Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(12)2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38928353

ABSTRACT

The lumen of the endoplasmic reticulum (ER) is usually considered an oxidative environment; however, oxidized thiol-disulfides and reduced pyridine nucleotides occur there parallelly, indicating that the ER lumen lacks components which connect the two systems. Here, we investigated the luminal presence of the thioredoxin (Trx)/thioredoxin reductase (TrxR) proteins, capable of linking the protein thiol and pyridine nucleotide pools in different compartments. It was shown that specific activity of TrxR in the ER is undetectable, whereas higher activities were measured in the cytoplasm and mitochondria. None of the Trx/TrxR isoforms were expressed in the ER by Western blot analysis. Co-localization studies of various isoforms of Trx and TrxR with ER marker Grp94 by immunofluorescent analysis further confirmed their absence from the lumen. The probability of luminal localization of each isoform was also predicted to be very low by several in silico analysis tools. ER-targeted transient transfection of HeLa cells with Trx1 and TrxR1 significantly decreased cell viability and induced apoptotic cell death. In conclusion, the absence of this electron transfer chain may explain the uncoupling of the redox systems in the ER lumen, allowing parallel presence of a reduced pyridine nucleotide and a probably oxidized protein pool necessary for cellular viability.


Subject(s)
Endoplasmic Reticulum , Oxidation-Reduction , Thioredoxin-Disulfide Reductase , Thioredoxins , Humans , Thioredoxins/metabolism , Thioredoxins/genetics , Endoplasmic Reticulum/metabolism , HeLa Cells , Thioredoxin-Disulfide Reductase/metabolism , Thioredoxin-Disulfide Reductase/genetics , Mitochondria/metabolism , Apoptosis , Cell Survival
2.
Cell Struct Funct ; 46(1): 1-9, 2021 Feb 19.
Article in English | MEDLINE | ID: mdl-33361684

ABSTRACT

The somatic haploidy is unstable in diplontic animals, but cellular processes determining haploid stability remain elusive. Here, we found that inhibition of mevalonate pathway by pitavastatin, a widely used cholesterol-lowering drug, drastically destabilized the haploid state in HAP1 cells. Interestingly, cholesterol supplementation did not restore haploid stability in pitavastatin-treated cells, and cholesterol inhibitor U18666A did not phenocopy haploid destabilization. These results ruled out the involvement of cholesterol in haploid stability. Besides cholesterol perturbation, pitavastatin induced endoplasmic reticulum (ER) stress, the suppression of which by a chemical chaperon significantly restored haploid stability in pitavastatin-treated cells. Our data demonstrate the involvement of the mevalonate pathway in the stability of the haploid state in human somatic cells through managing ER stress, highlighting a novel link between ploidy and ER homeostatic control.Key words: haploid, ER stress, Mevalonate pathway.


Subject(s)
Endoplasmic Reticulum Stress , Homeostasis , Cell Line , Cholesterol , Haploidy , Humans
3.
Antioxid Redox Signal ; 34(11): 875-889, 2021 04 10.
Article in English | MEDLINE | ID: mdl-31621376

ABSTRACT

Significance: Cardiovascular disorders are the most important cause of morbidity and mortality in the Western world. Monogenic developmental disorders of the heart and vessels are highly valuable to study the physiological and pathological processes in cardiovascular system homeostasis. The arterial tortuosity syndrome (ATS) is a rare, autosomal recessive connective tissue disorder showing lengthening, tortuosity, and stenosis of the large arteries, with a propensity for aneurysm formation. In histopathology, it associates with fragmentation and disorganization of elastic fibers in several tissues, including the arterial wall. ATS is caused by pathogenic variants in SLC2A10 encoding the facilitative glucose transporter (GLUT)10. Critical Issues: Although several hypotheses have been forwarded, the molecular mechanisms linking disrupted GLUT10 activity with arterial malformations are largely unknown. Recent Advances: The vascular and systemic manifestations and natural history of ATS patients have been largely delineated. GLUT10 was identified as an intracellular transporter of dehydroascorbic acid, which contributes to collagen and elastin cross-linking in the endoplasmic reticulum, redox homeostasis in the mitochondria, and global and gene-specific methylation/hydroxymethylation affecting epigenetic regulation in the nucleus. We revise here the current knowledge on ATS and the role of GLUT10 within the compartmentalization of ascorbate in physiological and diseased states. Future Directions: Centralization of clinical, treatment, and outcome data will enable better management for ATS patients. Establishment of representative animal disease models could facilitate the study of pathomechanisms underlying ATS. This might be relevant for other forms of vascular dysplasia, such as isolated aneurysm formation, hypertensive vasculopathy, and neovascularization. Antioxid. Redox Signal. 34, 875-889.


Subject(s)
Arteries/abnormalities , Ascorbic Acid/genetics , Glucose Transport Proteins, Facilitative/genetics , Homeostasis/genetics , Joint Instability/genetics , Skin Diseases, Genetic/genetics , Vascular Malformations/genetics , Animals , Arteries/metabolism , Arteries/pathology , Ascorbic Acid/metabolism , Ascorbic Acid/therapeutic use , Elastic Tissue/metabolism , Elastic Tissue/pathology , Humans , Joint Instability/metabolism , Joint Instability/pathology , Joint Instability/therapy , Mitochondria/drug effects , Mitochondria/genetics , Mutation/genetics , Oxidation-Reduction , Skin Diseases, Genetic/metabolism , Skin Diseases, Genetic/pathology , Skin Diseases, Genetic/therapy , Vascular Malformations/metabolism , Vascular Malformations/pathology , Vascular Malformations/therapy
4.
Int J Mol Sci ; 20(23)2019 Nov 24.
Article in English | MEDLINE | ID: mdl-31771288

ABSTRACT

Glucose is a basic nutrient in most of the creatures; its transport through biological membranes is an absolute requirement of life. This role is fulfilled by glucose transporters, mediating the transport of glucose by facilitated diffusion or by secondary active transport. GLUT (glucose transporter) or SLC2A (Solute carrier 2A) families represent the main glucose transporters in mammalian cells, originally described as plasma membrane transporters. Glucose transport through intracellular membranes has not been elucidated yet; however, glucose is formed in the lumen of various organelles. The glucose-6-phosphatase system catalyzing the last common step of gluconeogenesis and glycogenolysis generates glucose within the lumen of the endoplasmic reticulum. Posttranslational processing of the oligosaccharide moiety of glycoproteins also results in intraluminal glucose formation in the endoplasmic reticulum (ER) and Golgi. Autophagic degradation of polysaccharides, glycoproteins, and glycolipids leads to glucose accumulation in lysosomes. Despite the obvious necessity, the mechanism of glucose transport and the molecular nature of mediating proteins in the endomembranes have been hardly elucidated for the last few years. However, recent studies revealed the intracellular localization and functional features of some glucose transporters; the aim of the present paper was to summarize the collected knowledge.


Subject(s)
Glucose Transport Proteins, Facilitative/metabolism , Glucose/metabolism , Sodium-Glucose Transport Proteins/metabolism , Animals , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Glucose-6-Phosphatase/metabolism , Golgi Apparatus/metabolism , Humans
5.
Int J Mol Sci ; 20(20)2019 Oct 11.
Article in English | MEDLINE | ID: mdl-31614497

ABSTRACT

Besides the liver, which has always been considered the major source of endogenous glucose production in all post-absorptive situations, kidneys and intestines can also produce glucose in blood, particularly during fasting and under protein feeding. However, observations gained in different experimental animals have given ambiguous results concerning the presence of the glucose-6-phosphatase system in the small intestine. The aim of this study was to better define the species-related differences of this putative gluconeogenic organ in glucose homeostasis. The components of the glucose-6-phosphatase system (i.e., glucose-6-phosphate transporter and glucose-6-phosphatase itself) were analyzed in homogenates or microsomal fractions prepared from the small intestine mucosae and liver of rats, guinea pigs, and humans. Protein and mRNA levels, as well as glucose-6-phosphatase activities, were detected. The results showed that the glucose-6-phosphatase system is poorly represented in the small intestine of rats; on the other hand, significant expressions of glucose-6-phosphate transporter and of the glucose-6-phosphatase were found in the small intestine of guinea pigs and homo sapiens. The activity of the recently described fructose-6-phosphate transporter-intraluminal hexose isomerase pathway was also present in intestinal microsomes from these two species. The results demonstrate that the gluconeogenic role of the small intestine is highly species-specific and presumably dependent on feeding behavior (e.g., fructose consumption) and the actual state of metabolism.


Subject(s)
Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Intestine, Small/enzymology , Animals , Fructose/metabolism , Guinea Pigs , Humans , Microsomes/enzymology , Rats , Species Specificity
6.
Eur J Med Genet ; 62(8): 103669, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31082515

ABSTRACT

Preimplantation genetic testing for aneuploidy (PGT-A) is a suitable technique to identify euploid embryos, which have the highest potential to implant, thus increase the chance of a healthy live birth. The main indications of PGT-A are advanced maternal age, repeated implantation failure, repeated miscarriages and severe male infertility. Several studies have already proven that testing embryos for genetic abnormalities in the above cases results in higher implantation rate and reduced number of pregnancy loss. In spite of these - due to a legislative change in Hungary in 2015 - PGT-A was reclassified as an experimental procedure and its use became banned throughout the country. For this reason, after 4 years of successful practice, Hungarian patients were not able to participate in IVF procedure combined with PGT-A anymore. In this retrospective analysis, efficacy of PGT-A-based embryo selection was evaluated and was compared to the conventional morphology-based selection (MBS) in patients with advanced maternal age, between 2013 and 2017 at our private fertility clinic. PGT-A was performed with array comparative genomic hybridization. We found that implantation rate was significantly higher (43.62% vs. 27.88%; p = 0.0208) and miscarriage rate was significantly lower (17.07% vs. 37.93%; p = 0.0492) in the PGT-A group compared to the MBS group from 2013 to 2015. These outcomes were achieved with a significantly lower number of transferred embryos in the PGT-A group (1.25 vs. 1.58; p = 0.0003). In 2016-2017, the number of transferred embryos were significantly reduced in the MBS group as well (1.14 vs. 1.58; p < 0.0001). However, outcomes of the IVF treatments did not change significantly compared to the previous two years (2013-2015). Our results imply that PGT-A-based embryo selection is more efficient than morphology-based selection in patients with advanced maternal age. Therefore, prohibition of the use of PGT-A had significant consequences on the efficiency and safety of IVF treatment in the country.


Subject(s)
Abortion, Spontaneous/diagnosis , Aneuploidy , Fertilization in Vitro/methods , Preimplantation Diagnosis , Abortion, Spontaneous/epidemiology , Abortion, Spontaneous/genetics , Abortion, Spontaneous/pathology , Adult , Blastocyst/metabolism , Blastocyst/pathology , Comparative Genomic Hybridization , Embryo Transfer/methods , Female , Humans , Hungary/epidemiology , Patient Care , Pregnancy
7.
Oxid Med Cell Longev ; 2019: 8156592, 2019.
Article in English | MEDLINE | ID: mdl-30800210

ABSTRACT

Ascorbate requiring Fe2+/2-oxoglutarate-dependent dioxygenases located in the nucleoplasm have been shown to participate in epigenetic regulation of gene expression via histone and DNA demethylation. Transport of dehydroascorbic acid is impaired in the endomembranes of fibroblasts from arterial tortuosity syndrome (ATS) patients, due to the mutation in the gene coding for glucose transporter GLUT10. We hypothesized that altered nuclear ascorbate concentration might be present in ATS fibroblasts, affecting dioxygenase activity and DNA demethylation. Therefore, our aim was to characterize the subcellular distribution of vitamin C, the global and site-specific changes in 5-methylcytosine and 5-hydroxymethylcytosine levels, and the effect of ascorbate supplementation in control and ATS fibroblast cultures. Diminished nuclear accumulation of ascorbate was found in ATS fibroblasts upon ascorbate or dehydroascorbic acid addition. Analyzing DNA samples of cultured fibroblasts from controls and ATS patients, a lower global 5-hydroxymethylcytosine level was found in ATS fibroblasts, which could not be significantly modified by ascorbate addition. Investigation of the (hydroxy)methylation status of specific regions in six candidate genes related to ascorbate metabolism and function showed that ascorbate addition could stimulate hydroxymethylation and active DNA demethylation at the PPAR-γ gene region in control fibroblasts only. The altered DNA hydroxymethylation patterns in patient cells both at the global level and at specific gene regions accompanied with decreased nuclear accumulation of ascorbate suggests the epigenetic role of vitamin C in the pathomechanism of ATS. The present findings represent the first example for the role of vitamin C transport in epigenetic regulation suggesting that ATS is a compartmentalization disease.


Subject(s)
Arteries/abnormalities , Ascorbic Acid/metabolism , Cell Nucleus/metabolism , DNA Methylation/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Genome, Human , Joint Instability/genetics , Skin Diseases, Genetic/genetics , Vascular Malformations/genetics , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Cells, Cultured , Epigenesis, Genetic , Humans , Models, Biological , PPAR gamma/genetics , PPAR gamma/metabolism
8.
Int J Mol Sci ; 18(8)2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28829359

ABSTRACT

GLUT10 belongs to a family of transporters that catalyze the uptake of sugars/polyols by facilitated diffusion. Loss-of-function mutations in the SLC2A10 gene encoding GLUT10 are responsible for arterial tortuosity syndrome (ATS). Since subcellular distribution of the transporter is dubious, we aimed to clarify the localization of GLUT10. In silico GLUT10 localization prediction suggested its presence in the endoplasmic reticulum (ER). Immunoblotting showed the presence of GLUT10 protein in the microsomal, but not in mitochondrial fractions of human fibroblasts and liver tissue. An even cytosolic distribution with an intense perinuclear decoration of GLUT10 was demonstrated by immunofluorescence in human fibroblasts, whilst mitochondrial markers revealed a fully different decoration pattern. GLUT10 decoration was fully absent in fibroblasts from three ATS patients. Expression of exogenous, tagged GLUT10 in fibroblasts from an ATS patient revealed a strict co-localization with the ER marker protein disulfide isomerase (PDI). The results demonstrate that GLUT10 is present in the ER.


Subject(s)
Arteries/abnormalities , Endoplasmic Reticulum/metabolism , Fibroblasts/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Joint Instability/metabolism , Skin Diseases, Genetic/metabolism , Vascular Malformations/metabolism , Arteries/metabolism , Fluorescent Antibody Technique , Humans , Intracellular Space/metabolism , Joint Instability/genetics , Microsomes/metabolism , Protein Binding , Protein Transport , Skin Diseases, Genetic/genetics , Vascular Malformations/genetics
9.
Int J Mol Sci ; 18(1)2017 Jan 05.
Article in English | MEDLINE | ID: mdl-28067773

ABSTRACT

Accumulation of misfolded/unfolded proteins in the endoplasmic reticulum (ER) leads to the activation of three branches (Protein kinase (RNA)-like endoplasmic reticulum kinase [PERK], Inositol requiring protein 1 [IRE-1] and Activating trascription factor 6 [ATF6], respectively) of unfolded protein response (UPR). The primary role of UPR is to try to drive back the system to the former or a new homeostatic state by self-eating dependent autophagy, while excessive level of ER stress results in apoptotic cell death. Our study focuses on the role of PERK- and IRE-1-induced arms of UPR in life-or-death decision. Here we confirm that silencing of PERK extends autophagy-dependent survival, whereas the IRE-1-controlled apoptosis inducer is downregulated during ER stress. We also claim that the proper order of surviving and self-killing mechanisms is controlled by a positive feedback loop between PERK and IRE-1 branches. This regulatory network makes possible a smooth, continuous activation of autophagy with respect to ER stress, while the induction of apoptosis is irreversible and switch-like. Using our knowledge of molecular biological techniques and systems biological tools we give a qualitative description about the dynamical behavior of PERK- and IRE-1-controlled life-or-death decision. Our model claims that the two arms of UPR accomplish an altered upregulation of autophagy and apoptosis inducers during ER stress. Since ER stress is tightly connected to aging and age-related degenerative disorders, studying the signaling pathways of UPR and their role in maintaining ER proteostasis have medical importance.


Subject(s)
Endoplasmic Reticulum Stress/genetics , Signal Transduction/genetics , Systems Biology/methods , eIF-2 Kinase/genetics , Apoptosis/genetics , Autophagy/genetics , Blotting, Western , Cell Survival/genetics , Endoribonucleases/genetics , Endoribonucleases/metabolism , Feedback, Physiological , Gene Expression , HEK293 Cells , Homeostasis/genetics , Humans , Models, Biological , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Unfolded Protein Response/genetics , eIF-2 Kinase/metabolism
10.
FEBS Lett ; 590(11): 1630-40, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27153185

ABSTRACT

Loss-of-function mutations in the gene encoding GLUT10 are responsible for arterial tortuosity syndrome (ATS), a rare connective tissue disorder. In this study GLUT10-mediated dehydroascorbic acid (DAA) transport was investigated, supposing its involvement in the pathomechanism. GLUT10 protein produced by in vitro translation and incorporated into liposomes efficiently transported DAA. Silencing of GLUT10 decreased DAA transport in immortalized human fibroblasts whose plasma membrane was selectively permeabilized. Similarly, the transport of DAA through endomembranes was markedly reduced in fibroblasts from ATS patients. Re-expression of GLUT10 in patients' fibroblasts restored DAA transport activity. The present results demonstrate that GLUT10 is a DAA transporter and DAA transport is diminished in the endomembranes of fibroblasts from ATS patients.


Subject(s)
Arteries/abnormalities , Dehydroascorbic Acid/metabolism , Glucose Transport Proteins, Facilitative/genetics , Joint Instability/genetics , Skin Diseases, Genetic/genetics , Vascular Malformations/genetics , Ascorbic Acid/metabolism , Biological Transport/drug effects , Biological Transport/genetics , Cells, Cultured , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Glucose Transport Proteins, Facilitative/antagonists & inhibitors , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , RNA Interference , RNA, Small Interfering/pharmacology
11.
Free Radic Biol Med ; 94: 157-60, 2016 05.
Article in English | MEDLINE | ID: mdl-26928585

ABSTRACT

Cellular metabolism provides various sources of hydrogen peroxide (H2O2) in different organelles and compartments. The suitability of H2O2 as an intracellular signaling molecule therefore also depends on its ability to pass cellular membranes. The propensity of the membranous boundary of the endoplasmic reticulum (ER) to let pass H2O2 has been discussed controversially. In this essay, we challenge the recent proposal that the ER membrane constitutes a simple barrier for H2O2 diffusion and support earlier data showing that (i) ample H2O2 permeability of the ER membrane is a prerequisite for signal transduction, (ii) aquaporin channels are crucially involved in the facilitation of H2O2 permeation, and (iii) a proper experimental framework not prone to artifacts is necessary to further unravel the role of H2O2 permeation in signal transduction and organelle biology.


Subject(s)
Cell Membrane Permeability , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Hydrogen Peroxide/metabolism , Cell Membrane/chemistry , Diffusion , Endoplasmic Reticulum/chemistry , Hydrogen Peroxide/chemistry , Reactive Oxygen Species/metabolism , Signal Transduction
12.
Biochim Biophys Acta ; 1852(5): 826-38, 2015 May.
Article in English | MEDLINE | ID: mdl-25643868

ABSTRACT

Polyunsaturated fatty acids are susceptible to peroxidation and they yield various degradation products, including the main α,ß-unsaturated hydroxyalkenal, 4-hydroxy-2,3-trans-nonenal (HNE) in oxidative stress. Due to its high reactivity, HNE interacts with various macromolecules of the cell, and this general toxicity clearly contributes to a wide variety of pathological conditions. In addition, growing evidence suggests a more specific function of HNE in electrophilic signaling as a second messenger of oxidative/electrophilic stress. It can induce antioxidant defense mechanisms to restrain its own production and to enhance the cellular protection against oxidative stress. Moreover, HNE-mediated signaling can largely influence the fate of the cell through modulating major cellular processes, such as autophagy, proliferation and apoptosis. This review focuses on the molecular mechanisms underlying the signaling and regulatory functions of HNE. The role of HNE in the pathophysiology of cancer, cardiovascular and neurodegenerative diseases is also discussed.


Subject(s)
Aldehydes/metabolism , Cell Physiological Phenomena/physiology , Disease , Signal Transduction/physiology , Aldehydes/chemistry , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/physiopathology , Humans , Molecular Structure , Neoplasms/metabolism , Neoplasms/physiopathology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology
13.
Free Radic Biol Med ; 83: 331-40, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25678412

ABSTRACT

The endoplasmic reticulum (ER) is a metabolically active organelle, which has a central role in proteostasis by translating, modifying, folding, and occasionally degrading secretory and membrane proteins. The lumen of the ER represents a separate compartment of the eukaryotic cell, with a characteristic proteome and metabolome. Although the redox metabolome and proteome of the compartment have not been holistically explored, it is evident that proper redox conditions are necessary for the functioning of many luminal pathways. These redox conditions are defined by local oxidoreductases and the membrane transport of electron donors and acceptors. The main electron carriers of the compartment are identical with those of the other organelles: glutathione, pyridine and flavin nucleotides, ascorbate, and others. However, their composition, concentration, and redox state in the ER lumen can be different from those observed in other compartments. The terminal oxidases of oxidative protein folding generate and maintain an "oxidative environment" by oxidizing protein thiols and producing hydrogen peroxide. ER-specific mechanisms reutilize hydrogen peroxide as an electron acceptor of oxidative folding. These mechanisms, together with membrane and kinetic barriers, guarantee that redox systems in the reduced or oxidized state can be present simultaneously in the lumen. The present knowledge on the in vivo conditions of ER redox is rather limited; development of new genetically encoded targetable sensors for the measurement of the luminal state of redox systems other than thiol/disulfide will contribute to a better understanding of ER redox homeostasis.


Subject(s)
Endoplasmic Reticulum/physiology , Homeostasis/physiology , Hydrogen Peroxide/metabolism , Oxidants/metabolism , Animals , Humans , Oxidation-Reduction
14.
Biochim Biophys Acta ; 1843(9): 1909-16, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24907663

ABSTRACT

Beyond its general role as antioxidant, specific functions of ascorbate are compartmentalized within the eukaryotic cell. The list of organelle-specific functions of ascorbate has been recently expanded with the epigenetic role exerted as a cofactor for DNA and histone demethylases in the nucleus. Compartmentation necessitates the transport through intracellular membranes; members of the GLUT family and sodium-vitamin C cotransporters mediate the permeation of dehydroascorbic acid and ascorbate, respectively. Recent observations show that increased consumption and/or hindered entrance of ascorbate in/to a compartment results in pathological alterations partially resembling to scurvy, thus diseases of ascorbate compartmentation can exist. The review focuses on the reactions and transporters that can modulate ascorbate concentration and redox state in three compartments: endoplasmic reticulum, mitochondria and nucleus. By introducing the relevant experimental and clinical findings we make an attempt to coin the term of ascorbate compartmentation disease.


Subject(s)
Ascorbic Acid/metabolism , Cell Compartmentation , Disease , Animals , Gene Expression Regulation , Humans , Models, Biological , Organelles/metabolism
15.
FEBS Lett ; 586(19): 3354-9, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22819816

ABSTRACT

A phosphate-linked antiporter activity of the glucose-6-phosphate transporter (G6PT) has been recently described in liposomes including the reconstituded transporter protein. We directly investigated the mechanism of glucose-6-phosphate (G6P) transport in rat liver microsomal vesicles. Pre-loading with inorganic phosphate (Pi) did not stimulate G6P or Pi microsomal inward transport. Pi efflux from pre-loaded microsomes could not be enhanced by G6P or Pi addition. Rapid G6P or Pi influx was registered by light-scattering in microsomes not containing G6P or Pi. The G6PT inhibitor, S3483, blocked G6P transport irrespectively of experimental conditions. We conclude that hepatic G6PT functions as an uniporter.


Subject(s)
Antiporters/metabolism , Glucose-6-Phosphate/metabolism , Microsomes, Liver/metabolism , Monosaccharide Transport Proteins/metabolism , Animals , Biological Transport, Active , In Vitro Techniques , Kinetics , Light , Male , Permeability , Phosphates/metabolism , Rats , Rats, Sprague-Dawley , Scattering, Radiation
16.
Antioxid Redox Signal ; 16(10): 1088-99, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22369093

ABSTRACT

AIMS: Oxidative protein folding in the luminal compartment of endoplasmic reticulum (ER) is thought to be accompanied by the generation of H2O2, as side-product of disulfide bond formation. We aimed to examine the role of H2O2 produced in the lumen, which on one hand can lead to redox imbalance and hence can contribute to ER stress caused by overproduction of secretory proteins; on the other hand, as an excellent electron acceptor, H2O2 might serve as an additional pro-oxidant in physiological oxidative folding. RESULTS: Stimulation of H2O2 production in the hepatic ER resulted in a decrease in microsomal GSH and protein-thiol contents and in a redox shift of certain luminal oxidoreductases in mice. The oxidative effect, accompanied by moderate signs of ER stress and reversible dilation of ER cisternae, was prevented by concomitant reducing treatment. The imbalance also affected the redox state of pyridine nucleotides in the ER. Antibody producing cells artificially engineered with powerful luminal H2O2 eliminating system showed diminished secretion of mature antibody polymers, while incomplete antibody monomers/dimers were accumulated and/or secreted. INNOVATION: Evidence are provided by using in vivo models that hydrogen peroxide can promote disulfide bond formation in the ER. CONCLUSION: The results indicate that local H2O2 production promotes, while quenching of H2O2 impairs disulfide formation. The contribution of H2O2 to disulfide bond formation previously observed in vitro can be also shown in cellular and in vivo systems.


Subject(s)
Endoplasmic Reticulum/metabolism , Hydrogen Peroxide/metabolism , Protein Folding , Animals , Cell Line , Disulfides/chemistry , Guinea Pigs , Humans , Immunoglobulins/chemistry , Immunoglobulins/metabolism , Male , Mice , Oxidation-Reduction
17.
Antioxid Redox Signal ; 16(10): 1077-87, 2012 May 15.
Article in English | MEDLINE | ID: mdl-21854214

ABSTRACT

AIMS: The endoplasmic reticulum (ER) is involved in many functions, including protein folding, redox homeostasis, and Ca(2+) storage and signaling. To perform these multiple tasks, the ER is composed of distinct, specialized subregions, amongst which mitochondrial-associated ER membranes (MAM) emerge as key signaling hubs. How these multiple functions are integrated with one another in living cells remains unclear. RESULTS: Here we show that Ero1α, a key controller of oxidative folding and ER redox homeostasis, is enriched in MAM and regulates Ca(2+) fluxes. Downregulation of Ero1α by RNA interference inhibits mitochondrial Ca(2+) fluxes and modifies the activity of mitochondrial Ca(2+) uniporters. The overexpression of redox active Ero1α increases passive Ca(2+) efflux from the ER, lowering [Ca(2+)](ER) and mitochondrial Ca(2+) fluxes in response to IP3 agonists. INNOVATION: The unexpected observation that Ca(2+) fluxes are affected by either increasing or decreasing the levels of Ero1α reveals a pivotal role for this oxidase in the early secretory compartment and implies a strict control of its amounts. CONCLUSIONS: Taken together, our results indicate that the levels, subcellular localization, and activity of Ero1α coordinately regulate Ca(2+) and redox homeostasis and signaling in the early secretory compartment.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Membrane Glycoproteins/metabolism , Mitochondria/metabolism , Oxidoreductases/metabolism , Cell Line , Homeostasis , Humans , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Membrane Glycoproteins/genetics , Membrane Proteins/metabolism , Molecular Chaperones/metabolism , Oxidation-Reduction , Oxidoreductases/genetics , Protein Binding , Signal Transduction
18.
Antioxid Redox Signal ; 16(8): 772-80, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22142307

ABSTRACT

SIGNIFICANCE: The lumen of the endoplasmic reticulum (ER) constitutes a separate compartment with a special proteome and metabolome. The characteristic redox environment required for the optimal functioning of local pathways is defined by the redox couples of the main electron carriers. These molecules, glutathione, pyridine nucleotides, and ascorbic acid, are present within the ER, but their composition, concentration, and redox state are characteristically different from those observed in other subcellular compartments. Spatial and kinetic barriers contribute to the generation and maintenance of this special redox environment. RECENT ADVANCES: The ER redox has usually been considered from the perspective of oxidative protein folding, one of the major functions of the ER. Thus, the lumen has been described as a relatively oxidizing subcellular compartment. CRITICAL ISSUES: The ER redoxome has been scantily mapped. However, recent observations suggest that the redox systems in reduced and oxidized states are present simultaneously. The concerted actions of transmembrane uptake processes and local oxidoreductases as well as the absence of specific transport and enzyme activities maintain the oxidized state of the thiol-disulfide systems and the reduced state of the pyridine nucleotide redox systems. These states are prerequisites for the normal redox reactions localized in the ER. FUTURE DIRECTIONS: An outline of the interactions between the major electron carriers of the ER will contribute to a better understanding of human diseases related to ER redox homeostasis.


Subject(s)
Endoplasmic Reticulum/metabolism , Adenine Nucleotides/metabolism , Animals , Ascorbic Acid/metabolism , Electron Transport , Endoplasmic Reticulum Stress , Glutathione/metabolism , Humans , Oxidation-Reduction , Reactive Oxygen Species/metabolism
19.
Traffic ; 12(1): 1-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20716108

ABSTRACT

The endoplasmic reticulum (ER) is central for many essential cellular activities, such as folding, assembly and quality control of secretory and membrane proteins, disulfide bond formation, glycosylation, lipid biosynthesis, Ca(2+) storage and signaling. In addition, this multifunctional organelle integrates many adaptive and/or maladaptive signaling cues reporting on metabolism, proteostasis, Ca(2+) and redox homeostasis. We are beginning to understand how these functions and pathways are integrated with one another to regulate homeostasis at cell, tissue and organism levels. The mechanisms underlying the introduction of the proper set of disulfide bonds into secretory proteins (oxidative folding) are strictly related to redox homeostasis, ER stress sensing and signaling and provide a good example of the integration systems operative in the early secretory compartment.


Subject(s)
Oxidation-Reduction , Protein Folding , Signal Transduction , Animals , Cell Compartmentation , Cells/metabolism , Humans
20.
FEBS Lett ; 584(14): 2995-8, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20621831

ABSTRACT

Oxidative protein folding in the luminal compartment of the endoplasmic reticulum is thought to be mediated by a proteinaceous electron relay system composed by PDI and ER oxidoreductin 1 (Ero1), transferring electrons from the cysteinyl residues of substrate proteins to oxygen. However, recent observations revealed that Ero1 isoforms are dispensable. Endoplasmic reticulum is known as a generator and accumulator of low molecular weight oxidants; some of them have already been shown to promote oxidative folding. On the basis of these observations a new theory of oxidative folding is proposed where the oxidative power is provided by the stochastic contribution of prooxidants.


Subject(s)
Endoplasmic Reticulum/metabolism , Models, Biological , Animals , Oxidants/metabolism , Oxidation-Reduction , Oxides/metabolism , Oxygen/metabolism , Protein Folding , Protein Isoforms/metabolism , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...