Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 335
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Physiol Rev ; 98(1): 89-115, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29167332

ABSTRACT

Peroxisomes are highly dynamic intracellular organelles involved in a variety of metabolic functions essential for the metabolism of long-chain fatty acids, d-amino acids, and many polyamines. A byproduct of peroxisomal metabolism is the generation, and subsequent detoxification, of reactive oxygen and nitrogen species, particularly hydrogen peroxide (H2O2). Because of its relatively low reactivity (as a mild oxidant), H2O2 has a comparatively long intracellular half-life and a high diffusion rate, all of which makes H2O2 an efficient signaling molecule. Peroxisomes also have intricate connections to mitochondria, and both organelles appear to play important roles in regulating redox signaling pathways. Peroxisomal proteins are also subject to oxidative modification and inactivation by the reactive oxygen and nitrogen species they generate, but the peroxisomal LonP2 protease can selectively remove such oxidatively damaged proteins, thus prolonging the useful lifespan of the organelle. Peroxisomal homeostasis must adapt to the metabolic state of the cell, by a combination of peroxisome proliferation, the removal of excess or badly damaged organelles by autophagy (pexophagy), as well as by processes of peroxisome inheritance and motility. More recently the tumor suppressors ataxia telangiectasia mutate (ATM) and tuberous sclerosis complex (TSC), which regulate mTORC1 signaling, have been found to regulate pexophagy in response to variable levels of certain reactive oxygen and nitrogen species. It is now clear that any significant loss of peroxisome homeostasis can have devastating physiological consequences. Peroxisome dysregulation has been implicated in several metabolic diseases, and increasing evidence highlights the important role of diminished peroxisomal functions in aging processes.


Subject(s)
Homeostasis/physiology , Mitochondria/metabolism , Peroxisomes/metabolism , Proteostasis/physiology , Reactive Oxygen Species/metabolism , Animals , Homeostasis/drug effects , Humans , Hydrogen Peroxide/pharmacology , Peroxisomes/drug effects , Proteostasis/drug effects
2.
Nature ; 553(7687): 222-227, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29323298

ABSTRACT

Chromosomal translocations that generate in-frame oncogenic gene fusions are notable examples of the success of targeted cancer therapies. We have previously described gene fusions of FGFR3-TACC3 (F3-T3) in 3% of human glioblastoma cases. Subsequent studies have reported similar frequencies of F3-T3 in many other cancers, indicating that F3-T3 is a commonly occuring fusion across all tumour types. F3-T3 fusions are potent oncogenes that confer sensitivity to FGFR inhibitors, but the downstream oncogenic signalling pathways remain unknown. Here we show that human tumours with F3-T3 fusions cluster within transcriptional subgroups that are characterized by the activation of mitochondrial functions. F3-T3 activates oxidative phosphorylation and mitochondrial biogenesis and induces sensitivity to inhibitors of oxidative metabolism. Phosphorylation of the phosphopeptide PIN4 is an intermediate step in the signalling pathway of the activation of mitochondrial metabolism. The F3-T3-PIN4 axis triggers the biogenesis of peroxisomes and the synthesis of new proteins. The anabolic response converges on the PGC1α coactivator through the production of intracellular reactive oxygen species, which enables mitochondrial respiration and tumour growth. These data illustrate the oncogenic circuit engaged by F3-T3 and show that F3-T3-positive tumours rely on mitochondrial respiration, highlighting this pathway as a therapeutic opportunity for the treatment of tumours with F3-T3 fusions. We also provide insights into the genetic alterations that initiate the chain of metabolic responses that drive mitochondrial metabolism in cancer.


Subject(s)
Cell Respiration , Microtubule-Associated Proteins/genetics , Mitochondria/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Oncogene Proteins, Fusion/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Cell Respiration/drug effects , Cell Transformation, Neoplastic/drug effects , Female , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Male , Mice , Mitochondria/drug effects , Mitochondria/genetics , NIMA-Interacting Peptidylprolyl Isomerase/chemistry , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Organelle Biogenesis , Oxidative Phosphorylation/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisomes/drug effects , Peroxisomes/metabolism , Phosphorylation , Protein Biosynthesis , Reactive Oxygen Species/metabolism , Receptors, Estrogen/metabolism , Transcription, Genetic , Xenograft Model Antitumor Assays
3.
Biochem Biophys Res Commun ; 613: 47-52, 2022 07 12.
Article in English | MEDLINE | ID: mdl-35526488

ABSTRACT

Peroxisome proliferator-activated receptor α (PPARα) regulates fatty acid oxidation (FAO). Usually, very-long chain fatty acids are first activated by acyl-CoA synthetase (ACS) to generate acyl-CoA for oxidation by acyl-CoA oxidase (ACOX) in peroxisomes, and the resultant shorter chain fatty acids will be further oxidized in mitochondria. ACS long-chain family member 4 (ACSL4) preferentially uses arachidonic acid (AA) as substrates to synthesize arachidonoyl-CoA. Arachidonoyl-CoA is usually esterified into phospholipids. When AA is released by phospholipase A2 (PLA2) from phospholipids, it will be used for prostaglandin synthesis by cyclooxygenases (COX). In this study, when PPARα agonist WY-14,643 was mixed in liquid Lieber-DeCarli ethanol or control diets and fed to mice, liver PLA2, COX-2, and ACOX1 were induced but ACSL4 was inhibited, suggesting that AA released by PLA2 from phospholipid will be metabolized to prostaglandin via COX-2 instead of being synthesized into acyl-CoA by ACSL4. However, liver prostaglandin E2 (PGE2), a major component of prostaglandin, was not increased with the induced COX-2 but decreased by WY-14,643. ACOX1 specific inhibitor mixed in the liquid diets restored both the WY-14,643-suppressed liver TG and PGE2, but COX-2 specific inhibitor celecoxib mixed in the liquid diets reversed the WY-14,643-suppressed liver TG but not liver PGE2 contents. These results suggest that induction of PLA2, COX-2 and ACOX1 orchestrates to increase oxidation of AA/PGE2, which constitutes one new mechanism by which PPARα induces peroxisomal FAO and inhibits ethanol-induced liver fat accumulation.


Subject(s)
Acyl-CoA Oxidase , Cyclooxygenase 2 , Fatty Liver, Alcoholic , PPAR alpha , Phospholipases A2 , Pyrimidines , Acyl-CoA Oxidase/metabolism , Animals , Coenzyme A/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Fatty Acids/metabolism , Fatty Liver, Alcoholic/drug therapy , Fatty Liver, Alcoholic/metabolism , Lipid Metabolism/drug effects , Liver/metabolism , Mice , PPAR alpha/agonists , PPAR alpha/metabolism , Peroxisomes/drug effects , Peroxisomes/metabolism , Phospholipases A2/metabolism , Phospholipids/metabolism , Pyrimidines/pharmacology , Signal Transduction/drug effects
4.
J Cell Biochem ; 122(11): 1686-1700, 2021 11.
Article in English | MEDLINE | ID: mdl-34322908

ABSTRACT

Mitochondria and peroxisomes are metabolically interconnected and functionally active subcellular organelles. These two dynamic organelles, share a number of common biochemical functions such as ß-oxidation of fatty acids and detoxification of peroxides. The biogenesis and morphology of both these organelles in the mammalian cells is controlled by common transcription factors like PGC1α, and by a common fission machinery comprising of fission proteins like DRP1, Mff, and hFis1, respectively. In addition, the outer membrane mitochondria-anchored protein ligase (MAPL), the first mitochondrial SUMO E3 ligase with a RING-finger domain, also regulates mitochondrial morphology inducing mitochondrial fragmentation upon its overexpression. This fragmentation is dependent on both the RING domain of MAPL and the presence of the mitochondrial fission GTPase dynamin-related protein-1 (DRP1). Earlier studies have demonstrated that mitochondrial-derived vesicles are formed independently of the known mitochondrial fission GTPase, DRP1 are enriched for MAPL and are targeted to peroxisomes. The current study shows that MAPL regulates morphology of peroxisomes in a cell-type specific manner. Fascinatingly, the peroxisome elongation caused either due to silencing of DRP1 or by addition of polyunsaturated fatty acid, docosahexaenoic acid was blocked by overexpressing MAPL in mammalian cell lines. Furthermore, the transfection and colocalisation studies of MAPL with peroxisome membrane marker, PMP70, in different cell lines clearly revealed a cell-type specificity of transport of MAPL to peroxisomes. Previous work has placed the Vps35 (retromer component) as vital for delivery of MAPL to peroxisomes, placing the retromer as critical for the formation of MAPL-positive mitochondrial-derived vesicles. The results of polyethylene glycol-based cell-cell fusion assay signified that the enrichment of MAPL in peroxisomes is through vesicles and a retromer dependent phenomenon. Thus, a novel function for MAPL in peroxisomes is established to regulate peroxisome elongation and morphology under growth conditions and thus possibly modulate peroxisome fission.


Subject(s)
Peroxisomes/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , COS Cells , Chlorocebus aethiops , Docosahexaenoic Acids/pharmacology , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , Gene Expression , HeLa Cells , Hep G2 Cells , Humans , Mitochondrial Dynamics , Peroxisomes/drug effects , Peroxisomes/genetics , Ubiquitin-Protein Ligases/genetics
5.
Am J Physiol Heart Circ Physiol ; 320(5): H1813-H1821, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33666503

ABSTRACT

Although peroxisomes have been extensively studied in other cell types, their presence and function have gone virtually unexamined in cardiac myocytes. Here, in neonatal rat ventricular myocytes (NRVM) we showed that several known peroxisomal proteins co-localize to punctate structures with a morphology typical of peroxisomes. Surprisingly, we found that the peroxisomal protein, fatty acyl-CoA reductase 1 (FAR1), was upregulated by pharmacological and pathophysiological ER stress induced by tunicamycin (TM) and simulated ischemia-reperfusion (sI/R), respectively. Moreover, FAR1 induction in NRVM was mediated by the ER stress sensor, activating transcription factor 6 (ATF6). Functionally, FAR1 knockdown reduced myocyte death during oxidative stress induced by either sI/R or hydrogen peroxide (H2O2). Thus, Far1 is an ER stress-inducible gene, which encodes a protein that localizes to peroxisomes of cardiac myocytes, where it reduces myocyte viability during oxidative stress. Since FAR1 is critical for plasmalogen synthesis, these results imply that plasmalogens may exert maladaptive effects on the viability of myocytes exposed to oxidative stress.NEW & NOTEWORTHY The peroxisomal enzyme, FAR1, was shown to be an ER stress- and ATF6-inducible protein that localizes to peroxisomes in cardiac myocytes. FAR1 decreases myocyte viability during oxidative stress.


Subject(s)
Activating Transcription Factor 6/metabolism , Aldehyde Oxidoreductases/biosynthesis , Endoplasmic Reticulum Stress , Myocardial Reperfusion Injury/enzymology , Myocytes, Cardiac/enzymology , Peroxisomes/enzymology , Activating Transcription Factor 6/genetics , Aldehyde Oxidoreductases/genetics , Animals , Animals, Newborn , Cell Hypoxia , Cell Survival , Cells, Cultured , Endoplasmic Reticulum Stress/drug effects , Enzyme Induction , Hydrogen Peroxide/toxicity , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Oxidative Stress , Peroxisomes/drug effects , Peroxisomes/metabolism , Rats , Tunicamycin/toxicity
6.
J Cell Sci ; 131(2)2018 01 29.
Article in English | MEDLINE | ID: mdl-28183730

ABSTRACT

Ca2+ is a second messenger in many physiological and phytopathological processes. Peroxisomes are subcellular compartments with an active oxidative and nitrosative metabolism. Previous studies have demonstrated that peroxisomal nitric oxide (NO) generation is dependent on Ca2+ and calmodulin (CaM). Here, we used Arabidopsis thaliana transgenic seedlings expressing cyan fluorescent protein (CFP) containing a type 1 peroxisomal-targeting signal motif (PTS1; CFP-PTS1), which enables peroxisomes to be visualized in vivo, and also used a cell-permeable fluorescent probe for Ca2+ Analysis by confocal laser-scanning microscopy (CLSM) enabled us to visualize the presence of endogenous Ca2+ in the peroxisomes of both roots and guard cells. The presence of Ca2+ in peroxisomes and the import of CFP-PTS1 are drastically disrupted by both CaM antagonist and glutathione (GSH). Furthermore, the activity of three peroxisomal enzymes (catalase, glycolate oxidase and hydroxypyruvate reductase) containing PTS1 was clearly affected in these conditions, with a decrease of between 41 and 51%. In summary, data show that Ca2+ and CaM are strictly necessary for protein import and normal functionality of peroxisomal enzymes, including antioxidant and photorespiratory enzymes, as well as for NO production.


Subject(s)
Arabidopsis/metabolism , Calcium/metabolism , Calmodulin/antagonists & inhibitors , Peroxisomes/metabolism , Arabidopsis/cytology , Arabidopsis/drug effects , Arabidopsis/genetics , Arabidopsis Proteins/metabolism , Calmodulin/metabolism , Chloroplasts/drug effects , Chloroplasts/metabolism , Gene Expression Regulation, Plant/drug effects , Glutathione/pharmacology , Nitric Oxide/pharmacology , Peroxisomes/drug effects , Plant Cells/drug effects , Plant Cells/metabolism , Plant Roots/cytology , Plant Roots/drug effects , Plant Roots/metabolism , Protein Transport/drug effects
7.
Toxicol Appl Pharmacol ; 386: 114829, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31734319

ABSTRACT

Sodium valproate (SVP) is a first-line treatment for various forms of epilepsy; however, it can cause severe liver injury. Ginsenoside compound K (G-CK) is the main active ingredient of the traditional herbal medicine ginseng. According to our previous research, SVP-induced elevation of ALT and AST levels, as well as pathological changes of liver tissue, was believed to be significantly reversed by G-CK in LiCl-pilocarpine induced epileptic rats. Thus, we aimed to evaluate the protective effect of G-CK on hepatotoxicity caused by SVP. The rats treated with SVP showed liver injury with evident increases in hepatic index, transaminases activity, alkaline phosphatase level, hepatic triglyceride and lipid peroxidation; significant decreases in plasma albumin level and antioxidant capacity; and obvious changes in histopathological and subcellular structures. All of these changes could be mitigated by co-administration with G-CK. Proteomic analysis indicated that hepcidin, soluble epoxide hydrolase (sEH, UniProt ID P80299), and the peroxisome pathway were involved in the hepatoprotective effect of G-CK. Changes in protein expression of hepcidin and sEH were verified by ELISA and Western blot analysis, respectively. In addition, we observed that the hepatic iron rose in SVP group and decreased in the combination group. In summary, our findings demonstrate the clear hepatoprotective effect of G-CK against SVP-induced hepatotoxicity through the antioxidant effect, regulation of peroxisome pathway relying on sEH (P80299) downregulation, as well as regulation of iron homeostasis dependent on hepcidin upregulation.


Subject(s)
Antioxidants/pharmacology , Chemical and Drug Induced Liver Injury/drug therapy , Ginsenosides/pharmacology , Iron/metabolism , Peroxisomes/drug effects , Valproic Acid/toxicity , Animals , Biomarkers/blood , Chemical and Drug Induced Liver Injury/etiology , Homeostasis/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Microscopy, Electron, Transmission , Oxidative Stress/drug effects , Peroxisomes/metabolism , Rats , Rats, Sprague-Dawley , Valproic Acid/antagonists & inhibitors
8.
Cell Biol Int ; 44(3): 918-923, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31814220

ABSTRACT

Peroxisomicine A1 (PA1) is a potential antineoplastic agent with high and selective toxicity toward peroxisomes of tumor cells. Pexophagy is a selective autophagy process that degrades damaged peroxisomes; this process has been studied mainly in methylotrophic yeasts. There are two main modes of pexophagy in yeast: macropexophagy and micropexophagy. Previous studies showed that peroxisomes damaged by a prolonged exposition to PA1 are eliminated by macropexophagy. In this work, Candida boidinii was grown in methanol-containing media, and PA1 was added to the cultures at 2 µg/mL after they reached the mid-exponential growth phase. Samples were taken at 5, 10, 15, 20, and 25 min after the addition of PA1 and processed for ultrastructural analysis. Typical morphological characteristics of micropexophagy were observed: the direct engulfment of peroxisomes by the vacuolar membrane and the presence of the micropexophagic membrane apparatus (MIPA), which mediates the fusion between the opposing tips of the vacuole to complete sequestration of peroxisomes from the cytosol. In conclusion, here we report that, in addition to macropexophagy, peroxisomes damaged by PA1 can be eliminated by micropexophagy. This information is useful to deepen the knowledge of the mechanism of action of PA1 and of that of pexophagy per se.


Subject(s)
Anthracenes/pharmacology , Antineoplastic Agents/pharmacology , Candida/drug effects , Macroautophagy/drug effects , Microautophagy/drug effects , Peroxisomes/drug effects , Fungal Proteins/metabolism
9.
Biol Pharm Bull ; 43(9): 1382-1392, 2020.
Article in English | MEDLINE | ID: mdl-32879213

ABSTRACT

The effects of different dietary fats on hepatic fatty acid oxidation were compared in male ICR mice and Sprague-Dawley rats. Animals were fed diets containing 100 g/kg of either palm oil (saturated fat), safflower oil (rich in linoleic acid), an oil of evening primrose origin (γ-linolenic acid, GLA oil), perilla oil (α-linolenic acid) or fish oil (eicosapentaenoic and doxosahexaenoic acids) for 21 d. GLA, perilla and fish oils, compared with palm and safflower oils, increased the activity of fatty acid oxidation enzymes in both mice and rats, with some exceptions. In mice, GLA and fish oils greatly increased the peroxisomal palmitoyl-CoA oxidation rate, and the activity of acyl-CoA oxidase and enoyl-CoA hydratase to the same degree. The effects were much smaller with perilla oil. In rats, enhancing effects were more notable with fish oil than with GLA and perilla oils, excluding the activity of enoyl-CoA hydratase, and were comparable between GLA and perilla oils. In mice, strong enhancing effects of GLA oil, which were greater than with perilla oil and comparable to those of fish oil, were confirmed on mRNA levels of peroxisomal but not mitochondrial fatty acid oxidation enzymes. In rats, the effects of GLA and perilla oils on mRNA levels of peroxisomal and mitochondrial enzymes were indistinguishable, and lower than those observed with fish oil. Therefore, considerable diversity in the response to dietary polyunsaturated fats, especially the oil rich in γ-linolenic acid and fish oil, of hepatic fatty acid oxidation pathway exists between mice and rats.


Subject(s)
Dietary Fats/administration & dosage , Lipid Metabolism/drug effects , Liver/drug effects , gamma-Linolenic Acid/administration & dosage , Acyl-CoA Oxidase/metabolism , Animal Feed , Animals , Enoyl-CoA Hydratase/metabolism , Fish Oils/administration & dosage , Fish Oils/chemistry , Liver/cytology , Liver/enzymology , Male , Mice , Mice, Inbred ICR , Mitochondria/drug effects , Mitochondria/enzymology , Oxidation-Reduction/drug effects , Peroxisomes/drug effects , Peroxisomes/enzymology , Plant Oils/administration & dosage , Plant Oils/chemistry , Rats , Rats, Sprague-Dawley , Species Specificity
10.
J Cell Physiol ; 234(10): 18344-18348, 2019 08.
Article in English | MEDLINE | ID: mdl-30932193

ABSTRACT

This work analyzes the thermogenic flux induced by the very long-chain fatty acid (VLCFA) lignoceric acid (C24:0) in isolated peroxisomes. Specific metabolic alterations of peroxisomes are related to a variety of disorders, the most frequent one being the neurodegenerative inherited disease X-linked adrenoleukodystrophy (X-ALD). A peroxisomal transport protein is mutated in this disorder. Due to reduced catabolism and enhanced fatty acid (FA) elongation, VLCFA accumulates in plasma and in all tissues, contributing to the clinical manifestations of this disorder. During peroxisomal metabolism, heat is produced but it is considered lost. Instead, it is a form of energy that could play a role in molecular mechanisms of this pathology and other neurodegenerative disorders. The thermogenic flux induced by lignoceric acid (C24:0) was estimated by isothermal titration calorimetry in peroxisomes isolated from HepG2 cells and from fibroblasts obtained from patients with X-ALD and healthy subjects. Heat flux induced by lignoceric acid in HepG2 peroxisomes was exothermic, indicating normal peroxisomal metabolism. In X-ALD peroxisomes the heat flux was endothermic, indicating the requirement of heat/energy, possibly for cellular metabolism. In fibroblasts from healthy subjects, the effect was less pronounced than in HepG2, a kind of cell known to have greater FA metabolism than fibroblasts. Our hypothesis is that heat is not lost but it could act as an activator, for example on the heat-sensitive pathway related to TRVP2 receptors. To investigate this hypothesis we focused on peroxisomal metabolism, considering that impaired heat generation could contribute to the development of peroxisomal neurodegenerative disorders.


Subject(s)
Adrenoleukodystrophy/drug therapy , Fatty Acids/pharmacology , Fibroblasts/drug effects , Peroxisomes/drug effects , Thermogenesis/drug effects , Cell Line, Tumor , Hep G2 Cells , Humans , Lipid Metabolism/drug effects , Oxidation-Reduction/drug effects
11.
J Cell Biochem ; 120(3): 3243-3258, 2019 03.
Article in English | MEDLINE | ID: mdl-30362618

ABSTRACT

Zellweger spectrum disorder (ZSD) results from biallelic mutations in PEX genes required for peroxisome biogenesis. PEX1-G843D is a common hypomorphic allele in the patient population that is associated with milder disease. In prior work using a PEX1-G843D/null patient fibroblast line expressing a green fluorescent protein (GFP) reporter with a peroxisome-targeting signal (GFP-PTS1), we demonstrated that treatments with the chemical chaperone betaine and flavonoid acacetin diacetate recovered peroxisome functions. To identify more effective compounds for preclinical investigation, we evaluated 54 flavonoids using this cell-based phenotype assay. Diosmetin showed the most promising combination of potency and efficacy (EC50 2.5 µM). All active 5',7'-dihydroxyflavones showed greater average efficacy than their corresponding flavonols, whereas the corresponding flavanones, isoflavones, and chalcones tested were inactive. Additional treatment with the proteostasis regulator bortezomib increased the percentage of import-rescued cells over treatment with flavonoids alone. Cotreatments of diosmetin and betaine showed the most robust additive effects, as confirmed by three independent functional assays in primary PEX1-G843D patient cells, but neither agent was active alone or in combination in patient cells homozygous for the PEX1 c.2097_2098insT null allele. Moreover, diosmetin treatment increased PEX1, PEX6, and PEX5 protein levels in PEX1-G843D patient cells, but none of these proteins increased in PEX1 null cells. We propose that diosmetin acts as a pharmacological chaperone that improves the stability, conformation, and functions of PEX1/PEX6 exportomer complexes required for peroxisome assembly. We suggest that diosmetin, in clinical use for chronic venous disease, and related flavonoids warrant further preclinical investigation for the treatment of PEX1-G843D-associated ZSD.


Subject(s)
ATPases Associated with Diverse Cellular Activities/genetics , Alleles , Fibroblasts/metabolism , Flavonoids/pharmacology , Membrane Proteins/genetics , Peroxisomes/drug effects , Zellweger Syndrome/pathology , ATPases Associated with Diverse Cellular Activities/metabolism , Adenosine Triphosphate/metabolism , Cell Line , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Drug Therapy, Combination , Flavonoids/therapeutic use , Green Fluorescent Proteins/metabolism , Humans , Membrane Proteins/metabolism , Molecular Chaperones/pharmacology , Molecular Chaperones/therapeutic use , Peroxisomal Targeting Signals , Peroxisomes/metabolism , Signal Transduction/drug effects , Zellweger Syndrome/drug therapy
12.
Plant Physiol ; 176(1): 340-351, 2018 01.
Article in English | MEDLINE | ID: mdl-28931630

ABSTRACT

Peroxisomes are small organelles that house many oxidative reactions. Peroxisome proliferation is induced under multiple stress conditions, including salt stress; however, factors regulating this process are not well defined. We have identified a role for Arabidopsis (Arabidopsis thaliana) MAP KINASE17 (MPK17) in affecting peroxisome division in a manner that requires the known peroxisome division factor PEROXISOME AND MITOCHONDRIAL DIVISION FACTOR1 (PMD1). MPK17 and PMD1 are involved in peroxisome proliferation in response to NaCl stress. Additionally, we found that PMD1 is an actin-binding protein and that a functioning actin cytoskeleton is required for NaCl-induced peroxisome division. Our data suggest roles for MPK17 and PMD1 in influencing the numbers and cellular distribution of peroxisomes through the cytoskeleton-peroxisome connection. These findings expand our understanding of peroxisome division and potentially identify factors connecting the actin cytoskeleton and peroxisome proliferation.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Peroxisomes/metabolism , Sodium Chloride/pharmacology , Actins/metabolism , Arabidopsis/drug effects , Indoleacetic Acids/pharmacology , Models, Biological , Mutation/genetics , Peroxisomes/drug effects , Phenotype , Polymerization , Protein Binding/drug effects
13.
Mol Cell Neurosci ; 86: 65-71, 2018 01.
Article in English | MEDLINE | ID: mdl-29180229

ABSTRACT

Doxorubicin, a commonly used anti-neoplastic agent, causes severe neurotoxicity. Doxorubicin promotes thinning of the brain cortex and accelerates brain aging, leading to cognitive impairment. Oxidative stress induced by doxorubicin contributes to cellular damage. In addition to mitochondria, peroxisomes also generate reactive oxygen species (ROS) and promote cell senescence. Here, we investigated if doxorubicin affects peroxisomal homeostasis in neurons. We demonstrate that the number of peroxisomes is increased in doxorubicin-treated neurons and in the brains of mice which underwent doxorubicin-based chemotherapy. Pexophagy, the specific autophagy of peroxisomes, is downregulated in neurons, and peroxisomes produce more ROS. 2-hydroxypropyl-ß-cyclodextrin (HPßCD), an activator of the transcription factor TFEB, which regulates expression of genes involved in autophagy and lysosome function, mitigates damage of pexophagy and decreases ROS production induced by doxorubicin. We conclude that peroxisome-associated oxidative stress induced by doxorubicin may contribute to neurotoxicity, cognitive dysfunction, and accelerated brain aging in cancer patients and survivors. Peroxisomes might be a valuable new target for mitigating neuronal damage caused by chemotherapy drugs and for slowing down brain aging in general.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Doxorubicin/pharmacology , Neurons/drug effects , Oxidative Stress/drug effects , Peroxisomes/drug effects , Animals , Cells, Cultured , Female , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Frontal Lobe/ultrastructure , Mice , Neurons/metabolism , Neurons/ultrastructure , Oxidative Stress/physiology , Peroxisomes/metabolism , Peroxisomes/ultrastructure , Rats , Reactive Oxygen Species/metabolism
14.
Molecules ; 24(18)2019 Sep 11.
Article in English | MEDLINE | ID: mdl-31514417

ABSTRACT

Neurodegenerative diseases are characterized by oxidative stress, mitochondrial damage, and death of neuronal cells. To counteract such damage and to favor neurogenesis, neurotrophic factors could be used as therapeutic agents. Octadecaneuropeptide (ODN), produced by astrocytes, is a potent neuroprotective agent. In N2a cells, we studied the ability of ODN to promote neuronal differentiation. This parameter was evaluated by phase contrast microscopy, staining with crystal violet, cresyl blue, and Sulforhodamine 101. The effect of ODN on cell viability and mitochondrial activity was determined with fluorescein diacetate and DiOC6(3), respectively. The impact of ODN on the topography of mitochondria and peroxisomes, two tightly connected organelles involved in nerve cell functions and lipid metabolism, was evaluated by transmission electron microscopy and fluorescence microscopy: detection of mitochondria with MitoTracker Red, and peroxisome with an antibody directed against the ABCD3 peroxisomal transporter. The profiles in fatty acids, cholesterol, and cholesterol precursors were determined by gas chromatography, in some cases coupled with mass spectrometry. Treatment of N2a cells with ODN (10-14 M, 48 h) induces neurite outgrowth. ODN-induced neuronal differentiation was associated with modification of topographical distribution of mitochondria and peroxisomes throughout the neurites and did not affect cell viability and mitochondrial activity. The inhibition of ODN-induced N2a differentiation with H89, U73122, chelerythrine and U0126 supports the activation of a PKA/PLC/PKC/MEK/ERK-dependent signaling pathway. Although there is no difference in fatty acid profile between control and ODN-treated cells, the level of cholesterol and some of its precursors (lanosterol, desmosterol, lathosterol) was increased in ODN-treated cells. The ability of ODN to induce neuronal differentiation without cytotoxicity reinforces the interest for this neuropeptide with neurotrophic properties to overcome nerve cell damage in major neurodegenerative diseases.


Subject(s)
Cell Differentiation/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Diazepam Binding Inhibitor/pharmacology , Lipids/chemistry , Mitochondria/metabolism , Neuropeptides/pharmacology , Peptide Fragments/pharmacology , Peroxisomes/metabolism , Protein Kinase C/metabolism , Type C Phospholipases/metabolism , Animals , Cell Line, Tumor , Cell Survival/drug effects , MAP Kinase Signaling System/drug effects , Mice , Mitochondria/drug effects , Mitochondria/ultrastructure , Peroxisomes/drug effects , Peroxisomes/ultrastructure , Rhodamines/chemistry , Rhodamines/metabolism , Signal Transduction/drug effects
15.
J Biol Chem ; 292(37): 15312-15320, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28747436

ABSTRACT

The immunomodulatory receptor Siglec-3/CD33 influences risk for late-onset Alzheimer's disease (LOAD), an apparently human-specific post-reproductive disease. CD33 generates two splice variants: a full-length CD33M transcript produced primarily by the "LOAD-risk" allele and a shorter CD33m isoform lacking the sialic acid-binding domain produced primarily from the "LOAD-protective" allele. An SNP that modulates CD33 splicing to favor CD33m is associated with enhanced microglial activity. Individuals expressing more protective isoform accumulate less brain ß-amyloid and have a lower LOAD risk. How the CD33m isoform increases ß-amyloid clearance remains unknown. We report that the protection by the CD33m isoform may not be conferred by what it does but, rather, from what it cannot do. Analysis of blood neutrophils and monocytes and a microglial cell line revealed that unlike CD33M, the CD33m isoform does not localize to cell surfaces; instead, it accumulates in peroxisomes. Cell stimulation and activation did not mobilize CD33m to the surface. Thus, the CD33m isoform may neither interact directly with amyloid plaques nor engage in cell-surface signaling. Rather, production and localization of CD33m in peroxisomes is a way of diminishing the amount of CD33M and enhancing ß-amyloid clearance. We confirmed intracellular localization by generating a CD33m-specific monoclonal antibody. Of note, CD33 is the only Siglec with a peroxisome-targeting sequence, and this motif emerged by convergent evolution in toothed whales, the only other mammals with a prolonged post-reproductive lifespan. The CD33 allele that protects post-reproductive individuals from LOAD may have evolved by adaptive loss-of-function, an example of the less-is-more hypothesis.


Subject(s)
Alzheimer Disease/genetics , Genetic Predisposition to Disease , Macrophages/metabolism , Microglia/metabolism , Neutrophils/metabolism , Polymorphism, Single Nucleotide , Sialic Acid Binding Ig-like Lectin 3/metabolism , Alleles , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amino Acid Motifs , Bacterial Proteins/metabolism , Bacterial Proteins/toxicity , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane/pathology , Humans , Lipopolysaccharides/toxicity , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Microglia/cytology , Microglia/immunology , Microglia/pathology , N-Formylmethionine Leucyl-Phenylalanine/toxicity , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuraminidase/metabolism , Neuraminidase/toxicity , Neutrophil Activation/drug effects , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/pathology , Peroxisomes/drug effects , Peroxisomes/metabolism , Peroxisomes/pathology , Phylogeny , Protein Interaction Domains and Motifs , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Sorting Signals , Protein Transport/drug effects , Sialic Acid Binding Ig-like Lectin 3/chemistry , Sialic Acid Binding Ig-like Lectin 3/genetics
16.
J Cell Physiol ; 233(9): 7415-7423, 2018 09.
Article in English | MEDLINE | ID: mdl-29600563

ABSTRACT

Nitrogen-containing bisphosphonates including alendronate (ALN) are the current first line antiresorptive drug in treating osteoporosis. In our study, we found that ALN administration impaired the secretion of platelet derived growth factor-BB (PDGF-BB), the most important angiogenic cytokines produced by preosteoclast (POC), in both sham and ovariectomized (OVX) mice. To further understand this phenomenon, we induced bone marrow macrophages (BMMs) to POCs in vitro and detected the effects of ALN particularly in POCs. The proapoptotic effect of ALN in POCs was confirmed by flow cytometry. On the molecular level, we found that farnesyl diphosphate synthase (FDPS) inhibition of ALN led to peroxisomal dysfunction and up regulation of cytoprotective protein glucose-regulated protein (GRP) 78. Peroxisomal dysfunction further induced endoplasmic reticulum (ER) stress in POCs and finally resulted in cell apoptosis marked by reduced expression of B-cell lymphoma 2 (Bcl-2) and increased expressions of CCAAT/enhancer binding protein homologous protein (CHOP), Bcl2 associated X (Bax), and cleaved caspase-3. We concluded that ALN has no selectivity in inhibiting POC and mature osteoclast. For POCs, ALN inhibition of FDPS leads to peroxisomal dysfunction, which further mediates ER stress and finally causes cell apoptosis. Considering that decreased angiogenesis is also an important issue in treating osteoporosis, how to preserve pro-angiogenic POCs while depleting mature osteoclasts is a problem worthy to be solved.


Subject(s)
Alendronate/pharmacology , Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Osteoclasts/metabolism , Osteoclasts/pathology , Peroxisomes/metabolism , Animals , Becaplermin/metabolism , Cell Count , Cell Cycle Checkpoints/drug effects , Endoplasmic Reticulum Chaperone BiP , Female , Heat-Shock Proteins/metabolism , Mice, Inbred C57BL , Osteoclasts/drug effects , Osteoporosis/pathology , Ovariectomy , Peroxisomes/drug effects , Up-Regulation/drug effects
17.
Immunology ; 154(3): 434-451, 2018 07.
Article in English | MEDLINE | ID: mdl-29331024

ABSTRACT

Recent studies report that loss and dysfunction of mitochondria and peroxisomes contribute to the myelin and axonal damage in multiple sclerosis (MS). In this study, we investigated the efficacy of a combination of lovastatin and AMP-activated protein kinase (AMPK) activator (AICAR) on the loss and dysfunction of mitochondria and peroxisomes and myelin and axonal damage in spinal cords, relative to the clinical disease symptoms, using a mouse model of experimental autoimmune encephalomyelitis (EAE, a model for MS). We observed that lovastatin and AICAR treatments individually provided partial protection of mitochondria/peroxisomes and myelin/axons, and therefore partial attenuation of clinical disease in EAE mice. However, treatment of EAE mice with the lovastatin and AICAR combination provided greater protection of mitochondria/peroxisomes and myelin/axons, and greater improvement in clinical disease compared with individual drug treatments. In spinal cords of EAE mice, lovastatin-mediated inhibition of RhoA and AICAR-mediated activation of AMPK cooperatively enhanced the expression of the transcription factors and regulators (e.g. PPARα/ß, SIRT-1, NRF-1, and TFAM) required for biogenesis and the functions of mitochondria (e.g. OXPHOS, MnSOD) and peroxisomes (e.g. PMP70 and catalase). In summary, these studies document that oral medication with a combination of lovastatin and AICAR, which are individually known to have immunomodulatory effects, provides potent protection and repair of inflammation-induced loss and dysfunction of mitochondria and peroxisomes as well as myelin and axonal abnormalities in EAE. As statins are known to provide protection in progressive MS (Phase II study), these studies support that supplementation statin treatment with an AMPK activator may provide greater efficacy against MS.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Lovastatin/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Peroxisomes/drug effects , Peroxisomes/metabolism , Adenosine Triphosphate/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Biomarkers , Cell Line , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Gene Expression , Humans , Mice , Mitochondria/genetics , Mitochondria/ultrastructure , Peroxisomes/genetics , Peroxisomes/ultrastructure , Ribonucleotides/pharmacology , Spinal Cord/immunology , Spinal Cord/metabolism , Spinal Cord/pathology , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
18.
J Cell Sci ; 129(20): 3868-3877, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27587840

ABSTRACT

Export out of the endoplasmic reticulum (ER) involves the Sar1 and COPII machinery acting at ER exit sites (ERES). Whether and how cargo proteins are recruited upstream of Sar1 and COPII is unclear. Two models are conceivable, a recruitment model where cargo is actively transported through a transport factor and handed over to the Sar1 and COPII machinery in ERES, and a capture model, where cargo freely diffuses into ERES where it is captured by the Sar1 and COPII machinery. Using the novel secretion inhibitor FLI-06, we show that recruitment of the cargo VSVG to ERES is an active process upstream of Sar1 and COPII. Applying FLI-06 before concentration of VSVG in ERES completely abolishes its recruitment. In contrast, applying FLI-06 after VSVG concentration in ERES does not lead to dispersal of the concentrated VSVG, arguing that it inhibits recruitment to ERES as opposed to capture in ERES. FLI-06 also inhibits export out of the trans-Golgi network (TGN), suggesting that similar mechanisms might orchestrate cargo selection and concentration at the ER and TGN. FLI-06 does not inhibit autophagosome biogenesis and the ER-peroxisomal transport route, suggesting that these rely on different mechanisms.


Subject(s)
Endoplasmic Reticulum/metabolism , Quinolines/pharmacology , trans-Golgi Network/metabolism , Autophagosomes/drug effects , Autophagosomes/metabolism , Endocytosis/drug effects , Exocytosis/drug effects , HeLa Cells , Humans , Peroxisomes/drug effects , Peroxisomes/metabolism , Protein Folding/drug effects , Protein Transport/drug effects , trans-Golgi Network/drug effects
19.
J Cell Sci ; 129(21): 4057-4066, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27678487

ABSTRACT

Peroxisomal proteins carrying a type 1 peroxisomal targeting signal (PTS1) are recognized by the well-conserved cycling import receptor Pex5p. The yeast YMR018W gene encodes a Pex5p paralog and newly identified peroxin that is involved in peroxisomal import of a subset of matrix proteins. The new peroxin was designated Pex9p, and it interacts with the docking protein Pex14p and a subclass of PTS1-containing peroxisomal matrix enzymes. Unlike Pex5p, Pex9p is not expressed in glucose- or ethanol-grown cells, but it is strongly induced by oleate. Under these conditions, Pex9p acts as a cytosolic and membrane-bound peroxisome import receptor for both malate synthase isoenzymes, Mls1p and Mls2p. The inducible Pex9p-dependent import pathway provides a mechanism for the oleate-inducible peroxisomal targeting of malate synthases. The existence of two distinct PTS1 receptors, in addition to two PTS2-dependent import routes, contributes to the adaptive metabolic capacity of peroxisomes in response to environmental changes and underlines the role of peroxisomes as multi-purpose organelles. The identification of different import routes into peroxisomes contributes to the molecular understanding of how regulated protein targeting can alter the function of organelles according to cellular needs.


Subject(s)
Peroxisomes/metabolism , Protein Sorting Signals , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Malate Synthase/metabolism , Models, Biological , Oleic Acid/pharmacology , Peroxisomes/drug effects , Protein Binding/drug effects , Protein Sorting Signals/drug effects , Protein Transport/drug effects , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae Proteins/chemistry , Structural Homology, Protein , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism
20.
Toxicol Appl Pharmacol ; 353: 43-54, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29908243

ABSTRACT

Cisplatin is an alkylating agent that interferes with DNA replication and kills proliferating carcinogenic cells. Several studies have been conducted to attenuate the side effects of cisplatin; one such side effect in cancer patients undergoing cisplatin chemotherapy is ototoxicity. However, owing to a lack of understanding of the precise mechanism underlying cisplatin-induced side effects, management of cisplatin-induced ototoxicity remains unsolved. We investigated the protective effects of fenofibrate, a PPAR-α activator, on cisplatin-induced ototoxicity. Fenofibrate prevented cisplatin-induced loss of hair cells and improved cell viability; moreover, fenofibrate significantly attenuated the threshold of auditory brainstem responses (ABR) in cisplatin-injected mice. Fenofibrate significantly increased PPAR-α, PPAR-γ, and PGC-1α expression, which consequently resulted in increased number and functional enzyme levels of peroxisomes and mitochondria, and markedly decreased phospho-p53 (S15), activated caspase-3, cleaved-PARP, and NF-κB p65 nuclear translocation, which reduced NADPH oxidase isoform (NOX3 and NOX4) expression, thereby decreasing reactive oxygen species (ROS) production in cisplatin-treated tissues ex vivo. Taken together, these results indicate that fenofibrate rescues cisplatin-induced ototoxicity by maintaining peroxisome and mitochondria number and function, reducing inflammation, and decreasing ROS levels. Our findings suggest that fenofibrate administration might serve as an effective therapeutic agent against cisplatin-induced ototoxicity.


Subject(s)
Antineoplastic Agents/toxicity , Cisplatin/antagonists & inhibitors , Cisplatin/toxicity , Ear Diseases/chemically induced , Ear Diseases/prevention & control , Fenofibrate/therapeutic use , Hypolipidemic Agents/therapeutic use , Animals , Cell Line , Cell Survival/drug effects , Cochlea/pathology , Ear Diseases/pathology , Evoked Potentials, Auditory, Brain Stem/drug effects , Hair Cells, Auditory/pathology , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Mitochondria/drug effects , Peroxisomes/drug effects , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL