Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters

Publication year range
1.
Int J Cancer ; 136(9): 2078-90, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25302649

ABSTRACT

Ovarian cancer (OC) is caused by genetic aberrations in networks that control growth and survival. Importantly, aberrant cancer metabolism interacts with oncogenic signaling providing additional drug targets. Tumors overexpress the lipogenic enzyme fatty acid synthase (FASN) and are inhibited by FASN blockers, whereas normal cells are FASN-negative and FASN-inhibitor-resistant. Here, we demonstrate that this holds true when ovarian/oviductal cells reside in their autochthonous tissues, whereas in culture they express FASN and are FASN-inhibitor-sensitive. Upon subculture, nonmalignant cells cease growth, express senescence-associated ß-galactosidase, lose FASN and become FASN-inhibitor-resistant. Immortalized ovarian/oviductal epithelial cell lines­although resisting senescence­reveal distinct growth activities, which correlate with FASN levels and FASN drug sensitivities. Accordingly, ectopic FASN stimulates growth in these cells. Moreover, FASN levels and lipogenic activities affect cellular lipid composition as demonstrated by thin-layer chromatography. Correlation between proliferation and FASN levels was finally evaluated in cancer cells such as HOC-7, which contain subclones with variable differentiation/senescence and corresponding FASN expression/FASN drug sensitivity. Interestingly, senescent phenotypes can be induced in parental HOC-7 by differentiating agents. In OC cells, FASN drugs induce cell cycle blockade in S and/or G2/M and stimulate apoptosis, whereas in normal cells they only cause cell cycle deceleration without apoptosis. Thus, normal cells, although growth-inhibited, may survive and recover from FASN blockade, whereas malignant cells get extinguished. FASN expression and FASN drug sensitivity are directly linked to cell growth and correlate with transformation/differentiation/senescence only indirectly. FASN is therefore a metabolic marker of cell proliferation rather than a marker of malignancy and is a useful target for future drug development.


Subject(s)
Biomarkers, Tumor/genetics , Cell Proliferation/genetics , Fatty Acid Synthase, Type I/genetics , Ovarian Neoplasms/genetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle , Cell Line , Cell Line, Tumor , Epithelial Cells/drug effects , Female , Humans , Ovarian Neoplasms/drug therapy
2.
Proc Natl Acad Sci U S A ; 108(23): 9691-6, 2011 Jun 07.
Article in English | MEDLINE | ID: mdl-21593415

ABSTRACT

Brain-specific carnitine palmitoyltransferase-1 (CPT-1c) is implicated in CNS control of food intake. In this article, we explore the role of hypothalamic CPT-1c in leptin's anorexigenic actions. We first show that adenoviral overexpression of CPT-1c in hypothalamic arcuate nucleus of rats increases food intake and concomitantly up-regulates orexigenic neuropeptide Y (NPY) and Bsx (a transcription factor of NPY). Then, we demonstrate that this overexpression antagonizes the anorectic actions induced by central leptin or compound cerulenin (an inhibitor of fatty acid synthase). The overexpression of CPT-1c also blocks leptin-induced down-regulations of NPY and Bsx. Furthermore, the anorectic actions of central leptin or cerulenin are impaired in mice with brain CPT-1c deleted. Both anorectic effects require elevated levels of hypothalamic arcuate nucleus (Arc) malonyl-CoA, a fatty acid-metabolism intermediate that has emerged as a mediator in hypothalamic control of food intake. Thus, these data suggest that CPT-1c is implicated in malonyl-CoA action in leptin's hypothalamic anorectic signaling pathways. Moreover, ceramide metabolism appears to play a role in leptin's central control of feeding. Leptin treatment decreases Arc ceramide levels, with the decrease being important in leptin-induced anorectic actions and down-regulations of NPY and Bsx. Of interest, our data indicate that leptin impacts ceramide metabolism through malonyl-CoA and CPT-1c, and ceramide de novo biosynthesis acts downstream of both malonyl-CoA and CPT-1c in mediating their effects on feeding and expressions of NPY and Bsx. In summary, we provide insights into the important roles of malonyl-CoA, CPT-1c, and ceramide metabolism in leptin's hypothalamic signaling pathways.


Subject(s)
Brain/metabolism , Carnitine O-Palmitoyltransferase/metabolism , Ceramides/metabolism , Eating/drug effects , Leptin/pharmacology , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/physiology , Blotting, Western , Body Weight/drug effects , Carnitine O-Palmitoyltransferase/genetics , Cerulenin/pharmacology , Humans , Hypothalamus/drug effects , Hypothalamus/metabolism , Hypothalamus/physiology , Leptin/administration & dosage , Male , Malonyl Coenzyme A/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Transcription Factors/metabolism
3.
J Biol Chem ; 287(25): 21224-32, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22539351

