf2(-/+) mice, resulting in increased number and size of adenomas. Tg(FOXF2) mice, however, were partially resistant to intestinal neoplasia and developed fewer and smaller adenomas; Foxf2(-/+) mice developed 24-fold more tumors than Tg(FOXF2) mice. Epithelial cells of Foxf2(-/+) mice also had higher numbers of Lgr5(+) stem cells and greater amounts of crypt cell proliferation and expression of Myc (a target of Wnt signaling) than Tg(FOXF2) mice. Expression of Sfrp1, which encodes an extracellular inhibitor of Wnt, in fibroblasts increased with copy number of Foxf2. CONCLUSIONS: Foxf2 is a fibroblast factor that inhibits paracrine Wnt signaling and restricts the crypt stem cell niche in intestines of mice. Loss of Foxf2 promotes adenoma formation and growth.


Subject(s)
Adenoma/genetics , Colonic Neoplasms/genetics , Fibroblasts/metabolism , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , RNA, Neoplasm/genetics , Stem Cells/pathology , Adenoma/metabolism , Adenoma/pathology , Alleles , Animals , Cell Count , Cell Proliferation , Colon/metabolism , Colon/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Fibroblasts/pathology , Forkhead Transcription Factors/biosynthesis , Immunoblotting , Immunohistochemistry , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Neoplasms, Experimental , Polymerase Chain Reaction , Stem Cells/metabolism , Tumor Cells, Cultured , Wnt Proteins/metabolism , Wnt Signaling Pathway/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...