Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
iScience ; 27(1): 108670, 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38155767

ABSTRACT

Dysregulated cholesterol metabolism has been linked to neurodegeneration. We previously found that free, non-esterified, 7α,(25R)26-dihydroxycholesterol (7α,26-diHC), was significantly elevated in the cerebrospinal fluid of patients with Parkinson's disease (PD). In this study we investigated the role of 7α,26-diHC in midbrain dopamine (mDA) neuron development and survival. We report that 7α,26-diHC induces apoptosis and reduces the number of mDA neurons in hESC-derived cultures and in mouse progenitor cultures. Voriconazole, an oxysterol 7α-hydroxylase (CYP7B1) inhibitor, increases the number of mDA neurons and prevents the loss of mDA neurons induced by 7α,26-diHC. These effects are specific since neither 7α,26-diHC nor voriconazole alter the number of Islet1+ oculomotor neurons. Furthermore, our results suggest that elevated 24(S),25-epoxycholesterol, which has been shown to promote mDA neurogenesis, may be partially responsible for the effect of voriconazole on mDA neurons. These findings suggest that voriconazole, and/or other azole CYP7B1 inhibitors may have implications in PD therapy development.

2.
Front Aging Neurosci ; 13: 685594, 2021.
Article in English | MEDLINE | ID: mdl-34526889

ABSTRACT

Disordered cholesterol metabolism is linked to neurodegeneration. In this study we investigated the profile of cholesterol metabolites found in the cerebrospinal fluid (CSF) of Parkinson's disease (PD) patients. When adjustments were made for confounding variables of age and sex, 7α,(25R)26-dihydroxycholesterol and a second oxysterol 7α,x,y-trihydroxycholest-4-en-3-one (7α,x,y-triHCO), whose exact structure is unknown, were found to be significantly elevated in PD CSF. The likely location of the additional hydroxy groups on the second oxysterol are on the sterol side-chain. We found that CSF 7α-hydroxycholesterol levels correlated positively with depression in PD patients, while two presumptively identified cholestenoic acids correlated negatively with depression.

3.
Biomolecules ; 9(4)2019 04 13.
Article in English | MEDLINE | ID: mdl-31013940

ABSTRACT

Deficiency in cytochrome P450 (CYP) 7B1, also known as oxysterol 7α-hydroxylase, in humans leads to hereditary spastic paraplegia type 5 (SPG5) and in some cases in infants to liver disease. SPG5 is medically characterized by loss of motor neurons in the corticospinal tract. In an effort to gain a better understanding of the fundamental biochemistry of this disorder, we have extended our previous profiling of the oxysterol content of brain and plasma of Cyp7b1 knockout (-/-) mice to include, amongst other sterols, 25-hydroxylated cholesterol metabolites. Although brain cholesterol levels do not differ between wild-type (wt) and knockout mice, we find, using a charge-tagging methodology in combination with liquid chromatography-mass spectrometry (LC-MS) and multistage fragmentation (MSn), that there is a build-up of the CYP7B1 substrate 25-hydroxycholesterol (25-HC) in Cyp7b1-/- mouse brain and plasma. As reported earlier, levels of (25R)26-hydroxycholesterol (26-HC), 3ß-hydroxycholest-5-en-(25R)26-oic acid and 24S,25-epoxycholesterol (24S,25-EC) are similarly elevated in brain and plasma. Side-chain oxysterols including 25-HC, 26-HC and 24S,25-EC are known to bind to INSIG (insulin-induced gene) and inhibit the processing of SREBP-2 (sterol regulatory element-binding protein-2) to its active form as a master regulator of cholesterol biosynthesis. We suggest the concentration of cholesterol in brain of the Cyp7b1-/- mouse is maintained by balancing reduced metabolism, as a consequence of a loss in CYP7B1, with reduced biosynthesis. The Cyp7b1-/- mouse does not show a motor defect; whether the defect in humans is a consequence of less efficient homeostasis of cholesterol in brain has yet to be uncovered.


Subject(s)
Brain/metabolism , Cytochrome P450 Family 7/genetics , Hydroxycholesterols/metabolism , Spastic Paraplegia, Hereditary/metabolism , Steroid Hydroxylases/genetics , Animals , Cytochrome P450 Family 7/deficiency , Hydroxycholesterols/blood , Male , Mice , Spastic Paraplegia, Hereditary/blood , Spastic Paraplegia, Hereditary/genetics , Steroid Hydroxylases/deficiency
4.
J Biol Chem ; 294(11): 4169-4176, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30655290

ABSTRACT

The liver X receptors Lxrα/NR1H3 and Lxrß/NR1H2 are ligand-dependent nuclear receptors critical for midbrain dopaminergic (mDA) neuron development. We found previously that 24(S),25-epoxycholesterol (24,25-EC), the most potent and abundant Lxr ligand in the developing mouse midbrain, promotes mDA neurogenesis in vitro In this study, we demonstrate that 24,25-EC promotes mDA neurogenesis in an Lxr-dependent manner in the developing mouse midbrain in vivo and also prevents toxicity induced by the Lxr inhibitor geranylgeranyl pyrophosphate. Furthermore, using MS, we show that overexpression of human cholesterol 24S-hydroxylase (CYP46A1) increases the levels of both 24(S)-hydroxycholesterol (24-HC) and 24,25-EC in the developing midbrain, resulting in a specific increase in mDA neurogenesis in vitro and in vivo, but has no effect on oculomotor or red nucleus neurogenesis. 24-HC, unlike 24,25-EC, did not affect in vitro neurogenesis, indicating that the neurogenic effect of 24,25-EC on mDA neurons is specific. Combined, our results indicate that increased levels of 24,25-EC in vivo, by intracerebroventricular delivery in WT mice or by overexpression of its biosynthetic enzyme CYP46A1, specifically promote mDA neurogenesis. We propose that increasing the levels of 24,25-EC in vivo may be a useful strategy to combat the loss of mDA neurons in Parkinson's disease.


Subject(s)
Cholesterol 24-Hydroxylase/biosynthesis , Cholesterol/analogs & derivatives , Dopamine/metabolism , Mesencephalon/metabolism , Neurogenesis , Animals , Cells, Cultured , Cholesterol/biosynthesis , Female , Humans , Mice , Mice, Transgenic
5.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(2): 191-211, 2019 02.
Article in English | MEDLINE | ID: mdl-30471425

ABSTRACT

Cytochrome P450 (CYP) 27A1 is a key enzyme in both the acidic and neutral pathways of bile acid biosynthesis accepting cholesterol and ring-hydroxylated sterols as substrates introducing a (25R)26-hydroxy and ultimately a (25R)26-acid group to the sterol side-chain. In human, mutations in the CYP27A1 gene are the cause of the autosomal recessive disease cerebrotendinous xanthomatosis (CTX). Surprisingly, Cyp27a1 knockout mice (Cyp27a1-/-) do not present a CTX phenotype despite generating a similar global pattern of sterols. Using liquid chromatography - mass spectrometry and exploiting a charge-tagging approach for oxysterol analysis we identified over 50 cholesterol metabolites and precursors in the brain and circulation of Cyp27a1-/- mice. Notably, we identified (25R)26,7α- and (25S)26,7α-dihydroxy epimers of oxysterols and cholestenoic acids, indicating the presence of an additional sterol 26-hydroxylase in mouse. Importantly, our analysis also revealed elevated levels of 7α-hydroxycholest-4-en-3-one, which we found increased the number of oculomotor neurons in primary mouse brain cultures. 7α-Hydroxycholest-4-en-3-one is a ligand for the pregnane X receptor (PXR), activation of which is known to up-regulate the expression of CYP3A11, which we confirm has sterol 26-hydroxylase activity. This can explain the formation of (25R)26,7α- and (25S)26,7α-dihydroxy epimers of oxysterols and cholestenoic acids; the acid with the former stereochemistry is a liver X receptor (LXR) ligand that increases the number of oculomotor neurons in primary brain cultures. We hereby suggest that a lack of a motor neuron phenotype in some CTX patients and Cyp27a1-/- mice may involve increased levels of 7α-hydroxycholest-4-en-3-one and activation PXR, as well as increased levels of sterol 26-hydroxylase and the production of neuroprotective sterols capable of activating LXR.


Subject(s)
Cholestanetriol 26-Monooxygenase/physiology , Cholesterol/metabolism , Sterols/metabolism , Animals , Bile Acids and Salts/biosynthesis , Brain/metabolism , Cholestanetriol 26-Monooxygenase/genetics , Cholestenes/metabolism , Chromatography, Liquid , Cytochrome P-450 Enzyme System/metabolism , Hydroxylation , Lipid Metabolism/physiology , Liver X Receptors/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxysterols/metabolism , Pregnane X Receptor/metabolism , Tandem Mass Spectrometry , Xanthomatosis, Cerebrotendinous
6.
Stem Cell Reports ; 11(3): 651-664, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30146491

ABSTRACT

The development of midbrain dopaminergic (mDA) neurons is controlled by multiple morphogens and transcription factors. However, little is known about the role of extracellular matrix proteins in this process. Here we examined the function of roof plate-specific spondins (RSPO1-4) and the floor plate-specific, spondin 1 (SPON1). Only RSPO2 and SPON1 were expressed at high levels during mDA neurogenesis, and the receptor LGR5 was expressed by midbrain floor plate progenitors. Surprisingly, RSPO2, but not SPON1, specifically promoted the differentiation of mDA neuroblasts into mDA neurons in mouse primary cultures and embryonic stem cells (ESCs). In addition, RSPO2 was found to promote not only mDA differentiation, but also mDA neurogenesis in human ESCs. Our results thus uncover an unexpected function of the matricellular protein RSPO2 and suggest an application to improve mDA neurogenesis and differentiation in human stem cell preparations destined to cell replacement therapy or drug discovery for Parkinson disease.


Subject(s)
Dopaminergic Neurons/cytology , Human Embryonic Stem Cells/cytology , Intercellular Signaling Peptides and Proteins/metabolism , Mesencephalon/cytology , Neurogenesis , Thrombospondins/metabolism , Animals , Cells, Cultured , Dopaminergic Neurons/metabolism , Female , Human Embryonic Stem Cells/metabolism , Humans , Mesencephalon/metabolism , Mice
7.
Sci Rep ; 6: 26448, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27246266

ABSTRACT

Degeneration of dopamine neurons in the midbrain causes symptoms of the movement disorder, Parkinson disease. Dopamine neurons are generated from proliferating progenitor cells localized in the embryonic ventral midbrain. However, it remains unclear for how long cells with dopamine progenitor character are retained and if there is any potential for reactivation of such cells after cessation of normal dopamine neurogenesis. We show here that cells expressing Lmx1a and other progenitor markers remain in the midbrain aqueductal zone beyond the major dopamine neurogenic period. These cells express dopamine receptors, are located in regions heavily innervated by midbrain dopamine fibres and their proliferation can be stimulated by antagonizing dopamine receptors, ultimately leading to increased neurogenesis in vivo. Furthermore, treatment with dopamine receptor antagonists enhances neurogenesis in vitro, both from embryonic midbrain progenitors as well as from embryonic stem cells. Altogether our results indicate a potential for reactivation of resident midbrain cells with dopamine progenitor potential beyond the normal period of dopamine neurogenesis.

8.
F1000Prime Rep ; 7: 37, 2015.
Article in English | MEDLINE | ID: mdl-26097711

ABSTRACT

The development of the ventral midbrain is orchestrated by a number of cell-extrinsic and -intrinsic factors that control critical processes, such as the patterning of the neural tube along the main body axis and the specification of diverse neuronal cell types in distinct positions of the neural tube. Subsequently, the regulation of neurogenesis and survival- acquire particular relevance in order to define the final size of diverse neuronal populations. In a series of studies during the last few years, we have identified liver X receptors (LXRs) as critical regulators of ventral midbrain development. Moreover, specific cholesterol derivatives present in the midbrain or in the cerebrospinal fluid were identified as LXR ligands, capable of specifically and selectively regulating neurogenesis and the survival of distinct neuronal populations, including midbrain dopamine neurons. These studies have shown that cholesterol derivatives are an entirely new class of factors capable of regulating both neuronal survival and neurogenesis, thus providing a direct link between cholesterol metabolism and brain development. In addition, LXRs and cholesterol metabolism were found to play a critical role in regulating the balance between neuronal survival and death in diverse midbrain neuronal populations. In this review, we will focus on these two aspects and on the possible role of cholesterol metabolism and LXRs in neurodegeneration.

9.
J Clin Invest ; 124(11): 4829-42, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25271621

ABSTRACT

Cholestenoic acids are formed as intermediates in metabolism of cholesterol to bile acids, and the biosynthetic enzymes that generate cholestenoic acids are expressed in the mammalian CNS. Here, we evaluated the cholestenoic acid profile of mammalian cerebrospinal fluid (CSF) and determined that specific cholestenoic acids activate the liver X receptors (LXRs), enhance islet-1 expression in zebrafish, and increase the number of oculomotor neurons in the developing mouse in vitro and in vivo. While 3ß,7α-dihydroxycholest-5-en-26-oic acid (3ß,7α-diHCA) promoted motor neuron survival in an LXR-dependent manner, 3ß-hydroxy-7-oxocholest-5-en-26-oic acid (3ßH,7O-CA) promoted maturation of precursors into islet-1+ cells. Unlike 3ß,7α-diHCA and 3ßH,7O-CA, 3ß-hydroxycholest-5-en-26-oic acid (3ß-HCA) caused motor neuron cell loss in mice. Mutations in CYP7B1 or CYP27A1, which encode enzymes involved in cholestenoic acid metabolism, result in different neurological diseases, hereditary spastic paresis type 5 (SPG5) and cerebrotendinous xanthomatosis (CTX), respectively. SPG5 is characterized by spastic paresis, and similar symptoms may occur in CTX. Analysis of CSF and plasma from patients with SPG5 revealed an excess of the toxic LXR ligand, 3ß-HCA, while patients with CTX and SPG5 exhibited low levels of the survival-promoting LXR ligand 3ß,7α-diHCA. Moreover, 3ß,7α-diHCA prevented the loss of motor neurons induced by 3ß-HCA in the developing mouse midbrain in vivo.Our results indicate that specific cholestenoic acids selectively work on motor neurons, via LXR, to regulate the balance between survival and death.


Subject(s)
Cholestenes/cerebrospinal fluid , Motor Neurons/physiology , Orphan Nuclear Receptors/metabolism , Animals , Cell Survival , Cells, Cultured , Cholestenes/blood , Female , Humans , LIM-Homeodomain Proteins/metabolism , Liver X Receptors , Male , Mice, Inbred C57BL , Mice, Knockout , Paraparesis, Spastic/blood , Paraparesis, Spastic/cerebrospinal fluid , Transcription Factors/metabolism , Xanthomatosis, Cerebrotendinous/blood , Xanthomatosis, Cerebrotendinous/cerebrospinal fluid , Zebrafish
10.
Nat Chem Biol ; 9(2): 126-33, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23292650

ABSTRACT

Liver X receptors (Lxrα and Lxrß) are ligand-dependent nuclear receptors critical for ventral midbrain neurogenesis in vivo. However, no endogenous midbrain Lxr ligand has so far been identified. Here we used LC/MS and functional assays to identify cholic acid as a new Lxr ligand. Moreover, 24(S),25-epoxycholesterol (24,25-EC) was found to be the most potent and abundant Lxr ligand in the developing mouse midbrain. Both Lxr ligands promoted neural development in an Lxr-dependent manner in zebrafish in vivo. Notably, each ligand selectively regulated the development of distinct midbrain neuronal populations. Whereas cholic acid increased survival and neurogenesis of Brn3a-positive red nucleus neurons, 24,25-EC promoted dopaminergic neurogenesis. These results identify an entirely new class of highly selective and cell type-specific regulators of neurogenesis and neuronal survival. Moreover, 24,25-EC promoted dopaminergic differentiation of embryonic stem cells, suggesting that Lxr ligands may thus contribute to the development of cell replacement and regenerative therapies for Parkinson's disease.


Subject(s)
Mesencephalon/metabolism , Neurogenesis , Orphan Nuclear Receptors/metabolism , Animals , Brain Mapping/methods , Cell Differentiation , Cell Nucleus/metabolism , Cholesterol/analogs & derivatives , Cholesterol/metabolism , Cholic Acid/metabolism , Dopamine/metabolism , Dose-Response Relationship, Drug , Embryonic Stem Cells/cytology , Ligands , Liver X Receptors , Mice , Models, Biological , Time Factors , Transfection , Zebrafish
11.
Mol Cell Biol ; 33(1): 59-70, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23109420

ABSTRACT

Understanding the mechanisms that drive the differentiation of dopaminergic (DA) neurons is crucial for successful development of novel therapies for Parkinson's disease, in which DA neurons progressively degenerate. However, the mechanisms underlying the differentiation-promoting effects of Wnt5a on DA precursors are poorly understood. Here, we present the molecular and functional characterization of a signaling pathway downstream of Wnt5a, the Wnt/Dvl/Rac1 pathway. First, we characterize the interaction between Rac1 and Dvl and identify the N-terminal part of Dvl3 as necessary for Rac1 binding. Next, we show that Tiam1, a Rac1 guanosine exchange factor (GEF), is expressed in the ventral midbrain, interacts with Dvl, facilitates Dvl-Rac1 interaction, and is required for Dvl- or Wnt5a-induced activation of Rac1. Moreover, we show that Wnt5a promotes whereas casein kinase 1 (CK1), a negative regulator of the Wnt/Dvl/Rac1 pathway, abolishes the interactions between Dvl and Tiam1. Finally, using ventral midbrain neurosphere cultures, we demonstrate that the generation of DA neurons in culture is impaired after Tiam1 knockdown, indicating that Tiam1 is required for midbrain DA differentiation. In summary, our data identify Tiam1 as a novel regulator of DA neuron development and as a Dvl-associated and Rac1-specific GEF acting in the Wnt/Dvl/Rac1 pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Neuropeptides/metabolism , Phosphoproteins/metabolism , Wnt Proteins/metabolism , rac GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Casein Kinase I/metabolism , Cell Differentiation , Dishevelled Proteins , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/immunology , Humans , Immunoprecipitation , Mesencephalon/cytology , Mesencephalon/embryology , Mesencephalon/metabolism , Mice , Neuropeptides/genetics , Phosphoproteins/genetics , Signal Transduction , T-Lymphoma Invasion and Metastasis-inducing Protein 1 , Wnt Proteins/genetics , Wnt Signaling Pathway , Wnt-5a Protein , rac GTP-Binding Proteins/genetics , rac1 GTP-Binding Protein
12.
J Lipid Res ; 53(11): 2469-83, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22891291

ABSTRACT

Unesterified cholesterol is a major component of plasma membranes. In the brain of the adult, it is mostly found in myelin sheaths, where it plays a major architectural role. In the newborn mouse, little myelination of neurons has occurred, and much of this sterol comprises a metabolically active pool. In the current study, we have accessed this metabolically active pool and, using LC/MS, have identified cholesterol precursors and metabolites. Although desmosterol and 24S-hydroxycholesterol represent the major precursor and metabolite, respectively, other steroids, including the oxysterols 22-oxocholesterol, 22R-hydroxycholesterol, 20R,22R-dihydroxycholesterol, and the C(21)-neurosteroid progesterone, were identified. 24S,25-epoxycholesterol formed in parallel to cholesterol was also found to be a major sterol in newborn brain. Like 24S- and 22R-hydroxycholesterols, and also desmosterol, 24S,25-epoxycholesterol is a ligand to the liver X receptors, which are expressed in brain. The desmosterol metabolites (24Z),26-, (24E),26-, and 7α-hydroxydesmosterol were identified in brain for the first time.


Subject(s)
Chromatography, Liquid/methods , Mass Spectrometry/methods , Sterols/analysis , Animals , Animals, Newborn , Cholesterol/analysis , Desmosterol/analysis , Hydroxycholesterols/analysis , Mice
13.
PLoS One ; 6(2): e15786, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21347250

ABSTRACT

Dickkopf1 (Dkk1) is a Wnt/ß-catenin inhibitor that participates in many processes during embryonic development. One of its roles during embryogenesis is to induce head formation, since Dkk1-null mice lack head structures anterior to midbrain. The Wnt/ß-catenin pathway is also known to regulate different aspects of ventral midbrain (VM) dopaminergic (DA) neuron development and, in vitro, Dkk1-mediated inhibition of the Wnt/ß-catenin pathway improves the DA differentiation in mouse embryonic stem cells (mESC). However, the in vivo function of Dkk1 on the development of midbrain DA neurons remains to be elucidated. Here we examined Dkk1(+/-) embryos and found that Dkk1 is required for the differentiation of DA precursors/neuroblasts into DA neurons at E13.5. This deficit persisted until E17.5, when a defect in the number and distribution of VM DA neurons was detected. Furthermore, analysis of the few Dkk1(-/-) embryos that survived until E17.5 revealed a more severe loss of midbrain DA neurons and morphogenesis defects. Our results thus show that Dkk1 is required for midbrain DA differentiation and morphogenesis.


Subject(s)
Cell Differentiation , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Mesencephalon/cytology , Mesencephalon/growth & development , Morphogenesis , Animals , Cell Count , Gene Expression Regulation, Developmental , Intercellular Signaling Peptides and Proteins/deficiency , Intercellular Signaling Peptides and Proteins/genetics , Mesencephalon/metabolism , Mice , Mutation , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism
14.
J Biol Chem ; 285(7): 4666-79, 2010 Feb 12.
Article in English | MEDLINE | ID: mdl-19996111

ABSTRACT

In this study we have profiled the free sterol content of cerebrospinal fluid by a combination of charge tagging and liquid chromatography-tandem mass spectrometry. Surprisingly, the most abundant cholesterol metabolites were found to be C(27) and C(24) intermediates of the bile acid biosynthetic pathways with structures corresponding to 7alpha-hydroxy-3-oxocholest-4-en-26-oic acid (7.170 +/- 2.826 ng/ml, mean +/- S.D., six subjects), 3beta-hydroxycholest-5-en-26-oic acid (0.416 +/- 0.193 ng/ml), 7alpha,x-dihydroxy-3-oxocholest-4-en-26-oic acid (1.330 +/- 0.543 ng/ml), and 7alpha-hydroxy-3-oxochol-4-en-24-oic acid (0.172 +/- 0.085 ng/ml), and the C(26) sterol 7alpha-hydroxy-26-norcholest-4-ene-3,x-dione (0.204 +/- 0.083 ng/ml), where x is an oxygen atom either on the CD rings or more likely on the C-17 side chain. The ability of intermediates of the bile acid biosynthetic pathways to activate the liver X receptors (LXRs) and the farnesoid X receptor was also evaluated. The acidic cholesterol metabolites 3beta-hydroxycholest-5-en-26-oic acid and 3beta,7alpha-dihydroxycholest-5-en-26-oic acid were found to activate LXR in a luciferase assay, but the major metabolite identified in this study, i.e. 7alpha-hydroxy-3-oxocholest-4-en-26-oic acid, was not an LXR ligand. 7Alpha-hydroxy-3-oxocholest-4-en-26-oic acid is formed from 3beta,7alpha-dihydroxycholest-5-en-26-oic acid in a reaction catalyzed by 3beta-hydroxy-Delta(5)-C(27)-steroid dehydrogenase (HSD3B7), which may thus represent a deactivation pathway of LXR ligands in brain. Significantly, LXR activation has been found to reduce the symptoms of Alzheimer disease (Fan, J., Donkin, J., and Wellington C. (2009) Biofactors 35, 239-248); thus, cholesterol metabolites may play an important role in the etiology of Alzheimer disease.


Subject(s)
Bile Acids and Salts/metabolism , Brain/metabolism , Cerebrospinal Fluid/metabolism , Sterols/metabolism , Chenodeoxycholic Acid/analogs & derivatives , Chromatography, High Pressure Liquid , Humans , Liver X Receptors , Mass Spectrometry , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Orphan Nuclear Receptors/metabolism , Protein Binding , Receptors, Cytoplasmic and Nuclear/metabolism , Retinoid X Receptors/metabolism
15.
Cell Stem Cell ; 5(4): 409-19, 2009 Oct 02.
Article in English | MEDLINE | ID: mdl-19796621

ABSTRACT

Control over progenitor proliferation and neurogenesis remains a key challenge for stem cell neurobiology and a prerequisite for successful stem cell replacement therapies for neurodegenerative diseases like Parkinson's disease (PD). Here, we examined the function of two nuclear receptors, liver X receptors (Lxralpha and beta) and their ligands, oxysterols, as regulators of cell division, ventral midbrain (VM) neurogenesis, and dopaminergic (DA) neuron development. Deletion of Lxrs reduced cell cycle progression and VM neurogenesis, resulting in decreased DA neurons at birth. Activation of Lxrs with oxysterol ligands increased the number of DA neurons in mouse embryonic stem cells (ESCs) and in wild-type but not Lxralphabeta(-/-) VM progenitor cultures. Likewise, oxysterol treatment of human ESCs (hESCs) during DA differentiation increased neurogenesis and the number of mature DA neurons, while reducing proliferating progenitors. Thus, Lxr ligands may improve current hESC replacement strategies for PD by selectively augmenting the generation of DA neurons.


Subject(s)
Cholesterol/analogs & derivatives , Cholesterol/pharmacology , Embryonic Stem Cells/drug effects , Mesencephalon/cytology , Neurogenesis/drug effects , Orphan Nuclear Receptors/physiology , Animals , Cell Differentiation/drug effects , Dopamine/metabolism , Embryonic Stem Cells/cytology , Humans , Immunohistochemistry , In Situ Hybridization , Liver X Receptors , Mesencephalon/drug effects , Mice , Neurogenesis/genetics , Orphan Nuclear Receptors/genetics , Polymerase Chain Reaction
16.
Mol Biosyst ; 5(5): 529-41, 2009 May.
Article in English | MEDLINE | ID: mdl-19381367

ABSTRACT

In this study two regions of embryonic (E11) mouse central nervous system (CNS) have been profiled for their unesterified sterol content. Using high-performance liquid chromatography (HPLC)-mass spectrometry (MS) and tandem mass spectrometry (MS(n)) low levels of oxysterols (estimated 2-165 ng g(-1) wet weight) were identified in cortex (Ctx) and spinal cord (Sc). The identified oxysterols include 7 alpha-, 7 beta-, 22R-, 24S-, 25- and 27-hydroxycholesterol; 24,25- and 24,27-dihydroxycholesterol; and 24S,25-epoxycholesterol. Of these, 24S-hydroxycholesterol is biosynthesised exclusively in brain. In comparison to adult mouse where the 24S-hydroxycholesterol level is about 40 microg g(-1) in brain the level of 24S-hydroxycholesterol reported here (estimated 26 ng g(-1) in Ctx and 13 ng g(-1) in Sc) is extremely low. Interestingly, the level of 24S,25-epoxycholesterol in both CNS regions (estimated 165 ng g(-1) in Ctx and 91 ng g(-1) in Sc) is somewhat higher than the levels of the hydroxycholesterols. This oxysterol is formed in parallel to cholesterol via a shunt of the mevalonate pathway and its comparatively high abundance may be a reflection of a high rate of cholesterol synthesis at this stage of development. Levels of cholesterol (estimated 1.25 mg g(-1) in Ctx and 1.15 mg g(-1) in Sc) and its precursors were determined by gas chromatography-mass spectrometry (GC-MS). In both CNS regions cholesterol levels were found to be lower than those reported in the adult, but in relation to cholesterol the levels of cholesterol precursors were higher than found in adult indicating a high rate of cholesterol synthesis. In summary, our data provide evidence for the presence of endogenous oxysterols in two brain regions of the developing CNS. Moreover, while most of the enzymes involved in hydroxysterol synthesis are minimally active at E11, our results suggest that the mevalonate pathway is significantly active, opening up the possibility for a function of 24S,25-epoxycholesterol during brain development.


Subject(s)
Central Nervous System/embryology , Sterols/analysis , Animals , Brain/metabolism , Central Nervous System/metabolism , Cholesterol/analysis , Cholesterol/metabolism , Chromatography, High Pressure Liquid , Gas Chromatography-Mass Spectrometry , Mice , Spinal Cord/metabolism , Sterols/metabolism
17.
Brain Res Dev Brain Res ; 153(1): 39-51, 2004 Oct 15.
Article in English | MEDLINE | ID: mdl-15464216

ABSTRACT

Previously, this laboratory has shown that human foetal progenitor cells derived from ventral mesencephalon (VM) can be developmentally directed towards a dopaminergic lineage. In the present study, the effects are reported of several as yet untested differentiation/survival factors on the controlled conversion of neural progenitor cells to dopaminergic neurons. Positive immunoreactivity to tyrosine hydroxylase (TH) and raised levels of dopamine (DA) and its metabolite, 3,4-dihydroxyphenylacetic acid (DOPAC), secreted into culture medium, were used to indicate the presence of the dopaminergic neuronal phenotype, i.e., active TH. Incubation with retinoic acid (RA) (0.5 microM) lead to an increase in the number of cultured cells showing positive immunoreactivity for the neuronal marker, microtubule-associated protein (MAP)-2ab. A concomitant increase in TH-positive immunoreactivity was also demonstrated. The brain-derived neurotrophic factor (BDNF) (50 ng/ml), glial-derived neurotrophic factor (GDNF) (10 ng/ml) and interleukin-1 beta (IL-1 beta) (10 ng/ml) also had positive effects in promoting neural progenitor cell differentiation towards the dopaminergic phenotype in the presence of dopamine (10 microM) and forskolin (Fsk) (10 microM). There was no synergy in this effect when progenitor cells were incubated with all of these agents simultaneously. The trans-differentiation potential of the progenitor cells to be directed towards other neurotransmitter phenotypic lineages was also investigated. It was found that, with the right cocktails of agents, serotonin (Ser) (75 microM), acidic fibroblast growth factor (aFGF) (10 ng/ml), BDNF (50 ng/ml) and forskolin (10 microM), these same cells could be directed down the serotonergic cell lineage pathway (as judged by the appearance of tryptophan hydroxylase (TPH) positive immunoreactivity, and synthesis of 5-HT and its metabolites, secreted into the culture medium). However, no cocktail containing noradrenaline (10 nM-500 microM), BDNF (50 ng/ml) and forskolin (10 microM) was found which promoted differentiation towards the noradrenergic cell phenotype as judged by the absence of any TH or D beta H positive immunoreactivity, and no formation of 3,4-dihydroxyphenylethyleneglycol (DOPEG), the principal metabolite of noradrenaline. The controlled trans-differentiation potential of these cell could pave the way for development and harvesting of large numbers of neurons of the appropriate neurotransmitter phenotype for future transplantation therapies for the treatment of neurodegenerative diseases such as Parkinson's disease (PD) and Alzheimer's disease.


Subject(s)
Cell Differentiation/physiology , Mesencephalon/cytology , Methoxyhydroxyphenylglycol/analogs & derivatives , Neurons/cytology , Stem Cells/cytology , 3,4-Dihydroxyphenylacetic Acid/metabolism , Dopamine/metabolism , Fetus , Glial Cell Line-Derived Neurotrophic Factor , Growth Substances/pharmacology , Humans , Hydroxyindoleacetic Acid/metabolism , Immunohistochemistry , In Vitro Techniques , Interleukin-1/metabolism , Interleukin-1/pharmacology , Methoxyhydroxyphenylglycol/metabolism , Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacology , Neurons/drug effects , Neurons/physiology , Norepinephrine/metabolism , Serotonin/metabolism , Stem Cells/drug effects , Stem Cells/physiology , Tretinoin/metabolism , Tretinoin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...