Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 19 de 19
1.
Mol Cell Biol ; 23(2): 744-53, 2003 Jan.
Article En | MEDLINE | ID: mdl-12509471

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.


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
2.
Mol Genet Metab ; 74(1-2): 256-63, 2001.
Article En | MEDLINE | ID: mdl-11592822

X-linked adrenoleukodystrophy (X-ALD) is an inherited neurometabolic disorder associated with elevated levels of saturated unbranched very-long-chain fatty acids (VLCFA; C > 22:0) in plasma and tissues, and reduced VLCFA beta-oxidation in fibroblasts, white blood cells, and amniocytes from X-ALD patients. The X-ALD gene (ABCD1) at Xq28 encodes the adrenoleukodystrophy protein (ALDP) that is related to the peroxisomal ATP-binding cassette (ABCD) transmembrane half-transporter proteins. The function of ALDP is unknown and its role in VLCFA accumulation unresolved. Previously, our laboratory has shown that sodium 4-phenylbutyrate (4PBA) treatment of X-ALD fibroblasts results in increased peroxisomal VLCFA beta-oxidation activity and increased expression of the X-ALD-related protein, ALDRP, encoded by the ABCD2 gene. In this study, the effect of various pharmacological agents on VLCFA beta-oxidation in ALD mouse fibroblasts is tested. 4PBA, styrylacetate and benzyloxyacetate (structurally related to 4PBA), and trichostatin A (functionally related to 4PBA) increase both VLCFA (peroxisomal) and long-chain fatty acid [LCFA (peroxisomal and mitochondrial)] beta-oxidation. Isobutyrate, zaprinast, hydroxyurea, and 5-azacytidine had no effect on VLCFA or LCFA beta-oxidation. Lovastatin had no effect on fatty acid beta-oxidation under normal tissue culture conditions but did result in an increase in both VLCFA and LCFA beta-oxidation when ALD mouse fibroblasts were cultured in the absence of cholesterol. The effect of trichostatin A on peroxisomal VLCFA beta-oxidation is shown to be independent of an increase in ALDRP expression, suggesting that correction of the biochemical abnormality in X-ALD is not dependent on pharmacological induction of a redundant gene (ABCD2). These studies contribute to a better understanding of the role of ALDP in VLCFA accumulation and may lead to the development of more effective pharmacological therapies.


Adrenoleukodystrophy/genetics , Adrenoleukodystrophy/metabolism , Fatty Acids, Nonesterified/metabolism , Lipid Peroxidation/genetics , X Chromosome/genetics , Adrenoleukodystrophy/drug therapy , Adrenoleukodystrophy/enzymology , Animals , Cell Line , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Humans , Hydroxamic Acids/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lipid Peroxidation/drug effects , Lovastatin/pharmacology , Mice , Phenylbutyrates/pharmacology
3.
Expert Opin Investig Drugs ; 9(9): 1985-92, 2000 Sep.
Article En | MEDLINE | ID: mdl-11060787

Clinically, peroxisome biogenesis disorders (PBDs) are a group of lethal diseases with a continuum of severity of clinical symptoms ranging from the most severe form, Zellweger syndrome, to the milder forms, infantile Refsum disease and rhizomelic chondrodysplasia punctata. PBDs are characterised by a number of biochemical abnormalities including impaired degradation of peroxide, very long chain fatty acids, pipecolic acid, phytanic acid and xenobiotics and impaired synthesis of plasmalogens, bile acids, cholesterol and docosahexaenoic acid. Treatment of PBD patients as a group is problematic since a number of patients, especially those with Zellweger syndrome, have significant neocortical alterations in the brain at birth so that full recovery would be impossible even with postnatal therapy. To date, treatment of PBD patients has generally involved only supportive care and symptomatic therapy. However, the fact that some of the milder PBD patients live into the second decade has prompted research into possible treatments for these patients. A number of experimental therapies have been evaluated to determine whether or not correction of biochemical abnormalities through dietary supplementation and/or modification is of clinical benefit to PBD patients. Another approach has been pharmacological induction of peroxisomes in PBD patients to improve overall peroxisomal biochemical function. Well known rodent peroxisomal proliferators were found not to induce human peroxisomes. Recently, our laboratory demonstrated that sodium 4-phenylbutyrate induces peroxisome proliferation and improves biochemical function (very long chain fatty acid beta-oxidation rates and very long chain fatty acid and plasmalogens levels) in fibroblast cell lines from patients with milder PBD phenotypes. Dietary supplementation and/or modification and pharmacological induction of peroxisomes as treatment strategies for PBD patients will be the subject of this review.


Antineoplastic Agents/pharmacology , Dietary Supplements , Erucic Acids/therapeutic use , Peroxisomal Disorders/drug therapy , Phenylbutyrates/pharmacology , Triolein/therapeutic use , Zellweger Syndrome/drug therapy , Drug Combinations , Humans , Peroxisomal Disorders/diet therapy , Peroxisomal Disorders/therapy , Zellweger Syndrome/diet therapy
4.
Ann Neurol ; 47(3): 286-96, 2000 Mar.
Article En | MEDLINE | ID: mdl-10716247

Inherited aberrant peroxisome assembly results in a group of neurological diseases termed peroxisome biogenesis disorders (PBDs). PBDs include three major clinical phenotypes that represent a continuum of clinical features from the most severe form, Zellweger syndrome (ZS), through neonatal adrenoleukodystrophy (NALD) to the least severe form, infantile Refsum's disease (IRD). Somatic cell complementation studies have identified 13 PBD complementation groups, each representing a defect in a peroxisomal protein (peroxin) involved in peroxisome biogenesis. Most complementation groups include a range of clinical phenotypes. In this study, peroxisome numbers were determined in fibroblasts from 29 PBD (ZS, NALD, and IRD) patients, with various phenotypes from nine complementation groups, using antibodies against either a peroxisomal membrane protein (anti-Pex14p) or peroxisomal matrix proteins (anti-SKL). A correlation between the number of peroxisomes, determined with either antibody, and PBD phenotype was found, suggesting that induction of peroxisome number might have a favorable effect on PBD. After treatment of PBD fibroblasts with sodium 4-phenylbutyrate, a human peroxisome proliferator, there was an approximate twofold increase in peroxisome number. After 4-phenylbutyrate treatment, an increase in transcription of the adrenoleukodystrophy-related gene and the peroxin gene, PEX11alpha, was found in PBD fibroblasts. In NALD and IRD, but not ZS, fibroblasts there was an increase in very-long-chain fatty acid beta-oxidation and plasmalogen concentrations, and a decrease in very-long-chain fatty acid concentrations. These data suggest that pharmacological agents that induce peroxisome proliferation, such as 4-phenylbutyrate, may have therapeutic potential in the treatment of PBD patients with milder phenotypes (NALD and IRD).


Peroxisomal Disorders/genetics , Peroxisomes/genetics , Cells, Cultured , Child , Child, Preschool , Fibroblasts/pathology , Humans , Infant , Peroxisomal Disorders/pathology , Phenotype , Polymerase Chain Reaction , RNA, Messenger/analysis
5.
Arch Immunol Ther Exp (Warsz) ; 47(5): 281-7, 1999.
Article En | MEDLINE | ID: mdl-10604233

X-linked adrenoleukodystrophy (X-ALD) is an inherited neurodegenerative disease that affects approximately 1 in 25 000 males. It is characterized by elevated levels of saturated very long chain fatty acids (VLCFA), i.e., >C22:0, particularly in ganglioside and cholesterol ester fractions of brain white matter and adrenal cortex. Failure of peroxisomal very long chain fatty acyl-CoA synthetase (VLCS) to activate these VLCFA prevents their degradation by peroxisomal beta-oxidation. X-ALD maps to Xq28 and the gene encodes a peroxisomal membrane protein and not the gene for VLCS. The two most common forms of X-ALD are the cerebral (CER) form, with an inflammatory demyelinating reaction that resembles multiple sclerosis (MS), and adrenomyeloneuropathy (AMN), which involves the spinal cord and in which the inflammatory reaction is mild or absent. Investigations into the nature of the cerebral inflammatory demyelinating reaction in X-ALD will be the subject of this review.