ABSTRACT

The brain-specific isoform carnitine palmitoyltransferase 1C (CPT1C) has been implicated in the hypothalamic regulation of food intake and energy homeostasis. Nevertheless, its molecular function is not completely understood, and its role in other brain areas is unknown. We demonstrate that CPT1C is expressed in pyramidal neurons of the hippocampus and is located in the endoplasmic reticulum throughout the neuron, even inside dendritic spines. We used molecular, cellular, and behavioral approaches to determine CPT1C function. First, we analyzed the implication of CPT1C in ceramide metabolism. CPT1C overexpression in primary hippocampal cultured neurons increased ceramide levels, whereas in CPT1C-deficient neurons, ceramide levels were diminished. Correspondingly, CPT1C knock-out (KO) mice showed reduced ceramide levels in the hippocampus. At the cellular level, CPT1C deficiency altered dendritic spine morphology by increasing immature filopodia and reducing mature mushroom and stubby spines. Total protrusion density and spine head area in mature spines were unaffected. Treatment of cultured neurons with exogenous ceramide reverted the KO phenotype, as did ectopic overexpression of CPT1C, indicating that CPT1C regulation of spine maturation is mediated by ceramide. To study the repercussions of the KO phenotype on cognition, we performed the hippocampus-dependent Morris water maze test on mice. Results show that CPT1C deficiency strongly impairs spatial learning. All of these results demonstrate that CPT1C regulates the levels of ceramide in the endoplasmic reticulum of hippocampal neurons, and this is a relevant mechanism for the correct maturation of dendritic spines and for proper spatial learning.


Subject(s)
Carnitine O-Palmitoyltransferase/biosynthesis , Ceramides/metabolism , Dendrites/enzymology , Energy Metabolism/physiology , Gene Expression Regulation, Enzymologic/physiology , Lipid Metabolism/physiology , Nerve Tissue Proteins/biosynthesis , Pyramidal Cells/enzymology , Animals , Behavior, Animal/physiology , Carnitine O-Palmitoyltransferase/genetics , Cells, Cultured , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/genetics , Lipid Metabolism, Inborn Errors/enzymology , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/pathology , Maze Learning/physiology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Pyramidal Cells/cytology
4.
Am J Physiol Endocrinol Metab ; 305(3): E336-47, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23736540

ABSTRACT

Carnitine palmitoyltransferase-1 (CPT-1) liver isoform, or CPT-1a, is implicated in CNS control of food intake. However, the exact brain nucleus site(s) in mediating this action of CPT-1a has not been identified. In this report, we assess the role of CPT-1a in hypothalamic ventromedial nucleus (VMN). We stereotaxically injected an adenoviral vector containing CPT-1a coding sequence into the VMN of rats to induce overexpression and activation of CPT-1a. The VMN-selective activation of CPT-1a induced an orexigenic effect, suggesting CPT-1a in the VMN is involved in the central control of feeding. Intracerebroventricular administration of etomoxir, a CPT-1 inhibitor, decreases food intake. Importantly, in the animals with VMN overexpression of a CPT-1a mutant that antagonizes the CPT-1 inhibition by etomoxir, the anorectic response to etomoxir was attenuated. This suggests that VMN is involved in mediating the anorectic effect of central inhibition of CPT-1a. In contrast, arcuate nucleus (Arc) overexpression of the mutant did not alter etomoxir-induced inhibition of food intake, suggesting that Arc CPT-1a does not play significant roles in this anorectic action. Furthermore, in the VMN, CPT-1a appears to act downstream of hypothalamic malonyl-CoA action of feeding. Finally, we show that in the VMN CPT-1 activity was altered in concert with fasting and refeeding states, supporting a physiological role of CPT-1a in mediating the control of feeding. All together, CPT-1a in the hypothalamic VMN appears to play an important role in central control of food intake. VMN-selective modulation of CPT-1a activity may therefore be a promising strategy in controlling food intake and maintaining normal body weight.


Subject(s)
Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/physiology , Eating/physiology , Ventromedial Hypothalamic Nucleus/enzymology , Ventromedial Hypothalamic Nucleus/physiology , Acyl Coenzyme A/metabolism , Animals , Appetite Depressants/pharmacology , Arcuate Nucleus of Hypothalamus/metabolism , Blotting, Western , Body Weight/physiology , Carnitine/analogs & derivatives , Carnitine/metabolism , Dependovirus , Enzyme Activation/drug effects , Epoxy Compounds/pharmacology , Fasting/physiology , Genetic Vectors , Hypoglycemic Agents/pharmacology , Injections, Intraventricular , Male , Malonyl Coenzyme A/physiology , Rats , Rats, Sprague-Dawley
5.
Chirality ; 25(5): 281-7, 2013 May.
Article in English | MEDLINE | ID: mdl-23620264

ABSTRACT

C75 is a synthetic compound described as having antitumoral properties. It produces hypophagia and weight loss in rodents, limiting its use in cancer therapy but identifying it as a potential anti-obesity drug. C75 is a fatty acid synthase (FAS) inhibitor and, through its coenzyme A (CoA) derivative, it acts as a carnitine palmitoyltransferase (CPT) 1 inhibitor. Racemic mixtures of C75 have been used in all the previous studies; however, the potential different biological activities of C75 enantiomers have not been examined yet. To address this question we synthesized the two C75 enantiomers separately. Our results showed that (-)-C75 inhibits FAS activity in vitro and has a cytotoxic effect on tumor cell lines, without affecting food consumption. (+)-C75 inhibits CPT1 and its administration produces anorexia, suggesting that central inhibition of CPT1 is essential for the anorectic effect of C75. The differential activity of C75 enantiomers may lead to the development of potential new specific drugs for cancer and obesity.


Subject(s)
4-Butyrolactone/analogs & derivatives , Antineoplastic Agents/pharmacology , Appetite Depressants/pharmacology , 4-Butyrolactone/chemistry , 4-Butyrolactone/pharmacology , Animals , Antineoplastic Agents/chemistry , Appetite Depressants/chemistry , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Magnetic Resonance Spectroscopy , Male , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Electrospray Ionization , Stereoisomerism
6.
J Lipid Res ; 53(10): 2046-2056, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22847177

ABSTRACT

A novel lyase activity enzyme is characterized for the first time: HMG-CoA lyase-like1 (er-cHL), which is a close homolog of mitochondrial HMG-CoA lyase (mHL). Initial data show that there are nine mature transcripts for the novel gene HMGCLL1, although none of them has all its exons. The most abundant transcript is called "variant b," and it lacks exons 2 and 3. Moreover, a three-dimensional model of the novel enzyme is proposed. Colocalization studies show a dual location of the er-cHL in the endoplasmic reticulum (ER) and cytosol, but not in mitochondria or peroxisomes. Furthermore, the dissociation experiment suggests that it is a nonendoplasmic reticulum integral membrane protein. The kinetic parameters of er-cHL indicate that it has a lower V(max) and a higher substrate affinity than mHL. Protein expression and lyase activity were found in several tissues, and were particularly strong in lung and kidney. The occurrence of er-cHL in brain is surprising, as mHL has not been found there. Although mHL activity is clearly associated with energy metabolism, the results suggest that er-cHL is more closely related to another metabolic function, mostly at the pulmonary and brain level.


Subject(s)
Cytosol/enzymology , Endoplasmic Reticulum/enzymology , Oxo-Acid-Lyases/analysis , Oxo-Acid-Lyases/chemistry , Amino Acid Sequence , Cytosol/metabolism , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Mitochondria/enzymology , Mitochondria/metabolism , Molecular Sequence Data , Oxo-Acid-Lyases/genetics , Peroxisomes/enzymology , Peroxisomes/metabolism , Protein Splicing
7.
BMC Med Genet ; 13: 43, 2012 Jun 07.
Article in English | MEDLINE | ID: mdl-22676896

ABSTRACT

BACKGROUND: Cornelia de Lange syndrome (CdLS) is a dominantly inherited disorder characterized by facial dysmorphism, growth and cognitive impairment, limb malformations and multiple organ involvement. Mutations in NIPBL gene account for about 60% of patients with CdLS. This gene encodes a key regulator of the Cohesin complex, which controls sister chromatid segregation during both mitosis and meiosis. Turner syndrome (TS) results from the partial or complete absence of one of the X chromosomes, usually associated with congenital lymphedema, short stature, and gonadal dysgenesis. CASE PRESENTATION: Here we report a four-year-old female with CdLS due to a frameshift mutation in the NIPBL gene (c.1445_1448delGAGA), who also had a tissue-specific mosaic 45,X/46,XX karyotype. The patient showed a severe form of CdLS with craniofacial dysmorphism, pre- and post-natal growth delay, cardiovascular abnormalities, hirsutism and severe psychomotor retardation with behavioural problems. She also presented with minor clinical features consistent with TS, including peripheral lymphedema and webbed neck. The NIPBL mutation was present in the two tissues analysed from different embryonic origins (peripheral blood lymphocytes and oral mucosa epithelial cells). However, the percentage of cells with monosomy X was low and variable in tissues. These findings indicate that, ontogenically, the NIPBL mutation may have appeared before the mosaic monosomy X. CONCLUSIONS: The coexistence in several patients of these two rare disorders raises the issue of whether there is indeed a cause-effect association. The detailed clinical descriptions indicate predominant CdLS phenotype, although additional TS manifestations may appear in adolescence.


