Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
FASEB J ; 38(3): e23460, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38315443

ABSTRACT

Traumatic brain injury (TBI) is one of the leading causes of death worldwide. There are currently no effective treatments for TBI, and trauma survivors suffer from a variety of long-lasting health consequences. With nutritional support recently emerging as a vital step in improving TBI patients' outcomes, we sought to evaluate the potential therapeutic benefits of nutritional supplements derived from bovine thymus gland, which can deliver a variety of nutrients and bioactive molecules. In a rat model of controlled cortical impact (CCI), we determined that animals supplemented with a nuclear fraction of bovine thymus (TNF) display greatly improved performance on beam balance and spatial memory tests following CCI. Using RNA-Seq, we identified an array of signaling pathways that are modulated by TNF supplementation in rat hippocampus, including those involved in the process of autophagy. We further show that bovine thymus-derived extracts contain antigens found in neural tissues and that supplementation of rats with thymus extracts induces production of serum IgG antibodies against neuronal and glial antigens, which may explain the enhanced animal recovery following CCI through possible oral tolerance mechanism. Collectively, our data demonstrate, for the first time, the potency of a nutritional supplement containing nuclear fraction of bovine thymus in enhancing the functional recovery from TBI.


Subject(s)
Brain Injuries, Traumatic , Thymus Extracts , Humans , Rats , Animals , Cattle , Thymus Extracts/pharmacology , Thymus Extracts/therapeutic use , Brain Injuries, Traumatic/drug therapy , Neurons , Neuroglia , Hippocampus , Disease Models, Animal
2.
PLoS One ; 15(7): e0223395, 2020.
Article in English | MEDLINE | ID: mdl-32645024

ABSTRACT

Development of the cerebral cortex may be influenced by the composition of the maternal gut microbiota. To test this possibility, we administered probiotic Lactococcus lactis in drinking water to mouse dams from day 10.5 of gestation until pups reached postnatal day 1 (P1). Pups were assessed in a battery of behavioral tests starting at 10 weeks old. We found that females, but not males, exposed to probiotic during prenatal development spent more time in the center of the open field and displayed decreased freezing time in cue associated learning, compared to controls. Furthermore, we found that probiotic exposure changed the density of cortical neurons and increased the density of blood vessels in the cortical plate of P1 pups. Sex-specific differences were observed in the number of mitotic neural progenitor cells, which were increased in probiotic exposed female pups. In addition, we found that probiotic treatment in the latter half of pregnancy significantly increased plasma oxytocin levels in mouse dams, but not in the offspring. These results suggest that exposure of naïve, unstressed dams to probiotic may exert sex-specific long-term effects on cortical development and anxiety related behavior in the offspring.


Subject(s)
Anxiety/prevention & control , Cerebral Cortex/drug effects , Lactococcus lactis , Prenatal Exposure Delayed Effects/psychology , Probiotics/pharmacology , Animals , Animals, Newborn , Cell Count , Cerebral Cortex/blood supply , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Fear , Female , Learning , Male , Mice , Mice, Inbred C57BL , Neurons/cytology , Oxytocin/metabolism , Pregnancy , Sex Characteristics
3.
FASEB J ; 33(8): 9194-9209, 2019 08.
Article in English | MEDLINE | ID: mdl-31091977

ABSTRACT

Adequate supply of choline, an essential nutrient, is necessary to support proper brain development. Whether prenatal choline availability plays a role in development of the visual system is currently unknown. In this study, we addressed the role of in utero choline supply for the development and later function of the retina in a mouse model. We lowered choline availability in the maternal diet during pregnancy and assessed proliferative and differentiation properties of retinal progenitor cells (RPCs) in the developing prenatal retina, as well as visual function in adult offspring. We report that low choline availability during retinogenesis leads to persistent retinal cytoarchitectural defects, ranging from focal lesions with displacement of retinal neurons into subretinal space to severe hypocellularity and ultrastructural defects in photoreceptor organization. We further show that low choline availability impairs timely differentiation of retinal neuronal cells, such that the densities of early-born retinal ganglion cells, amacrine and horizontal cells, as well as cone photoreceptor precursors, are reduced in low choline embryonic d 17.5 retinas. Maintenance of higher proportions of RPCs that fail to exit the cell cycle underlies aberrant neuronal differentiation in low choline embryos. Increased RPC cell cycle length, and associated reduction in neurofibromin 2/Merlin protein, an upstream regulator of the Hippo signaling pathway, at least in part, explain aberrant neurogenesis in low choline retinas. Furthermore, we find that animals exposed to low choline diet in utero exhibit a significant degree of intraindividual variation in vision, characterized by marked functional discrepancy between the 2 eyes in individual animals. Together, our findings demonstrate, for the first time, that choline availability plays an essential role in the regulation of temporal progression of retinogenesis and provide evidence for the importance of adequate supply of choline for proper development of the visual system.-Trujillo-Gonzalez, I., Friday, W. B., Munson, C. A., Bachleda, A., Weiss, E. R., Alam, N. M., Sha, W., Zeisel, S. H., Surzenko, N. Low availability of choline in utero disrupts development and function of the retina.


Subject(s)
Choline Deficiency/embryology , Retina/abnormalities , Animals , Cell Cycle/physiology , Cell Differentiation/physiology , Choline/administration & dosage , Choline/metabolism , Choline Deficiency/physiopathology , Diet , Down-Regulation , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurofibromin 2/genetics , Neurofibromin 2/metabolism , Neurogenesis/physiology , Pregnancy , Retina/embryology , Retina/physiopathology , Retinal Cone Photoreceptor Cells/ultrastructure , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/physiology , Stem Cells/cytology , Stem Cells/physiology
4.
FASEB J ; 33(3): 3601-3612, 2019 03.
Article in English | MEDLINE | ID: mdl-30521373

ABSTRACT

Choline availability modulates neurogenesis and cerebral cortex development through the regulation of neural progenitor cell (NPC) proliferative and differentiation capacity. In this study, we demonstrated that cortical NPC self-renewal is controlled by choline via the expression of a microRNA (miR-129-5p), whose role in the developing brain has not been examined, and which, in turn, inhibits synthesis of the epidermal growth factor receptor (EGFR) protein. Specifically, we found that low choline (LC) availability led to the upregulation of miR-129-5p expression in cortical NPCs in vitro and in vivo, causing the downregulation of EGFR and thereby disrupting NPC self-renewal and cortical neurogenesis. Furthermore, in response to LC availability, methylation potential (the S-adenosylmethionine: S-adenosylhomocysteine ratio) in the developing brain was reduced. Restoring methylation potential in LC cortical NPCs led to the re-establishment of normal miR-129-5p expression. We concluded that inhibiting miR-129-5p function and restoring EGFR protein levels in vivo is sufficient to reverse LC-induced defects in cortical NPC self-renewal. For the first time, to our knowledge, we have identified the molecular links that explain how a change in the availability of the diet metabolite choline impacts the essential cellular processes underlying brain development.-Trujillo-Gonzalez, I., Wang, Y., Friday, W. B., Vickers, K. C., Toth, C. L., Molina-Torres, L., Surzenko, N., Zeisel, S. H. MicroRNA-129-5p is regulated by choline availability and controls EGF receptor synthesis and neurogenesis in the cerebral cortex.


Subject(s)
Cerebral Cortex/physiology , Choline/genetics , ErbB Receptors/genetics , MicroRNAs/genetics , Neurogenesis/genetics , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Down-Regulation/genetics , Mice , Mice, Inbred C57BL , Stem Cells/physiology , Up-Regulation/genetics
5.
FASEB J ; 30(4): 1566-78, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26700730

ABSTRACT

Maternal diets low in choline, an essential nutrient, increase the risk of neural tube defects and lead to low performance on cognitive tests in children. However, the consequences of maternal dietary choline deficiency for the development and structural organization of the cerebral cortex remain unknown. In this study, we fed mouse dams either control (CT) or low-choline (LC) diets and investigated the effects of choline on cortical development in the offspring. As a result of a low choline supply between embryonic day (E)11 and E17 of gestation, the number of 2 types of cortical neural progenitor cells (NPCs)-radial glial cells and intermediate progenitor cells-was reduced in fetal brains (P< 0.01). Furthermore, the number of upper layer cortical neurons was decreased in the offspring of dams fed an LC diet at both E17 (P< 0.001) and 4 mo of age (P< 0.001). These effects of LC maternal diet were mediated by a decrease in epidermal growth factor receptor (EGFR) signaling in NPCs related to the disruption of EGFR posttranscriptional regulation. Our findings describe a novel mechanism whereby low maternal dietary intake of choline alters brain development.-Wang, Y., Surzenko, N., Friday, W. B., Zeisel, S. H. Maternal dietary intake of choline in mice regulates development of the cerebral cortex in the offspring.


Subject(s)
Cerebral Cortex/drug effects , Choline/pharmacology , Fetal Development/drug effects , Maternal Nutritional Physiological Phenomena , Animals , Blotting, Western , Cell Count , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Choline/administration & dosage , Choline Deficiency/physiopathology , Diet , ErbB Receptors/genetics , ErbB Receptors/metabolism , Female , Fetal Development/physiology , Immunohistochemistry , Mice, Inbred C57BL , Mice, Transgenic , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Pregnancy , Pregnancy Complications/physiopathology , Reverse Transcriptase Polymerase Chain Reaction
6.
Dev Cell ; 26(1): 59-72, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23867227

ABSTRACT

Cone photoreceptors carry out phototransduction in daylight conditions and provide the critical first step in color vision. Despite their importance, little is known about the developmental mechanisms involved in their generation, particularly how they are determined relative to rod photoreceptors, the cells that initiate vision in dim light. Here, we report the identification of a cis-regulatory module (CRM) for the thyroid hormone receptor beta (Thrb) gene, an early cone marker. We found that ThrbCRM1 is active in progenitor cells biased to the production of cones and an interneuronal cell type, the horizontal cell (HC). Molecular analysis of ThrbCRM1 revealed that it is combinatorially regulated by the Otx2 and Onecut1 transcription factors. Onecut1 is sufficient to induce cells with the earliest markers of cones and HCs. Conversely, interference with Onecut1 transcriptional activity leads to precocious rod development, suggesting that Onecut1 is critically important in defining cone versus rod fates.


Subject(s)
Hepatocyte Nuclear Factor 6/metabolism , Otx Transcription Factors/metabolism , Retina/cytology , Retinal Cone Photoreceptor Cells/cytology , Retinal Rod Photoreceptor Cells/cytology , Animals , Cell Lineage , Chick Embryo , Chickens/metabolism , Electroporation/methods , Gene Expression Regulation, Developmental , Hepatocyte Nuclear Factor 6/genetics , Mice , Mice, Knockout , Otx Transcription Factors/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Regulatory Elements, Transcriptional , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Stem Cells/metabolism , Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Receptors beta/metabolism , Transcription, Genetic
7.
Development ; 140(7): 1445-56, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23462474

ABSTRACT

Within discrete regions of the developing mammalian central nervous system, small subsets of glia become specialized to function as neural stem cells. As a result of their self-renewal and neurogenic capacity, these cells later serve to replenish neurons and glia during persistent or injury-induced adult neurogenesis. SOX2, an HMG box transcription factor, plays an essential role in the maintenance of both embryonic and adult neural progenitors. It is unclear, however, which biological mechanisms regulated by SOX2 are required for neural stem cell maintenance. In this study, we address this question through genetic analysis of SOX2 function in differentiating postnatal Müller glia, a cell type that maintains neurogenic capacity in the adult retina. By utilizing molecular analysis and real-time imaging, we show that two progenitor characteristics of nascent Müller glia - their radial morphology and cell cycle quiescence - are disrupted following conditional genetic ablation of Sox2 in the mouse postnatal retina, leading to Müller cell depletion and retinal degeneration. Moreover, we demonstrate that genetic induction of the Notch signaling pathway restores Müller glial cell identity to Sox2 mutant cells, but does not secure their quiescent state. Collectively, these results uncouple the roles of SOX2 and the Notch signaling pathway in the postnatal retina, and uncover a novel role for SOX2 in preventing the depletion of postnatal Müller glia through terminal cell division.


Subject(s)
Neuroglia/physiology , Retina/cytology , SOXB1 Transcription Factors/physiology , Stem Cells/physiology , Animals , Animals, Newborn , Cell Cycle Checkpoints/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/genetics , Neurogenesis/physiology , Neuroglia/cytology , Neuroglia/metabolism , Retina/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Stem Cells/metabolism
8.
Proc Natl Acad Sci U S A ; 109(20): 7882-7, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22543161

ABSTRACT

Previous lineage analyses have shown that retinal progenitor cells (RPCs) are multipotent throughout development, and expression-profiling studies have shown a great deal of molecular heterogeneity among RPCs. To determine if the molecular heterogeneity predicts that an RPC will produce particular types of progeny, clonal lineage analysis was used to investigate the progeny of a subset of RPCs, those that express the basic helix-loop-helix transcription factor, Olig2. The embryonic Olig2(+) RPCs underwent terminal divisions, producing small clones with primarily two of the five cell types being made by the pool of RPCs at that time. The later, postnatal Olig2(+) RPCs also made terminal divisions, which were biased toward production of rod photoreceptors and amacrine cell interneurons. These data indicate that the multipotent progenitor pool is made up of distinctive types of RPCs, which have biases toward producing subsets of retinal neurons in a terminal division, with the types of neurons produced varying over time. This strategy is similar to that of the developing Drosophila melanogaster ventral nerve cord, with the Olig2(+) cells behaving as ganglion mother cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Cell Lineage/physiology , Nerve Tissue Proteins/metabolism , Retina/cytology , Stem Cells/metabolism , Animals , Electroporation , Immunohistochemistry , In Situ Hybridization , Mice , Oligodendrocyte Transcription Factor 2
SELECTION OF CITATIONS
SEARCH DETAIL