ATP-Binding Cassette Transporters/physiology , Adrenoleukodystrophy/pathology , Brain/pathology , Demyelinating Diseases/pathology , Membrane Proteins/physiology , ATP Binding Cassette Transporter, Subfamily D, Member 1 , ATP-Binding Cassette Transporters/genetics , Adrenoleukodystrophy/epidemiology , Adrenoleukodystrophy/genetics , Adrenoleukodystrophy/metabolism , Age of Onset , Brain Chemistry , Child , Cytokines/physiology , Demyelinating Diseases/epidemiology , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Fatty Acids/metabolism , Genetic Heterogeneity , Genetic Predisposition to Disease , Genotype , HLA Antigens/genetics , Humans , Major Histocompatibility Complex , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Peroxisomes/chemistry , Peroxisomes/enzymology , Phenotype , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/physiology , X Chromosome
6.
Mol Genet Metab ; 68(1): 32-42, 1999 Sep.
Article En | MEDLINE | ID: mdl-10479480

Activation of fatty acids, catalyzed by acyl-coenzyme A (acyl-CoA) synthetases, is required for their subsequent metabolism. Peroxisomes and microsomes contain very-long-chain acyl-CoA synthetases (VLCSs) capable of activating fatty acids with a chain length of 22 or more carbons. Decreased peroxisomal VLCS activity is, in part, responsible for the biochemical pathology in X-linked adrenoleukodystrophy (X-ALD), illustrating the importance of VLCSs in cellular fatty acid homeostasis. We previously cloned two human genes encoding proteins homologous to rat peroxisomal VLCS; one (hVLCS) is the human ortholog to the rat VLCS gene and another (hVLCS-H1) encodes a related heart-specific protein. Here, we report the cloning of a third gene (hVLCS-H2) and characterization of its protein product. The hVLCS-H2 gene is located on human chromosome 19 and encodes a 690-amino-acid protein. The amino acid sequence of hVLCS-H2 is 44-45% identical and 67-69% similar to those of both hVLCS and hVLCS-H1. COS-1 cells transiently overexpressing hVLCS-H2 activated the very-long-chain fatty acid lignocerate (C24:0) at a rate >1.5-fold higher than that of nontransfected cells (P < 0.002). The hVLCS-H2-dependent activation of long- and branched-chain fatty acids following transient transfection was less striking. However, hVLCS-H2-dependent acyl-CoA synthetase activity with long- and very-long-chain fatty acid substrates was detected in COS-1 cells stably expressing hVLCS-H2. For all substrates tested (C18:0, C20:0, C24:0, C26:0), the hVLCS-H2 catalyzed activity was significantly increased (P < 0.01 to P < 0.0001). By both Northern analysis and reverse transcription polymerase chain reaction, hVLCS-H2 is expressed primarily in liver. Indirect immunofluorescence of COS-1 cells or human hepatoma-derived HepG2 cells expressing epitope-tagged hVLCS-H2 revealed that the protein was associated with the endoplasmic reticulum but not with peroxisomes. Thus, the primary role of hVLCS-H2 is likely to be in fatty acid elongation or complex lipid synthesis rather than in degradation.


Coenzyme A Ligases/genetics , Liver/enzymology , Repressor Proteins , Saccharomyces cerevisiae Proteins , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , COS Cells , Chromosome Mapping , Chromosomes, Human, Pair 19/genetics , Cloning, Molecular , Coenzyme A Ligases/metabolism , DNA, Complementary/chemistry , DNA, Complementary/genetics , Endoplasmic Reticulum/chemistry , Fluorescent Antibody Technique, Indirect , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Microbodies/chemistry , Molecular Sequence Data , RNA/genetics , RNA/metabolism , Sequence Analysis, DNA , Tissue Distribution , Tumor Cells, Cultured
7.
Nat Med ; 4(11): 1261-8, 1998 Nov.
Article En | MEDLINE | ID: mdl-9809549

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.


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
8.
Hum Mol Genet ; 7(2): 239-47, 1998 Feb.
Article En | MEDLINE | ID: mdl-9425230

X-Linked adrenoleukodystrophy (X-ALD) is a neurodegenerative disorder characterized by reduced peroxisomal very long chain fatty acid (VLCFA) beta-oxidation. The X - ALD gene product (ALDP) is a peroxisomal transmembrane protein with an ATP binding cassette (ABC). ALDP and three other ABC proteins (PMP70, ALDR, P70R) localize to the peroxisomal membrane. The function of this family of peroxisomal membrane proteins is unknown. We used complementation studies to begin analysis of their role in VLCFA beta-oxidation and on the peroxisomal membrane. Expression of either ALDP or PMP70 restores VLCFA beta-oxidation in X-ALD fibroblasts, indicating overlapping functions. Their expression also restores peroxisome biogenesis in cells that are deficient in the peroxisomal membrane protein Pex2p. Thus it is likely that complex protein interactions are involved in the function and biogenesis of peroxisomal membranes that may contribute to disease heterogeneity.


ATP-Binding Cassette Transporters/physiology , Adrenoleukodystrophy/metabolism , Membrane Proteins/deficiency , Membrane Proteins/physiology , Microbodies/metabolism , ATP Binding Cassette Transporter, Subfamily D, Member 1 , ATP-Binding Cassette Transporters/genetics , Adrenoleukodystrophy/genetics , Adrenoleukodystrophy/pathology , Cells, Cultured , Cosmids , Fatty Acids/metabolism , Fibroblasts/metabolism , Genetic Complementation Test , Humans , Male , Membrane Proteins/genetics , Oxidation-Reduction , Peroxisomal Biogenesis Factor 2 , X Chromosome/genetics
9.
J Neuroimmunol ; 75(1-2): 174-82, 1997 May.
Article En | MEDLINE | ID: mdl-9143252

The two most common forms of X-linked adrenoleukodystrophy (X-ALD) are the cerebral forms (CER) with an inflammatory demyelinating reaction that resembles multiple sclerosis, and adrenomyeloneuropathy (AMN) which involves primarily the spinal cord and in which the inflammatory reaction is mild or absent. We found no significant association between the childhood cerebral form (CCER) or AMN and the human leukocyte (HLA) class I and Class II antigens including the class II DR2 haplotypes associated with multiple sclerosis. Inflammatory cytokine (tumor necrosis factor-alpha, interleukin-1 beta, interleukin-4, interleukin-6 and interferon-gamma) gene expression was increased in multiple sclerosis brain lesions, as has been reported previously, but much less so in CER brain lesions. These findings suggest that the pathogenesis of the inflammatory response in X-ALD differs from that in multiple sclerosis.


Adrenoleukodystrophy/metabolism , Cytokines/metabolism , Demyelinating Diseases/metabolism , Encephalitis/metabolism , HLA Antigens/metabolism , Multiple Sclerosis/metabolism , X Chromosome , Adrenoleukodystrophy/genetics , Blood Cells/immunology , HLA Antigens/classification , Humans , Lymphocytes/immunology , Tissue Distribution , Tumor Necrosis Factor-alpha/metabolism
10.
Mol Endocrinol ; 9(10): 1380-95, 1995 Oct.
Article En | MEDLINE | ID: mdl-8544846

Rat insulin-like growth factor I (IGF-I) mRNAs contain multiple 5'-untranslated regions due to the use of leader exons transcribed from several transcription initiation sites and to alternative splicing within leader exon 1. Synthetic RNAs with 5'-ends corresponding to the use of exon 1 transcription initiation sites were translated in vitro into prepro-IGF-I peptides initiated at a Met-48 codon in exon 1 or a Met-22 codon in exon 3, and RNAs with a 5'-end corresponding to the major exon 2 transcription start site were translated into a prepro-IGF-I peptide initiated at a Met-32 codon in exon 2. All forms of prepro-IGF-I were processed by canine pancreatic microsomes, suggesting that all these prepeptides function as signal peptides. The translational efficiency of IGF-I RNAs was inversely proportional to the length of the 5'-untranslated region. Mutation of the first of three upstream AUG codons in exon 1, which potentially initiates a 14-amino acid open reading frame, did not affect prepro-IGF-I translation. The other two AUG codons are immediately followed by stop codons. The absence of both upstream AUG codons in a completely spliced exon 1-derived RNA enhanced the in vitro and in vivo translatability of this RNA as compared with the full-length RNA. Mutation of the downstream initiation codon in particular increased translational efficiency in vitro and in intact cells, suggesting that an inefficient reinitiation event at the Met-48 codon contributes to the poorer translation of IGF-I mRNAs in which these upstream AUGUGA motifs occur. We conclude that IGF-I mRNAs potentially encode multiple forms of preproIGF and that specific differences in their 5'-untranslated regions provide a molecular basis for translational control of IGF-I biosynthesis.


