Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Cell Rep ; 21(1): 47-59, 2017 Oct 03.
Article in English | MEDLINE | ID: mdl-28978483

ABSTRACT

During development, chromatin-modifying enzymes regulate both the timely establishment of cell-type-specific gene programs and the coordinated repression of alternative cell fates. To dissect the role of one such enzyme, the intellectual-disability-linked lysine demethylase 5C (Kdm5c), in the developing and adult brain, we conducted parallel behavioral, transcriptomic, and epigenomic studies in Kdm5c-null and forebrain-restricted inducible knockout mice. Together, genomic analyses and functional assays demonstrate that Kdm5c plays a critical role as a repressor responsible for the developmental silencing of germline genes during cellular differentiation and in fine-tuning activity-regulated enhancers during neuronal maturation. Although the importance of these functions declines after birth, Kdm5c retains an important genome surveillance role preventing the incorrect activation of non-neuronal and cryptic promoters in adult neurons.


Subject(s)
Gene Expression Regulation, Developmental , Neurons/metabolism , Oxidoreductases, N-Demethylating/genetics , Prosencephalon/metabolism , Transcription, Genetic , Animals , DNA-Binding Proteins , Doublecortin Domain Proteins , Enhancer Elements, Genetic , Female , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Histone Demethylases , Histones/genetics , Histones/metabolism , Male , Maze Learning , Mice , Mice, Knockout , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/pathology , Neuropeptides/genetics , Neuropeptides/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oxidoreductases, N-Demethylating/deficiency , Prosencephalon/pathology , Signal Transduction
2.
Cell Rep ; 14(5): 1000-1009, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26804915

ABSTRACT

Mutations in a number of chromatin modifiers are associated with human neurological disorders. KDM5C, a histone H3 lysine 4 di- and tri-methyl (H3K4me2/3)-specific demethylase, is frequently mutated in X-linked intellectual disability (XLID) patients. Here, we report that disruption of the mouse Kdm5c gene recapitulates adaptive and cognitive abnormalities observed in XLID, including impaired social behavior, memory deficits, and aggression. Kdm5c-knockout brains exhibit abnormal dendritic arborization, spine anomalies, and altered transcriptomes. In neurons, Kdm5c is recruited to promoters that harbor CpG islands decorated with high levels of H3K4me3, where it fine-tunes H3K4me3 levels. Kdm5c predominantly represses these genes, which include members of key pathways that regulate the development and function of neuronal circuitries. In summary, our mouse behavioral data strongly suggest that KDM5C mutations are causal to XLID. Furthermore, our findings suggest that loss of KDM5C function may impact gene expression in multiple regulatory pathways relevant to the clinical phenotypes.


Subject(s)
Genes, X-Linked , Histones/metabolism , Intellectual Disability/genetics , Aggression , Animals , Brain/pathology , CpG Islands , Dendritic Spines/pathology , Disease Models, Animal , Gene Expression Regulation , Histone Demethylases , Lysine/metabolism , Memory , Methylation , Mice, Knockout , Oxidoreductases, N-Demethylating/deficiency , Oxidoreductases, N-Demethylating/metabolism , Promoter Regions, Genetic , Social Behavior , Transcription, Genetic
4.
Am J Physiol Heart Circ Physiol ; 301(5): H1862-71, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21873498

ABSTRACT

Histone methylation, a determinant of chromatin structure and gene transcription, was thought to be irreversible, but recent evidence suggests that lysine-specific demethylase-1 (LSD1, Kdm1a) induces demethylation of histone H3 lysine 4 (H3K4) or H3K9 and thereby alters gene transcription. We previously demonstrated a human LSD1 phenotype associated with salt-sensitive hypertension. To test the hypothesis that LSD1 plays a role in the regulation of blood pressure (BP) via vascular mechanisms and gene transcription, we measured BP and examined vascular function and endothelial nitric oxide (NO) synthase (eNOS) expression in thoracic aorta of male wild-type (WT) and heterozygous LSD1 knockout mice (LSD1(+/-)) fed either a liberal salt (HS; 4% NaCl) or restricted salt diet (LS; 0.08% NaCl). BP was higher in LSD1(+/-) than WT mice on the HS diet but not different between LSD1(+/-) and WT mice on the LS diet. Further examination of the mechanisms of this salt-sensitive hypertension in LSD1(+/-) mice on the HS diet demonstrated that plasma renin activity and plasma levels and urinary excretion of aldosterone were less in LSD1(+/-) than WT, suggesting suppressed renin-angiotensin-aldosterone system. In contrast, phenylephrine (Phe)-induced aortic contraction was greater in LSD1(+/-) than WT mice on the HS diet. Treatment of aortic rings with 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ; a blocker of guanylate cyclase) enhanced Phe contraction in LSD1(+/-) compared with WT mice on the HS diet. Acetylcholine (Ach)-induced relaxation was less in LSD1(+/-) than WT mice on the HS diet. Endothelium removal or pretreatment with N(ω)-nitro-L-arginine methyl ester (blocker of NOS) or ODQ abolished Ach-induced relaxation in aorta of WT but had minimal effect in LSD1(+/-). Vascular relaxation to sodium nitroprusside, an exogenous NO donor and guanylate cyclase activator, was decreased in LSD1(+/-) vs. WT mice on the HS diet. RT-PCR and Western blots revealed decreased eNOS mRNA expression and eNOS and guanylate cyclase protein in the heart and aorta of LSD1(+/-) compared with WT mice on HS diet. Thus, during the HS diet, LSD1 deficiency is associated with hypertension, enhanced vascular contraction, and reduced relaxation via NO-cGMP pathway. The data support a role for LSD1-mediated histone demethylation in the regulation of NOS/guanylate cyclase gene expression, vascular function, and BP during the HS diet.


Subject(s)
Blood Pressure , Cyclic GMP/metabolism , Hypertension/enzymology , Muscle, Smooth, Vascular/enzymology , Nitric Oxide/metabolism , Oxidoreductases, N-Demethylating/deficiency , Sodium Chloride, Dietary , Vasoconstriction , Aldosterone/blood , Aldosterone/urine , Animals , Aorta, Thoracic/enzymology , Aorta, Thoracic/physiopathology , Blood Pressure/drug effects , Blood Pressure/genetics , Blotting, Western , Disease Models, Animal , Dose-Response Relationship, Drug , Genotype , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , Histone Demethylases , Hypertension/etiology , Hypertension/genetics , Hypertension/physiopathology , Male , Mice , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Oxidoreductases, N-Demethylating/genetics , Phenotype , Real-Time Polymerase Chain Reaction , Renin/blood , Signal Transduction , Vasoconstriction/drug effects , Vasoconstriction/genetics , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology
5.
Pediatr Infect Dis J ; 30(11): 1001-3, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21633320

ABSTRACT

Efavirenz, used in treating pediatric human immunodeficiency virus infection, has central nervous system side effects. We report on a 5-year-old girl with perinatally acquired human immunodeficiency virus infection, presenting with new onset absence seizures after starting treatment with efavirenz. Plasma efavirenz values were above therapeutic range. The child was homozygous for the CYP2B6-516T/T genotype, which is associated with poor efavirenz clearance. Seizures abated after efavirenz discontinuation.


Subject(s)
Anti-HIV Agents/adverse effects , Aryl Hydrocarbon Hydroxylases/genetics , Benzoxazines/adverse effects , Epilepsy, Absence/genetics , HIV Infections/genetics , HIV/physiology , Oxidoreductases, N-Demethylating/genetics , Alkynes , Anti-HIV Agents/blood , Aryl Hydrocarbon Hydroxylases/deficiency , Benzoxazines/blood , Child, Preschool , Cyclopropanes , Cytochrome P-450 CYP2B6 , Epilepsy, Absence/blood , Epilepsy, Absence/chemically induced , Epilepsy, Absence/physiopathology , Epilepsy, Absence/virology , Female , Genetic Predisposition to Disease , Genotype , HIV/drug effects , HIV Infections/blood , HIV Infections/drug therapy , HIV Infections/physiopathology , HIV Infections/virology , Homozygote , Humans , Oxidoreductases, N-Demethylating/deficiency , Polymorphism, Genetic , South Africa , Viral Load/drug effects
6.
Mol Cell Biol ; 30(20): 4851-63, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20713442

ABSTRACT

Lysine-specific demethylase 1 (LSD1), which demethylates mono- and dimethylated histone H3-Lys4 as part of a complex including CoREST and histone deacetylases (HDACs), is essential for embryonic development in the mouse beyond embryonic day 6.5 (e6.5). To determine the role of LSD1 during this early period of embryogenesis, we have generated loss-of-function gene trap mice and conditional knockout embryonic stem (ES) cells. Analysis of postimplantation gene trap embryos revealed that LSD1 expression, and therefore function, is restricted to the epiblast. Conditional deletion of LSD1 in mouse ES cells, the in vitro counterpart of the epiblast, revealed a reduction in CoREST protein and associated HDAC activity, resulting in a global increase in histone H3-Lys56 acetylation, but not H3-Lys4 methylation. Despite this biochemical perturbation, ES cells with LSD1 deleted proliferate normally and retain stem cell characteristics. Loss of LSD1 causes the aberrant expression of 588 genes, including those coding for transcription factors with roles in anterior/posterior patterning and limb development, such as brachyury, Hoxb7, Hoxd8, and retinoic acid receptor γ (RARγ). The gene coding for brachyury, a key regulator of mesodermal differentiation, is a direct target gene of LSD1 and is overexpressed in e6.5 Lsd1 gene trap embryos. Thus, LSD1 regulates the expression and appropriate timing of key developmental regulators, as part of the LSD1/CoREST/HDAC complex, during early embryonic development.


Subject(s)
Gene Expression Profiling , Nerve Tissue Proteins/metabolism , Oxidoreductases, N-Demethylating/metabolism , Repressor Proteins/metabolism , Animals , Base Sequence , Cell Differentiation , Cell Line , Co-Repressor Proteins , DNA Primers/genetics , Embryonic Development/genetics , Embryonic Development/physiology , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Fetal Proteins/genetics , Fetal Proteins/metabolism , Gene Expression Regulation, Developmental , Histone Deacetylases/metabolism , Histone Demethylases , Mice , Mice, Knockout , Oxidoreductases, N-Demethylating/deficiency , Oxidoreductases, N-Demethylating/genetics , Pregnancy , Protein Stability , RNA, Messenger/genetics , RNA, Messenger/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
7.
Nature ; 461(7262): 415-8, 2009 Sep 17.
Article in English | MEDLINE | ID: mdl-19727073

ABSTRACT

Differential DNA methylation of the paternal and maternal alleles regulates the parental origin-specific expression of imprinted genes in mammals. The methylation imprints are established in male and female germ cells during gametogenesis, and the de novo DNA methyltransferase DNMT3A and its cofactor DNMT3L are required in this process. However, the mechanisms underlying locus- and parental-specific targeting of the de novo DNA methylation machinery in germline imprinting are poorly understood. Here we show that amine oxidase (flavin-containing) domain 1 (AOF1), a protein related to the lysine demethylase KDM1 (also known as LSD1), functions as a histone H3 lysine 4 (H3K4) demethylase and is required for de novo DNA methylation of some imprinted genes in oocytes. AOF1, now renamed lysine demethylase 1B (KDM1B) following a new nomenclature, is highly expressed in growing oocytes where genomic imprints are established. Targeted disruption of the gene encoding KDM1B had no effect on mouse development and oogenesis. However, oocytes from KDM1B-deficient females showed a substantial increase in H3K4 methylation and failed to set up the DNA methylation marks at four out of seven imprinted genes examined. Embryos derived from these oocytes showed biallelic expression or biallelic suppression of the affected genes and died before mid-gestation. Our results suggest that demethylation of H3K4 is critical for establishing the DNA methylation imprints during oogenesis.


Subject(s)
DNA Methylation , Genomic Imprinting , Histones/metabolism , Mothers , Oxidoreductases, N-Demethylating/metabolism , Alleles , Animals , Embryo Loss/genetics , Embryo, Mammalian/metabolism , Female , Gene Expression Regulation, Developmental , Genes, Developmental/genetics , Male , Mice , NIH 3T3 Cells , Oocytes/metabolism , Oxidoreductases, N-Demethylating/deficiency , Oxidoreductases, N-Demethylating/genetics
8.
Nat Genet ; 41(1): 125-9, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19098913

ABSTRACT

Histone methylation and DNA methylation cooperatively regulate chromatin structure and gene activity. How these two systems coordinate with each other remains unclear. Here we study the biological function of lysine-specific demethylase 1 (LSD1, also known as KDM1 and AOF2), which has been shown to demethylate histone H3 on lysine 4 (H3K4) and lysine 9 (H3K9). We show that LSD1 is required for gastrulation during mouse embryogenesis. Notably, targeted deletion of the gene encoding LSD1 (namely, Aof2) in embryonic stem (ES) cells induces progressive loss of DNA methylation. This loss correlates with a decrease in DNA methyltransferase 1 (Dnmt1) protein, as a result of reduced Dnmt1 stability. Dnmt1 protein is methylated in vivo, and its methylation is enhanced in the absence of LSD1. Furthermore, Dnmt1 can be methylated by Set7/9 (also known as KMT7) and demethylated by LSD1 in vitro. Our findings suggest that LSD1 demethylates and stabilizes Dnmt1, thus providing a previously unknown mechanistic link between the histone and DNA methylation systems.


Subject(s)
DNA Methylation , Oxidoreductases, N-Demethylating/metabolism , Animals , Cell Differentiation , Cell Proliferation , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , Embryonic Development , Embryonic Stem Cells/cytology , Embryonic Stem Cells/enzymology , Histone Demethylases , Mice , Oxidoreductases, N-Demethylating/deficiency , Substrate Specificity
10.
Mol Cell ; 27(4): 562-72, 2007 Aug 17.
Article in English | MEDLINE | ID: mdl-17707228

ABSTRACT

Gfi-1 and Gfi-1b are homologous transcriptional repressors involved in diverse developmental contexts, including hematopoiesis and oncogenesis. Transcriptional repression by Gfi proteins requires the conserved SNAG domain. To elucidate the function of Gfi proteins, we purified Gfi-1b complexes and identified interacting proteins. Prominent among these is the corepressor CoREST, the histone demethylase LSD1, and HDACs 1 and 2. CoREST and LSD1 associate with Gfi-1/1b via the SNAG repression domain. Gfi-1b further recruits these cofactors to the majority of target gene promoters in vivo. Inhibition of CoREST and LSD1 perturbs differentiation of erythroid, megakaryocytic, and granulocytic cells as well as primary erythroid progenitors. LSD1 depletion derepresses Gfi targets in lineage-specific patterns, accompanied by enhanced histone 3 lysine 4 methylation at the respective promoters. Overall, we show that chromatin regulatory proteins CoREST and LSD1 mediate transcriptional repression by Gfi proteins. Lineage-restricted deployment of these cofactors through interaction with Gfi proteins controls hematopoietic differentiation.


Subject(s)
Cell Differentiation/genetics , DNA-Binding Proteins/metabolism , Epigenesis, Genetic , Hematopoiesis/genetics , Nerve Tissue Proteins/metabolism , Oxidoreductases, N-Demethylating/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Lineage , Co-Repressor Proteins , DNA Methylation , DNA-Binding Proteins/chemistry , Erythroid Cells/cytology , Histone Deacetylases/metabolism , Histone Demethylases , Histones/metabolism , Mice , Models, Genetic , Molecular Sequence Data , Multiprotein Complexes/metabolism , Oxidoreductases, N-Demethylating/deficiency , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , RNA Interference , Repressor Proteins/chemistry , Transcription Factors/chemistry
11.
Nature ; 448(7154): 718-22, 2007 Aug 09.
Article in English | MEDLINE | ID: mdl-17687328

ABSTRACT

Histone methylation is crucial for regulating chromatin structure, gene transcription and the epigenetic state of the cell. LSD1 is a lysine-specific histone demethylase that represses transcription by demethylating histone H3 on lysine 4 (ref. 1). The LSD1 complex contains a number of proteins, all of which have been assigned roles in events upstream of LSD1-mediated demethylation apart from BHC80 (also known as PHF21A), a plant homeodomain (PHD) finger-containing protein. Here we report that, in contrast to the PHD fingers of the bromodomain PHD finger transcription factor (BPTF) and inhibitor of growth family 2 (ING2), which bind methylated H3K4 (H3K4me3), the PHD finger of BHC80 binds unmethylated H3K4 (H3K4me0), and this interaction is specifically abrogated by methylation of H3K4. The crystal structure of the PHD finger of BHC80 bound to an unmodified H3 peptide has revealed the structural basis of the recognition of H3K4me0. Knockdown of BHC80 by RNA inhibition results in the de-repression of LSD1 target genes, and this repression is restored by the reintroduction of wild-type BHC80 but not by a PHD-finger mutant that cannot bind H3. Chromatin immunoprecipitation showed that BHC80 and LSD1 depend reciprocally on one another to associate with chromatin. These findings couple the function of BHC80 to that of LSD1, and indicate that unmodified H3K4 is part of the 'histone code'. They further raise the possibility that the generation and recognition of the unmodified state on histone tails in general might be just as crucial as post-translational modifications of histone for chromatin and transcriptional regulation.


Subject(s)
Gene Silencing , Histone Deacetylases/metabolism , Histones/metabolism , Lysine/metabolism , Oxidoreductases, N-Demethylating/metabolism , Amino Acid Sequence , Chromatin/chemistry , Chromatin/metabolism , HeLa Cells , Histone Deacetylases/chemistry , Histone Deacetylases/deficiency , Histone Deacetylases/genetics , Histone Demethylases , Humans , Methylation , Models, Molecular , Molecular Sequence Data , Oxidoreductases, N-Demethylating/deficiency , Oxidoreductases, N-Demethylating/genetics , Protein Structure, Tertiary , RNA Interference , Zinc Fingers
12.
J Biol Chem ; 282(21): 15471-5, 2007 May 25.
Article in English | MEDLINE | ID: mdl-17409384

ABSTRACT

The lysine-specific demethylase 1 (LSD1), a component of several histone deacetylase complexes, plays an important role in chromatin remodeling and transcriptional regulation. Here, we generated multiple cell lines in which LSD1 is inducibly expressed or knocked down and found that LSD1 is required for cell proliferation. In addition, we found that deficiency in LSD1 leads to a partial cell cycle arrest in G(2)/M and sensitizes cells to growth suppression induced by DNA damage or MDM2 inhibition in a p53-dependent manner. We also showed that LSD1 deficiency delays p53 stabilization induced by DNA damage, leading to a delayed induction of p21 and MDM2. Finally, we performed a microarray study and identified several novel LSD1 target genes, including S100A8, which encodes a calcium-binding protein, and DEK, a proto-oncogene. Taken together, we uncovered that LSD1 has a pro-oncogenic function by modulating pro-survival gene expression and p53 transcriptional activity.


Subject(s)
Cell Division/physiology , Chromatin Assembly and Disassembly/physiology , G2 Phase/physiology , Oxidoreductases, N-Demethylating/metabolism , Tumor Suppressor Protein p53/metabolism , Calgranulin A/biosynthesis , Calgranulin A/genetics , Cell Line, Tumor , Chromosomal Proteins, Non-Histone/biosynthesis , Chromosomal Proteins, Non-Histone/genetics , DNA Damage/genetics , Gene Expression Profiling , Histone Demethylases , Humans , Oligonucleotide Array Sequence Analysis , Oncogene Proteins/biosynthesis , Oncogene Proteins/genetics , Oxidoreductases, N-Demethylating/deficiency , Poly-ADP-Ribose Binding Proteins , Proto-Oncogene Mas , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/genetics
13.
Nature ; 446(7138): 882-7, 2007 Apr 19.
Article in English | MEDLINE | ID: mdl-17392792

ABSTRACT

Precise control of transcriptional programmes underlying metazoan development is modulated by enzymatically active co-regulatory complexes, coupled with epigenetic strategies. One thing that remains unclear is how specific members of histone modification enzyme families, such as histone methyltransferases and demethylases, are used in vivo to simultaneously orchestrate distinct developmental gene activation and repression programmes. Here, we report that the histone lysine demethylase, LSD1--a component of the CoREST-CtBP co-repressor complex--is required for late cell-lineage determination and differentiation during pituitary organogenesis. LSD1 seems to act primarily on target gene activation programmes, as well as in gene repression programmes, on the basis of recruitment of distinct LSD1-containing co-activator or co-repressor complexes. LSD1-dependent gene repression programmes can be extended late in development with the induced expression of ZEB1, a Krüppel-like repressor that can act as a molecular beacon for recruitment of the LSD1-containing CoREST-CtBP co-repressor complex, causing repression of an additional cohort of genes, such as Gh, which previously required LSD1 for activation. These findings suggest that temporal patterns of expression of specific components of LSD1 complexes modulate gene regulatory programmes in many mammalian organs.


Subject(s)
Down-Regulation/genetics , Gene Expression Regulation, Developmental , Oxidoreductases, N-Demethylating/metabolism , Animals , Cell Differentiation , Growth Hormone/genetics , Histone Demethylases , Homeodomain Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Lactotrophs/metabolism , Mice , Oxidoreductases, N-Demethylating/deficiency , Oxidoreductases, N-Demethylating/genetics , Pituitary Gland/cytology , Pituitary Gland/metabolism , Transcriptional Activation , Zinc Finger E-box-Binding Homeobox 1
14.
Am J Hum Genet ; 68(4): 839-47, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11231903

ABSTRACT

Dimethylglycine dehydrogenase (DMGDH) (E.C. number 1.5.99.2) is a mitochondrial matrix enzyme involved in the metabolism of choline, converting dimethylglycine to sarcosine. Sarcosine is then transformed to glycine by sarcosine dehydrogenase (E.C. number 1.5.99.1). Both enzymes use flavin adenine dinucleotide and folate in their reaction mechanisms. We have identified a 38-year-old man who has a lifelong condition of fishlike body odor and chronic muscle fatigue, accompanied by elevated levels of the muscle form of creatine kinase in serum. Biochemical analysis of the patient's serum and urine, using (1)H-nuclear magnetic resonance NMR spectroscopy, revealed that his levels of dimethylglycine were much higher than control values. The cDNA and the genomic DNA for human DMGDH (hDMGDH) were then cloned, and a homozygous A-->G substitution (326 A-->G) was identified in both the cDNA and genomic DNA of the patient. This mutation changes a His to an Arg (H109R). Expression analysis of the mutant cDNA indicates that this mutation inactivates the enzyme. We therefore confirm that the patient described here represents the first reported case of a new inborn error of metabolism, DMGDH deficiency.


Subject(s)
Metabolism, Inborn Errors/enzymology , Metabolism, Inborn Errors/genetics , Oxidoreductases, N-Demethylating/deficiency , Oxidoreductases, N-Demethylating/genetics , Point Mutation/genetics , Sarcosine/analogs & derivatives , Adult , Amino Acid Sequence , Amino Acid Substitution/genetics , Base Sequence , Black People/genetics , Blotting, Western , Cell Line , Chronic Disease , Cloning, Molecular , Creatine Kinase/blood , DNA Mutational Analysis , Dimethylglycine Dehydrogenase , Expressed Sequence Tags , Fatigue/complications , Fatigue/enzymology , Fatigue/genetics , Fatigue/metabolism , Humans , Magnetic Resonance Spectroscopy , Male , Metabolism, Inborn Errors/complications , Metabolism, Inborn Errors/metabolism , Mitochondria/enzymology , Mitochondrial Proteins , Molecular Sequence Data , Odorants , Oxidoreductases, N-Demethylating/chemistry , Phenotype , Sarcosine/blood , Sarcosine/urine
15.
Clin Chem ; 45(4): 459-64, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10102904

ABSTRACT

BACKGROUND: A38-year-old man presented with a history of fish odor (since age 5) and unusual muscle fatigue with increased serum creatine kinase. Our aim was to identify the metabolic error in this new condition. METHODS: We used 1H NMR spectroscopy to study serum and urine from the patient. RESULTS: The concentration of N, N-dimethylglycine (DMG) was increased approximately 100-fold in the serum and approximately 20-fold in the urine. The presence of DMG as a storage product was confirmed by use of 13C NMR spectroscopy and gas chromatography-mass spectrometry. The high concentration of DMG was caused by a deficiency of the enzyme dimethylglycine dehydrogenase (DMGDH). A homozygous missense mutation was found in the DMGDH gene of the patient. CONCLUSIONS: DMGDH deficiency must be added to the differential diagnosis of patients complaining of a fish odor. This deficiency is the first inborn error of metabolism discovered by use of in vitro 1H NMR spectroscopy of body fluids.


Subject(s)
Metabolism, Inborn Errors/enzymology , Oxidoreductases, N-Demethylating/genetics , Adult , Dimethylglycine Dehydrogenase , Gas Chromatography-Mass Spectrometry , Humans , Magnetic Resonance Spectroscopy , Male , Metabolism, Inborn Errors/blood , Metabolism, Inborn Errors/physiopathology , Metabolism, Inborn Errors/urine , Mitochondrial Proteins , Mutation, Missense , Odorants , Oxidoreductases, N-Demethylating/deficiency , Oxidoreductases, N-Demethylating/urine , Sarcosine/analogs & derivatives , Sarcosine/urine
17.
Pediatr Surg Int ; 12(2-3): 196-7, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9156859

ABSTRACT

A case of congenital portal-systemic shunting due to an intrahepatic connection diagnosed by ultrasound scanning and color Doppler in an 8-month-old girl is reported. She began to manifest trimethylaminuria 3 years later. At 7 years of age, she is asymptomatic without therapeutic measures except for diet. This is the seventh reported case and the third in a child to our knowledge.


Subject(s)
Arteriovenous Malformations/surgery , Liver/blood supply , Methylamines/urine , Portal Vein/abnormalities , Ultrasonography, Doppler, Color , Vena Cava, Inferior/abnormalities , Arteriovenous Malformations/diagnostic imaging , Child , Child, Preschool , Female , Follow-Up Studies , Humans , Infant , Metabolism, Inborn Errors/complications , Oxidoreductases, N-Demethylating/deficiency , Palliative Care , Portal Vein/diagnostic imaging , Vena Cava, Inferior/diagnostic imaging
18.
FEMS Microbiol Lett ; 143(2-3): 151-8, 1996 10 01.
Article in English | MEDLINE | ID: mdl-8837467

ABSTRACT

Gram-positive thermophilic Bacillus species contain cytochrome caa3-type cytochrome c oxidase as a terminal oxidase in the respiratory chain. To identify alternative oxidases, we isolated B. stearothermophilus mutants defective in the caa3-type oxidase activity. One mutant contained little cytochrome a and had low cytochrome c oxidase activity. However, growth and the respiratory activity of membranes in the presence of NADH were close to normal, suggesting that the mutant contains an alternative electron transfer pathway. A novel oxidase was isolated from the membrane fraction of the mutant. The enzyme is a cytochrome bd-type quinol oxidase composed of two subunits of 52 and 40 kDa, whose N-terminal regions show sequence similarity to polypeptides of the bd-type oxidase from Escherichia coli and Azotobacter vinelandii. This is the first report of a bd-type terminal oxidase purified from a Gram-positive bacterium.


Subject(s)
Electron Transport Chain Complex Proteins , Electron Transport Complex IV/genetics , Escherichia coli Proteins , Geobacillus stearothermophilus/enzymology , Geobacillus stearothermophilus/genetics , Mutation , Oxidoreductases/metabolism , Amino Acid Sequence , Cytochrome b Group , Cytochrome-c Oxidase Deficiency , Cytochromes/metabolism , Electron Transport , Electron Transport Complex IV/metabolism , Geobacillus stearothermophilus/growth & development , Molecular Sequence Data , Molecular Weight , Oxidoreductases/chemistry , Oxidoreductases/genetics , Oxidoreductases, N-Demethylating/deficiency , Oxidoreductases, N-Demethylating/genetics , Oxidoreductases, N-Demethylating/metabolism , Protein Conformation , Sequence Homology, Amino Acid
19.
J Inherit Metab Dis ; 18(3): 306-12, 1995.
Article in English | MEDLINE | ID: mdl-7474897

ABSTRACT

Trimethylaminuria is an autosomal recessive disorder involving deficient N-oxidation of the dietary-derived amine trimethylamine (TMA). TMA, a volatile tertiary amine, accumulates and is excreted in urine of patients with deficient TMA oxidase activity. Treatment strategies for this condition are limited. We report a new stable-isotope dilution method for rapid sequential analysis of TMA concentrations and the clinical and biochemical response to treatment with metronidazole.


Subject(s)
Metabolism, Inborn Errors/diagnosis , Metabolism, Inborn Errors/drug therapy , Methylamines/urine , Metronidazole/therapeutic use , Adult , Child , Female , Humans , Male , Mass Spectrometry , Metabolism, Inborn Errors/urine , Middle Aged , Odorants , Oxidoreductases, N-Demethylating/deficiency , Radioisotope Dilution Technique
20.
Am J Med Genet ; 45(3): 335-9, 1993 Feb 01.
Article in English | MEDLINE | ID: mdl-8434620

ABSTRACT

We report on an individual with trimethylaminuria, Prader-Willi syndrome, and del(15) (q11q13). To our knowledge, such an association has never been reported. Skin sores secondary to choline-rich foods and amenable to dietary control have not been described in trimethylaminuria, although they are seen in some patients with Prader-Willi syndrome. Pathogenesis, clinical diagnosis, and management of reported cases with trimethylaminuria are reviewed. Serious social and behavioral problems may result from strong body odor. Amelioration of the "fish odor" by dietary choline restriction makes trimethylaminuria detection important. Association of trimethylaminuria with Prader-Willi syndrome and del(15) (q11q13) in this patient is of particular interest. It may represent a contiguous gene syndrome, or deletion of the normal allele leading to expression of a single recessive trimethylaminuria gene, or an unrelated association, such as in Noonan syndrome. However, recent development of mapping of flavin-containing monooxygenase 2 (FMO2), the likely enzyme that is defective in fish odor syndrome, to chromosome 1q probably excludes pathogenetic association of fish odor syndrome with the Prader-Willi syndrome.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 15 , Metabolism, Inborn Errors/urine , Methylamines/urine , Prader-Willi Syndrome/genetics , Animals , Child , Choline/administration & dosage , Diet , Female , Fishes , Humans , Odorants , Oxidoreductases, N-Demethylating/deficiency , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL
...