Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Nat Genet ; 17(2): 185-9, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9326939

ABSTRACT

Refsum disease is an autosomal recessive disorder characterized by retinitis pigmentosa, peripheral polyneuropathy, cerebellar ataxia and increased cerebrospinal fluid protein. Biochemically, the disorder is defined by two related properties: pronounced accumulation of phytanic acid and selective loss of the peroxisomal dioxygenase required for alpha-hydroxylation of phytanoyl-CoA2. Decreased phytanic-acid oxidation is also observed in human cells lacking PEX7, the receptor for the type-2 peroxisomal targetting signal (PTS2; refs 3,4), suggesting that the enzyme defective in Refsum disease is targetted to peroxisomes by a PTS2. We initially identified the human PAHX and mouse Pahx genes as expressed sequence tags (ESTs) capable of encoding PTS2 proteins. Human PAHX is targetted to peroxisomes, requires the PTS2 receptor for peroxisomal localization, interacts with the PTS2 receptor in the yeast two-hybrid assay and has intrinsic phytanoyl-CoA alpha-hydroxylase activity that requires the dioxygenase cofactor iron and cosubstrate 2-oxoglutarate. Radiation hybrid data place PAHX on chromosome 10 between the markers D10S249 and D10S466, a region previously implicated in Refsum disease by homozygosity mapping. We find that both Refsum disease patients examined are homozygous for inactivating mutations in PAHX, demonstrating that mutations in PAHX can cause Refsum disease.


Subject(s)
Mixed Function Oxygenases/genetics , Refsum Disease/genetics , Amino Acid Sequence , Animals , Base Sequence , Chromosome Mapping , Chromosomes, Human, Pair 10/genetics , DNA Mutational Analysis , DNA Primers/genetics , DNA, Complementary/genetics , Gene Expression , Homozygote , Humans , Mice , Microbodies/metabolism , Mixed Function Oxygenases/metabolism , Molecular Sequence Data , Peroxisomal Targeting Signal 2 Receptor , Polymerase Chain Reaction , Receptors, Cytoplasmic and Nuclear/metabolism , Refsum Disease/metabolism , Sequence Homology, Amino Acid , Species Specificity
2.
Nat Med ; 4(11): 1261-8, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9809549

ABSTRACT

As more functional redundancy in mammalian cells is discovered, enhanced expression of genes involved in alternative pathways may become an effective form of gene therapy. X-linked adrenoleukodystrophy (X-ALD) is a peroxisomal disorder with impaired very-long-chain fatty acid metabolism. The X-ALD gene encodes a peroxisomal membrane protein (ALDP) that is part of a small family of related peroxisomal membrane proteins. We show that 4-phenylbutyrate treatment of cells from both X-ALD patients and X-ALD knockout mice results in decreased levels of and increased beta-oxidation of very-long-chain fatty acids; increased expression of the peroxisomal protein ALDRP; and induction of peroxisome proliferation. We also demonstrate that ALDP and ALDRP are functionally related, by ALDRP cDNA complementation of X-ALD fibroblasts. Finally, we demonstrate the in vivo efficacy of dietary 4-phenylbutyrate treatment through its production of a substantial reduction of very-long-chain fatty acid levels in the brain and adrenal glands of X-ALD mice.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Adrenoleukodystrophy/genetics , Adrenoleukodystrophy/therapy , Genetic Therapy , Proteins/genetics , X Chromosome , ATP Binding Cassette Transporter, Subfamily D , ATP Binding Cassette Transporter, Subfamily D, Member 1 , Animals , Cell Line , Cells, Cultured , DNA Primers , Fibroblasts/cytology , Fibroblasts/drug effects , Humans , Lymphocytes/cytology , Lymphocytes/drug effects , Mice , Mice, Knockout , Microbodies/drug effects , Microbodies/physiology , Microbodies/ultrastructure , Multigene Family , Phenylbutyrates/pharmacology , Reverse Transcriptase Polymerase Chain Reaction
3.
J Cell Biol ; 73(2): 332-53, 1977 May.
Article in English | MEDLINE | ID: mdl-192733

ABSTRACT

The nonproliferating chicken liver cell culture system described yields cell monolayers with morphological and lipogenic properties characteristic of the physiological-nutritional state of donor animals. Synthesis and secretion of fatty acid, cholesterol, and very low density lipoprotein (VLDL) occur at in vivo rates and respond to hormones and agents which affect these processes in vivo. Cells derived from fed chickens maintain high rates of synthesis of fatty acid and cholesterol for several days if insulin is present in the medium. High rates of fatty acid synthesis are correlated with the appearance of membrane-enclosed triglyceride-rich vesicles in the cytoplasm; deletion of insulin causes a decrease (T1/2 = 22 h) in fatty acid synthetic activity. Addition of glucagon or cyclic AMP (cAMP) causes an immediate cessation of fatty acid synthesis and blocks the appearance of the triglyceride-rich vesicles. Fatty acid synthesis in liver cells prepared from fasted chickens is less than 5% that of cells from fed animals. After 2-3 days in culture with serum-free medium containing insulin +/- triiodothyronine, fatty acid synthesis is restored to normal; glucagon or dibutyryl cAMP blocks this recovery. Liver cells derived from estradiol-treated chickens synthesize and secrete VLDL for at least 48 h in culture. Electron micrographs of these cells reveal more extensive development of the rough endoplasmic reticulum and Golgi complex compared to cells from untreated chickens. Whereas [3H]leucine incorporation into total protein is unaffected by estrogen treatment, [3H]leucine incorporation into cellular and secreted immunoprecipitable VLDL is markedly increased indicating specific activation of VLDL apopeptide synthesis; 8-10% of the labeled protein synthesized and secreted is VLDL. Dodecyl sulfate-acrylamide gel electrophoresis of immunoprecipitated 3H-VLDL reveals three major apopepetides of 300,000, 11,000, and 8,000 daltons corresponding to those of purified chicken VLDL.


Subject(s)
Estradiol/pharmacology , Glucagon/pharmacology , Insulin/pharmacology , Lipids/biosynthesis , Lipoproteins, VLDL/metabolism , Liver/cytology , Bucladesine/pharmacology , Cells, Cultured , Cholesterol/biosynthesis , Cyclic AMP/pharmacology , Endoplasmic Reticulum/ultrastructure , Fatty Acids/biosynthesis , Golgi Apparatus/ultrastructure , Lipoproteins, VLDL/biosynthesis , Liver/metabolism , Protein Biosynthesis , Triiodothyronine/pharmacology
4.
J Inherit Metab Dis ; 32(1): 109-19, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19127411

ABSTRACT

Zellweger spectrum disorders (ZSD) are diagnosed by biochemical assay in blood, urine and cultured fibroblasts and PEX gene mutation identification. In most cases studies in fibroblasts corroborate results obtained in body fluids. In 1996 Clayton and colleagues described a 10-year old girl with evidence of a peroxisome disorder, based on elevated bile acid metabolites and phytanate. At the time it was not possible to distinguish whether she had a ZSD or a single peroxisomal protein defect. Studies in our laboratory showed that she also had elevated plasma pipecolate, supporting the former diagnosis. Despite the abnormal metabolites detected in blood (phytanate, bile acid intermediates and pipecolate), analysis of multiple peroxisomal pathways in fibroblasts yielded normal results. In addition, she had a milder clinical phenotype than usually associated with ZSD. Since complementation analysis to determine the gene defect was not possible, we screened this patient following the PEX Gene Screen algorithm (PGS). The PGS provides a template for sequencing PEX gene exons independent of complementation analysis. Two mutations in PEX10 were identified, a frameshift mutation inherited from her father and a de novo missense mutation in a conserved functional domain on the other allele. This case highlights that molecular analysis may be essential to the diagnosis of patients at the milder end of the ZSD spectrum. Furthermore, it supports the concept that some tissues are less affected by certain PEX gene defects than brain and liver.


Subject(s)
Fibroblasts/metabolism , Peroxisomes/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Zellweger Syndrome/diagnosis , Adolescent , Amino Acid Sequence , Base Sequence , Cells, Cultured , Female , Humans , Molecular Sequence Data , Mutation/physiology , Pedigree , Peroxins , Peroxisomes/chemistry , Receptors, Cytoplasmic and Nuclear/metabolism , Zellweger Syndrome/genetics , Zellweger Syndrome/metabolism
5.
J Clin Invest ; 83(3): 771-7, 1989 Mar.
Article in English | MEDLINE | ID: mdl-2921319

ABSTRACT

Peroxisomal function was evaluated in a male infant with clinical features of neonatal adrenoleukodystrophy. Very long chain fatty acid levels were elevated in both plasma and fibroblasts, and beta-oxidation of very long chain fatty acids in cultured fibroblasts was significantly impaired. Although the level of the bile acid intermediate trihydroxycoprostanoic acid was slightly elevated in plasma, phytanic acid and L-pipecolic acid levels were normal, as was plasmalogen synthesis in cultured fibroblasts. The latter three parameters distinguish this case from classical neonatal adrenoleukodystrophy. In addition, electron microscopy and catalase subcellular distribution studies revealed that, in contrast to neonatal adrenoleukodystrophy, peroxisomes were present in the patient's tissues. Immunoblot studies of peroxisomal beta-oxidation enzymes revealed that the bifunctional enzyme (enoyl-CoA hydratase/3-hydroxyacyl-CoA dehydrogenase) was deficient in postmortem liver samples, whereas acyl-CoA oxidase and the mature form of beta-ketothiolase were present. Density gradient centrifugation of fibroblast homogenates confirmed that intact peroxisomes were present. Immunoblots of fibroblasts peroxisomal fractions showed that they contained acyl-CoA oxidase and beta-ketothiolase, but bifunctional enzyme was not detected. Northern analysis, however, revealed that mRNA coding for the bifunctional enzyme was present in the patient's fibroblasts. These results indicate that the primary biochemical defect in this patient is a deficiency of peroxisomal bifunctional enzyme. It is of interest that the phenotype of this patient resembled neonatal adrenoleukodystrophy and would not have been distinguished from this disorder by clinical study alone.


Subject(s)
3-Hydroxyacyl CoA Dehydrogenases/deficiency , Enoyl-CoA Hydratase/deficiency , Hydro-Lyases/deficiency , Isomerases , Microbodies/enzymology , Multienzyme Complexes/deficiency , 3-Hydroxyacyl CoA Dehydrogenases/genetics , Adrenoleukodystrophy , Brain/pathology , Cell Fractionation , Cells, Cultured , Cholic Acids/metabolism , Diagnosis, Differential , Enoyl-CoA Hydratase/genetics , Fatty Acids/blood , Fatty Acids/metabolism , Fibroblasts/analysis , Fibroblasts/metabolism , Fibroblasts/ultrastructure , Humans , Immunoblotting , Infant, Newborn , Liver/enzymology , Male , Microbodies/pathology , Multienzyme Complexes/genetics , Oxidation-Reduction , Peroxisomal Bifunctional Enzyme , RNA, Messenger/analysis
6.
Mol Cell Biol ; 23(2): 744-53, 2003 Jan.
Article in English | MEDLINE | ID: mdl-12509471

ABSTRACT

Peroxisomal disorders have been associated with malfunction of peroxisomal metabolic pathways, but the pathogenesis of these disorders is largely unknown. X-linked adrenoleukodystrophy (X-ALD) is associated with elevated levels of very-long-chain fatty acids (VLCFA; C(>22:0)) that have been attributed to reduced peroxisomal VLCFA beta-oxidation activity. Previously, our laboratory and others have reported elevated VLCFA levels and reduced peroxisomal VLCFA beta-oxidation in human and mouse X-ALD fibroblasts. In this study, we found normal levels of peroxisomal VLCFA beta-oxidation in tissues from ALD mice with elevated VLCFA levels. Treatment of ALD mice with pharmacological agents resulted in decreased VLCFA levels without a change in VLCFA beta-oxidation activity. These data indicate that ALDP does not determine the rate of VLCFA beta-oxidation and that VLCFA levels are not determined by the rate of VLCFA beta-oxidation. The rate of peroxisomal VLCFA beta-oxidation in human and mouse fibroblasts in vitro is affected by the rate of mitochondrial long-chain fatty acid beta-oxidation. We hypothesize that ALDP facilitates the interaction between peroxisomes and mitochondria, resulting, when ALDP is deficient in X-ALD, in increased VLCFA accumulation despite normal peroxisomal VLCFA beta-oxidation in ALD mouse tissues. In support of this hypothesis, mitochondrial structural abnormalities were observed in adrenal cortical cells of ALD mice.


Subject(s)
ATP-Binding Cassette Transporters/physiology , Adrenoleukodystrophy/genetics , Mitochondria , ATP Binding Cassette Transporter, Subfamily D, Member 1 , Adrenal Glands/ultrastructure , Animals , Cell Line , Cell Separation , Cells, Cultured , Fatty Acids/metabolism , Fibroblasts/metabolism , Flow Cytometry , Humans , Mice , Microscopy, Electron , Mitochondria/metabolism , Mutation , Oxygen/metabolism , Peroxisomes/metabolism , Time Factors , Tissue Distribution
7.
Biochim Biophys Acta ; 1214(3): 288-94, 1994 Oct 06.
Article in English | MEDLINE | ID: mdl-7918611

ABSTRACT

alpha-Oxidation of the branched-chain fatty acid, phytanic acid, is defective in patients with Refsum's disease, the disorders of peroxisome biogenesis (e.g., Zellweger syndrome), and in rhizomelic chondrodysplasia punctata. 3H-Release from [2,3-3H]phytanic acid, which is impaired in cultured skin fibroblasts from these patients, was investigated in rat liver peroxisomes. Cofactors necessary for optimal 3H-release, ATP, Mg2+, and coenzyme A, were also necessary for optimal acyl-CoA synthetase activity, suggesting that the substrate for 3H-release might be phytanoyl-CoA. 5,8,11,14-Eicosatetraynoic acid (ETYA), an inhibitor of long-chain acyl-CoA synthetase activity, blocked phytanoyl-CoA synthesis as well as 3H-release from [2,3-3H]phytanic acid in a dose-dependent manner. However, this inhibitor had little effect on 3H-release from [2,3-3H]phytanoyl-CoA. Tetradecylglycidic acid (TDGA) inhibited 3H-release from [2,3-3H]phytanic acid in peroxisomal but not in mitochondrial fractions from rat liver. This agent inhibited 3H-release from [2,3-3H]phytanic acid and [2,3-3H]phytanoyl-CoA equally. In contrast to ETYA, which appeared to decrease 3H-release as a consequence of synthetase inhibition, TDGA appeared to act directly on the enzyme catalyzing 3H-release. This enzyme was partially purified from rat liver. The purified enzyme, which did not possess phytanoyl-CoA synthetase activity, catalyzed tritium release from [2,3-3H]phytanoyl-CoA. This enzyme catalyzed 3H-release from [2,3-3H]phytanic acid only if a source of phytanoyl-CoA synthetase was present. We conclude that in rat liver peroxisomes, phytanic acid must be activated to its coenzyme A derivative prior to subsequent alpha-oxidation.


Subject(s)
Coenzyme A/metabolism , Liver/metabolism , Microbodies/metabolism , Phytanic Acid/analogs & derivatives , Phytanic Acid/metabolism , Repressor Proteins , Saccharomyces cerevisiae Proteins , 5,8,11,14-Eicosatetraynoic Acid/pharmacology , Animals , Coenzyme A/antagonists & inhibitors , Coenzyme A Ligases/antagonists & inhibitors , Coenzyme A Ligases/metabolism , Female , Liver/enzymology , Oxidation-Reduction , Phytanic Acid/antagonists & inhibitors , Protons , Rats , Tritium
8.
Biochim Biophys Acta ; 840(3): 401-8, 1985 Jul 05.
Article in English | MEDLINE | ID: mdl-3924116

ABSTRACT

125I-labeled 1-(p-hydroxyphenyl) 2-guanidinoethane (N-guanyltyramine), previously used to assay for the bacterial toxin choleragen (Mekalanos, J.J., Collier, R.J. and Romig, W.R. (1979) J. Biol. Chem. 254, 5849-5854) was utilized to identify NAD:arginine ADPribosyltransferases in animal tissues. The use of this radiolabelled ADPribose acceptor, rather than radiolabelled NAD, would bypass the problem posed by the almost ubiquitous presence of enzymes that degrade NAD. With a homogeneous ADPribosyltransferase from turkey erythrocytes, NAD and 125I-labeled guanyltyramine as ADPribose acceptor, formation of ADPribosyl 125I-guanyltyramine was linear with time and enzyme concentration. The product was indistinguishable on both thin-layer and high-performance liquid chromatography from that formed by choleragen. Using 125I-guanyltyramine, ADPribosyltransferase activity was also demonstrated in crude turkey erythrocyte cytosolic and membrane fractions. When rat liver was fractionated, apparent activity was detected primarily in the microsomes. The NAD-dependent product of the microsomal reaction was, however, distinguished from the turkey erythrocyte transferase product by thin-layer and DEAE-Sephadex chromatography; this product had a retention time identical to that of free 125I on high-performance liquid chromatography. In addition to NAD, the microsomal deiodinase activity was supported by NADH, NADP and NADPH. Phenyl boronate selectively bound ADPribosyl 125I-guanyltyramine and other metabolites of 125I-guanyltyramine which were formed by microsomes in a NAD-dependent process. These metabolites were distinguished from ADPribosyl 125I-guanyltyramine by high-performance liquid chromatography. These results indicate that in some cases, for example, turkey erythrocyte cytosolic and membrane fractions, 125I-guanyltyramine can be used to quantify ADPribosyltransferases in crude mixtures, whereas in others, for example, rat liver microsomes, high-performance liquid chromatographic analysis must be used to identify products.


Subject(s)
Pentosyltransferases/analysis , ADP Ribose Transferases , Adenosine Diphosphate Ribose , Animals , Erythrocytes/enzymology , Male , Microsomes, Liver/enzymology , NAD , NADP , Rats , Substrate Specificity , Turkeys , Tyramine/analogs & derivatives
9.
Exp Hematol ; 27(11): 1675-81, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10560915

ABSTRACT

Human bone marrow contains mesenchymal stem cells (MSCs) that can differentiate into various cells of mesenchymal origin. We developed an efficient method of isolating and culture expanding a homogenous population of MSCs from bone marrow and determined that MSCs express alpha-L-iduronidase, arylsulfatase-A and B, glucocerebrosidase, and adrenoleukodystrophy protein. These findings raised the possibility that MSCs may be useful in the treatment of storage disorders. To determine if donor derived MSCs are transferred to the recipients with lysosomal or peroxisomal storage diseases by allogeneic hematopoietic stem cell (HSC) transplantation, we investigated bone marrow derived MSCs of 13 patients 1-14 years after allogeneic transplantation. Highly purified MSCs were genotyped either by fluorescence in situ hybridization using probes for X and Y-chromosomes in gender mis-matched recipients or by radiolabeled PCR amplification of polymorphic simple sequence repeats. Phenotype was determined by the measurement of disease specific protein/enzyme activity in purified MSCs. We found that MSCs isolated from recipients of allogeneic HSC transplantation are not of donor genotype and have persistent phenotypic defects despite successful donor type hematopoietic engraftment. Whether culture expanded normal MSCs can be successfully transplanted into patients with storage diseases and provide therapeutic benefit needs to be determined.


Subject(s)
Hematopoietic Stem Cell Transplantation , Lysosomal Storage Diseases/therapy , Mesoderm/cytology , Peroxisomal Disorders/therapy , Case-Control Studies , Child , Child, Preschool , Humans , Infant , Phenotype , Polymorphism, Genetic , Transplantation, Homologous , Treatment Outcome
10.
Eur J Cell Biol ; 76(4): 237-45, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9765053

ABSTRACT

The mutant Chinese hamster ovary (CHO) cell line Z78/C has defective peroxisome assembly due to a missense mutation in PEX2, the gene which encodes the 35 kDa peroxisomal integral membrane protein. In humans, PEX2 mutations are responsible for complementation group 10 of the human peroxisome biogenesis disorders (PBD), a genetically heterogeneous group of lethal, autosomal recessive diseases including the Zellweger syndrome and related phenotypes. To develop additional cellular models for Zellweger syndrome, we produced a series of new mutant CHO cell clones in the same complementation group as Z78/C (Z2, Z7, Z22, and Z105). As expected, expression of human PEX2 restores peroxisomal biogenesis in all of these clones. Surprisingly, expression of the human 70 kDa peroxisomal membrane protein (PMP70) also restores peroxisome biogenesis in these same CHO cell clones. We confirmed this effect of PMP70 expression on peroxisome biogenesis by determining the subcellular latency of catalase, the immunohistochemical localization of catalase and the beta-oxidation of very long chain fatty acids (VLCFA). By contrast, expression of a mutant allele of PMP70 identified in a patient with Zellweger syndrome did not restore peroxisome biogenesis in the PEX2-deficient CHO cell clones. Our results indicate that overexpression of PMP70 suppresses the phenotype of PEX2 gene mutations. These observations suggest a functional interaction between PEX2 and PMP70 in the peroxisome membrane.