Subject(s)
De Lange Syndrome/genetics , Frameshift Mutation , Mosaicism , Proteins/genetics , Turner Syndrome/genetics , Cell Cycle Proteins , Child , Female , Humans , Lymphocytes , Mouth Mucosa , Severity of Illness Index
8.
Hepatology ; 53(3): 821-32, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21319201

ABSTRACT

UNLABELLED: Obesity-induced insulin resistance is associated with both ectopic lipid deposition and chronic, low-grade adipose tissue inflammation. Despite their excess fat, obese individuals show lower fatty-acid oxidation (FAO) rates. This has raised the question of whether burning off the excess fat could improve the obese metabolic phenotype. Here we used human-safe nonimmunoreactive adeno-associated viruses (AAV) to mediate long-term hepatic gene transfer of carnitine palmitoyltransferase 1A (CPT1A), the key enzyme in fatty-acid ß-oxidation, or its permanently active mutant form CPT1AM, to high-fat diet-treated and genetically obese mice. High-fat diet CPT1A- and, to a greater extent, CPT1AM-expressing mice showed an enhanced hepatic FAO which resulted in increased production of CO(2) , adenosine triphosphate, and ketone bodies. Notably, the increase in hepatic FAO not only reduced liver triacylglyceride content, inflammation, and reactive oxygen species levels but also systemically affected a decrease in epididymal adipose tissue weight and inflammation and improved insulin signaling in liver, adipose tissue, and muscle. Obesity-induced weight gain, increase in fasting blood glucose and insulin levels, and augmented expression of gluconeogenic genes were restored to normal only 3 months after AAV treatment. Thus, CPT1A- and, to a greater extent, CPT1AM-expressing mice were protected against obesity-induced weight gain, hepatic steatosis, diabetes, and obesity-induced insulin resistance. In addition, genetically obese db/db mice that expressed CPT1AM showed reduced glucose and insulin levels and liver steatosis. CONCLUSION: A chronic increase in liver FAO improves the obese metabolic phenotype, which indicates that AAV-mediated CPT1A expression could be a potential molecular therapy for obesity and diabetes.


Subject(s)
Carnitine O-Palmitoyltransferase/administration & dosage , Diabetes Mellitus/therapy , Fatty Acids/metabolism , Liver/metabolism , Obesity/therapy , Animals , Carnitine O-Palmitoyltransferase/genetics , Dependovirus/genetics , Dietary Fats/administration & dosage , Fatty Liver/metabolism , Fatty Liver/therapy , Genetic Therapy , Humans , Insulin Resistance/physiology , Male , Mice , Mice, Obese , Obesity/complications , Oxidation-Reduction , Triglycerides/metabolism
9.
Mol Biol Rep ; 39(4): 4777-85, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21952825

ABSTRACT

The genes HMGCS2 and HMGCL encode the two main enzymes for ketone-body synthesis, mitochondrial HMG-CoA synthase and HMG-CoA lyase. Here, we identify and describe possible splice variants of these genes in human tissues. We detected an alternative transcript of HMGCS2 carrying a deletion of exon 4, and two alternative transcripts of HMGCL with deletions of exons 5 and 6, and exons 5, 6 and 7, respectively. All splice variants maintained the reading frame. However, Western blot studies and overexpression measurements in eukaryotic or prokaryotic cell models did not reveal HL or mHS protein variants. Both genes showed a similar distribution of the inactive variants in different tissues. Surprisingly, the highest percentages were found in tissues where almost no ketone bodies are synthesized: heart, skeletal muscle and brain. Our results suggest that alternative splicing might coordinately block the two main enzymes of ketogenesis in specific human tissues.


Subject(s)
Alternative Splicing/genetics , Biosynthetic Pathways/genetics , Hydroxymethylglutaryl-CoA Synthase/genetics , Ketone Bodies/biosynthesis , Mitochondria/enzymology , Mitochondria/genetics , Oxo-Acid-Lyases/genetics , Blotting, Western , Computational Biology , HEK293 Cells , Humans , Hydroxymethylglutaryl-CoA Synthase/chemistry , Hydroxymethylglutaryl-CoA Synthase/metabolism , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Oxo-Acid-Lyases/chemistry , Oxo-Acid-Lyases/metabolism , Protein Structure, Secondary , Real-Time Polymerase Chain Reaction
10.
Am J Physiol Regul Integr Comp Physiol ; 301(1): R209-17, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21508288

ABSTRACT

Hypothalamic fatty acid metabolism is involved in central nervous system controls of feeding and energy balance. Malonyl-CoA, an intermediate of fatty acid biosynthesis, is emerging as a significant player in these processes. Notably, hypothalamic malonyl-CoA has been implicated in leptin's feeding effect. Leptin treatment increases malonyl-CoA level in the hypothalamic arcuate nucleus (Arc), and this increase is required for leptin-induced decrease in food intake. However, the intracellular downstream mediators of malonyl-CoA's feeding effect have not been identified. A primary biochemical action of malonyl-CoA is the inhibition of the acyltransferase activity of carnitine palmitoyltransferase-1 (CPT-1). In the hypothalamus, the predominant isoform of CPT-1 that possesses the acyltransferase activity is CPT-1 liver type (CPT-1a). To address the role of CPT-1a in malonyl-CoA's anorectic action, we used a recombinant adenovirus expressing a mutant CPT-1a that is insensitive to malonyl-CoA inhibition. We show that Arc overexpression of the mutant CPT-1a blocked the malonyl-CoA-mediated inhibition of CPT-1 activity. However, the overexpression of this mutant did not affect the anorectic actions of leptin or central cerulenin for which an increase in Arc malonyl-CoA level is also required. Thus, CPT-1a does not appear to be involved in the malonyl-CoA's anorectic actions induced by leptin. Furthermore, long-chain fatty acyl-CoAs, substrates of CPT-1a, dissociate from malonyl-CoA's actions in the Arc under different feeding states. Together, our results suggest that Arc intracellular mechanisms of malonyl-CoA's anorectic actions induced by leptin are independent of CPT-1a. The data suggest that target(s), rather than CPT-1a, mediates malonyl-CoA action on feeding.


Subject(s)
Appetite Regulation/physiology , Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Hypothalamus/physiology , Leptin/physiology , Malonyl Coenzyme A/physiology , Acyltransferases/physiology , Animals , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/physiology , Cerulenin/metabolism , Energy Metabolism/physiology , Male , Models, Animal , Mutation/genetics , Rats , Rats, Sprague-Dawley
11.
J Biol Chem ; 284(34): 22840-52, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19553674

ABSTRACT

In addition to its essential role in permitting mitochondrial import and oxidation of long chain fatty acids, carnitine also functions as an acyl group acceptor that facilitates mitochondrial export of excess carbons in the form of acylcarnitines. Recent evidence suggests carnitine requirements increase under conditions of sustained metabolic stress. Accordingly, we hypothesized that carnitine insufficiency might contribute to mitochondrial dysfunction and obesity-related impairments in glucose tolerance. Consistent with this prediction whole body carnitine diminution was identified as a common feature of insulin-resistant states such as advanced age, genetic diabetes, and diet-induced obesity. In rodents fed a lifelong (12 month) high fat diet, compromised carnitine status corresponded with increased skeletal muscle accumulation of acylcarnitine esters and diminished hepatic expression of carnitine biosynthetic genes. Diminished carnitine reserves in muscle of obese rats was accompanied by marked perturbations in mitochondrial fuel metabolism, including low rates of complete fatty acid oxidation, elevated incomplete beta-oxidation, and impaired substrate switching from fatty acid to pyruvate. These mitochondrial abnormalities were reversed by 8 weeks of oral carnitine supplementation, in concert with increased tissue efflux and urinary excretion of acetylcarnitine and improvement of whole body glucose tolerance. Acetylcarnitine is produced by the mitochondrial matrix enzyme, carnitine acetyltransferase (CrAT). A role for this enzyme in combating glucose intolerance was further supported by the finding that CrAT overexpression in primary human skeletal myocytes increased glucose uptake and attenuated lipid-induced suppression of glucose oxidation. These results implicate carnitine insufficiency and reduced CrAT activity as reversible components of the metabolic syndrome.


Subject(s)
Aging/physiology , Carnitine/physiology , Mitochondria, Muscle/metabolism , Overnutrition/physiopathology , Vitamin B Complex/physiology , Animals , Biological Transport/drug effects , Blotting, Western , Carnitine/analogs & derivatives , Carnitine/deficiency , Carnitine/metabolism , Carnitine/pharmacology , Carnitine O-Acetyltransferase/genetics , Carnitine O-Acetyltransferase/physiology , Cells, Cultured , Dietary Fats/adverse effects , Glucose Intolerance , Glucose Tolerance Test , Humans , Lipid Metabolism/drug effects , Male , Mitochondria, Muscle/drug effects , Mixed Function Oxygenases/genetics , Oxidative Phosphorylation , Random Allocation , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Vitamin B Complex/pharmacology , gamma-Butyrobetaine Dioxygenase
12.
Am J Med Genet A ; 152A(4): 924-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20358602

ABSTRACT

Cornelia de Lange syndrome (CdLS) manifests facial dysmorphic features, growth and cognitive impairment, and limb malformations. Mutations in three genes (NIPBL, SMC1A, and SMC3) of the cohesin complex and its regulators have been found in affected patients. Here, we present clinical and molecular characterization of 30 unrelated patients with CdLS. Eleven patients had mutations in NIPBL (37%) and three patients had mutations in SMC1A (10%), giving an overall rate of mutations of 47%. Several patients shared the same mutation in NIPBL (p.R827GfsX2) but had variable phenotypes, indicating the influence of modifiers in CdLS. Patients with NIPBL mutations had a more severe phenotype than those with mutations in SMC1A or those without identified mutations. However, a high incidence of palate defects was noted in patients with SMC1A mutations. In addition, we observed a similar phenotype in both male and female patients with SMC1A mutations. Finally, we report the first patient with an SMC1A mutation and the Sandifer complex.


Subject(s)
Cell Cycle Proteins/genetics , Chondroitin Sulfate Proteoglycans/genetics , Chromosomal Proteins, Non-Histone/genetics , De Lange Syndrome/genetics , Mutation/genetics , Proteins/genetics , Alleles , Cohort Studies , Female , Genotype , Humans , Male , Phenotype
13.
J Inherit Metab Dis ; 33(4): 405-10, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20532825

ABSTRACT

3-Hydroxy-3-methylglutaric aciduria is a rare human autosomal recessive disorder caused by deficiency of 3-hydroxy-3-methylglutaryl CoA lyase (HL). This mitochondrial enzyme catalyzes the common final step of leucine degradation and ketogenesis. Acute symptoms include vomiting, seizures and lethargy, accompanied by metabolic acidosis and hypoketotic hypoglycaemia. Such organs as the liver, brain, pancreas, and heart can also be involved. However, the pathophysiology of this disease is only partially understood. We measured mRNA levels, protein expression and enzyme activity of human HMG-CoA lyase from liver, kidney, pancreas, testis, heart, skeletal muscle, and brain. Surprisingly, the pancreas is, after the liver, the tissue with most HL activity. However, in heart and adult brain, HL activity was not detected in the mitochondrial fraction. These findings contribute to our understanding of the enzyme function and the consequences of its deficiency and suggest the need for assessment of pancreatic damage in these patients.


Subject(s)
Acids/urine , Gene Expression Regulation, Enzymologic , Meglutol/metabolism , Oxo-Acid-Lyases/genetics , Oxo-Acid-Lyases/metabolism , Point Mutation , Aged , Brain/enzymology , Enzyme Activation , Humans , Kidney/enzymology , Liver/enzymology , Male , Muscle, Skeletal/enzymology , Myocardium/enzymology , Organ Specificity , Pancreas/enzymology , RNA, Messenger/metabolism , Testis/enzymology
14.
Hum Mutat ; 30(3): E520-9, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19177531

ABSTRACT

3-Hydroxy-3-methylglutaric aciduria is a rare autosomal recessive genetic disorder that affects ketogenesis and L-leucine catabolism. The clinical acute symptoms include vomiting, convulsions, metabolic acidosis, hypoketotic hypoglycaemia and lethargy. To date, 33 mutations in 100 patients have been reported in the HMGCL gene. In this study 10 new mutations in 24 patients are described. They include: 5 missense mutations: c.109G>A, c.425C>T, c.521G>A, c.575T>C and c.598A>T, 2 nonsense mutations: c.242G>A and c.559G>T, one small deletion: c.853delC, and 2 mutations in intron regions: c.497+4A>G and c.750+1G>A. Two prevalent mutations were detected, 109G>T (E37X) in 38% of disease alleles analyzed and c.504_505delCT in 10% of them. Although patients are mainly of European origin (71%) and mostly Spanish (54%), the group is ethnically diverse and includes, for the first time, patients from Pakistan, Palestine and Ecuador. We also present a simple, efficient method to express the enzyme and we analyze the possible functional effects of missense mutations. The finding that all identified missense mutations cause a >95% decrease in the enzyme activity, indicates that the disease appears only in very severe genotypes."


Subject(s)
Meglutol/metabolism , Metabolism, Inborn Errors/genetics , Mutation , Oxo-Acid-Lyases/genetics , Alleles , Amino Acid Sequence , Arabs/genetics , Catalytic Domain/genetics , DNA Mutational Analysis , Ecuador , Europe , Gene Frequency , Genotype , Humans , Kinetics , Metabolism, Inborn Errors/ethnology , Metabolism, Inborn Errors/pathology , Models, Molecular , Molecular Sequence Data , Mutation, Missense , Oxo-Acid-Lyases/chemistry , Oxo-Acid-Lyases/metabolism , Pakistan , Protein Structure, Tertiary , Sequence Homology, Amino Acid
15.
J Lipid Res ; 50(9): 1789-99, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19429947

ABSTRACT

Carnitine palmitoyltransferase 1 (CPT1) catalyzes the first step in long-chain fatty acid import into mitochondria, and it is believed to be rate limiting for beta-oxidation of fatty acids. However, in muscle, other proteins may collaborate with CPT1. Fatty acid translocase/CD36 (FAT/CD36) may interact with CPT1 and contribute to fatty acid import into mitochondria in muscle. Here, we demonstrate that another membrane-bound fatty acid binding protein, fatty acid transport protein 1 (FATP1), collaborates with CPT1 for fatty acid import into mitochondria. Overexpression of FATP1 using adenovirus in L6E9 myotubes increased both fatty acid oxidation and palmitate esterification into triacylglycerides. Moreover, immunocytochemistry assays in transfected L6E9 myotubes showed that FATP1 was present in mitochondria and coimmunoprecipitated with CPT1 in L6E9 myotubes and rat skeletal muscle in vivo. The cooverexpression of FATP1 and CPT1 also enhanced mitochondrial fatty acid oxidation, similar to the cooverexpression of FAT/CD36 and CPT1. However, etomoxir, an irreversible inhibitor of CPT1, blocked all these effects. These data reveal that FATP1, like FAT/CD36, is associated with mitochondria and has a role in mitochondrial oxidation of fatty acids.


