Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.153
Filter
1.
J Biol Chem ; 298(12): 102654, 2022 12.
Article in English | MEDLINE | ID: mdl-36441026

ABSTRACT

The cytochrome-b5 reductase (CYB5R) family of flavoproteins is known to regulate reduction-oxidation (redox) balance in cells. The five enzyme members are highly compartmentalized at the subcellular level and function as "redox switches" enabling the reduction of several substrates, such as heme and coenzyme Q. Critical insight into the physiological and pathophysiological significance of CYB5R enzymes has been gleaned from several human genetic variants that cause congenital disease and a broad spectrum of chronic human diseases. Among the CYB5R genetic variants, CYB5R3 is well-characterized and deficiency in expression and activity is associated with type II methemoglobinemia, cancer, neurodegenerative disorders, diabetes, and cardiovascular disease. Importantly, pharmacological and genetic-based strategies are underway to target CYB5R3 to circumvent disease onset and mitigate severity. Despite our knowledge of CYB5R3 in human health and disease, the other reductases in the CYB5R family have been understudied, providing an opportunity to unravel critical function(s) for these enzymes in physiology and disease. In this review, we aim to provide the broad scientific community an up-to-date overview of the molecular, cellular, physiological, and pathophysiological roles of CYB5R proteins.


Subject(s)
Cytochrome-B(5) Reductase , Methemoglobinemia , Humans , Cytochrome-B(5) Reductase/genetics , Cytochrome-B(5) Reductase/metabolism , Cytochromes b5/metabolism , Methemoglobinemia/congenital , Methemoglobinemia/genetics , Oxidation-Reduction , Homeostasis , Cytochrome Reductases/metabolism
2.
Sci Rep ; 8(1): 15478, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30341307

ABSTRACT

The periplasmic reduction of the electron acceptors nitrate (Em +420 mV) and trimethylamine-N-oxide (TMAO; Em +130 mV) by Nap and Tor reductases is widespread in Gram-negative bacteria and is usually considered to be driven by non-energy conserving quinol dehydrogenases. The Epsilonproteobacterium Campylobacter jejuni can grow by nitrate and TMAO respiration and it has previously been assumed that these alternative pathways of electron transport are independent of the proton-motive menaquinol-cytochrome c reductase complex (QcrABC) that functions in oxygen-linked respiration. Here, we show that a qcrABC deletion mutant is completely deficient in oxygen-limited growth on both nitrate and TMAO and is unable to reduce these oxidants with physiological electron donors. As expected, the mutant grows normally on fumarate under oxygen-limited conditions. Thus, the periplasmic Nap and Tor reductases receive their electrons via QcrABC in C. jejuni, explaining the general absence of NapC and TorC quinol dehydrogenases in Epsilonproteobacteria. Moreover, the specific use of menaquinol (Em -75 mV) coupled with a Qcr complex to drive reduction of nitrate or TMAO against the proton-motive force allows the process to be electrogenic with a H+/2e- ratio of 2. The results have general implications for the role of Qcr complexes in bacterial oxygen-independent respiration and growth.


Subject(s)
Campylobacter jejuni/enzymology , Campylobacter jejuni/metabolism , Cytochrome Reductases/metabolism , Electron Transport , Methylamines/metabolism , Nitrates/metabolism , Campylobacter jejuni/growth & development , Cytochrome Reductases/deficiency , Gene Deletion , Oxidation-Reduction
3.
Apoptosis ; 23(11-12): 695-706, 2018 12.
Article in English | MEDLINE | ID: mdl-30259216

ABSTRACT

Mitochondria are the center of energy metabolism in the cell and the preferential target of various toxicants and ischemic injury. Renal ischemia-reperfusion (I/R) injury triggers proximal tubule injury and the mitochondria are believed to be the primary subcellular target of I/R injury. The promotion of mitochondrial biogenesis (MB) is critical for the prevention I/R injury. The results of our previous study showed that augmenter of liver regeneration (ALR) has anti-apoptotic and anti-oxidant functions. However, the modulatory mechanism of ALR remains unclear and warrants further investigation. To gain further insight into the role of ALR in MB, human kidney (HK)-2 cells were treated with lentiviruses carrying ALR short interfering RNA (siRNA) and a model of hypoxia reoxygenation (H/R) injury in vitro was created. We observed that knockdown of ALR promoted apoptosis of renal tubular cells and aggravated mitochondrial injury, as evidenced by the decrease in the mitochondrial respiratory proteins adenosine triphosphate (ATP) synthase subunit ß, cytochrome c oxidase subunit 1, and nicotinamide adenine dinucleotide dehydrogenase (ubiquinone) beta subcomplex 8. Meanwhile, the production of reactive oxygen species was increased and ATP levels were decreased significantly in HK-2 cells, as compared with the siRNA/control group (p < 0.05). In addition, the mitochondrial DNA copy number and membrane potential were markedly decreased. Furthermore, critical transcriptional regulators of MB (i.e., peroxisome proliferator-activated receptor-gamma coactivator 1 alpha, mitochondrial transcription factor A, sirtuin-1, and nuclear respiratory factor-1) were depleted in the siRNA/ALR group. Taken together, these findings unveil essential roles of ALR in the inhibition of renal tubular cell apoptosis and attenuation of mitochondrial dysfunction by promoting MB in AKI.


Subject(s)
Cytochrome Reductases/metabolism , Kidney/pathology , Mitochondria/pathology , Organelle Biogenesis , Reperfusion Injury/pathology , Adenosine Triphosphate/metabolism , Apoptosis , Cell Line, Transformed , Cytochrome Reductases/antagonists & inhibitors , Cytochrome Reductases/genetics , DNA, Mitochondrial/metabolism , Gene Expression Regulation , Gene Silencing , Humans , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Mitochondria/genetics , Mitochondria/metabolism , Oxidative Stress , Oxidoreductases Acting on Sulfur Group Donors , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism
4.
Circulation ; 138(16): 1693-1705, 2018 10 16.
Article in English | MEDLINE | ID: mdl-29739755

ABSTRACT

BACKGROUND: In addition to enhanced proinflammatory signaling, impaired resolution of vascular inflammation plays a key role in atherosclerosis. Proresolving lipid mediators formed through the 12/15 lipoxygenase pathways exert protective effects against murine atherosclerosis. n-3 Polyunsaturated fatty acids, including eicosapentaenoic acid (EPA), serve as the substrate for the formation of lipid mediators, which transduce potent anti-inflammatory and proresolving actions through their cognate G-protein-coupled receptors. The aim of this study was to identify signaling pathways associated with EPA supplementation and lipid mediator formation that mediate atherosclerotic disease progression. METHODS: Lipidomic plasma analysis were performed after EPA supplementation in Apoe-/- mice. Erv1/Chemr23-/- xApoe-/- mice were generated for the evaluation of atherosclerosis, phagocytosis, and oxidized low-density lipoprotein uptake. Histological and mRNA analyses were done on human atherosclerotic lesions. RESULTS: Here, we show that EPA supplementation significantly attenuated atherosclerotic lesion growth induced by Western diet in Apoe-/- mice and was associated with local cardiovascular n-3 enrichment and altered lipoprotein metabolism. Our systematic plasma lipidomic analysis identified the resolvin E1 precursor 18-monohydroxy EPA as a central molecule formed during EPA supplementation. Targeted deletion of the resolvin E1 receptor Erv1/Chemr23 in 2 independent hyperlipidemic murine models was associated with proatherogenic signaling in macrophages, increased oxidized low-density lipoprotein uptake, reduced phagocytosis, and increased atherosclerotic plaque size and necrotic core formation. We also demonstrate that in macrophages the resolvin E1-mediated effects in oxidized low-density lipoprotein uptake and phagocytosis were dependent on Erv1/Chemr23. When analyzing human atherosclerotic specimens, we identified ERV1/ChemR23 expression in a population of macrophages located in the proximity of the necrotic core and demonstrated augmented ERV1/ChemR23 mRNA levels in plaques derived from statin users. CONCLUSIONS: This study identifies 18-monohydroxy EPA as a major plasma marker after EPA supplementation and demonstrates that the ERV1/ChemR23 receptor for its downstream mediator resolvin E1 transduces protective effects in atherosclerosis. ERV1/ChemR23 signaling may represent a previously unrecognized therapeutic pathway to reduce atherosclerotic cardiovascular disease.


Subject(s)
Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Eicosapentaenoic Acid/pharmacology , Lipoproteins, LDL/metabolism , Macrophages/drug effects , Phagocytosis/drug effects , Plaque, Atherosclerotic , Receptors, G-Protein-Coupled/agonists , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cytochrome Reductases/genetics , Cytochrome Reductases/metabolism , Diet, Western , Disease Models, Animal , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/blood , Eicosapentaenoic Acid/metabolism , Genetic Predisposition to Disease , Humans , Macrophages/metabolism , Macrophages/pathology , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Necrosis , Oxidoreductases Acting on Sulfur Group Donors , Phenotype , Receptors, Chemokine , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, LDL/genetics , Receptors, LDL/metabolism , Signal Transduction/drug effects
5.
HPB (Oxford) ; 19(12): 1091-1103, 2017 12.
Article in English | MEDLINE | ID: mdl-28941575

ABSTRACT

BACKGROUND: The Associating Liver Partition and Portal Ligation for Staged Hepatectomy (ALPPS) depends on a significant inter-stages kinetic growth rate (KGR). Liver regeneration is highly energy-dependent. The metabolic adaptations in ALPPS are unknown. AIMS: i) Assess bioenergetics in both stages of ALPPS (T1 and T2) and compare them with control patients undergoing minor (miHp) and major hepatectomy (MaHp), respectively; ii) Correlate findings in ALPPS with volumetric data; iii) Investigate expression of genes involved in liver regeneration and energy metabolism. METHODS: Five patients undergoing ALPPS, five controls undergoing miHp and five undergoing MaHp. Assessment of remnant liver bioenergetics in T1, T2 and controls. Analysis of gene expression and protein content in ALPPS. RESULTS: Mitochondrial function was worsened in T1 versus miHp; and in T2 versus MaHp (p < 0.05); but improved from T1 to T2 (p < 0.05). Liver bioenergetics in T1 strongly correlated with KGR (p < 0.01). An increased expression of genes associated with liver regeneration (STAT3, ALR) and energy metabolism (PGC-1α, COX, Nampt) was found in T2 (p < 0.05). CONCLUSION: Metabolic capacity in ALPPS is worse than in controls, improves between stages and correlates with volumetric growth. Bioenergetic adaptations in ALPPS could serve as surrogate markers of liver reserve and as target for energetic conditioning.


Subject(s)
Energy Metabolism , Hepatectomy/methods , Liver Regeneration , Liver/surgery , Mitochondria, Liver/metabolism , Portal Vein/surgery , Aged , Case-Control Studies , Cyclooxygenase 1/genetics , Cyclooxygenase 1/metabolism , Cytochrome Reductases/genetics , Cytochrome Reductases/metabolism , Cytokines/genetics , Cytokines/metabolism , Energy Metabolism/genetics , Female , Gene Expression Regulation , Hepatectomy/adverse effects , Humans , Ligation , Liver/metabolism , Liver/pathology , Liver Regeneration/genetics , Male , Middle Aged , Nicotinamide Phosphoribosyltransferase/genetics , Nicotinamide Phosphoribosyltransferase/metabolism , Oxidoreductases Acting on Sulfur Group Donors , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Time Factors , Treatment Outcome
6.
Lab Invest ; 97(12): 1400-1411, 2017 12.
Article in English | MEDLINE | ID: mdl-28825695

ABSTRACT

Hepatocellular carcinoma (HCC) is highly chemoresistant and therefore challenges both physicians and patients. Augmenter of liver regeneration (ALR), previously also known as 'hepatic stimulator substance', is reported to inhibit the epithelial-mesenchymal transition (EMT) in HCC, one of the frequent events that occur in cancer metastasis, suggesting that ALR is involved in HCC. In this study, we report for the first time that the transfection of ALR enhances the antitumor effect of chemotherapy with doxorubicin, a typical anticancer drug, on HCC in vitro and in vivo. The efflux of doxorubicin from ALR-transfected HCC cells is efficiently suppressed. This implies the intracellular retention of doxorubicin in tumor cells, which is at least partly attributable to the effective inhibition of ABCB1 and ABCG2 transporter expression in ALR-expressing cells. The downregulation of ALR expression by short hairpin RNA diminishes the antitumor effect of ALR. We further demonstrate that ALR inhibits the AKT/Snail signaling pathway, resulting in the downregulation of ABCB1 and ABCG2 expression. In conclusion, our results suggest that ALR is a potential chemotherapeutic agent against HCC.


Subject(s)
ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/metabolism , Cytochrome Reductases , Doxorubicin/pharmacology , Liver Neoplasms/metabolism , ATP Binding Cassette Transporter, Subfamily B/analysis , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/analysis , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Animals , Cytochrome Reductases/genetics , Cytochrome Reductases/metabolism , Cytochrome Reductases/pharmacology , Hep G2 Cells , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Oxidoreductases Acting on Sulfur Group Donors , Xenograft Model Antitumor Assays
7.
Anticancer Res ; 37(7): 3379-3383, 2017 07.
Article in English | MEDLINE | ID: mdl-28668825

ABSTRACT

BACKGROUND/AIM: Hepatocellular carcinoma is a substantial healthcare burden with high prevalence and poor prognosis. As such, efforts are continually made to uncover molecules relevant in cancer biology, that are exploitable as targets for therapy. The mitochondrion is the powerhouse of the cell and exhibits altered functionality in the malignant state, including aberrant regulation of apoptosis and cellular respiration. Augmenter of liver regeneration (ALR) is a multifunctional mitochondrial protein that demonstrates anti-oxidative and anti-apoptotic properties and plays a key role in liver regeneration. MATERIALS AND METHODS: The present study systematically reviews the available literature on the role of ALR in cancer. RESULTS: Systematic search of PubMed resulted in 12 studies discussing ALR in multiple types of cancer. More specifically, ALR appears to be up-regulated in malignant cells and tissues. Furthermore, treatment of cells with exogenous ALR shows an anti-apoptotic effect while silencing or inhibiting ALR decreases cell and tumor survival. CONCLUSION: ALR clearly plays a role in cancer biology and demonstrates potential as a therapeutic target.


Subject(s)
Cytochrome Reductases/metabolism , Liver Regeneration/physiology , Mitochondrial Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Apoptosis/physiology , Humans , Mitochondria/metabolism , Mitochondria/pathology , Oxidoreductases Acting on Sulfur Group Donors , Up-Regulation/physiology
8.
Proc Natl Acad Sci U S A ; 114(28): 7426-7431, 2017 07 11.
Article in English | MEDLINE | ID: mdl-28652330

ABSTRACT

The recent discovery of small molecules targeting the cytochrome bc1 :aa3 in Mycobacterium tuberculosis triggered interest in the terminal respiratory oxidases for antituberculosis drug development. The mycobacterial cytochrome bc1 :aa3 consists of a menaquinone:cytochrome c reductase (bc1 ) and a cytochrome aa3 -type oxidase. The clinical-stage drug candidate Q203 interferes with the function of the subunit b of the menaquinone:cytochrome c reductase. Despite the affinity of Q203 for the bc1 :aa3 complex, the drug is only bacteriostatic and does not kill drug-tolerant persisters. This raises the possibility that the alternate terminal bd-type oxidase (cytochrome bd oxidase) is capable of maintaining a membrane potential and menaquinol oxidation in the presence of Q203. Here, we show that the electron flow through the cytochrome bd oxidase is sufficient to maintain respiration and ATP synthesis at a level high enough to protect M. tuberculosis from Q203-induced bacterial death. Upon genetic deletion of the cytochrome bd oxidase-encoding genes cydAB, Q203 inhibited mycobacterial respiration completely, became bactericidal, killed drug-tolerant mycobacterial persisters, and rapidly cleared M. tuberculosis infection in vivo. These results indicate a synthetic lethal interaction between the two terminal respiratory oxidases that can be exploited for anti-TB drug development. Our findings should be considered in the clinical development of drugs targeting the cytochrome bc1 :aa3 , as well as for the development of a drug combination targeting oxidative phosphorylation in M. tuberculosis.


Subject(s)
Mycobacterium tuberculosis/metabolism , Oxidoreductases/chemistry , Synthetic Lethal Mutations , Adenosine Triphosphate/chemistry , Animals , Antineoplastic Agents/pharmacology , Antitubercular Agents/pharmacology , Cytochrome Reductases/metabolism , Diarylquinolines/pharmacology , Electron Transport , Electron Transport Complex IV/metabolism , Gene Deletion , Humans , Inflammation , Mice , Mice, Inbred BALB C , Mitochondrial Proteins , Mycobacterium Infections/microbiology , Mycobacterium bovis , Mycobacterium tuberculosis/genetics , Oxidative Phosphorylation , Oxidoreductases/genetics , Oxygen/chemistry , Plant Proteins , THP-1 Cells
9.
Exp Mol Pathol ; 102(3): 428-433, 2017 06.
Article in English | MEDLINE | ID: mdl-28506765

ABSTRACT

The acute-phase response (APR) is an inflammatory process triggered mainly by IL-6 in response to neoplasm, tissue injury, infection or inflammation. Signaling of IL-6 is transduced by activating STAT3 which rapidly results in production of acute-phase proteins (APPs) such as fibrinogen ß (FGB) and haptoglobin (HP). Augmenter of liver regeneration (ALR), a hepatotrophic factor supporting liver regeneration, was reported to be upregulated after liver damage. In this study we analyzed the role of ALR for IL-6 signaling and APR. Thus, we investigated the expression and release of APPs in human liver cells under conditions of increased exogenous or endogenous ALR. HepG2 cells and ALR-reexpressing HepG2 cells were treated with IL-6 in the presence or absence of exogenous ALR for different time points. The mRNA expression and release of both FGB and HP were measured by RT-PCR and ELISA. We found that exogenously applied ALR attenuated the IL-6-induced mRNA expression and protein secretion of both FGB and HP. In contrast, IL-6 stimulation in HepG2 cells which re-express ALR, revealed elevated APR shown by increased mRNA expression and secretion of FGB and HP. Furthermore, we found that ALR-mediated regulation of IL-6-induced APP production is accompanied by altered STAT3 activity. While exogenous ALR reduced the IL-6-induced phosphorylation of STAT3, endogenous ALR enhanced STAT3 activity in liver cells. In conclusion, ALR, dependent on its localization, changes APR at least in part, by modifying STAT3 activation. This study shows a dual signaling of ALR and suggests that ALR is pivotal for the regulation of APR, a crucial event in liver injury and regeneration.


Subject(s)
Acute-Phase Reaction/genetics , Cytochrome Reductases/metabolism , Hepatocytes/metabolism , STAT3 Transcription Factor/metabolism , Acute-Phase Reaction/pathology , Cytochrome Reductases/genetics , Fibrinogen/genetics , Fibrinogen/metabolism , Haptoglobins/genetics , Haptoglobins/metabolism , Hep G2 Cells , Humans , Interleukin-6/pharmacology , Liver/metabolism , Liver Regeneration , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Oxidoreductases Acting on Sulfur Group Donors , Phosphorylation , Protein Inhibitors of Activated STAT/genetics , Protein Inhibitors of Activated STAT/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT3 Transcription Factor/genetics , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , Up-Regulation
10.
Neuropharmacology ; 121: 30-38, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28431972

ABSTRACT

Alterations in mitochondrial functions have been hypothesized to participate in the pathogenesis of depression, because brain bioenergetic abnormalities have been detected in depressed patients by neuroimaging in vivo studies. However, this hypothesis is not clearly demonstrated in experimental studies: some suggest that antidepressants are inhibitors of mitochondrial metabolism, while others observe the opposite. In this study, the effects of 21-day treatment with desipramine (15 mg/kg) and fluoxetine (10 mg/kg) were examined on the energy metabolism of rat hippocampus, evaluating the catalytic activity of regulatory enzymes of mitochondrial energy-yielding metabolic pathways. Because of the micro-heterogeneity of brain mitochondria, we have distinguished between (a) non-synaptic mitochondria (FM) of neuronal perikaryon (post-synaptic compartment) and (b) intra-synaptic light (LM) and heavy (HM) mitochondria (pre-synaptic compartment). Desipramine and fluoxetine changed the catalytic activity of specific enzymes in the different types of mitochondria: (a) in FM, both drugs enhanced cytochrome oxidase and glutamate dehydrogenase, (b) in LM, the overall bioenergetics was unaffected and (c) in HM only desipramine increased malate dehydrogenase and decreased the activities of Electron Transport Chain Complexes. These results integrate the pharmacodynamic features of desipramine and fluoxetine at subcellular level, overcoming the previous conflicting data about the effects of antidepressants on brain energy metabolism, mainly referred to whole brain homogenates or to bulk of cerebral mitochondria. With the differentiation in non-synaptic and intra-synaptic mitochondria, this study demonstrates that desipramine and fluoxetine lead to adjustments in the mitochondrial bioenergetics respect to the energy requirements of pre- and post-synaptic compartments.


Subject(s)
Antidepressive Agents/pharmacology , Desipramine/pharmacology , Energy Metabolism/drug effects , Fluoxetine/pharmacology , Hippocampus , Mitochondria/drug effects , Analysis of Variance , Animals , Cytochrome Reductases/metabolism , Electron Transport Complex IV/metabolism , Glutamate Dehydrogenase/metabolism , Hippocampus/drug effects , Hippocampus/enzymology , Hippocampus/ultrastructure , Male , Rats , Rats, Sprague-Dawley
11.
J Immunol ; 198(2): 718-728, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27994073

ABSTRACT

Unresolved inflammation is key in linking metabolic dysregulation and the immune system in type 2 diabetes. Successful regulation of acute inflammation requires biosynthesis of specialized proresolving lipid mediators, such as E-series resolvin (RvE) 1, and activation of cognate G protein-coupled receptors. RvE1 binds to leukotriene B4 (BLT-1) on neutrophils and to ERV-1/ChemR23 on monocyte/macrophages. We show novel actions of RvE1 and expression patterns of neutrophil receptors in type 2 diabetes. Neutrophils from healthy subjects express functional BLT-1, low levels of minimally functional ERV-1, and inversed coexpression when compared to neutrophils from type 2 diabetes subjects. Stimulation with TNF-α or LPS increased the expression of ERV-1 by healthy and diabetic neutrophils. RvE1 counteracted LPS and TNF-α induction of ERV-1 overexpression and endogenous diabetic overexpression, activating phagocytosis and resolution signals. Functional ERV-1 was determined by phosphorylation of the signaling protein ribosomal S6. Receptor-antagonism experiments revealed that the increase in phosphorylation of ribosomal S6 was mediated by BLT-1 in healthy subject neutrophils and by ERV-1 in diabetes. Metabololipidomics reveal a proinflammatory profile in diabetic serum. Cell phagocytosis is impaired in type 2 diabetes and requires RvE1 for activation. The dose of RvE1 required to activate resolution signals in type 2 diabetic neutrophils was significantly higher than in healthy controls. RvE1 rescues the dysregulation seen on neutrophil receptor profile and, following a therapeutic dosage, activates phagocytosis and resolution signals in type 2 diabetes. These findings reveal the importance of resolution receptors in health, disease, and dysregulation of inflammation in type 2 diabetes.


Subject(s)
Cytochrome Reductases/metabolism , Diabetes Mellitus, Type 2/metabolism , Eicosapentaenoic Acid/analogs & derivatives , Neutrophils/metabolism , Receptors, Leukotriene B4/metabolism , Adult , Cells, Cultured , Chromatography, Liquid , Cytochrome Reductases/immunology , Diabetes Mellitus, Type 2/immunology , Eicosapentaenoic Acid/immunology , Eicosapentaenoic Acid/metabolism , Female , Humans , Inflammation/immunology , Inflammation/metabolism , Male , Middle Aged , Neutrophils/immunology , Oxidoreductases Acting on Sulfur Group Donors , Phagocytosis/immunology , Polymerase Chain Reaction , Receptors, Leukotriene B4/immunology , Tandem Mass Spectrometry , Transcriptome
12.
Acta Biochim Pol ; 63(3): 527-31, 2016.
Article in English | MEDLINE | ID: mdl-27390784

ABSTRACT

INTRODUCTION: The pathogenesis of chronic hepatitis B depends on both, the immune response and oxidative stress. AIM OF THE STUDY: To assess the hepatic expression of miR-122 and the antioxidant genes: HMOX-1, NQO1 and GFER1, in liver biopsy specimens obtained from patients with chronic hepatitis B, with regard to selected clinical and histological parameters, using RT-PCR. RESULTS: The study group comprised 34 HBV-infected patients. Statistically significant associations were found between lower hepatic expression of HMOX-1 and greater severity of liver inflammation (p=0.04). However, significantly higher expression of NQO1 was observed in patients with advanced liver fibrosis (p=0.035). Hepatic expression of miR-122 in HBV patients was not associated with viral load or liver injury. CONCLUSION: The hepatic expression of HMOX-1and NQO1 may be associated with liver injuries in chronic hepatitis B. However, hepatic expression of miR-122 does not seem to correspond to progression of the liver disease.


Subject(s)
Hepatitis B, Chronic/metabolism , Liver/metabolism , MicroRNAs/metabolism , Adult , Cytochrome Reductases/metabolism , Female , Gene Expression , Heme Oxygenase-1/metabolism , Hepatitis B, Chronic/genetics , Hepatitis B, Chronic/virology , Humans , Liver/pathology , Male , MicroRNAs/genetics , Middle Aged , NAD(P)H Dehydrogenase (Quinone)/metabolism , Oxidoreductases Acting on Sulfur Group Donors , Viral Load , Young Adult
13.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 31(8): 1017-21, 2015 Aug.
Article in Chinese | MEDLINE | ID: mdl-26271971

ABSTRACT

OBJECTIVE: To investigate the effects of over-expression of 23 kDa augmenter of liver regeneration (ALR) on cell proliferation and apoptosis in the normal human hepatic cell line LO2. METHODS: The recombinant plasmid expressing 23 kDa ALR (pcDNA6/23 kDa ALR) was constructed and transfected into LO2 cells with MegaTran 1.0 transfection reagent. The expressions of ALR mRNA and protein in LO2 cells were detected by real-time quantitative PCR and Western blotting, respectively; MTS assay was used to detect the cell proliferation of LO2 cells; cell cycle and apoptosis of LO2 cells were measured by flow cytometry. RESULTS: The recombinant expression plasmid pcDNA6/23 kDa ALR was constructed successfully, and the expression of the target protein 23 kDa ALR increased significantly in the transfected cells. Compared with pcDNA6-myc/HisA group, the transient transfection of pcDNA6/23 kDa ALR into LO2 cells promoted cell proliferation and inhibited cell apoptosis induced by H2O2, however, no significant differences were detected in G0 phase and S phase. CONCLUSION: The over-expression of 23 kDa ALR in LO2 cells promoted the cell proliferation and enhanced cell resistance to H2O2.


Subject(s)
Apoptosis/drug effects , Cell Proliferation , Cytochrome Reductases/metabolism , Hydrogen Peroxide/pharmacology , Apoptosis/genetics , Blotting, Western , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line , Cytochrome Reductases/genetics , Flow Cytometry , Gene Expression , Hep G2 Cells , Humans , Liver/cytology , Liver/drug effects , Liver/metabolism , Oxidants/pharmacology , Oxidoreductases Acting on Sulfur Group Donors , Reverse Transcriptase Polymerase Chain Reaction
14.
World J Gastroenterol ; 21(17): 5250-8, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25954098

ABSTRACT

AIM: To investigate the role of autophagy in the anti-apoptotic effect of augmenter of liver regeneration (ALR). METHODS: Autophagy was induced through serum deprivation. An ALR-expressing plasmid was transfected into HepG2 cells, and autophagic flux was determined using fluorescence microscopy, electron microscopy, Western blot and quantitative polymerase chain reaction (qPCR) assays. After ALR-expressing plasmid transfection, an autophagy inhibitor [3-methyladenine (3-MA)] was added to HepG2 cells, and apoptosis was observed using fluorescence microscopy and flow cytometry. RESULTS: Autophagy was activated in HepG2 cells, peaking at 24 h after serum deprivation. Microtubule-associated protein light chain three-II levels were higher in HepG2 cells treated with ALR than in control cells, fluorescence microscopy, electron microscopy and qPCR studies showed the similar trend, and p62 levels showed the opposite trend, which indicated that ALR may play an important role in increasing autophagy flux. The numbers of apoptotic cells were substantially higher in HepG2 cells treated with both ALR and 3-MA than in cells treated with ALR alone. Therefore, the protective effect of ALR was significantly attenuated or abolished when autophagy was inhibited, indicating that the anti-apoptotic effect of ALR may be related to autophagy. CONCLUSION: ALR protects cells from apoptosis partly through increased autophagy in HepG2 cells and may be valuable as a new therapeutic treatment for liver disease.


Subject(s)
Autophagy , Cytochrome Reductases/metabolism , Hepatocytes/enzymology , Adenine/analogs & derivatives , Adenine/pharmacology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Beclin-1 , Cytochrome Reductases/genetics , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/ultrastructure , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Oxidoreductases Acting on Sulfur Group Donors , Signal Transduction , Time Factors , Transfection , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism
15.
Orv Hetil ; 156(13): 503-9, 2015 Mar 29.
Article in Hungarian | MEDLINE | ID: mdl-25796277

ABSTRACT

ALR is a mystic protein. It has a so called "long" 22 kDa and a "short" 15 kDa forms. It has been described after partial hepatectomy and it has just been considered as a key protein of liver regeneration. At the beginning of the 21st century it has been revealed that the "long" form is localized in the mitochondrial intermembrane space and it is an element of the mitochondrial protein import and disulphide relay system. Several proteins of the substrates of the mitochondrial disulphide relay system are necessary for the proper function of the mitochondria, thus any mutation of the ALR gene leads to mitochondrial diseases. The "short" form of ALR functions as a secreted extracellular growth factor and it promotes the protection, regeneration and proliferation of hepatocytes. The results gained on the recently generated conditional ALR mutant mice suggest that ALR can play an important role in the pathogenesis of alcoholic and non-alcoholic steatosis. Since the serum level of ALR is modified in several liver diseases it can be a promising marker molecule in laboratory diagnostics.


Subject(s)
Cytochrome Reductases/physiology , Hepatocytes/metabolism , Liver Regeneration , Mitochondria, Liver/metabolism , Animals , Cytochrome Reductases/genetics , Cytochrome Reductases/metabolism , Hepatectomy , Humans , Mutation , Oxidoreductases Acting on Sulfur Group Donors
16.
Apoptosis ; 20(4): 423-32, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25633409

ABSTRACT

Oxidative stress plays an important role in cellular destruction. Augmenter of liver regeneration (ALR) is an anti-apoptotic factor that is expressed in all mammalian cells and functions as an anti-oxidant by stimulating the expression of a secretory isoform of clusterin and inhibiting reactive oxygen species (ROS) generation. Previous work from our group showed that ALR expression is upregulated in acute kidney injury (AKI) rats, and recombinant human ALR reduces tubular injury. In the present study, we used small interfering RNA (siRNA) silencing of ALR to examine its role in H2O2 induced mitochondrial injury and apoptosis. Knockdown of ALR increased ROS levels, reduced mitochondrial membrane potential, and increased the release of mitochondrial proteins and the rate of apoptosis in response to H2O2. In addition, the ratio of Bax/Bcl-2 was increased in siRNA/ALR groups treated with H2O2. These data confirm the protective role of ALR against oxidative stress-induced mitochondrial injury and suggest a potential mechanism underlying the protective role of ALR in AKI.


Subject(s)
Acute Kidney Injury/enzymology , Apoptosis , Cytochrome Reductases/metabolism , Hydrogen Peroxide/metabolism , Kidney Tubules, Proximal/enzymology , Oxidative Stress , Acute Kidney Injury/genetics , Acute Kidney Injury/metabolism , Acute Kidney Injury/physiopathology , Cytochrome Reductases/genetics , Humans , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/physiopathology , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/metabolism , Oxidoreductases Acting on Sulfur Group Donors
17.
Biochem J ; 464(3): 449-59, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25269795

ABSTRACT

Erv1 (essential for respiration and viability 1) is an FAD-dependent thiol oxidase of the Erv/ALR (augmenter of liver regeneration) sub-family. It is an essential component of the mitochondrial import and assembly (MIA) pathway, playing an important role in the oxidative folding of the mitochondrial intermembrane space (IMS) proteins and linking the MIA pathway to the mitochondrial respiratory chain via cytochrome c (cyt c). The importance of the Erv/ALR enzymes was also demonstrated in a recent study where a single mutation in the human ALR (R194H) leads to autosomal recessive myopathy [Di Fonzo, Ronchi, Lodi, Fassone, Tigano, Lamperti, Corti, Bordoni, Fortunato, Nizzardo et al. (2009) Am. J. Hum. Genet. 84, 594-604]. However, the molecular mechanism of the disease is still unclear. In the present study, we use yeast Erv1 as a model to provide clear evidence for a progressive functional defect in the catalytic activity of the corresponding Erv1 R182H mutant. We show that the FAD cofactor was released from Erv1 R182H during its catalytic cycle, which led to the inactivation of the enzyme. We also characterized the effects of the mutation on the folding and stability of Erv1 and tested our in vitro findings in vivo using a yeast genetic approach. The results of the present study allow us to provide a model for the functional defect in Erv1 R182H, which could potentially be extended to human ALR R194H and provides insights into the molecular basis of autosomal recessive myopathy.


Subject(s)
Cytochrome Reductases/genetics , Cytochrome Reductases/metabolism , Muscular Diseases/genetics , Mutation, Missense , Amino Acid Sequence , Amino Acid Substitution , Catalysis , Catalytic Domain/genetics , Coenzymes/metabolism , Cytochrome Reductases/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Oxidoreductases Acting on Sulfur Group Donors , Protein Binding , Protein Structure, Tertiary/genetics , Sequence Homology, Amino Acid
19.
J Biol Chem ; 289(15): 10345-10358, 2014 Apr 11.
Article in English | MEDLINE | ID: mdl-24500710

ABSTRACT

Mitochondrial amidoxime reducing component (mARC) proteins are molybdopterin-containing enzymes of unclear physiological function. Both human isoforms mARC-1 and mARC-2 are able to catalyze the reduction of nitrite when they are in the reduced form. Moreover, our results indicate that mARC can generate nitric oxide (NO) from nitrite when forming an electron transfer chain with NADH, cytochrome b5, and NADH-dependent cytochrome b5 reductase. The rate of NO formation increases almost 3-fold when pH was lowered from 7.5 to 6.5. To determine if nitrite reduction is catalyzed by molybdenum in the active site of mARC-1, we mutated the putative active site cysteine residue (Cys-273), known to coordinate molybdenum binding. NO formation was abolished by the C273A mutation in mARC-1. Supplementation of transformed Escherichia coli with tungsten facilitated the replacement of molybdenum in recombinant mARC-1 and abolished NO formation. Therefore, we conclude that human mARC-1 and mARC-2 are capable of catalyzing reduction of nitrite to NO through reaction with its molybdenum cofactor. Finally, expression of mARC-1 in HEK cells using a lentivirus vector was used to confirm cellular nitrite reduction to NO. A comparison of NO formation profiles between mARC and xanthine oxidase reveals similar Kcat and Vmax values but more sustained NO formation from mARC, possibly because it is not vulnerable to autoinhibition via molybdenum desulfuration. The reduction of nitrite by mARC in the mitochondria may represent a new signaling pathway for NADH-dependent hypoxic NO production.


Subject(s)
Coenzymes/metabolism , Metalloproteins/metabolism , Mitochondria/enzymology , Mitochondrial Proteins/metabolism , Nitric Oxide Synthase/metabolism , Nitrite Reductases/metabolism , Oxidoreductases/metabolism , Pteridines/metabolism , Amino Acid Sequence , Cytochrome Reductases/metabolism , Cytochromes b5/metabolism , Electron Transport , HEK293 Cells , Humans , Hydrogen-Ion Concentration , Kinetics , Molecular Sequence Data , Molybdenum/metabolism , Molybdenum Cofactors , Nitric Oxide/metabolism , Nitrites/metabolism , Oxygen/metabolism , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Xanthine Oxidase/metabolism
20.
Biochemistry ; 52(46): 8323-32, 2013 Nov 19.
Article in English | MEDLINE | ID: mdl-24147449

ABSTRACT

Augmenter of liver regeneration is a member of the ERV family of small flavin-dependent sulfhydryl oxidases that contain a redox-active CxxC disulfide bond in redox communication with the isoalloxazine ring of bound FAD. These enzymes catalyze the oxidation of thiol substrates with the reduction of molecular oxygen to hydrogen peroxide. This work studies the catalytic mechanism of the short, cytokine form of augmenter of liver regeneration (sfALR) using model thiol substrates of the enzyme. The redox potential of the proximal disulfide in sfALR was found to be approximately 57 mV more reducing than the flavin chromophore, in agreement with titration experiments. Rapid reaction studies show that dithiothreitol (DTT) generates a transient mixed disulfide intermediate with sfALR signaled by a weak charge-transfer interaction between the thiolate of C145 and the oxidized flavin. The subsequent transfer of reducing equivalents to the flavin ring is relatively slow, with a limiting apparent rate constant of 12.4 s(-1). However, reoxidation of the reduced flavin by molecular oxygen is even slower (2.3 s(-1) at air saturation) and thus largely limits turnover at 5 mM DTT. The nature of the charge-transfer complexes observed with DTT was explored using a range of simple monothiols to mimic the initial nucleophilic attack on the proximal disulfide. While ß-mercaptoethanol is a very poor substrate of sfALR (∼0.3 min(-1) at 100 mM thiol), it rapidly generates a mixed disulfide intermediate allowing the thiolate of C145 to form a strong charge-transfer complex with the flavin. Unlike the other monothiols tested, glutathione is unable to form charge-transfer complexes and is an undetectable substrate of the oxidase. These data are rationalized on the basis of the stringent steric requirements for thiol-disulfide exchange reactions. The inability of the relatively bulky glutathione to attain the in-line geometry required for efficient disulfide exchange in sfALR may be physiologically important in preventing the oxidase from catalyzing the potentially harmful oxidation of intracellular glutathione.


Subject(s)
Cytochrome Reductases/metabolism , Oxidoreductases/metabolism , Catalysis , Cytochrome Reductases/genetics , Disulfides/chemistry , Flavins/chemistry , Flavins/metabolism , Humans , Mercaptoethanol/metabolism , Models, Molecular , Oxidation-Reduction , Oxidoreductases/genetics , Oxidoreductases Acting on Sulfur Group Donors , Oxygen/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...