Subject(s)
ATP-Binding Cassette Transporters , Membrane Proteins/genetics , Microbodies/chemistry , Animals , Blotting, Northern , CHO Cells , Cloning, Molecular , Cricetinae , DNA, Complementary , Gene Expression Regulation, Enzymologic , Humans , Membrane Proteins/analysis , Membrane Proteins/metabolism , Microbodies/genetics , Mutagenesis , Oxidation-Reduction , Peroxisomal Biogenesis Factor 2 , Phenotype , Protein Binding/physiology , RNA, Messenger/analysis
11.
Endocrinology ; 131(6): 2839-45, 1992 Dec.
Article in English | MEDLINE | ID: mdl-1446622

ABSTRACT

We investigated the effects of 8 days of LH withdrawal on rat Leydig cell peroxisomal volume, total and intraperoxisomal catalase and sterol carrier protein-2 (SCP2) contents, and LH-stimulated testosterone secretion in vitro. Three groups of adult male Sprague-Dawley rats, i.e. control, TE-implanted (testosterone-17 beta-estradiol-filled Silastic implants to suppress LH), and TELH-implanted (TE-implanted and LH replacement via Alzet mini osmotic pumps), were used. After 8 days, Leydig cell organelle volumes (stereology), intraperoxisomal catalase and SCP2 contents (immunocytochemistry), LH-stimulated testosterone secretion by isolated Leydig cells in vitro (determined by RIA), and total catalase and SCP2 contents in equal numbers of Leydig cells (immunoblot analyses) were determined. Results showed that the TELH-implanted rats were identical to controls in every parameter tested. Testis volume and Leydig cell number per testis in control and TE-implanted rats were not significantly different; however, reductions (P < 0.05) were observed in the average volume of a Leydig cell (one third of controls) and the volume of Leydig cells per testis. All Leydig cell organelle volumes tested were significantly lower in TE-implanted rats than in the controls; however, the volumes of smooth endoplasmic reticulum (SER) and peroxisomes were the most reduced (lowered to one sixth of control values). LH-stimulated testosterone secretion per Leydig cell in vitro correlated well with these changes in the volumes of Leydig cell SER and peroxisomes. Intraperoxisomal catalase in Leydig cells was unchanged in TE-implanted rats, although immunoblotting demonstrated a loss of total catalase content (which reflected the reduction in the volume of peroxisomes). SCP2 in Leydig cells of TE-implanted rats was undetectable with immunoblot analysis (explained by the reductions in Leydig cell peroxisome volume and intraperoxisomal SCP2). These results demonstrate that the organelles SER and peroxisomes and the protein SCP2 in Leydig cells are more LH dependent than the other organelles (e.g. mitochondria, lysosomes) and protein catalase, respectively. Moreover, the findings of this study are consistent with the hypothesis that Leydig cell peroxisomes play a significant role in testosterone production.


Subject(s)
Carrier Proteins/metabolism , Leydig Cells/metabolism , Leydig Cells/ultrastructure , Luteinizing Hormone/administration & dosage , Microbodies/ultrastructure , Plant Proteins , Sterols/metabolism , Animals , Catalase/metabolism , Estradiol/pharmacology , Immunoblotting , Leydig Cells/drug effects , Luteinizing Hormone/pharmacology , Male , Microbodies/enzymology , Organelles/drug effects , Organelles/ultrastructure , Rats , Rats, Sprague-Dawley , Testis/anatomy & histology , Testis/drug effects , Testosterone/metabolism , Testosterone/pharmacology
12.
Endocrinology ; 127(6): 2947-54, 1990 Dec.
Article in English | MEDLINE | ID: mdl-2249635

ABSTRACT

The aim of the present study was to investigate the effects of a single injection of LH on rat Leydig cell peroxisome volume and peroxisomal sterol carrier protein-2 (SCP2) content. Sexually mature Sprague-Dawley rats (n = 5) were injected sc with 500 micrograms LH and euthanized, and trunk blood was collected at 0, 0.5, 1, 2, and 3 h. Additionally, LH-treated rats were whole body perfused-fixed, and their testes were processed for qualitative and quantitative histochemical and immunocytochemical studies at 0, 0.5, 1, and 2 h. Peroxisomes were identified by cytochemical staining for catalase activity with the alkaline 3,3'-diaminobenzidine tetrahydrochloride method. Catalase and SCP2 were immunolocalized in Leydig cell organelles via 10-nm AuroProbe EM protein-A gold particles. Peak plasma testosterone concentrations were observed 1 and 2 h after the single sc LH injection. The average volume of a Leydig cell was unchanged by the LH treatment at all time points tested. Similarly, the absolute volumes of smooth endoplasmic reticulum and mitochondria per Leydig cell were unchanged at all time points tested. By contrast, the absolute volume of peroxisomes per Leydig cell increased 3-fold 0.5 h after LH injection (P less than 0.01) and then returned to control values by 2 h. The absolute volume of negative bodies (single membrane-bound cytoplasmic organelles lacking catalase) per Leydig cell was elevated above the control value 0.5 and 1 h after LH injection. Western blot analysis demonstrated a single protein at 14 and 60 kDa with anti-SCP2 and anticatalase, respectively, for both homogenates obtained from liver and purified Leydig cells. Quantitative immunocytochemical studies demonstrated that the gold particle density representing SCP2 over peroxisomes increased 5-fold 0.5 h after the LH injection (P less than 0.01) and then returned to control values by 2 h. In contrast, the gold particle density representing catalase over peroxisomes was not different in control and LH-injected groups. We conclude that a single sc injection of LH causes a rapid, specific, and transient increase in both the volume of peroxisomes and the peroxisomal content of SCP2 in Leydig cells.


Subject(s)
Carrier Proteins/metabolism , Leydig Cells/ultrastructure , Luteinizing Hormone/pharmacology , Microbodies/ultrastructure , Plant Proteins , Sterols/metabolism , Animals , Antibodies , Carrier Proteins/isolation & purification , Immunoblotting , Kinetics , Leydig Cells/drug effects , Leydig Cells/metabolism , Male , Microbodies/drug effects , Microbodies/metabolism , Microscopy, Electron , Molecular Weight , Rats , Rats, Inbred Strains , Reference Values , Testosterone/blood , Testosterone/metabolism
13.
FEBS Lett ; 481(3): 240-4, 2000 Sep 22.
Article in English | MEDLINE | ID: mdl-11007971

ABSTRACT

Salinity and hyperosmotic stress are environmental factors that severely affect the growth and development of plants. Adaptation to these stresses is known to be a complex multistep process, but a rise in cytoplasmic Ca(2+) and increased polyphosphoinositide turnover have now been identified as being amongst the early events leading to the development of tolerance. To determine whether a causal link exists between these two events we have investigated the effects of several salts and osmotic agents on levels of inositol(1, 4,5)trisphosphate (Ins(1,4,5)P(3)) in plant cells. Our data show that salts as well as osmotic agents induce a rapid and up to 15-fold increase in cellular Ins(1,4,5)P(3) levels. The increase in Ins(1,4,5)P(3) occurs in a dose-dependent manner and levels remain elevated for at least 10 min. These data indicate that increased Ins(1,4,5)P(3) production is a common response to salt and hyperosmotic stresses in plants and that it may play an important role in the processes leading to stress tolerance.


Subject(s)
Daucus carota/metabolism , Inositol 1,4,5-Trisphosphate/biosynthesis , Sodium Chloride/pharmacology , Adaptation, Physiological/drug effects , Adaptation, Physiological/physiology , Calcium/physiology , Cell Fractionation , Cell Membrane/drug effects , Cell Membrane/enzymology , Cell Membrane/physiology , Daucus carota/drug effects , Daucus carota/enzymology , Daucus carota/physiology , Enzyme Activation/drug effects , Hydrogen-Ion Concentration , Inositol 1,4,5-Trisphosphate/physiology , Osmolar Concentration , Osmotic Pressure/drug effects , Type C Phospholipases/metabolism
14.
FEBS Lett ; 476(3): 145-9, 2000 Jul 07.
Article in English | MEDLINE | ID: mdl-10913602

ABSTRACT

Localised alterations in cytoplasmic Ca(2+) levels are an integral part of the response of eukaryotic cells to a plethora of external stimuli. Due to the large size of nuclear pores, it has generally been assumed that intranuclear Ca(2+) levels reflect the prevailing cytoplasmic Ca(2+) levels. Using nuclei prepared from carrot (Daucus carota L.) cells, we now show that Ca(2+) can be transported across nuclear membranes in an ATP-dependent manner and that over 95% of Ca(2+) is accumulated into a pool releasable by the Ca(2+) ionophore A.23187. ATP-dependent nuclear Ca(2+) uptake did not occur in the presence of ADP or ADPgammaS and was abolished by orthovanadate. Confocal microscopy of nuclei loaded with dextran-linked Indo-1 showed that the initial ATP-induced rise in [Ca(2+)] occurs in the nuclear periphery. The occurrence of ATP-dependent Ca(2+) uptake in plant nuclei suggests that alterations of intranuclear Ca(2+) levels may occur independently of cytoplasmic [Ca(2+)] changes.


Subject(s)
Adenosine Triphosphate/metabolism , Calcium/metabolism , Cell Nucleus/metabolism , Plants/metabolism , Biological Transport, Active , Cytoplasm/metabolism , Daucus carota/metabolism , Kinetics , Microscopy, Confocal , Nuclear Envelope/metabolism
15.
Neurology ; 36(3): 357-61, 1986 Mar.
Article in English | MEDLINE | ID: mdl-3951702

ABSTRACT

Monounsaturated fatty acids, especially oleic acid (C18:1), decreased the content of saturated very-long-chain fatty acids (VLFA) in cultured skin fibroblasts from patients with adrenoleukodystrophy (ALD) and controls. When confluent ALD fibroblasts were incubated with oleic acid for 5 days in lipid-free medium, which eliminates uptake of exogenous VLFA, the mean cell content of C26:0 was decreased by 33.7 +/- 10.1%. Oleic acid inhibited C26:0 synthesis in ALD fibroblasts by 58% and total fatty acid synthesis by 68 to 78%. Therefore, the elevated C26:0 levels in ALD cells may be lowered by inhibiting fatty acid biosynthesis, and inhibition of saturated VLFA synthesis by oleate may be useful in treating ALD.


Subject(s)
Adrenoleukodystrophy/metabolism , Diffuse Cerebral Sclerosis of Schilder/metabolism , Fatty Acids/metabolism , Fibroblasts/metabolism , Oleic Acids/pharmacology , Animals , Cattle , Cell Line , Female , Humans , In Vitro Techniques , Male , Oleic Acid , Skin/metabolism
16.
Neurology ; 38(7): 1100-7, 1988 Jul.
Article in English | MEDLINE | ID: mdl-3386829

ABSTRACT

Neonatal hypotonia, seizures beginning at 5 days, and severe retardation were noted in a girl with normal karyotype and biochemical evidence of impaired adrenal function. Postmortem examination at 14 months revealed malformative and destructive lesions of central gray and white matter, atrophy of adrenal cortex with striated adrenocortical cells, hepatic fibrosis, and PAS-positive macrophages in several organs. Pathologically and clinically, this patient most closely approximated neonatal adrenoleukodystrophy (ALD) and differed strikingly from X-linked childhood ALD. In contrast, biochemical changes resembled the abnormalities observed in X-linked ALD and differed from those in the neonatal form. The very-long-chain fatty acid accumulation characteristic of both disorders was demonstrated, but unlike neonatal ALD, the levels or metabolism of plasmalogens, pipecolic acid, phytanic acid, and bile acid intermediates were normal, and peroxisomes in a liver biopsy specimen were present in normal number and appeared enlarged. While the case resembles the recently reported entity of peroxisomal acyl-CoA oxidase deficiency, assignment to this category was excluded by immunoblot studies on postmortem liver, which revealed normal amounts of this enzyme. Correlation of clinical, morphologic, and biochemical data suggests that this case is an example of a so-far undescribed entity, and reinforces the concept that the phenotypic spectrum of peroxisomal disorders is wider than realized.


Subject(s)
Adrenoleukodystrophy/genetics , Diffuse Cerebral Sclerosis of Schilder/genetics , Fatty Acids/metabolism , Intellectual Disability/physiopathology , Liver/ultrastructure , Microbodies/ultrastructure , Seizures/physiopathology , X Chromosome , Adrenoleukodystrophy/complications , Adrenoleukodystrophy/physiopathology , Catalase/metabolism , Cells, Cultured , Fatty Acids/blood , Female , Humans , Infant, Newborn , Intellectual Disability/complications , Liver/metabolism , Liver/pathology , Microbodies/metabolism , Plasmalogens/biosynthesis , Reference Values , Seizures/complications , Seizures/congenital , Skin/metabolism
17.
Cell Biochem Biophys ; 32 Spring: 333-7, 2000.
Article in English | MEDLINE | ID: mdl-11330068

ABSTRACT

X-linked adrenoleukodystrophy (X-ALD) is characterized biochemically by elevated levels of saturated very long-chain fatty acids (VLCFAs) in plasma and tissues. In X-ALD, peroxisomal very-long-chain acyl-CoA synthetase (VLCS) fails to activate VLCFAs, preventing their degradation via beta-oxidation. However, the product of the defective XALD gene (ALDP) is not a VLCS, but rather a peroxisomal membrane protein (PMP). Disruption of either or both of two yeast PMP genes related to the XALD gene did not produce a biochemical phenotype resembling that found in X-ALD fibroblasts. The authors identified a candidate yeast VLCS gene (the FAT1 locus) by its homology to rat liver VLCS. Disruption of this gene decreased VLCS activity, but had no effect on long-chain acyl-CoA synthetase activity. In FAT1-disruption strains, VLCS activity was reduced to 30-40% of wild-type in both a microsome-rich 27,000 g supernatant fraction and a peroxisome- and mitochondria-rich pellet fraction of yeast spheroplast homogenates. Separation of the latter organelles by density gradient centrifugation revealed that VLCS activity was peroxisomal and not mitochondrial. VLCS gene-disruption strains had increased cellular VLCFA levels, compared to wild-type yeast. The extent of both the decrease in peroxisomal VLCS activity and the VLCFA accumulation in this yeast model resembles that observed in cells from X-ALD patients. Characterization of the gene(s) responsible for the residual peroxisomal VLCS activity may suggest new therapeutic approaches in X-ALD.


Subject(s)
Adrenoleukodystrophy/genetics , Coenzyme A Ligases/genetics , Saccharomyces cerevisiae/genetics , Adrenoleukodystrophy/etiology , Coenzyme A Ligases/metabolism , Gene Deletion , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Fungal , Genes, Fungal , Humans
18.
J Mol Neurosci ; 16(2-3): 333-5, 2001.
Article in English | MEDLINE | ID: mdl-11478387

ABSTRACT

A primary goal of the international workshop "Brain Uptake and Utilization of Fatty Acids" was to identify research areas that would benefit from further investigation. The major themes for future research are presented below: (1) Elucidating the role of the developing and mature cerebrovascular endothelium (CVE) in the uptake of fatty acids (FA) into the brain. (2) Clarifying the role of diffusion and receptor-mediated uptake of FAs by various brain cell membranes and protein-mediated shuttling of FAs between the CVE and various brain cells and tissues. (3) Illuminating the mechanisms of intermediate metabolism and the roles of polyunsaturated fatty acids (PUFA) in astrocytes, neurons and oligodendrocytes. Of special interest are the long-chain omega-3 PUFA and their derivatives, such as lipoproteins, phospholipids and plasmalogens, that have been associated with various disease states (such as those listed in [5], below). (4) Elucidating the role of gene expression on long-chain omega-3 PUFA incorporation in membranes and the regulatory role these and other PUFA have on gene expression in the brain. (5) Elucidating the recently identified roles of long-chain omega-3 PUFA in mood disorders, schizophrenia, stroke, peroxisomal biogenesis disorders, Huntington's disease, other neurodegenerative disorders and disorders of oxidative stress. (6) Undertaking placebo-controlled clinical trials to assess the therapeutic potential of omega-3 PUFA in the above disorders. (7) Developing new, and utilizing existing animal models in the above studies. (8) Developing noninvasive imaging and tagging methods for quantifying the migration and distribution of PUFA and their derivatives in the brain. (9) Applying multi-disciplinary collaborations among biophysicists, physiologists and molecular biologists to the resolution of the above.


Subject(s)
Brain/metabolism , Fatty Acids/metabolism , Animals , Biological Transport , Carrier Proteins/metabolism , Cell Membrane/metabolism , Docosahexaenoic Acids/metabolism , Fatty Acids, Unsaturated/metabolism , Forecasting , Humans , Membrane Lipids/metabolism , Mice , Models, Animal , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Peroxisomes/metabolism , Rats , Research , Zellweger Syndrome/metabolism
19.
J Mol Neurosci ; 16(2-3): 87-92; discussion 151-7, 2001.
Article in English | MEDLINE | ID: mdl-11478388

ABSTRACT

The brain is rich in diverse fatty acids saturated, monounsaturated and polyunsaturated fatty acids with chain lengths ranging from less than 16 to more than 24 carbons that make up the complex lipids present in this organ. While some fatty acids are derived from endogenous synthesis, others must come from exogenous sources. The mechanism(s) by which fatty acids enter cells has been the subject of much debate. While some investigators argue for a protein-mediated process, others suggest that simple diffusion is sufficient. In the brain, uptake is further complicated by the presence of the blood-brain barrier. Brain fatty acid homeostasis is disturbed in many human disorders, as typified by the peroxisomal biogenesis diseases. A workshop designed to bring together researchers from varied backgrounds to discuss these issues in an open forum was held in March, 2000. In addition to assessing the current state of knowledge, areas requiring additional investigation were identified and recommendations for future research were made. A brief overview of the invited talks is presented here.


Subject(s)
Brain/metabolism , Fatty Acids/metabolism , Animals , Dietary Fats/pharmacokinetics , Docosahexaenoic Acids/metabolism , Energy Metabolism , Fatty Acids/pharmacokinetics , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-6 , Fatty Acids, Unsaturated/metabolism , Fatty Acids, Unsaturated/pharmacokinetics , Humans , Membrane Lipids/metabolism , Neurons/metabolism , Peroxisomal Disorders/metabolism , Peroxisomal Disorders/pathology , Peroxisomes/metabolism , Peroxisomes/pathology , Zellweger Syndrome/metabolism
20.
Am J Med Genet ; 57(2): 239-45, 1995 Jun 05.
Article in English | MEDLINE | ID: mdl-7668338

ABSTRACT

The neuronal ceroid-lipofuscinoses (NCL), also known as Batten disease, are a not uncommon group of disorders affecting infants, children, and young adults. The abnormal ultrastructural profiles seen in NCL are used for standard diagnosis; however, they can be missed, and are also found in other neurodegenerative conditions. Furthermore, there is an overlap between the types of inclusion profiles among the different forms of NCL. Therefore, a more specific and biochemically-based marker is necessary to confirm the diagnosis of NCL. Antibodies raised against the storage material from the ovine form of NCL (mitochondrial ATP synthase subunit c) were utilized to determine whether NCL could be distinguished from other metabolic-neurodegenerative disorders. By immunoblotting and immunohistochemistry, several brain samples of well-evaluated NCL cases confirmed increased accumulations in all NCL cases except in the brain of an infantile-onset NCL patient. The immunoblot studies of skin fibroblasts and brain were sensitive but not highly specific to NCL, due to the recognition of this material in normal controls as well as in other neurogenetic diseases. Immunocytochemistry of skin fibroblasts clearly distinguished LINCL and JNCL cases from controls, and with further refinement has the potential for becoming a diagnostic tool.


Subject(s)
Brain/pathology , Neuronal Ceroid-Lipofuscinoses/diagnosis , Proton-Translocating ATPases/analysis , Adult , Animals , Brain/enzymology , Child , Diagnosis, Differential , Fibroblasts/enzymology , Fibroblasts/pathology , Humans , Immunoblotting , Immunoenzyme Techniques , Immunohistochemistry , Infant , Neuronal Ceroid-Lipofuscinoses/enzymology , Neuronal Ceroid-Lipofuscinoses/pathology , Sheep , Skin/enzymology , Skin/pathology
SELECTION OF CITATIONS
SEARCH DETAIL