Subject(s)
Fatty Acid Transport Proteins/metabolism , Fatty Acids/metabolism , Mitochondria/metabolism , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Animals , CD36 Antigens/metabolism , Carnitine O-Palmitoyltransferase/metabolism , Cell Line , Coenzyme A Ligases/metabolism , Gene Expression Regulation , Humans , Immunoprecipitation , Lipid Metabolism , Male , Mice , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Oxidation-Reduction , Protein Transport , Rats
16.
FEMS Microbiol Lett ; 267(2): 159-66, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17156120

ABSTRACT

The wine yeast Saccharomyces cerevisiae is central in the production of aroma compounds during fermentation. Some of the most important yeast-derived aroma compounds produced are esters. The esters ethyl acetate and isoamyl acetate are formed from alcohols and acetyl-CoA in a reaction catalysed by alcohol acetyltransferases. The pool of acetyl-CoA available in yeast cells could play a key role in the development of ester aromas. Carnitine acetyltransferases catalyse the reversible reaction between carnitine and acetyl-CoA to form acetylcarnitine and free CoA. This reaction is important in transferring activated acetyl groups to the mitochondria and in regulating the acetyl-CoA/CoA pools within the cell. We investigated the effect of overexpressing CAT2, which encodes the major mitochondrial and peroxisomal carnitine acetyltransferase, on the formation of esters and other flavour compounds during fermentation. We also overexpressed a modified CAT2 that results in a protein that localizes to the cytosol. In general, the overexpression of both forms of CAT2 resulted in a reduction in ester concentrations, especially in ethyl acetate and isoamyl acetate. We hypothesize that overproduction of Cat2p favours the formation of acetylcarnitine and CoA and therefore limits the precursor for ester production. Carnitine acetyltransferase expression could potentially to be used successfully in order to modulate wine flavour.


Subject(s)
Carnitine O-Acetyltransferase/metabolism , Odorants , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/enzymology , Wine , Acetates/metabolism , Carnitine O-Acetyltransferase/chemistry , Carnitine O-Acetyltransferase/genetics , Esters/metabolism , Fermentation , Pentanols/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
17.
Eur J Med Chem ; 131: 207-221, 2017 May 05.
Article in English | MEDLINE | ID: mdl-28324785

ABSTRACT

C75 is a synthetic anticancer drug that inhibits fatty acid synthase (FAS) and shows a potent anorexigenic side effect. In order to find new cytotoxic compounds that do not impact food intake, we synthesized a new family of C75 derivatives. The most promising anticancer compound among them was UB006 ((4SR,5SR)-4-(hydroxymethyl)-3-methylene-5-octyldihydrofuran-2(3H)-one). The effects of this compound on cytotoxicity, food intake and body weight were studied in UB006 racemic mixture and in both its enantiomers separately. The results showed that both enantiomers inhibit FAS activity and have potent cytotoxic effects in several tumour cell lines, such as the ovarian cell cancer line OVCAR-3. The (-)-UB006 enantiomer's cytotoxic effect on OVCAR-3 was 40-fold higher than that of racemic C75, and 2- and 38-fold higher than that of the racemic mixture and its opposite enantiomer, respectively. This cytotoxic effect on the OVCAR-3 cell line involves mechanisms that reduce mitochondrial respiratory capacity and ATP production, DDIT4/REDD1 upregulation, mTOR activity inhibition, and caspase-3 activation, resulting in apoptosis. In addition, central and peripheral administration of (+)-UB006 or (-)-UB006 into rats and mice did not affect food intake or body weight. Altogether, our data support the discovery of a new potential anticancer compound (-)-UB006 that has no anorexigenic side effects.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Fatty Acid Synthases/antagonists & inhibitors , Furans/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Body Weight/drug effects , Cell Survival/drug effects , Cells, Cultured , Eating/drug effects , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Fatty Acid Synthases/metabolism , Furans/chemistry , Humans , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley
18.
Diabetes ; 54(2): 462-71, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15677504

ABSTRACT

Carnitine palmitoyltransferase I, which is expressed in the pancreas as the liver isoform (LCPTI), catalyzes the rate-limiting step in the transport of fatty acids into the mitochondria for their oxidation. Malonyl-CoA derived from glucose metabolism regulates fatty acid oxidation by inhibiting LCPTI. To examine directly whether the availability of long-chain fatty acyl-CoA (LC-CoA) affects the regulation of insulin secretion in the beta-cell and whether malonyl-CoA may act as a metabolic coupling factor in the beta-cell, we infected INS(832/13) cells and rat islets with an adenovirus encoding a mutant form of LCPTI (Ad-LCPTI M593S) that is insensitive to malonyl-CoA. In Ad-LCPTI M593S-infected INS(832/13) cells, LCPTI activity increased sixfold. This was associated with enhanced fatty acid oxidation, at any glucose concentration, and a 60% suppression of glucose-stimulated insulin secretion (GSIS). In isolated rat islets in which LCPTI M593S was overexpressed, GSIS decreased 40%. The impairment of GSIS in Ad-LCPTI M593S-infected INS(832/13) cells was not recovered when cells were incubated with 0.25 mmol/l palmitate, indicating the deep metabolic influence of a nonregulated fatty acid oxidation system. At high glucose concentration, overexpression of a malonyl-CoA-insensitive form of LCPTI reduced partitioning of exogenous palmitate into lipid esterification products and decreased protein kinase C activation. Moreover, LCPTI M593S expression impaired K(ATP) channel-independent GSIS in INS(832/13) cells. The LCPTI M593S mutant caused more pronounced alterations in GSIS and lipid partitioning (fat oxidation, esterification, and the level of nonesterified palmitate) than LCPTI wt in INS(832/13) cells that were transduced with these constructs. The results provide direct support for the hypothesis that the malonyl-CoA/CPTI interaction is a component of a metabolic signaling network that controls insulin secretion.


Subject(s)
Carnitine O-Palmitoyltransferase/metabolism , Glucose/pharmacology , Insulin/metabolism , Islets of Langerhans/metabolism , Malonyl Coenzyme A/metabolism , Animals , Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Cell Line, Tumor , Insulin Secretion , Insulinoma , Islets of Langerhans/drug effects , Kinetics , Malonyl Coenzyme A/pharmacology , Pancreatic Neoplasms , Rats
19.
Biochem J ; 379(Pt 3): 777-84, 2004 May 01.
Article in English | MEDLINE | ID: mdl-14711372

ABSTRACT

CPT I (carnitine palmitoyltransferase I) catalyses the conversion of palmitoyl-CoA into palmitoylcarnitine in the presence of L-carnitine, facilitating the entry of fatty acids into mitochondria. We propose a 3-D (three-dimensional) structural model for L-CPT I (liver CPT I), based on the similarity of this enzyme to the recently crystallized mouse carnitine acetyltransferase. The model includes 607 of the 773 amino acids of L-CPT I, and the positions of carnitine, CoA and the palmitoyl group were assigned by superposition and docking analysis. Functional analysis of this 3-D model included the mutagenesis of several amino acids in order to identify putative catalytic residues. Mutants D477A, D567A and E590D showed reduced L-CPT I activity. In addition, individual mutation of amino acids forming the conserved Ser685-Thr686-Ser687 motif abolished enzyme activity in mutants T686A and S687A and altered K(m) and the catalytic efficiency for carnitine in mutant S685A. We conclude that the catalytic residues are His473 and Asp477, while Ser687 probably stabilizes the transition state. Several conserved lysines, i.e. Lys455, Lys505, Lys560 and Lys561, were also mutated. Only mutants K455A and K560A showed decreases in activity of 50%. The model rationalizes the finding of nine natural mutations in patients with hereditary L-CPT I deficiencies.


Subject(s)
Carnitine O-Acetyltransferase/chemistry , Carnitine O-Palmitoyltransferase/chemistry , Amino Acid Sequence , Amino Acids/metabolism , Animals , Binding Sites , Blotting, Western , Carnitine/metabolism , Carnitine O-Palmitoyltransferase/deficiency , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Coenzyme A/metabolism , Crystallization , Crystallography, X-Ray , Humans , Kinetics , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation, Missense/genetics , Palmitoyl Coenzyme A/metabolism , Protein Conformation , Rats , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism
20.
Diabetes ; 63(10): 3242-52, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24848071

ABSTRACT

Rigorous control of substrate oxidation by humoral factors is essential for maintaining metabolic homeostasis. During feeding and fasting cycles, carbohydrates and fatty acids are the two primary substrates in oxidative metabolism. Here, we report a novel role for the peptide hormone adropin in regulating substrate oxidation preferences. Plasma levels of adropin increase with feeding and decrease upon fasting. A comparison of whole-body substrate preference and skeletal muscle substrate oxidation in adropin knockout and transgenic mice suggests adropin promotes carbohydrate oxidation over fat oxidation. In muscle, adropin activates pyruvate dehydrogenase (PDH), which is rate limiting for glucose oxidation and suppresses carnitine palmitoyltransferase-1B (CPT-1B), a key enzyme in fatty acid oxidation. Adropin downregulates PDH kinase-4 (PDK4) that inhibits PDH, thereby increasing PDH activity. The molecular mechanisms of adropin's effects involve acetylation (suggesting inhibition) of the transcriptional coactivator PGC-1α, downregulating expression of Cpt1b and Pdk4. Increased PGC-1α acetylation by adropin may be mediated by inhibiting Sirtuin-1 (SIRT1), a PGC-1α deacetylase. Altered SIRT1 and PGC-1α activity appear to mediate aspects of adropin's metabolic actions in muscle. Similar outcomes were observed in fasted mice treated with synthetic adropin. Together, these results suggest a role for adropin in regulating muscle substrate preference under various nutritional states.


Subject(s)
Fasting/metabolism , Fatty Acids/metabolism , Muscle, Skeletal/metabolism , Proteins/metabolism , Animals , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Intercellular Signaling Peptides and Proteins , Male , Mice , Mice, Knockout , Oxidation-Reduction , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphorylation , Proteins/genetics , Sirtuin 1/genetics , Sirtuin 1/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL