Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Front Aging Neurosci ; 11: 96, 2019.
Article in English | MEDLINE | ID: mdl-31139073

ABSTRACT

Down syndrome (DS), a genetic disorder caused by partial or complete triplication of chromosome 21, is the most common genetic cause of intellectual disability. DS mouse models and cell lines display defects in cellular adaptive stress responses including autophagy, unfolded protein response, and mitochondrial bioenergetics. We tested the ability of hydroxyurea (HU), an FDA-approved pharmacological agent that activates adaptive cellular stress response pathways, to improve the cognitive function of Ts65Dn mice. The chronic HU treatment started at a stage when early mild cognitive deficits are present in this model (∼3 months of age) and continued until a stage of advanced cognitive deficits in untreated mice (∼5-6 months of age). The HU effects on cognitive performance were analyzed using a battery of water maze tasks designed to detect changes in different types of memory with sensitivity wide enough to detect deficits as well as improvements in spatial memory. The most common characteristic of cognitive deficits observed in trisomic mice at 5-6 months of age was their inability to rapidly acquire new information for long-term storage, a feature akin to episodic-like memory. On the background of severe cognitive impairments in untreated trisomic mice, HU-treatment produced mild but significant benefits in Ts65Dn by improving memory acquisition and short-term retention of spatial information. In control mice, HU treatment facilitated memory retention in constant (reference memory) as well as time-variant conditions (episodic-like memory) implicating a robust nootropic effect. This was the first proof-of-concept study of HU treatment in a DS model, and indicates that further studies are warranted to assess a window to optimize timing and dosage of the treatment in this pre-clinical phase. Findings of this study indicate that HU has potential for improving memory retention and cognitive flexibility that can be harnessed for the amelioration of cognitive deficits in normal aging and in cognitive decline (dementia) related to DS and other neurodegenerative diseases.

2.
Neurobiol Aging ; 72: 121-133, 2018 12.
Article in English | MEDLINE | ID: mdl-30245242

ABSTRACT

Alzheimer's disease (AD) is an age-related neurodegenerative disorder characterized by accumulation of amyloid ß-peptide (Aß) plaques in the brain and decreased cognitive function leading to dementia. We tested if hydroxyurea (HU), a ribonucleotide reductase inhibitor known to activate adaptive cellular stress responses and ameliorate abnormalities associated with several genetic disorders, could protect rat hippocampal neurons against oxidative-, excitatory-, mitochondrial-, and Aß-induced stress and if HU treatment could improve learning and memory in the APP/PS1 mouse model of AD. HU treatment attenuated the loss of cell viability induced by treatment of hippocampal neurons with hydrogen peroxide, glutamate, rotenone, and Aß1-42. HU treatment attenuated reductions of mitochondrial reserve capacity, maximal respiration, and cellular adenosine triphosphate content induced by hydrogen peroxide treatment. In vivo, treatment of APP/PS1 mice with HU (45 mg/kg/d) improved spatial memory performance in the hippocampus-dependent Morris water maze task without reducing Aß levels. HU provides neuroprotection against toxic insults including Aß, improves mitochondrial bioenergetics, and improves spatial memory in an AD mouse model. HU may offer a new therapeutic approach to delay cognitive decline in AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides , Enzyme Inhibitors/pharmacology , Hippocampus/drug effects , Hormesis/drug effects , Hydroxyurea/pharmacology , Mitochondria/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Spatial Memory/drug effects , Stress, Physiological/drug effects , Animals , Behavior, Animal/drug effects , Cells, Cultured , Disease Models, Animal , Enzyme Inhibitors/administration & dosage , Female , Hydroxyurea/administration & dosage , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Neuroprotective Agents/administration & dosage , Rats , Rats, Sprague-Dawley
3.
Proc Natl Acad Sci U S A ; 111(43): 15550-5, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25313065

ABSTRACT

Autism spectrum disorder (ASD), characterized by both impaired communication and social interaction, and by stereotypic behavior, affects about 1 in 68, predominantly males. The medico-economic burdens of ASD are enormous, and no recognized treatment targets the core features of ASD. In a placebo-controlled, double-blind, randomized trial, young men (aged 13-27) with moderate to severe ASD received the phytochemical sulforaphane (n = 29)--derived from broccoli sprout extracts--or indistinguishable placebo (n = 15). The effects on behavior of daily oral doses of sulforaphane (50-150 µmol) for 18 wk, followed by 4 wk without treatment, were quantified by three widely accepted behavioral measures completed by parents/caregivers and physicians: the Aberrant Behavior Checklist (ABC), Social Responsiveness Scale (SRS), and Clinical Global Impression Improvement Scale (CGI-I). Initial scores for ABC and SRS were closely matched for participants assigned to placebo and sulforaphane. After 18 wk, participants receiving placebo experienced minimal change (<3.3%), whereas those receiving sulforaphane showed substantial declines (improvement of behavior): 34% for ABC (P < 0.001, comparing treatments) and 17% for SRS scores (P = 0.017). On CGI-I, a significantly greater number of participants receiving sulforaphane had improvement in social interaction, abnormal behavior, and verbal communication (P = 0.015-0.007). Upon discontinuation of sulforaphane, total scores on all scales rose toward pretreatment levels. Dietary sulforaphane, of recognized low toxicity, was selected for its capacity to reverse abnormalities that have been associated with ASD, including oxidative stress and lower antioxidant capacity, depressed glutathione synthesis, reduced mitochondrial function and oxidative phosphorylation, increased lipid peroxidation, and neuroinflammmation.


Subject(s)
Child Development Disorders, Pervasive/drug therapy , Isothiocyanates/therapeutic use , Adolescent , Adult , Humans , Isothiocyanates/adverse effects , Male , Placebos , Social Behavior , Sulfoxides , Treatment Outcome , Young Adult
4.
Hum Mol Genet ; 21(19): 4237-52, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22752410

ABSTRACT

Various small molecule pharmacologic agents with different known functions produce similar outcomes in diverse Mendelian and complex disorders, suggesting that they may induce common cellular effects. These molecules include histone deacetylase inhibitors, 4-phenylbutyrate (4PBA) and trichostatin A, and two small molecules without direct histone deacetylase inhibitor activity, hydroxyurea (HU) and sulforaphane. In some cases, the therapeutic effects of histone deacetylase inhibitors have been attributed to an increase in expression of genes related to the disease-causing gene. However, here we show that the pharmacological induction of mitochondrial biogenesis was necessary for the potentially therapeutic effects of 4PBA or HU in two distinct disease models, X-linked adrenoleukodystrophy and sickle cell disease. We hypothesized that a common cellular response to these four molecules is induction of mitochondrial biogenesis and peroxisome proliferation and activation of the stress proteome, or adaptive cell survival response. Treatment of human fibroblasts with these four agents induced mitochondrial and peroxisomal biogenesis as monitored by flow cytometry, immunofluorescence and/or western analyses. In treated normal human fibroblasts, all four agents induced the adaptive cell survival response: heat shock, unfolded protein, autophagic and antioxidant responses and the c-jun N-terminal kinase pathway, at the transcriptional and translational levels. Thus, activation of the evolutionarily conserved stress proteome and mitochondrial biogenesis may be a common cellular response to such small molecule therapy and a common basis of therapeutic action in various diseases. Modulation of this novel therapeutic target could broaden the range of treatable diseases without directly targeting the causative genetic abnormalities.


Subject(s)
Adrenoleukodystrophy/drug therapy , Drug Therapy , Hydroxamic Acids/therapeutic use , Hydroxyurea/therapeutic use , Phenylbutyrates/therapeutic use , Proteome/metabolism , Thiocyanates/therapeutic use , Adrenoleukodystrophy/genetics , Adrenoleukodystrophy/metabolism , Adrenoleukodystrophy/physiopathology , Cell Line , Humans , Isothiocyanates , Mitochondrial Turnover/drug effects , Proteome/genetics , Small Molecule Libraries/therapeutic use , Sulfoxides
5.
J Neurol ; 259(7): 1440-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22218650

ABSTRACT

X-linked adrenoleukodystrophy (XALD), a neurological disorder caused by mutations in the peroxisomal membrane protein gene ABCD1, presents as a rapidly progressing, inflammatory cerebral demyelination (cerebral cases) or a slowly progressing, distal axonopathy (non-cerebral cases). Specific ABCD1 defects do not explain this significant phenotypic variation. Patients have increased plasma and tissue very long chain fatty acid levels and increased cellular oxidative stress and oxidative damage. Superoxide dismutase 2 (SOD2), at candidate modifier locus 6q25.3, detoxifies superoxide radicals protecting against oxidative stress and damage. We tested an SOD2 variant C47T (Ala16Val) associated with reduced enzymatic activity as a potential modifier gene of cerebral demyelinating disease by comparing 117 cerebral XALD cases with 105 non-cerebral XALD cases. The hypoactive valine allele of the variant was associated with cerebral disease under a dominant model in the full data set (p = 0.04; ORT* = 1.90, 95% CI 1.01-3.56) and the non-childhood cerebral disease subset (p = 0.03; ORT* = 2.47, 95% CI 1.08-5.61). Three tag SNPs were genotyped to test for additional SNP or haplotype associations. A common haplotype, GTAC, which included the SOD2 valine allele, was associated with cerebral disease in the full data set (p = 0.03; OR = 1.75, 95% CI 1.11-2.75) and the non-childhood cerebral disease subset (p = 0.008; OR = 2.20, 95% CI 1.27-3.83). There was no association between childhood cerebral XALD and the C47T variant or the GTAC haplotype. Thus, reduced SOD2 activity may contribute to the development of cerebral demyelination in adolescent and adult XALD patients.


Subject(s)
Adrenoleukodystrophy/genetics , Genetic Predisposition to Disease/genetics , Phenotype , Polymorphism, Single Nucleotide/genetics , Superoxide Dismutase/genetics , Adolescent , Adult , Age of Onset , Case-Control Studies , Chi-Square Distribution , Child , Child, Preschool , Gene Frequency , Genotype , Humans , Male , Young Adult
6.
Lab Invest ; 87(3): 261-72, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17260006

ABSTRACT

X-linked adreno-leukodystrophy is a progressive, systemic peroxisomal disorder that primarily affects the adrenal cortex, as well as myelin and axons of the central nervous system. Marked phenotypic heterogeneity does not correlate with disease-causing mutations in ABCD1, which encodes a peroxisomal membrane protein that is a member of the ABC transmembrane transporter proteins. The precise physiological functions of ABCD1 and ABCD2, a closely related peroxisomal membrane half-transporter, are unknown. The abcd1 knockout mouse does not develop the inflammatory demyelination so typical and devastating in adreno-leukodystrophy, but it does display the same lamellae and lipid profiles in adrenocortical cells under the electron microscope as the human patients. The adrenocortical cells in the mouse also exhibit immunohistochemical evidence of oxidative stress at 12 weeks but no evidence of oxidative damage. To better understand the pathogenesis of this complex disease, we evaluate the adrenal lesion of the abcd1 knockout mouse as a function of normal aging, dietary or therapeutic manipulations, and abcd genotype. The loss of abcd2 causes oxidative stress in the adrenal at 12 weeks, as judged by increased immunoreactivity for the mitochondrial manganese superoxide dismutase, in both the inner cortex and medulla. The loss of abcd2 (n=20), but not abcd1 (n=27), results in the spontaneous and premature deposition of ceroid, a known end-product of oxidative damage, predominantly in adrenal medullary cells. These data indicate that the loss of abcd2 results in greater oxidative stress in murine adrenal cells than the loss of abcd1, providing a clue to its cellular function. We also find that the adrenocortical lesion of the abcd1 knockout mouse does not produce functional impairment at ten to nineteen months or overt hypocortisolism at any age, nor does it progress histologically; these and other data align this mouse model closer to human female heterozygotes than to male ALD or AMN hemizygotes.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Adrenal Glands/metabolism , Chemokines, CC/physiology , Oxidative Stress , Peroxisomes/metabolism , ATP Binding Cassette Transporter, Subfamily D , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Chemokine CCL22 , Chemokines, CC/genetics , Chemokines, CC/metabolism , Dehydroepiandrosterone Sulfate/administration & dosage , Drug Combinations , Erucic Acids/administration & dosage , Immunohistochemistry , In Situ Hybridization , Male , Mice , Mice, Knockout , Triolein/administration & dosage
7.
Exp Hematol ; 34(9): 1151-61, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16939808

ABSTRACT

OBJECTIVE: In vivo, several drugs have been shown to increase fetal hemoglobin (HbF), including 5-azacytidine (AZA), sodium butyrate (SB), and hydroxyurea (HU). Studies in K562 cells suggest that cyclic guanosine monophosphate (cGMP) is required for HbF induction; however, the role of cyclic nucleotides in HbF induction in primary erythroid cultures has not been established. METHODS: CD34-selected peripheral blood monocytes cultured in a semi-solid serum-free system that mimics in vivo F-cell production are utilized to explore the role of cyclic adenosine monophosphate (cAMP) and cGMP in HbF induction in response to HU, AZA, and SB. RESULTS: In serum-free CD34 cultures, HU, SB, and AZA all markedly stimulate FNRBC production up to 30-fold, associated with induction of gamma-globin mRNA and total HbF protein. Guanylate cyclase inhibition results in only minimal blunting of HbF induction by each agent. In contrast, adenylate cyclase inhibition markedly reduces HU, SB, and AZA-mediated FNRBC induction and gamma-globin mRNA induction. The adenylate cyclase activator forskolin modestly induces FNRBC production and augments the action of standard induction agents. HU, AZA, and SB, however, fail to significantly stimulate adenylate cyclase themselves. CONCLUSIONS: In human CD34(+) cultures, cAMP production is required for full induction of HbF by HU, SB, and AZA, while perturbation of cGMP production has only minimal effects. These findings are in marked contrast to data in K562 cells where cGMP production is critical for HbF induction while cAMP stimulation blunts HbF response, and suggest that these agents may share a common induction pathway.


Subject(s)
Antigens, CD34 , Antisickling Agents/pharmacology , Azacitidine/analogs & derivatives , Butyrates/pharmacology , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Enzyme Inhibitors/pharmacology , Fetal Hemoglobin/biosynthesis , Hydroxyurea/pharmacology , Monocytes/metabolism , Adenylyl Cyclases/metabolism , Azacitidine/pharmacology , Colforsin/pharmacology , Decitabine , Erythroid Cells/cytology , Erythroid Cells/metabolism , Humans , K562 Cells , Monocytes/cytology , RNA, Messenger/biosynthesis
8.
J Neuropathol Exp Neurol ; 64(12): 1067-79, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16319717

ABSTRACT

X-linked adreno-leukodystrophy is a progressive, systemic peroxisomal disorder that affects primarily nervous system myelin and axons as well as the adrenal cortex. Several divergent clinical phenotypes can occur in the same family; thus, there is no correlation between the clinical phenotype and the mutation in the ABCD1 gene in this disease. The most urgent and unresolved clinical issue is the fulminant inflammatory (immune) demyelination of the central nervous system in which a variety of cellular participants, cytokines, and chemokines are noted. A knockout mouse model exhibits mitochondrial deficits and axonal degeneration, but not inflammatory demyelination. To determine whether oxidative stress and damage might play a pathogenic role, we assessed standard biochemical and immunohistochemical markers of such activity both in our knockout mouse model and patients. We find that oxidative stress, as judged by increased immunoreactivity for the mitochondrial manganese-superoxide dismutase, is present in the knockout mouse liver, adrenal cortex, and renal cortex, tissues that normally express high levels of ABCD1 but no evidence of oxidative damage. The brain does not exhibit either oxidative stress or damage. On the other hand, both the human adrenal cortex and brain show evidence of oxidative stress (e.g. hemoxygenase-1 and manganese-superoxide dismutase) and oxidative damage, particularly from lipid peroxidation (4-hydroxynonenal and malondialdehyde). The presence of nitrotyrosylated proteins is strong circumstantial evidence for the participation of the highly toxic peroxynitrite molecule, whereas the demonstration of interferon gamma and interleukin-12 is indicative of a TH1 response in the inflammatory demyelinative lesions of the cerebral phenotype. These differences between the adreno-leukodystrophy mouse and human patients are intriguing and may provide a clue to the phenotypic divergence in this disease.


Subject(s)
Adrenoleukodystrophy/etiology , Oxidative Stress , Adrenal Cortex/enzymology , Adrenal Cortex/metabolism , Adrenal Cortex/pathology , Adrenoleukodystrophy/metabolism , Adrenoleukodystrophy/pathology , Animals , Biochemistry/methods , Biomarkers/metabolism , Brain/metabolism , Brain/pathology , Chemokine CCL22 , Chemokines, CC/deficiency , Humans , Immunohistochemistry , Interferon-gamma/metabolism , Interleukin-12/metabolism , Kidney Cortex/enzymology , Kidney Cortex/pathology , Liver/enzymology , Liver/pathology , Mice , Mice, Knockout , Mitochondria/enzymology , Mitochondria/pathology , Superoxide Dismutase/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism
9.
Mol Genet Metab ; 83(1-2): 117-27, 2004.
Article in English | MEDLINE | ID: mdl-15464426

ABSTRACT

The principal biochemical abnormality in the neurodegenerative disorder X-linked adrenoleukodystrophy (X-ALD) is elevated plasma and tissue levels of very long-chain fatty acids (VLCFA). Enzymes with very long-chain acyl-CoA synthetase (VLACS) activity are required for VLCFA metabolism, including degradation by peroxisomal beta-oxidation or incorporation into complex lipids, and may also participate in VLCFA synthesis. Two enzymes with VLACS activity, ACSVL1 and BG1, were investigated for their potential role in X-ALD biochemical pathology. Skin fibroblast mRNA levels for ACSVL1, an enzyme previously shown to be in peroxisomes and to participate in VLCFA beta-oxidation, were not significantly different between normal controls, patients with childhood cerebral X-ALD, and patients with adrenomyeloneuropathy. Similar results were obtained with mRNA for BG1, a non-peroxisomal enzyme that is highly expressed in nervous system, adrenal gland, and testis, the principal tissues pathologically affected in X-ALD. No significant differences in the immunohistochemical staining patterns of tissues expressing either ACSVL1 or BG1 were observed when wild-type and X-ALD mice were compared. Western blot analysis of BG1 protein levels showed no differences between fibroblasts from controls, cerebral X-ALD, or adrenomyeloneuropathy patients. BG1 protein levels were similar in wild-type and X-ALD mouse brain, spinal cord, testis, and adrenal gland. We hypothesized that one function of BG1 was to direct VLCFA into the cholesterol ester synthesis pathway. However, BG1 depletion in Neuro2a cells using RNA interference did not decrease incorporation of labeled VLCFA into cholesterol esters. We conclude that the role, if any, of ACSVL1 and BG1 in X-ALD biochemical pathology is indirect.


Subject(s)
Adrenoleukodystrophy/enzymology , Coenzyme A Ligases/physiology , ATP Binding Cassette Transporter, Subfamily D, Member 1 , ATP-Binding Cassette Transporters/genetics , Adrenoleukodystrophy/pathology , Animals , Cells, Cultured , Cholesterol Esters/biosynthesis , Coenzyme A Ligases/genetics , Fatty Acids/chemistry , Fatty Acids/metabolism , Fibroblasts/enzymology , Gene Expression Regulation , Humans , Mice , Mice, Mutant Strains , Reference Values , Skin/cytology , Skin/enzymology
10.
J Biol Chem ; 278(47): 47070-8, 2003 Nov 21.
Article in English | MEDLINE | ID: mdl-12975357

ABSTRACT

Acyl-CoA synthetases play a pivotal role in fatty acid metabolism, providing activated substrates for fatty acid catabolic and anabolic pathways. Acyl-CoA synthetases comprise numerous proteins with diverse substrate specificities, tissue expression patterns, and subcellular localizations, suggesting that each enzyme directs fatty acids toward a specific metabolic fate. We reported that hBG1, the human homolog of the acyl-CoA synthetase mutated in the Drosophila mutant "bubblegum," belongs to a previously unidentified enzyme family and is capable of activating both long- and very long-chain fatty acid substrates. We now report that when overexpressed, hBG1 can activate diverse saturated, monosaturated, and polyunsaturated fatty acids. Using in situ hybridization and immunohistochemistry, we detected expression of mBG1, the mouse homolog of hBG1, in cerebral cortical and cerebellar neurons and in steroidogenic cells of the adrenal gland, testis, and ovary. The expression pattern and ability of BG1 to activate very long-chain fatty acids implicates this enzyme in the pathogenesis of X-linked adrenoleukodystrophy. In neuron-derived Neuro2a cells, mBG1 co-sedimented with mitochondria and was found in small vesicular structures located in close proximity to mitochondria. RNA interference was used to decrease mBG1 expression in Neuro2a cells and led to a 30-35% decrease in activation and beta-oxidation of the long-chain fatty acid, palmitate. These results suggest that in Neuro2a cells, mBG1-activated long-chain fatty acids are directed toward mitochondrial degradation. mBG1 appears to play a minor role in very long-chain fatty acid activation in these cells, indicating that other acyl-CoA synthetases are necessary for very long-chain fatty acid metabolism in Neuro2a cells.


Subject(s)
Coenzyme A Ligases/physiology , Fatty Acids/metabolism , Neurons/enzymology , Adrenoleukodystrophy/etiology , Animals , COS Cells , Coenzyme A Ligases/genetics , DNA, Complementary , Humans , Mice , Mice, Inbred Strains , Microscopy, Fluorescence , Mitochondria/enzymology , Mitochondria/ultrastructure , Neurons/ultrastructure , Oxidation-Reduction , RNA, Messenger/analysis , RNA, Messenger/metabolism , Subcellular Fractions , Substrate Specificity , Tissue Distribution , Transfection
11.
Hum Mol Genet ; 12(10): 1145-54, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12719378

ABSTRACT

X-linked adrenoleukodystrophy (X-ALD) is a neurodegenerative and endocrine disorder resulting from mutations in ABCD1 which encodes a peroxisomal membrane protein in the ATP binding cassette superfamily. The biochemical signature of X-ALD is increased levels of saturated very long-chain fatty acids (VLCFA; carbon chains of 22 or more) in tissues and plasma that has been associated with decreased peroxisomal very long-chain acyl-CoA synthetase (VLCS) activity and decreased peroxisomal VLCFA beta-oxidation. It has been hypothesized that ABCD1, which has no demonstrable VLCS activity itself, has an indirect effect on peroxisomal VLCS activity and VLCFA beta-oxidation by transporting fatty acid substrates, VLCS protein or some required co-factor into peroxisomes. Here we report the characterization of a Vlcs knockout mouse that exhibits decreased peroxisomal VLCS activity and VLCFA beta-oxidation but does not accumulate VLCFA. The XALD/Vlcs double knockout mouse has the biochemical abnormalities observed in the individual knockout mice but does not display a more severe X-ALD phenotype. These data lead us to conclude that (1) VLCFA levels are independent of peroxisomal fatty acid beta-oxidation, (2) there is no ABCD1/VLCS interaction and (3) the common severe forms of X-ALD cannot be modeled by decreasing peroxisomal VLCS activity in the XALD mouse.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Adrenoleukodystrophy/genetics , Coenzyme A Ligases/deficiency , Repressor Proteins , Saccharomyces cerevisiae Proteins , ATP Binding Cassette Transporter, Subfamily D, Member 1 , ATP-Binding Cassette Transporters/metabolism , Adrenoleukodystrophy/enzymology , Animals , Brain/enzymology , Coenzyme A Ligases/genetics , Fatty Acids/metabolism , Humans , Kidney/enzymology , Liver/enzymology , Mice , Mice, Knockout
13.
Mol Cell Biol ; 22(23): 8226-40, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12417726

ABSTRACT

The PEX11 peroxisomal membrane proteins promote peroxisome division in multiple eukaryotes. As part of our effort to understand the molecular and physiological functions of PEX11 proteins, we disrupted the mouse PEX11alpha gene. Overexpression of PEX11alpha is sufficient to promote peroxisome division, and a class of chemicals known as peroxisome proliferating agents (PPAs) induce the expression of PEX11alpha and promote peroxisome division. These observations led to the hypothesis that PPAs induce peroxisome abundance by enhancing PEX11alpha expression. The phenotypes of PEX11alpha(-/-) mice indicate that this hypothesis remains valid for a novel class of PPAs that act independently of peroxisome proliferator-activated receptor alpha (PPARalpha) but is not valid for the classical PPAs that act as activators of PPARalpha. Furthermore, we find that PEX11alpha(-/-) mice have normal peroxisome abundance and that cells lacking both PEX11alpha and PEX11beta, a second mammalian PEX11 gene, have no greater defect in peroxisome abundance than do cells lacking only PEX11beta. Finally, we report the identification of a third mammalian PEX11 gene, PEX11gamma, and show that it too encodes a peroxisomal protein.


Subject(s)
Membrane Proteins/genetics , Peroxisome Proliferators/pharmacology , Peroxisomes/drug effects , Peroxisomes/metabolism , Phenylbutyrates/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Antineoplastic Agents/pharmacology , Diet , Fatty Acids/chemistry , Fatty Acids/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation , Gene Targeting , Liver/cytology , Liver/metabolism , Membrane Proteins/chemistry , Membrane Proteins/classification , Membrane Proteins/metabolism , Mice , Mice, Inbred Strains , Mice, Knockout , Mitochondria/ultrastructure , Molecular Sequence Data , Oxidation-Reduction , Peroxisome Proliferators/administration & dosage , Peroxisomes/ultrastructure , Phenotype , Phylogeny , Plasmalogens/metabolism , Sequence Alignment , Tissue Distribution
14.
J Biol Chem ; 277(32): 28765-73, 2002 Aug 09.
Article in English | MEDLINE | ID: mdl-12048192

ABSTRACT

X-linked adrenoleukodystrophy (X-ALD) is a neurodegenerative disorder characterized by accumulation of very long-chain fatty acids (VLCFA). This accumulation has been attributed to decreased VLCFA beta-oxidation and peroxisomal very long-chain acyl-CoA synthetase (VLCS) activity. The X-ALD gene, ABCD1, encodes a peroxisomal membrane ATP binding cassette transporter, ALDP, that is hypothesized to affect VLCS activity in peroxisomes by direct interaction with the VLCS enzyme. Recently, a VLCS gene that encodes a protein with significant sequence identity to known rat and human peroxisomal VLCS protein has been identified in mice. We find that the mouse VLCS gene (Vlcs) encodes an enzyme (Vlcs) with VLCS activity that localizes to peroxisomes and is expressed in X-ALD target tissues. We show that the expression of Vlcs in the peroxisomes of X-ALD mouse fibroblasts improves VLCFA beta-oxidation in these cells, implying a role for this enzyme in the biochemical abnormality of X-ALD. X-ALD mice, which accumulate VLCFA in tissues, show no change in the expression of Vlcs, the subcellular localization of Vlcs, or general peroxisomal VLCS activity. These observations imply that ALDP is not necessary for the proper expression or localization of Vlcs protein, and the control of VLCFA levels does not depend on the direct interaction of Vlcs and ALDP.


Subject(s)
Adrenoleukodystrophy/enzymology , Adrenoleukodystrophy/genetics , Coenzyme A Ligases/biosynthesis , Repressor Proteins , Saccharomyces cerevisiae Proteins , Amino Acid Sequence , Animals , Blotting, Northern , Blotting, Western , COS Cells , Catalase/metabolism , Cells, Cultured , Cloning, Molecular , Coenzyme A Ligases/genetics , DNA, Complementary/metabolism , Fibroblasts/metabolism , Immunohistochemistry , Liver/enzymology , Mice , Microscopy, Fluorescence , Microsomes, Liver/enzymology , Molecular Sequence Data , Peroxisomes/metabolism , Phenotype , Protein Binding , Reverse Transcriptase Polymerase Chain Reaction , Subcellular Fractions/metabolism , Tissue Distribution , Transfection
15.
J Biol Chem ; 277(27): 24771-9, 2002 Jul 05.
Article in English | MEDLINE | ID: mdl-11980911

ABSTRACT

Bile acids are synthesized de novo in the liver from cholesterol and conjugated to glycine or taurine via a complex series of reactions involving multiple organelles. Bile acids secreted into the small intestine are efficiently reabsorbed and reutilized. Activation by thioesterification to CoA is required at two points in bile acid metabolism. First, 3alpha,7alpha,12alpha-trihydroxy-5beta-cholestanoic acid, the 27-carbon precursor of cholic acid, must be activated to its CoA derivative before side chain cleavage via peroxisomal beta-oxidation. Second, reutilization of cholate and other C24 bile acids requires reactivation prior to re-conjugation. We reported previously that homolog 2 of very long-chain acyl-CoA synthetase (VLCS) can activate cholate (Steinberg, S. J., Mihalik, S. J., Kim, D. G., Cuebas, D. A., and Watkins, P. A. (2000) J. Biol. Chem. 275, 15605-15608). We now show that this enzyme also activates chenodeoxycholate, the secondary bile acids deoxycholate and lithocholate, and 3alpha,7alpha,12alpha-trihydroxy-5beta-cholestanoic acid. In contrast, VLCS activated 3alpha,7alpha,12alpha-trihydroxy-5beta-cholestanoate, but did not utilize any of the C24 bile acids as substrates. We hypothesize that the primary function of homolog 2 is in the reactivation and recycling of C24 bile acids, whereas VLCS participates in the de novo synthesis pathway. Results of in situ hybridization, topographic orientation, and inhibition studies are consistent with the proposed roles of these enzymes in bile acid metabolism.


Subject(s)
Bile Acids and Salts/metabolism , Coenzyme A Ligases/metabolism , Repressor Proteins , Saccharomyces cerevisiae Proteins , Bile Acids and Salts/biosynthesis , Chenodeoxycholic Acid/metabolism , Cholic Acid/pharmacology , Cloning, Molecular , Coenzyme A Ligases/antagonists & inhibitors , Coenzyme A Ligases/genetics , DNA Primers , Humans , Kinetics , Liver/enzymology , Molecular Sequence Data , Open Reading Frames , Polymerase Chain Reaction , Recombinant Proteins/metabolism , Stereoisomerism , Substrate Specificity
16.
Am J Hum Genet ; 70(6): 1520-31, 2002 Jun.
Article in English | MEDLINE | ID: mdl-11992258

ABSTRACT

X-linked adrenoleukodystrophy (X-ALD) results from mutations in ABCD1. ABCD1 resides on Xq28 and encodes an integral peroxisomal membrane protein (ALD protein [ALDP]) that is of unknown function and that belongs to the ATP-binding cassette-transporter superfamily. Individuals with ABCD1 mutations accumulate very-long-chain fatty acids (VLCFA) (carbon length >22). Childhood cerebral X-ALD is the most devastating form of the disease. These children have the earliest onset (age 7.2 +/- 1.7 years) among the clinical phenotypes for ABCD1 mutations, but onset does not occur at <3 years of age. Individuals with either peroxisomal biogenesis disorders (PBD) or single-enzyme deficiencies (SED) in the peroxisomal beta-oxidation pathway--disorders such as acyl CoA oxidase deficiency and bifunctional protein deficiency--also accumulate VLCFA, but they present during the neonatal period. Until now, it has been possible to distinguish unequivocally between individuals with these autosomal recessively inherited syndromes and individuals with ABCD1 mutations, on the basis of the clinical presentation and measurement of other biochemical markers. We have identified three newborn boys who had clinical symptoms and initial biochemical results consistent with PBD or SED. In further study, however, we showed that they lacked ALDP, and we identified deletions that extended into the promoter region of ABCD1 and the neighboring gene, DXS1357E. Mutations in DXS1357E and the ABCD1 promoter region have not been described previously. We propose that the term "contiguous ABCD1 DXS1357E deletion syndrome" (CADDS) be used to identify this new contiguous-gene syndrome. The three patients with CADDS who are described here have important implications for genetic counseling, because individuals with CADDS may previously have been misdiagnosed as having an autosomal recessive PBD or SED


Subject(s)
Adrenoleukodystrophy/genetics , Chemokines, CC/genetics , Infant, Newborn, Diseases/genetics , Peroxisomal Disorders/physiopathology , Proteins/genetics , Sequence Deletion/genetics , X Chromosome/genetics , ATP Binding Cassette Transporter, Subfamily D, Member 1 , ATP-Binding Cassette Transporters/genetics , Adrenoleukodystrophy/diagnosis , Adrenoleukodystrophy/metabolism , Adrenoleukodystrophy/physiopathology , Age of Onset , Chemokine CCL22 , Child , Child, Preschool , Exons/genetics , Female , Fibroblasts , Genetic Complementation Test , Heterozygote , Humans , Infant , Infant, Newborn , Infant, Newborn, Diseases/diagnosis , Infant, Newborn, Diseases/metabolism , Infant, Newborn, Diseases/physiopathology , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Peroxisomal Disorders/diagnosis , Peroxisomal Disorders/genetics , Peroxisomal Disorders/metabolism , Peroxisomes/metabolism , Peroxisomes/pathology , Phenotype , Prenatal Diagnosis , Promoter Regions, Genetic/genetics , Syndrome
17.
Blood ; 85(4): 1111-7, Feb. 15, 1995.
Article in English | MedCarib | ID: med-5870

ABSTRACT

Five factors have been shown to influence the 20-fold variation of fetal hemoglobin (Hb F) levels in sickle cell anemia (SS): age, sex, the O-globin gene number, á-globin haplotypes, and an X-linked locus that regulates the production of Hb F-containing erythrocytes (F cells), i.e., the F-cell production (FCP) locus. To determine the relative importance of these factors, we studied 257 Jamaican SS subjects from a cohort group identified by newborn screening and from a sib pair study. Linear regression analyses showed that each variable, when analyzed alone, had a significant association with Hb F levels (P < 0.05). Multiple regression analysis, including all variables, showed that the FCP locus is the strongest predictor, accounting for 40 percent of Hb F variation. á-Globin haplotypes, O-globin genes, and age accounted for less than 10 percent of the variation. The association between the á-globin haplotypes and Hb F levels becomes apparent if the influence of the FCP locus is removed by analyzing only individuals with the same FCP phenotype. Thus, the FCP locus is the most important factor identified to date in determining Hb F levels. The variation within each FCP phenotype is modulated by factors associated with the three common á-globin haplotypes and other as yet unidentified factor(s).(AU)


Subject(s)
Humans , Infant, Newborn , Infant , Child, Preschool , Child , Adolescent , Adult , Fetal Hemoglobin/genetics , Anemia, Sickle Cell/genetics , Genetic Variation , Anemia, Sickle Cell/blood , Cohort Studies
18.
Blood ; 64(5): 1053-8, Nov. 1984.
Article in English | MedCarib | ID: med-14716

ABSTRACT

Levels of fetal hemoglobin (HbF) bearing reticulocytes (F reticulocytes) range from 2 percent to 50 percent in patients with sickle cell (SS) anemia. To learn whether any portion of such variation in F cell production is regulated by loci genetically separable from the á- globin gene cluster, percentages of F reticulocytes were compared in 59 sib pairs composed solely of SS members, including 40 pairs from Jamaica and 19 from the United States. We reasoned that differences in F reticulocyte levels might arise (1) from any of several kinds of artifact, (2) via half-sib status, or (3) because one or more genes regulating F cell production segregate separately from ás. We minimized the role of artifact by assay of fresh samples from 84 SS individuals, including both members of 38 sib pairs. In 78 of the 84 subjects, serial values for percent F reticulocytes fell within 99.9 percent confidence limits or were alike by t test (Po .05). This left 32 sib pairs for which F reticulocyte levels in each member were reproducible. When sib-sib comparisons were limited to these 32 pairs, percentages of F reticulocytes were grossly dissimilar within 12 Jamaican and 3 American sibships. Within them, the probability that sibs were alike was always ó .005 and usually ó 10 to the 4th power. We next minimized the contribution of half-sibs among Jamaicans by a combination of paternity testing and sib-sib comparison of á-globin region DNA restriction fragment length polymorphisms, especially among discordant pairs. We thereafter concluded that at least seven to eight Jamaican pairs were composed of reproducibly discordant full sibs. There is thus little doubt that there are genes regulating between-patient differences in F cell production that are separate from the á-globin gene cluster. Still unanswered is (1) whether or not these genes are actually linked to á to the s power, (2) why F reticulocyte levels in Americans tend to be lower than in Jamaicans, and (3) whether or not differences in F cell production among SS patients are regulated by several major loci or by only one (AU)


Subject(s)
Humans , Child , Adolescent , Adult , Anemia, Sickle Cell/genetics , Fetal Hemoglobin/analysis , Gene Expression Regulation , Alleles , Anemia, Sickle Cell/blood , Comparative Study , Family , Reticulocytes/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...