Insulin-Like Growth Factor I/genetics , Protein Biosynthesis , Protein Sorting Signals/genetics , RNA, Messenger/metabolism , Animals , Base Sequence , Dogs , Insulin-Like Growth Factor I/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Sorting Signals/metabolism , Rats , Sequence Analysis
11.
J Neuroimmunol ; 61(2): 161-9, 1995 Sep.
Article En | MEDLINE | ID: mdl-7593551

The two most common forms of X-linked adrenoleukodystrophy (X-ALD), the childhood cerebral form (CCER) and the adult form, adrenomyeloneuropathy (AMN), arise from the same mutations in the X-ALD gene at Xq28. These two forms are distinguished by the degree of cerebral inflammation. Segregation analysis suggests that an autosomal modifying gene may be a major determinant of phenotype in X-ALD. Thus, a modifying gene could be involved in initiating or promoting the inflammatory response. In this study we detected a difference in tumor necrosis factor-alpha (TNF-alpha) bioactivity, but not TNF-alpha protein levels, in serum from some advanced CCER patients. Early-stage CCER patients and AMN patients were in the normal range. Allelic differences in TNF-alpha or levels of soluble TNF receptor did not account for bioactivity differences or phenotypic heterogeneity in X-ALD.


Adrenoleukodystrophy/physiopathology , Tumor Necrosis Factor-alpha/genetics , Adolescent , Alleles , Base Sequence , Child , DNA Primers/chemistry , DNA, Complementary/genetics , Humans , Interleukin-1/pharmacology , Introns , Lymphocyte Activation , Male , Molecular Sequence Data , Phytohemagglutinins/pharmacology , Polymorphism, Single-Stranded Conformational , Receptors, Tumor Necrosis Factor/metabolism
12.
Ann Neurol ; 38(3): 472-7, 1995 Sep.
Article En | MEDLINE | ID: mdl-7668838

The clinical distinction between patients with a disorder of peroxisome assembly (e.g., Zellweger syndrome) and those with a defect in a peroxisomal fatty acid beta-oxidation enzyme can be difficult. We studied 29 patients suspected of belonging to the latter group. Using complementation analysis, 24 were found to be deficient in enoylcoenzyme A hydratase/3-hydroxyacylcoenzyme A dehydrogenase bifunctional enzyme and 5 were deficient in acyl-CoA oxidase. Elevated plasma very long-chain fatty acids (VLCFA), impaired fibroblast VLCFA beta-oxidation, decreased fibroblast phytanic acid oxidation, normal plasmalogen synthesis, normal plasma L-pipecolic acid level, and normal subcellular catalase distribution were characteristic findings in both disorders. The elevation in plasma VLCFA levels and impairment in fibroblast VLCFA beta-oxidation were more severe in bifunctional-deficient than in oxidase-deficient patients. The clinical course in bifunctional deficiency (profound hypotonia, neonatal seizures, dysmorphic features, age at death approximately 9 months) was more severe than in oxidase deficiency (moderate hypotonia without dysmorphic features, development of a leukodystrophy, age at death approximately 4 yr). Based on these findings, accurate early diagnosis of these deficiencies of peroxisomal beta-oxidation enzymes is possible.


Multienzyme Complexes/deficiency , Oxidoreductases/deficiency , Zellweger Syndrome/enzymology , Acyl-CoA Oxidase , Child, Preschool , Humans , Infant
13.
Article En | MEDLINE | ID: mdl-7881809

Female turkey poults were hypophysectomized at 4-5 weeks of age. Beginning at 6 weeks of age, 20 hypophysectomized and 20 intact birds received a daily intramuscular injection of natural chicken growth hormone (cGH, 100 micrograms/kg body weight) or vehicle for 12 days. Blood samples were taken from each bird just before injection and 4 hr post-injection at 6 and 12 days of treatment. Hypophysectomy reduced the growth rate of turkey poults to 75% of that of intact controls, significantly reduced carcass protein and ash percentages, and significantly lower plasma concentrations of GH, insulin-like growth factor-I, triiodothyronine, thyroxine, insulin, glucose, triglycerides, and non-esterified fatty acids. Hypophysectomy was without effect on liver GH receptor binding activity, but increased liver 5'-monodeiodinase activity. Daily cGH injection had no effect on the average daily gain of either hypophysectomized or intact poults when compared to vehicle-injected controls over 12 days of treatment. Daily cGH administration increased plasma insulin-like growth factor-I levels in intact and hypophysectomized turkeys, and increased plasma triiodothyronine, insulin, glucose, and triglyceride concentrations in hypophysectomized birds, but not in intact birds. Responses of young turkeys to hypophysectomy and GH replacement were consistent with the known metabolic role of GH in other species, but the influence of GH on growth appears to be of less importance in poultry than in mammals.


Body Composition/drug effects , Growth Hormone/pharmacology , Hormones/blood , Hypophysectomy , Turkeys/physiology , Animals , Chickens , Female , Growth/drug effects , Growth/physiology , Growth Hormone/blood , Liver/drug effects , Liver/enzymology , Organ Size/drug effects , Weight Gain/drug effects
14.
Genomics ; 19(1): 60-7, 1994 Jan 01.
Article En | MEDLINE | ID: mdl-8188243

Enoyl-CoA hydratase:3-hydroxyacyl-CoA dehydrogenase bifunctional enzyme is one of the four enzymes of the peroxisomal beta-oxidation pathway. Here, we report the full-length human cDNA sequence and the localization of the corresponding gene on chromosome 3q26.3-3q28. The cDNA sequence spans 3779 nucleotides with an open reading frame of 2169 nucleotides. The tripeptide SKL at the carboxy terminus, known to serve as a peroxisomal targeting signal, is present. DNA sequence comparison of the coding region showed an 80% homology between human and rat bifunctional enzyme cDNA. The 3' noncoding sequence contains 117 nucleotides homologous to an Alu repeat. Based on sequence comparison, we propose that these nucleotides are a free left Alu arm with 86% homology to the Alu-J family. RNA analysis shows one band with highest intensity in liver and kidney. This cDNA will allow in-depth studies of molecular defects in patients with defective peroxisomal bifunctional enzyme. Moreover, it will also provide a means for studying the regulation of peroxisomal beta-oxidation in humans.


3-Hydroxyacyl CoA Dehydrogenases/genetics , Chromosomes, Human, Pair 3 , Enoyl-CoA Hydratase/genetics , Genes , Isomerases/genetics , Multienzyme Complexes/genetics , Amino Acid Sequence , Animals , Base Sequence , Blotting, Northern , Chromosome Mapping , DNA, Complementary/genetics , Humans , Introns , Kidney/enzymology , Liver/enzymology , Microbodies/enzymology , Molecular Sequence Data , Open Reading Frames , Organ Specificity , Peroxisomal Bifunctional Enzyme , Rats/genetics , Repetitive Sequences, Nucleic Acid , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity
15.
Biochem Med Metab Biol ; 49(2): 228-42, 1993 Apr.
Article En | MEDLINE | ID: mdl-8484962

Complementation analysis, using peroxisomal beta-oxidation of very long chain fatty acids (VLCFA) as the criterion for complementation, is useful in the study of patients who are suspected of having a single enzyme defect in the peroxisomal beta-oxidation pathway. Laboratory findings for these patients include elevated plasma VLCFA and impaired VLCFA oxidation in fibroblasts. Some of these patients have slightly abnormal phytanic acid oxidation in fibroblasts. In addition, elevated levels of bile acid intermediates have been reported in some cases. Plasmalogen synthesis, pipecolic acid levels, and subcellular distribution of catalase are normal. Using complementation analysis, we show that six patients, who were suspected of having a single enzyme defect in the peroxisomal beta-oxidation pathway, are deficient in peroxisomal bifunctional enzyme [enoyl-CoA hydratase (EC 4.2.1.17)/3-hydroxyacyl-CoA dehydrogenase (EC 1.1.1.35)] activity. This group of six patients, deficient in bifunctional enzyme activity, may be subdivided into two complementation groups. It would appear that patients in each of these two groups are deficient in only one of the bifunctional enzyme activities.


3-Hydroxyacyl CoA Dehydrogenases/deficiency , Enoyl-CoA Hydratase/deficiency , Fatty Acids/metabolism , Microbodies/enzymology , Adrenoleukodystrophy/enzymology , Cell Line , Humans , Immunoblotting , Oxidation-Reduction , Phytanic Acid/metabolism , Zellweger Syndrome/enzymology
16.
Domest Anim Endocrinol ; 8(4): 611-20, 1991 Oct.
Article En | MEDLINE | ID: mdl-1786708

The purpose of this study was to determine if exogenous insulin-like growth factor-I (IGF-I) would improve growth rate or body composition of young broiler chickens. Broiler cockerels were given a daily intramuscular (im) injection of sodium acetate buffer (buffer control), 100 or 200 micrograms recombinant-derived human IGF-I (rhIGF-I) per kg body weight from 11 to 24 days of age. Exogenous IGF-I did not affect the average daily gain, average daily feed consumption, or the gain-to-feed ratio of broiler chickens. Although daily injection of 200 micrograms/kg of rhIGF-I reduced (P less than 0.05) body ash content, there was no significant effect of IGF-I treatment on either body fat or protein content. Plasma GH levels were depressed (P less than 0.05) by chronic treatment with rhIGF-I. In contrast, plasma levels of T3 and T4 were not affected by rhIGF-I treatment. The half-life of rhIGF-I in plasma was determined at 25 days of age in naive control or chronically-injected chickens after a single intravenous dose of 50 micrograms rhIGF-I/kg. We found a single compartment, first-order disappearance pattern of rhIGF-I from chicken plasma. The half-life (t1/2) of rhIGF-I in plasma was similar (t1/2 = 32.5 min) for naive controls (injected once) or chronically-treated chickens which had received a daily injection of rhIGF-I (100 or 200 micrograms/kg) for 14 d. These data indicate that daily injection of IGF-I cannot be used to enhance growth performance or body composition of broiler chickens when given during the early growth period. The depression of plasma GH levels in rhIGF-I-injected chickens supports a negative-feedback role of IGF-I on pituitary GH secretion.


Chickens/growth & development , Insulin-Like Growth Factor I/pharmacology , Animals , Body Composition/drug effects , Chickens/anatomy & histology , Dose-Response Relationship, Drug , Eating/drug effects , Half-Life , Humans , Injections, Intramuscular/veterinary , Injections, Intravenous/veterinary , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/pharmacokinetics , Male , Recombinant Proteins/administration & dosage , Recombinant Proteins/pharmacokinetics , Recombinant Proteins/pharmacology , Weight Gain/drug effects
17.
Biochem Biophys Res Commun ; 172(1): 364-9, 1990 Oct 15.
Article En | MEDLINE | ID: mdl-2222480

Complementation studies, using fused cell lines from patients with peroxisomal disorders, have shown correction of defective plasmalogen synthesis and phytanic acid oxidation as well as an increase in the number of peroxisomes. At least six complementation groups have been reported. We demonstrate here that complementing cell lines also acquire the ability to oxidize very long chain fatty acids (VLCFA), and that complementation groups defined with this technique are identical to those reported previously when plasmalogen synthesis was used as the criterion for complementation. This VLCFA complementation technique is of particular value in the study of patients in whom defective VLCFA is the only or major enzymatic defect, and we show complementation between cell lines from two patients each with an isolated defect in one of the peroxisomal fatty acid beta-oxidation enzymes.


Fatty Acids, Nonesterified/metabolism , Metabolism, Inborn Errors/genetics , Microbodies/metabolism , Skin/metabolism , Cell Fusion , Cell Line , Cells, Cultured , Fibroblasts/metabolism , Genetic Complementation Test , Humans , Metabolism, Inborn Errors/metabolism , Oxidation-Reduction , Reference Values
18.
Gen Comp Endocrinol ; 79(3): 446-58, 1990 Sep.
Article En | MEDLINE | ID: mdl-2272466

The main purpose of this study was to examine the relationship between insulin-like growth factor-I (IGF-I) and growth hormone (GH) during embryonic and posthatching development of broiler chickens. Two heterologous assays were validated for measurement of IGF-I in chicken and turkey plasma. A radioreceptor assay (RRA), utilizing microsomal membranes prepared from human placenta, was modified and validated for measurement of IGF peptide (mainly IGF-I). A double-antibody radioimmunoassay (RIA) was validated for measurement of immunoreactive IGF-I levels in chicken and turkey plasma. In both assay systems, recombinant-derived human IGF-I was used for standards and trace hormone. Hypophysectomy in turkey poults reduced plasma levels of IGF (RRA) by 35% and IGF-I (RIA) by 59% as compared to intact control turkeys. In Experiment 1, 14 chicken embryos were bled at 15, 17, 19, and 21 days of incubation and at 1 week of age to determine plasma levels of IGF-I and GH. Plasma IGF levels (RRA) remained constant during late incubation, but increased significantly (P less than 0.05) at 1 week of age. Plasma IGF-I levels (RIA) declined 2 days before hatching; however, plasma levels of IGF-I were sharply elevated (P less than 0.05) at 1 week of age. Plasma GH concentrations were low in embryos and were greatly elevated (P less than 0.05) at hatching (21 days of incubation) and at 1 week of age. In Experiment 2, 12 different broiler cockerels were weighed and then bled by cardiac puncture each week from hatching (1 day of age) to 7 weeks of age. The plasma profiles of IGF, IGF-I, GH, triiodothyronine (T3), and thyroxine (T4) were each compared to relative growth rate by analysis of covariance. Plasma IGF and IGF-I levels increased progressively from 0 to 3 weeks of age and were maintained in a plateau from 3 to 7 weeks of age. Plasma GH levels reached a peak at 4 weeks of age, but declined sharply thereafter, while IGF and IGF-I levels remained elevated. Plasma T3 concentrations were progressively increased and reached peak concentrations at 3 weeks of age, while plasma T4 levels increased only at 6 and 7 weeks of age. There was a high correlation (P less than 0.01) between relative growth rate and age-related changes in plasma levels of IGF (r = 0.96), IGF-I (r = 0.97), and T3 (r = 0.94); however, there was no correlation between relative growth rate and changes in plasma GH or T4.(ABSTRACT TRUNCATED AT 400 WORDS)


Aging/physiology , Chickens/growth & development , Insulin-Like Growth Factor I/biosynthesis , Animals , Growth Hormone/blood , Hypophysectomy , Pituitary Gland/metabolism , Radioimmunoassay , Radioligand Assay , Somatomedins/biosynthesis , Thyroxine/blood , Triiodothyronine/blood
19.
Proc Soc Exp Biol Med ; 192(2): 127-34, 1989 Nov.
Article En | MEDLINE | ID: mdl-2510176

The effects of thyroid manipulation on growth, feed efficiency, and plasma hormone levels were determined in rapidly growing chickens. Beginning at 3 weeks of age, eight broiler cockerels were provided with control feed (CF) or feed containing either 1 ppm of triiodothyronine (T3), 1 ppm of thyroxine (T4), 0.3% propylthiouracil (PTU), or 5 ppm of thyrotropin-releasing hormone (TRH) for 3 weeks. Blood samples were taken at 4, 5, and 6 weeks for determination of plasma levels of growth hormone, insulin-like growth factor, T3, T4, insulin, glucagon, glucose, and nonesterified fatty acids. Dietary TRH increased (P less than 0.05) the growth rate of chickens by 14% when compared with the CF group. Plasma growth hormone levels were reduced (P less than 0.05) 65% by dietary T3 and 33% by treatment with either T4 or TRH when compared with the CF group. Plasma insulin-like growth factor levels were 16% lower (P less than 0.05) in PTU-fed birds than the other treatment groups. Plasma T3 levels were elevated (P less than 0.05) 3-fold by dietary T3 and 38% by TRH whereas plasma T3 in the PTU group was 38% below the average of CF birds. Plasma T4 levels were increased (P less than 0.05) by 12-fold in T4-fed birds, decreased 48% in TRH-fed birds, and nondetectable in birds treated with either T3 or PTU. Compared with the other treatments, dietary PTU increased (P less than 0.01) plasma insulin levels 4.3-fold whereas TRH provided a 2.7-fold increase in plasma insulin. Plasma glucagon levels were 26% higher (P less than 0.05) in T3-fed birds than those fed either T4 or PTU. These observations indicate that thyroid activity plays an important role in regulating secretion of GH and the pancreatic hormones. Furthermore, our study demonstrates the potential use of TRH as an orally active growth promoter for poultry.


Chickens/growth & development , Glucagon/metabolism , Insulin/metabolism , Thyrotropin-Releasing Hormone/pharmacology , Aging , Animals , Diet , Glucagon/blood , Insulin/blood , Insulin Secretion , Male , Propylthiouracil/administration & dosage , Propylthiouracil/pharmacology , Reference Values , Thyrotropin-Releasing Hormone/administration & dosage , Thyroxine/administration & dosage , Thyroxine/blood , Thyroxine/pharmacology , Triiodothyronine/administration & dosage , Triiodothyronine/blood , Triiodothyronine/pharmacology
...