Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
1.
Cell ; 154(3): 651-63, 2013 Aug 01.
Article in English | MEDLINE | ID: mdl-23911327

ABSTRACT

Vessel sprouting by migrating tip and proliferating stalk endothelial cells (ECs) is controlled by genetic signals (such as Notch), but it is unknown whether metabolism also regulates this process. Here, we show that ECs relied on glycolysis rather than on oxidative phosphorylation for ATP production and that loss of the glycolytic activator PFKFB3 in ECs impaired vessel formation. Mechanistically, PFKFB3 not only regulated EC proliferation but also controlled the formation of filopodia/lamellipodia and directional migration, in part by compartmentalizing with F-actin in motile protrusions. Mosaic in vitro and in vivo sprouting assays further revealed that PFKFB3 overexpression overruled the pro-stalk activity of Notch, whereas PFKFB3 deficiency impaired tip cell formation upon Notch blockade, implying that glycolysis regulates vessel branching.


Subject(s)
Endothelial Cells/metabolism , Glycolysis , Neovascularization, Physiologic , Phosphofructokinase-2/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Endothelial Cells/cytology , Female , Gene Deletion , Gene Silencing , Humans , Male , Mice , Mice, Inbred C57BL , Phosphofructokinase-2/genetics , Pseudopodia/metabolism , Zebrafish
2.
Proc Natl Acad Sci U S A ; 120(43): e2301733120, 2023 Oct 24.
Article in English | MEDLINE | ID: mdl-37862382

ABSTRACT

Retinal pigment epithelium (RPE) cells have to phagocytose shed photoreceptor outer segments (POS) on a daily basis over the lifetime of an organism, but the mechanisms involved in the digestion and recycling of POS lipids are poorly understood. Although it was frequently assumed that peroxisomes may play an essential role, this was never investigated. Here, we show that global as well as RPE-selective loss of peroxisomal ß-oxidation in multifunctional protein 2 (MFP2) knockout mice impairs the digestive function of lysosomes in the RPE at a very early age, followed by RPE degeneration. This was accompanied by prolonged mammalian target of rapamycin activation, lipid deregulation, and mitochondrial structural anomalies without, however, causing oxidative stress or energy shortage. The RPE degeneration caused secondary photoreceptor death. Notably, the deterioration of the RPE did not occur in an Mfp2/rd1 mutant mouse line, characterized by absent POS shedding. Our findings prove that peroxisomal ß-oxidation in the RPE is essential for handling the polyunsaturated fatty acids present in ingested POS and shed light on retinopathy in patients with peroxisomal disorders. Our data also have implications for gene therapy development as they highlight the importance of targeting the RPE in addition to the photoreceptor cells.


Subject(s)
Lysosomes , Retinal Pigment Epithelium , Mice , Humans , Animals , Retinal Pigment Epithelium/metabolism , Lysosomes/metabolism , Phagocytosis/genetics , Oxidative Stress , Mice, Knockout , Mammals
3.
Hum Mol Genet ; 31(21): 3729-3740, 2022 10 28.
Article in English | MEDLINE | ID: mdl-35652444

ABSTRACT

Rabenosyn (RBSN) is a conserved endosomal protein necessary for regulating internalized cargo. Here, we present clinical, genetic, cellular and biochemical evidence that two distinct RBSN missense variants are responsible for a novel Mendelian disorder consisting of progressive muscle weakness, facial dysmorphisms, ophthalmoplegia and intellectual disability. Using exome sequencing, we identified recessively acting germline alleles p.Arg180Gly and p.Gly183Arg, which are both situated in the FYVE domain of RBSN. We find that these variants abrogate binding to its cognate substrate phosphatidylinositol 3-phosphate (PI3P) and thus prevent its translocation to early endosomes. Although the endosomal recycling pathway was unaltered, mutant p.Gly183Arg patient fibroblasts show accumulation of cargo tagged for lysosomal degradation. Our results suggest that these variants are separation-of-function alleles, which cause a delay in endosomal maturation without affecting cargo recycling. We conclude that distinct germline mutations in RBSN cause non-overlapping phenotypes with specific and discrete endolysosomal cellular defects.


Subject(s)
Endosomes , Intellectual Disability , Vesicular Transport Proteins , Humans , Alleles , Endosomes/genetics , Endosomes/metabolism , Intellectual Disability/genetics , Lysosomes/genetics , Lysosomes/metabolism , Mutation , Protein Transport/genetics , Vesicular Transport Proteins/genetics
4.
Crit Care ; 25(1): 252, 2021 07 17.
Article in English | MEDLINE | ID: mdl-34274000

ABSTRACT

BACKGROUND: Muscle weakness is a complication of critical illness which hampers recovery. In critically ill mice, supplementation with the ketone body 3-hydroxybutyrate has been shown to improve muscle force and to normalize illness-induced hypocholesterolemia. We hypothesized that altered cholesterol homeostasis is involved in development of critical illness-induced muscle weakness and that this pathway can be affected by 3-hydroxybutyrate. METHODS: In both human critically ill patients and septic mice, the association between circulating cholesterol concentrations and muscle weakness was assessed. In septic mice, the impact of 3-hydroxybutyrate supplementation on cholesterol homeostasis was evaluated with use of tracer technology and through analysis of markers of cholesterol metabolism and downstream pathways. RESULTS: Serum cholesterol concentrations were lower in weak than in non-weak critically ill patients, and in multivariable analysis adjusting for baseline risk factors, serum cholesterol was inversely correlated with weakness. In septic mice, plasma cholesterol correlated positively with muscle force. In septic mice, exogenous 3-hydroxybutyrate increased plasma cholesterol and altered cholesterol homeostasis, by normalization of plasma mevalonate and elevation of muscular, but not hepatic, expression of cholesterol synthesis genes. In septic mice, tracer technology revealed that 3-hydroxybutyrate was preferentially taken up by muscle and metabolized into cholesterol precursor mevalonate, rather than TCA metabolites. The 3-hydroxybutyrate protection against weakness was not related to ubiquinone or downstream myofiber mitochondrial function, whereas cholesterol content in myofibers was increased. CONCLUSIONS: These findings point to a role for low cholesterol in critical illness-induced muscle weakness and to a protective mechanism-of-action for 3-hydroxybutyrate supplementation.


Subject(s)
Cholesterol/analysis , Homeostasis/drug effects , 3-Hydroxybutyric Acid , Aged , Aged, 80 and over , Animals , Cholesterol/metabolism , Critical Illness/therapy , Disease Models, Animal , Female , Humans , Lipid Metabolism/drug effects , Male , Mice , Mice, Inbred C57BL/metabolism , Mice, Inbred C57BL/physiology , Middle Aged , Multivariate Analysis , Muscle Weakness/physiopathology
5.
Nature ; 520(7546): 192-197, 2015 Apr 09.
Article in English | MEDLINE | ID: mdl-25830893

ABSTRACT

The metabolism of endothelial cells during vessel sprouting remains poorly studied. Here we report that endothelial loss of CPT1A, a rate-limiting enzyme of fatty acid oxidation (FAO), causes vascular sprouting defects due to impaired proliferation, not migration, of human and murine endothelial cells. Reduction of FAO in endothelial cells did not cause energy depletion or disturb redox homeostasis, but impaired de novo nucleotide synthesis for DNA replication. Isotope labelling studies in control endothelial cells showed that fatty acid carbons substantially replenished the Krebs cycle, and were incorporated into aspartate (a nucleotide precursor), uridine monophosphate (a precursor of pyrimidine nucleoside triphosphates) and DNA. CPT1A silencing reduced these processes and depleted endothelial cell stores of aspartate and deoxyribonucleoside triphosphates. Acetate (metabolized to acetyl-CoA, thereby substituting for the depleted FAO-derived acetyl-CoA) or a nucleoside mix rescued the phenotype of CPT1A-silenced endothelial cells. Finally, CPT1 blockade inhibited pathological ocular angiogenesis in mice, suggesting a novel strategy for blocking angiogenesis.


Subject(s)
Carbon/metabolism , Endothelial Cells/metabolism , Fatty Acids/chemistry , Fatty Acids/metabolism , Nucleotides/biosynthesis , Acetic Acid/pharmacology , Adenosine Triphosphate/metabolism , Animals , Blood Vessels/cytology , Blood Vessels/drug effects , Blood Vessels/metabolism , Blood Vessels/pathology , Carnitine O-Palmitoyltransferase/antagonists & inhibitors , Carnitine O-Palmitoyltransferase/deficiency , Carnitine O-Palmitoyltransferase/genetics , Carnitine O-Palmitoyltransferase/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Citric Acid Cycle , DNA/biosynthesis , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Gene Silencing , Glucose/metabolism , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Mice , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Nucleotides/chemistry , Nucleotides/pharmacology , Oxidation-Reduction/drug effects , Retinopathy of Prematurity/drug therapy , Retinopathy of Prematurity/metabolism , Retinopathy of Prematurity/pathology
6.
Adv Exp Med Biol ; 1299: 19-30, 2020.
Article in English | MEDLINE | ID: mdl-33417204

ABSTRACT

Peroxisomes are multifunctional organelles best known for their role in cellular lipid and hydrogen peroxide metabolism. In this chapter, we review and discuss the diverse functions of this organelle in brain physiology and neurodegeneration, with a particular focus on oxidative stress. We first briefly summarize what is known about the various nexuses among peroxisomes, the central nervous system, oxidative stress, and neurodegenerative disease. Next, we provide a comprehensive overview of the complex interplay among peroxisomes, oxidative stress, and neurodegeneration in patients suffering from primary peroxisomal disorders. Particular examples that are discussed include the prototypic Zellweger spectrum disorders and X-linked adrenoleukodystrophy, the most prevalent peroxisomal disorder. Thereafter, we elaborate on secondary peroxisome dysfunction in more common neurodegenerative disorders, including Alzheimer's disease, Parkinson's disease, and multiple sclerosis. Finally, we highlight some issues and challenges that need to be addressed to progress towards therapies and prevention strategies preserving, normalizing, or improving peroxisome activity in patients suffering from neurodegenerative conditions.


Subject(s)
Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Oxidative Stress , Peroxisomes/metabolism , Peroxisomes/pathology , Adrenoleukodystrophy/metabolism , Adrenoleukodystrophy/pathology , Alzheimer Disease , Humans , Multiple Sclerosis , Parkinson Disease , Zellweger Syndrome/metabolism , Zellweger Syndrome/pathology
7.
Mol Cell Biochem ; 456(1-2): 53-62, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30604065

ABSTRACT

The retinal pathology in peroxisomal disorders suggests that peroxisomes are important to maintain retinal homeostasis and function. These ubiquitous cell organelles are mainly involved in lipid metabolism, which comprises α- and ß-oxidation and ether lipid synthesis. Although peroxisomes were extensively studied in liver, their role in the retina still remains to be elucidated. As a first step in gaining more insight into the role of peroxisomes in retinal physiology, we performed immunohistochemical stainings, immunoblotting and enzyme activity measurements to reveal the distribution of peroxisomes and peroxisomal lipid metabolizing enzymes in the murine retina. Whereas peroxisomes were detected in every retinal layer, we found a clear differential distribution of the peroxisomal lipid metabolizing enzymes in the neural retina compared to the retinal pigment epithelium. In particular, the ABC transporters that transfer lipid substrates into the organelle as well as several enzymes of the ß-oxidation pathway were enriched either in the neural retina or in the retinal pigment epithelium. In conclusion, our results strongly indicate that peroxisome function varies between different regions in the murine retina.


Subject(s)
Eye Proteins/metabolism , Lipid Metabolism/physiology , Peroxisomes/enzymology , Retina/enzymology , ATP-Binding Cassette Transporters/metabolism , Animals , Mice
8.
Crit Care ; 23(1): 236, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-31262340

ABSTRACT

BACKGROUND: ICU-acquired weakness is a debilitating consequence of prolonged critical illness that is associated with poor outcome. Recently, premorbid obesity has been shown to protect against such illness-induced muscle wasting and weakness. Here, we hypothesized that this protection was due to increased lipid and ketone availability. METHODS: In a centrally catheterized, fluid-resuscitated, antibiotic-treated mouse model of prolonged sepsis, we compared markers of lipolysis and fatty acid oxidation in lean and obese septic mice (n = 117). Next, we compared markers of muscle wasting and weakness in septic obese wild-type and adipose tissue-specific ATGL knockout (AAKO) mice (n = 73), in lean septic mice receiving either intravenous infusion of lipids or standard parenteral nutrition (PN) (n = 70), and in lean septic mice receiving standard PN supplemented with either the ketone body 3-hydroxybutyrate or isocaloric glucose (n = 49). RESULTS: Obese septic mice had more pronounced lipolysis (p ≤ 0.05), peripheral fatty acid oxidation (p ≤ 0.05), and ketogenesis (p ≤ 0.05) than lean mice. Blocking lipolysis in obese septic mice caused severely reduced muscle mass (32% loss vs. 15% in wild-type, p < 0.001) and specific maximal muscle force (59% loss vs. 0% in wild-type; p < 0.001). In contrast, intravenous infusion of lipids in lean septic mice maintained specific maximal muscle force up to healthy control levels (p = 0.6), whereas this was reduced with 28% in septic mice receiving standard PN (p = 0.006). Muscle mass was evenly reduced with 29% in both lean septic groups (p < 0.001). Lipid administration enhanced fatty acid oxidation (p ≤ 0.05) and ketogenesis (p < 0.001), but caused unfavorable liver steatosis (p = 0.01) and a deranged lipid profile (p ≤ 0.01). Supplementation of standard PN with 3-hydroxybutyrate also attenuated specific maximal muscle force up to healthy control levels (p = 0.1), but loss of muscle mass could not be prevented (25% loss in both septic groups; p < 0.001). Importantly, this intervention improved muscle regeneration markers (p ≤ 0.05) without the unfavorable side effects seen with lipid infusion. CONCLUSIONS: Obesity-induced muscle protection during sepsis is partly mediated by elevated mobilization and metabolism of endogenous fatty acids. Furthermore, increased availability of ketone bodies, either through ketogenesis or through parenteral infusion, appears to protect against sepsis-induced muscle weakness also in the lean.


Subject(s)
Adipose Tissue/physiopathology , Lipolysis/physiology , Muscle Weakness/etiology , Sepsis/complications , Animals , Disease Models, Animal , Fatty Acids/metabolism , Fatty Acids/pharmacokinetics , Ketones/metabolism , Lipid Metabolism/physiology , Male , Mice , Muscle Weakness/metabolism , Muscle Weakness/physiopathology , Obesity/physiopathology , Protective Factors , Sepsis/metabolism , Sepsis/physiopathology
9.
J Biol Chem ; 292(43): 17718-17730, 2017 10 27.
Article in English | MEDLINE | ID: mdl-28887304

ABSTRACT

Tetratricopeptide repeat (TPR) domains are ubiquitous structural motifs that mediate protein-protein interactions. For example, the TPR domains in the peroxisomal import receptor PEX5 enable binding to a range of type 1 peroxisomal targeting signal motifs. A homolog of PEX5, tetratricopeptide repeat-containing Rab8b-interacting protein (TRIP8b), binds to and functions as an auxiliary subunit of hyperpolarization-activated cyclic nucleotide (HCN)-gated channels. Given the similarity between TRIP8b and PEX5, this difference in function raises the question of what mechanism accounts for their binding specificity. In this report, we found that the cyclic nucleotide-binding domain and the C terminus of the HCN channel are critical for conferring specificity to TRIP8b binding. We show that TRIP8b binds the HCN cyclic nucleotide-binding domain through a 37-residue domain and the HCN C terminus through the TPR domains. Using a combination of fluorescence polarization- and co-immunoprecipitation-based assays, we establish that binding at either site increases affinity at the other. Thus, allosteric coupling of the TRIP8b TPR domains both promotes binding to HCN channels and limits binding to type 1 peroxisomal targeting signal substrates. These results raise the possibility that other TPR domains may be similarly influenced by allosteric mechanisms as a general feature of protein-protein interactions.


Subject(s)
Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Protein Subunits/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Allosteric Regulation/physiology , Binding Sites , HEK293 Cells , Humans , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/genetics , Protein Subunits/genetics , Receptors, Cytoplasmic and Nuclear/genetics
10.
Immunology ; 153(3): 342-356, 2018 03.
Article in English | MEDLINE | ID: mdl-28940384

ABSTRACT

Macrophage activation is characterized by pronounced metabolic adaptation. Classically activated macrophages show decreased rates of mitochondrial fatty acid oxidation and oxidative phosphorylation and acquire a glycolytic state together with their pro-inflammatory phenotype. In contrast, alternatively activated macrophages require oxidative phosphorylation and mitochondrial fatty acid oxidation for their anti-inflammatory function. Although it is evident that mitochondrial metabolism is regulated during macrophage polarization and essential for macrophage function, little is known on the regulation and role of peroxisomal ß-oxidation during macrophage activation. In this study, we show that peroxisomal ß-oxidation is strongly decreased in classically activated bone-marrow-derived macrophages (BMDM) and mildly induced in alternatively activated BMDM. To examine the role of peroxisomal ß-oxidation in macrophages, we used Mfp2-/- BMDM lacking the key enzyme of this pathway. Impairment of peroxisomal ß-oxidation in Mfp2-/- BMDM did not cause lipid accumulation but rather an altered distribution of lipid species with very-long-chain fatty acids accumulating in the triglyceride and phospholipid fraction. These lipid alterations in Mfp2-/- macrophages led to decreased inflammatory activation of Mfp2-/- BMDM and peritoneal macrophages evidenced by impaired production of several inflammatory cytokines and chemokines, but did not affect anti-inflammatory polarization. The disturbed inflammatory responses of Mfp2-/- macrophages did not affect immune cell infiltration, as mice with selective elimination of MFP2 from myeloid cells showed normal monocyte and neutrophil influx upon challenge with zymosan. Together, these data demonstrate that peroxisomal ß-oxidation is involved in fine-tuning the phenotype of macrophages, probably by influencing the dynamic lipid profile during macrophage polarization.


Subject(s)
Homeostasis/immunology , Inflammation/immunology , Lipids/immunology , Macrophages/immunology , Animals , Cytokines/immunology , Macrophage Activation/immunology , Mice , Monocytes/immunology , Neutrophils/immunology , Oxidation-Reduction , Oxidative Phosphorylation , Phenotype
11.
Mol Ther ; 25(11): 2513-2525, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28822689

ABSTRACT

A causal role of hypercholesterolemia in non-ischemic heart failure has never been demonstrated. Adeno-associated viral serotype 8 (AAV8)-low-density lipoprotein receptor (AAV8-LDLr) gene transfer was performed in LDLr-deficient mice without and with pressure overload induced by transverse aortic constriction (TAC). AAV8-LDLr gene therapy resulted in an 82.8% (p < 0.0001) reduction of plasma cholesterol compared with controls. Mortality rate was lower (p < 0.05) in AAV8-LDLr TAC mice compared with control TAC mice (hazard ratio for mortality 0.457, 95% confidence interval [CI] 0.237-0.882) during 8 weeks of follow-up. AAV8-LDLr gene therapy attenuated cardiac hypertrophy, reduced interstitial and perivascular fibrosis, and decreased lung congestion in TAC mice. Cardiac function, quantified by invasive hemodynamic measurements and magnetic resonance imaging, was significantly improved 8 weeks after sham operation or after TAC in AAV8-LDLr mice compared with respective control groups. Myocardial protein levels of mammalian target of rapamycin and of acetyl-coenzyme A carboxylase were strikingly decreased following cholesterol lowering in mice without and with pressure overload. AAV8-LDLr therapy potently reduced cardiac glucose uptake and counteracted metabolic remodeling following pressure overload. Furthermore, oxidative stress and myocardial apoptosis were decreased following AAV8-LDLr therapy in mice with pressure overload. In conclusion, cholesterol-lowering gene therapy potently counteracts structural and metabolic remodeling, and enhances cardiac function.


Subject(s)
Cardiomegaly/therapy , Cardiomyopathies/therapy , Cholesterol/metabolism , Genetic Therapy/methods , Genetic Vectors/metabolism , Receptors, LDL/genetics , Acetyl-CoA C-Acetyltransferase/genetics , Acetyl-CoA C-Acetyltransferase/metabolism , Animals , Aorta/surgery , Biomarkers/metabolism , Cardiomegaly/etiology , Cardiomegaly/metabolism , Cardiomegaly/mortality , Cardiomyopathies/etiology , Cardiomyopathies/metabolism , Cardiomyopathies/mortality , Constriction, Pathologic/complications , Constriction, Pathologic/metabolism , Constriction, Pathologic/pathology , Dependovirus/genetics , Dependovirus/metabolism , Female , Gene Expression , Genetic Vectors/administration & dosage , Genetic Vectors/chemistry , Heart Function Tests , Hemodynamics , Mice , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Receptors, LDL/deficiency , Survival Analysis , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
12.
J Lipid Res ; 58(1): 42-59, 2017 01.
Article in English | MEDLINE | ID: mdl-27881717

ABSTRACT

1-Deoxysphingolipids (deoxySLs) are atypical sphingolipids that are elevated in the plasma of patients with type 2 diabetes and hereditary sensory and autonomic neuropathy type 1 (HSAN1). Clinically, diabetic neuropathy and HSAN1 are very similar, suggesting the involvement of deoxySLs in the pathology of both diseases. However, very little is known about the biology of these lipids and the underlying pathomechanism. We synthesized an alkyne analog of 1-deoxysphinganine (doxSA), the metabolic precursor of all deoxySLs, to trace the metabolism and localization of deoxySLs. Our results indicate that the metabolism of these lipids is restricted to only some lipid species and that they are not converted to canonical sphingolipids or fatty acids. Furthermore, exogenously added alkyne-doxSA [(2S,3R)-2-aminooctadec-17-yn-3-ol] localized to mitochondria, causing mitochondrial fragmentation and dysfunction. The induced mitochondrial toxicity was also shown for natural doxSA, but not for sphinganine, and was rescued by inhibition of ceramide synthase activity. Our findings therefore indicate that mitochondrial enrichment of an N-acylated doxSA metabolite may contribute to the neurotoxicity seen in diabetic neuropathy and HSAN1. Hence, we provide a potential explanation for the characteristic vulnerability of peripheral nerves to elevated levels of deoxySLs.


Subject(s)
Diabetes Mellitus, Type 2/blood , Diabetic Neuropathies/blood , Hereditary Sensory and Autonomic Neuropathies/blood , Sphingolipids/blood , Animals , Diabetes Mellitus, Type 2/pathology , Diabetic Neuropathies/pathology , Hereditary Sensory and Autonomic Neuropathies/pathology , Humans , Lipids/blood , Male , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Oxidoreductases/metabolism , Peripheral Nerves/metabolism , Peripheral Nerves/pathology , Sphingolipids/chemical synthesis , Sphingolipids/pharmacology
13.
Biochim Biophys Acta ; 1863(5): 956-70, 2016 May.
Article in English | MEDLINE | ID: mdl-26453805

ABSTRACT

The peroxisomal compartment in hepatocytes hosts several essential metabolic conversions. These are defective in peroxisomal disorders that are either caused by failure to import the enzymes in the organelle or by mutations in the enzymes or in transporters needed to transfer the substrates across the peroxisomal membrane. Hepatic pathology is one of the cardinal features in disorders of peroxisome biogenesis and peroxisomal ß-oxidation although it only rarely determines the clinical fate. In mouse models of these diseases liver pathologies also occur, although these are not always concordant with the human phenotype which might be due to differences in diet, expression of enzymes and backup mechanisms. Besides the morphological changes, we overview the impact of peroxisome malfunction on other cellular compartments including mitochondria and the ER. We further focus on the metabolic pathways that are affected such as bile acid formation, and dicarboxylic acid and branched chain fatty acid degradation. It appears that the association between deregulated metabolites and pathological events remains unclear.


Subject(s)
Liver/metabolism , Membrane Proteins/deficiency , Peroxisomal Disorders/metabolism , Peroxisomes/metabolism , Animals , Disease Models, Animal , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Gene Expression Regulation , Humans , Liver/pathology , Membrane Proteins/chemistry , Membrane Proteins/genetics , Metabolic Networks and Pathways/genetics , Mice , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Mutation , Peroxisomal Disorders/genetics , Peroxisomal Disorders/pathology , Peroxisomes/pathology , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport , Signal Transduction
14.
Gastroenterology ; 150(4): 875-87.e9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26752109

ABSTRACT

BACKGROUND & AIMS: Histamine sensitizes the nociceptor transient reporter potential channel V1 (TRPV1) and has been shown to contribute to visceral hypersensitivity in animals. We investigated the role of TRPV1 in irritable bowel syndrome (IBS) and evaluated if an antagonist of histamine receptor H1 (HRH1) could reduce symptoms of patients in a randomized placebo-controlled trial. METHODS: By using live calcium imaging, we compared activation of submucosal neurons by the TRPV1 agonist capsaicin in rectal biopsy specimens collected from 9 patients with IBS (ROME 3 criteria) and 15 healthy subjects. The sensitization of TRPV1 by histamine, its metabolite imidazole acetaldehyde, and supernatants from biopsy specimens was assessed by calcium imaging of mouse dorsal root ganglion neurons. We then performed a double-blind trial of patients with IBS (mean age, 31 y; range, 18-65 y; 34 female). After a 2-week run-in period, subjects were assigned randomly to groups given either the HRH1 antagonist ebastine (20 mg/day; n = 28) or placebo (n = 27) for 12 weeks. Rectal biopsy specimens were collected, barostat studies were performed, and symptoms were assessed (using the validated gastrointestinal symptom rating scale) before and after the 12-week period. Patients were followed up for an additional 2 weeks. Abdominal pain, symptom relief, and health-related quality of life were assessed on a weekly basis. The primary end point of the study was the effect of ebastine on the symptom score evoked by rectal distension. RESULTS: TRPV1 responses of submucosal neurons from patients with IBS were potentiated compared with those of healthy volunteers. Moreover, TRPV1 responses of submucosal neurons from healthy volunteers could be potentiated by their pre-incubation with histamine; this effect was blocked by the HRH1 antagonist pyrilamine. Supernatants from rectal biopsy specimens from patients with IBS, but not from the healthy volunteers, sensitized TRPV1 in mouse nociceptive dorsal root ganglion neurons via HRH1; this effect could be reproduced by histamine and imidazole acetaldehyde. Compared with subjects given placebo, those given ebastine had reduced visceral hypersensitivity, increased symptom relief (ebastine 46% vs placebo 13%; P = .024), and reduced abdominal pain scores (ebastine 39 ± 23 vs placebo 62 ± 22; P = .0004). CONCLUSIONS: In studies of rectal biopsy specimens from patients, we found that HRH1-mediated sensitization of TRPV1 is involved in IBS. Ebastine, an antagonist of HRH1, reduced visceral hypersensitivity, symptoms, and abdominal pain in patients with IBS. Inhibitors of this pathway might be developed as a new treatment approach for IBS. ClinicalTrials.gov no: NCT01144832.


Subject(s)
Analgesics/therapeutic use , Butyrophenones/therapeutic use , Gastrointestinal Agents/therapeutic use , Histamine H1 Antagonists/therapeutic use , Irritable Bowel Syndrome/drug therapy , Neurons/drug effects , Pain Threshold/drug effects , Piperidines/therapeutic use , Receptors, Histamine H1/drug effects , Rectum/innervation , TRPV Cation Channels/metabolism , Abdominal Pain/metabolism , Abdominal Pain/physiopathology , Abdominal Pain/prevention & control , Adolescent , Adult , Aged , Analgesics/adverse effects , Belgium , Biopsy , Butyrophenones/adverse effects , Calcium Signaling/drug effects , Double-Blind Method , Female , Gastrointestinal Agents/adverse effects , Histamine H1 Antagonists/adverse effects , Humans , Irritable Bowel Syndrome/diagnosis , Irritable Bowel Syndrome/metabolism , Irritable Bowel Syndrome/physiopathology , Male , Middle Aged , Neurons/metabolism , Pain Measurement , Piperidines/adverse effects , Quality of Life , Receptor Cross-Talk/drug effects , Receptors, Histamine H1/metabolism , Remission Induction , Surveys and Questionnaires , Time Factors , Treatment Outcome , Young Adult
15.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(9): 972-990, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28629946

ABSTRACT

2-Hydroxyacyl-CoA lyase (HACL1) is a key enzyme of the peroxisomal α-oxidation of phytanic acid. To better understand its role in health and disease, a mouse model lacking HACL1 was investigated. Under normal conditions, these mice did not display a particular phenotype. However, upon dietary administration of phytol, phytanic acid accumulated in tissues, mainly in liver and serum of KO mice. As a consequence of phytanic acid (or a metabolite) toxicity, KO mice displayed a significant weight loss, absence of abdominal white adipose tissue, enlarged and mottled liver and reduced hepatic glycogen and triglycerides. In addition, hepatic PPARα was activated. The central nervous system of the phytol-treated mice was apparently not affected. In addition, 2OH-FA did not accumulate in the central nervous system of HACL1 deficient mice, likely due to the presence in the endoplasmic reticulum of an alternate HACL1-unrelated lyase. The latter may serve as a backup system in certain tissues and account for the formation of pristanic acid in the phytol-fed KO mice. As the degradation of pristanic acid is also impaired, both phytanoyl- and pristanoyl-CoA levels are increased in liver, and the ω-oxidized metabolites are excreted in urine. In conclusion, HACL1 deficiency is not associated with a severe phenotype, but in combination with phytanic acid intake, the normal situation in man, it might present with phytanic acid elevation and resemble a Refsum like disorder.


Subject(s)
Enoyl-CoA Hydratase/deficiency , Enoyl-CoA Hydratase/metabolism , Lyases/metabolism , Phytol/pharmacology , Animals , Disease Models, Animal , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Fatty Acids/pharmacology , Female , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Knockout , Oxidation-Reduction , PPAR alpha/metabolism , Phytanic Acid/pharmacology
16.
Cell Biochem Funct ; 35(3): 144-155, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28370438

ABSTRACT

The presence of peroxisomes in mammalian intestine has been revealed formerly by catalase staining combined with electron microscopy. Despite the central role of intestine in lipid uptake and the established importance of peroxisomes in different lipid-related pathways, few data are available on the physiological role of peroxisomes in intestinal metabolism, more specifically, α-, ß-oxidation, and etherlipid synthesis. Hence, the peroxisomal compartment was analyzed in more detail in mouse intestine. On the basis of immunohistochemistry, the organelles are mainly confined to the epithelial cells. The expression of the classical peroxisome marker catalase was highest in the proximal part of jejunum and decreased along the tract. PEX14 showed a similar profile, but was still substantial expressed in large intestinal epithelium. Immunoblotting of epithelial cells, isolated from the different segments, showed also such gradient for some enzymes, ie, catalase, ACOX1, and D-specific multifunctional protein 2, and for the ABCD1 transporter, being high in small and low or absent in large intestine. Other peroxisomal enzymes (PHYH, HACL1, and ACAA1), the ABCD2 and ABCD3 transporters, and peroxins PEX13 and PEX14, however, did not follow this pattern, displaying rather constant signals throughout the intestinal epithelium. The small intestine displayed the highest peroxisomal ß-oxidation activity and is particularly active on dicarboxylic acids. Etherlipid synthesis was high in the large intestine, and colonic cells had the highest content of plasmalogens. Overall, these data suggest that peroxisomes exert different functions according to the intestinal segment.


Subject(s)
Fatty Acids/metabolism , Intestinal Mucosa/metabolism , Peroxisomes/metabolism , Animals , Catalase/metabolism , Cells, Cultured , Female , Intestinal Mucosa/cytology , Male , Mice , Mice, Inbred C57BL , Oxidation-Reduction , Signal Transduction
17.
Int J Mol Sci ; 18(7)2017 07 18.
Article in English | MEDLINE | ID: mdl-28718833

ABSTRACT

Studies evaluating the effects of high-saturated fat diets on cardiac function are most often confounded by diet-induced obesity and by systemic insulin resistance. We evaluated whether coconut oil, containing C12:0 and C14:0 as main fatty acids, aggravates pressure overload-induced cardiomyopathy induced by transverse aortic constriction (TAC) in C57BL/6 mice. Mortality rate after TAC was higher (p < 0.05) in 0.2% cholesterol 10% coconut oil diet-fed mice than in standard chow-fed mice (hazard ratio 2.32, 95% confidence interval 1.16 to 4.64) during eight weeks of follow-up. The effects of coconut oil on cardiac remodeling occurred in the absence of weight gain and of systemic insulin resistance. Wet lung weight was 1.76-fold (p < 0.01) higher in coconut oil mice than in standard chow mice. Myocardial capillary density (p < 0.001) was decreased, interstitial fibrosis was 1.88-fold (p < 0.001) higher, and systolic and diastolic function was worse in coconut oil mice than in standard chow mice. Myocardial glucose uptake was 1.86-fold (p < 0.001) higher in coconut oil mice and was accompanied by higher myocardial pyruvate dehydrogenase levels and higher acetyl-CoA carboxylase levels. The coconut oil diet increased oxidative stress. Myocardial triglycerides and free fatty acids were lower (p < 0.05) in coconut oil mice. In conclusion, coconut oil aggravates pressure overload-induced cardiomyopathy.


Subject(s)
Cardiomyopathies/pathology , Coconut Oil/adverse effects , Insulin Resistance , Myocardium/pathology , Obesity/pathology , Pressure , Animals , Aorta/pathology , Body Weight , Capillaries/pathology , Cardiomyopathies/blood , Cardiomyopathies/complications , Cardiomyopathies/physiopathology , Cholesterol , Constriction, Pathologic , Diastole , Diet , Female , Fibrosis , Glucose/metabolism , Hemodynamics , Hypertrophy, Right Ventricular/complications , Hypertrophy, Right Ventricular/pathology , Hypertrophy, Right Ventricular/physiopathology , Kaplan-Meier Estimate , Lung/pathology , Mice, Inbred C57BL , Oxidative Stress , Signal Transduction , Systole , Transforming Growth Factor beta1/metabolism
18.
Molecules ; 22(10)2017 10 13.
Article in English | MEDLINE | ID: mdl-29027957

ABSTRACT

Circulating heptadecanoic acid (C17:0) is reported to be a pathology risk/prognosis biomarker and a dietary biomarker. This pathology relationship has been shown to be reliably predictive even when independent of dietary contributions, suggesting that the endogenous biosynthesis of C17:0 is related to the pathological aetiology. Little is known about C17:0 biosynthesis, which tissues contribute to the circulating levels, and how C17:0 is related to pathology. Hacl1+/- mice were mated to obtain Hacl1-/- and Hacl1+/+ control mice. At 14 weeks, they were anesthetized for tissue collection and fatty acid analysis. Compared to Hacl1+/+, C15:0 was not significantly affected in any Hacl1-/- tissues. However, the Hacl1-/- plasma and liver C17:0 levels were significantly lower: ~26% and ~22%, respectively. No significant differences were seen in the different adipose tissues. To conclude, Hacl1 plays a significant role in the liver and plasma levels of C17:0, providing evidence it can be endogenously biosynthesized via alpha-oxidation. The strong inverse association of C17:0 with pathology raises the question whether there is a direct link between α-oxidation and these diseases. Currently, there is no clear evidence, warranting further research into the role of α-oxidation in relation to metabolic diseases.


Subject(s)
Carbon-Carbon Lyases/physiology , Enoyl-CoA Hydratase/genetics , Fatty Acids/genetics , Liver/enzymology , Peroxisomes/enzymology , Adipose Tissue/enzymology , Animals , Carbon-Carbon Lyases/genetics , Fatty Acids/biosynthesis , Humans , Mice , Mice, Knockout , Oxidation-Reduction , Peroxisomes/genetics , Tissue Distribution/genetics
19.
Traffic ; 15(1): 94-103, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24118911

ABSTRACT

Peroxisome maintenance depends on the import of nuclear-encoded proteins from the cytosol. The vast majority of these proteins is destined for the peroxisomal lumen and contains a C-terminal peroxisomal targeting signal, called PTS1. This targeting signal is recognized in the cytosol by the receptor PEX5. After docking at the peroxisomal membrane and release of the cargo into the organelle matrix, PEX5 is recycled to the cytosol through a process requiring monoubiquitination of an N-terminal, cytosolically exposed cysteine residue (Cys11 in the human protein). At present, the reason why a cysteine, and not a lysine residue, is the target of ubiquitination remains unclear. Here, we provide evidence that PTS1 protein import into human fibroblasts is a redox-sensitive process. We also demonstrate that Cys11 in human PEX5 functions as a redox switch that regulates PEX5 activity in response to intracellular oxidative stress. Finally, we show that exposure of human PEX5 to oxidized glutathione results in a ubiquitination-deficient PEX5 molecule, and that substitution of Cys11 by a lysine can counteract this effect. In summary, these findings reveal that the activity of PEX5, and hence PTS1 import, is controlled by the redox state of the cytosol. The potential physiological implications of these findings are discussed.


Subject(s)
Oxidative Stress , Peroxisomes/metabolism , Protein Sorting Signals , Receptors, Cytoplasmic and Nuclear/metabolism , Cell Line , Cysteine/genetics , Cysteine/metabolism , Cytosol/metabolism , Glutathione/metabolism , Humans , Oxidation-Reduction , Peroxisome-Targeting Signal 1 Receptor , Protein Transport , Receptors, Cytoplasmic and Nuclear/chemistry , Receptors, Cytoplasmic and Nuclear/genetics , Ubiquitination
20.
Biochim Biophys Acta ; 1853(2): 285-98, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25450972

ABSTRACT

The tight interrelationship between peroxisomes and mitochondria is illustrated by their cooperation in lipid metabolism, antiviral innate immunity and shared use of proteins executing organellar fission. In addition, we previously reported that disruption of peroxisome biogenesis in hepatocytes severely impacts on mitochondrial integrity, primarily damaging the inner membrane. Here we investigated the molecular impairments of the dysfunctional mitochondria in hepatocyte selective Pex5 knockout mice. First, by using blue native electrophoresis and in-gel activity stainings we showed that the respiratory complexes were differentially affected with reduction of complexes I and III and incomplete assembly of complex V, whereas complexes II and IV were normally active. This resulted in impaired oxygen consumption in cultured Pex5(-/-) hepatocytes. Second, mitochondrial DNA was depleted causing an imbalance in the expression of mitochondrial- and nuclear-encoded subunits of the respiratory chain complexes. Third, mitochondrial membranes showed increased permeability and fluidity despite reduced content of the polyunsaturated fatty acid docosahexaenoic acid. Fourth, the affected mitochondria in peroxisome deficient hepatocytes displayed increased oxidative stress. Acute deletion of PEX5 in vivo using adeno-Cre virus phenocopied these effects, indicating that mitochondrial perturbations closely follow the loss of functional peroxisomes in time. Likely to compensate for the functional impairments, the volume of the mitochondrial compartment was increased several folds. This was not driven by PGC-1α but mediated by activation of PPARα, possibly through c-myc overexpression. In conclusion, loss of peroxisomal metabolism in hepatocytes perturbs the mitochondrial inner membrane, depletes mitochondrial DNA and causes mitochondrial biogenesis independent of PGC-1α.


Subject(s)
DNA, Mitochondrial/metabolism , Hepatocytes/metabolism , Mitochondria/metabolism , Peroxisomes/metabolism , Transcription Factors/metabolism , Animals , Cell Compartmentation , Cell Proliferation , Cell Respiration , Electron Transport , Gene Deletion , Hepatocytes/ultrastructure , Lipids/chemistry , Membrane Fluidity , Mice, Knockout , Mitochondria/ultrastructure , Oxidation-Reduction , Oxidative Phosphorylation , Oxidative Stress , PPAR alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Peroxisome-Targeting Signal 1 Receptor , Protein Subunits/metabolism , Receptors, Cytoplasmic and Nuclear/deficiency , Receptors, Cytoplasmic and Nuclear/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL