Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.823
1.
Biochem Pharmacol ; 197: 114939, 2022 03.
Article En | MEDLINE | ID: mdl-35114188

Kidney injury often causes anemia due to a lack of production of the erythroid growth factor erythropoietin (EPO) in the kidneys. Roxadustat is one of the first oral medicines inducing EPO production in patients with renal anemia by activating hypoxia-inducible factors (HIFs), which are activators of EPO gene expression. In this study, to develop prodrugs of roxadustat with improved permeability through cell membrane, we investigated the effects of 8 types of esterification on the pharmacokinetics and bioactivity of roxadustat using Hep3B hepatoma cells that HIF-dependently produce EPO. Mass spectrometry of cells incubated with the esterified roxadustat derivatives revealed that the designed compounds were deesterified after being taken up by cells and showed low cytotoxicity compared to the original compound. Esterification prolonged the effective duration of roxadustat with respect to EPO gene induction and HIF activation in cells transiently exposed to the compounds. In the kidneys and livers of mice, both of which are unique sites of EPO production, a majority of the methyl-esterified roxadustat was deesterified within 6 h after drug administration. The deesterified roxadustat derivative was continuously detectable in plasma and urine for at least 48 h after administration, while the administered compound became undetectable 24 h after administration. Additionally, we confirmed that methyl-esterified roxadustat activated erythropoiesis in mice by inducing Epo mRNA expression exclusively in renal interstitial cells, which have intrinsic EPO-producing potential. These data suggest that esterification could lead to the development of roxadustat prodrugs with improvements in cell membrane permeability, effective duration and cytotoxicity.


Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Survival/drug effects , Glycine/analogs & derivatives , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Intracellular Membranes/metabolism , Isoquinolines/metabolism , Isoquinolines/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/agonists , Cell Survival/physiology , Dose-Response Relationship, Drug , Esterification/drug effects , Esterification/physiology , Glycine/metabolism , Glycine/pharmacology , Humans , Intracellular Membranes/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Time Factors , Treatment Outcome , Tumor Cells, Cultured
2.
Toxicol Lett ; 357: 73-83, 2022 Mar 01.
Article En | MEDLINE | ID: mdl-34999165

MeHg, an environmental toxicant, is highly toxic to the central nervous system. Recent studies have reported that LMP is an important way in the lysosomal damage. However, the role and molecular mechanism of LMP in MeHg-induced neurotoxicity remain unknown. To study MeHg-induced LMP, we used 10µM MeHg to treat SH-SY5Y cells and 2µM MeHg to treat rat cerebral cortical neurons. Acridine orange (AO) staining and analysis of cathepsin B (CTSB) release were used to determine LMP. We found that MeHg reduced red AO fluorescence and induced CTSB release from lysosomes to the cytoplasm in a time-dependent manner. Moreover, pretreatment with the CTSB inhibitor alleviated cytotoxicity in neuronal cells. These results indicate MeHg induces LMP and subsequent CTSB-dependent cytotoxicity in neuronal cells. Bax is a pore-forming protein, which is involved in mitochondrial outer membrane permeabilization. Intriguingly, we demonstrated that MeHg induced Bax to translocate to lysosomes by using immunofluorescence and Western blot analysis of subcellular fractions. Furthermore, downregulating Bax expression suppressed MeHg-induced LMP. Bax subcellular localization is regulated by protein interaction with the cytoplasmic 14-3-3. Our previous study demonstrated that JNK participated in neurotoxicity through regulating protein interaction. In the current study, we showed that JNK dissociated Bax-14-3-3 complex to facilitate Bax lysosomal translocation. Finally, inhibition of the JNK/Bax pathway could alleviate MeHg-induced cytotoxicity in neuronal cells. The present study implies that inhibiting lysosomal damage (LMP)-related signaling might alleviate MeHg neurotoxicity.


Cell Membrane Permeability/drug effects , Intracellular Membranes/drug effects , Lysosomes/drug effects , MAP Kinase Signaling System , Methylmercury Compounds/toxicity , Neurons/drug effects , bcl-2-Associated X Protein/metabolism , Animals , Cell Line , Cells, Cultured , Hazardous Substances/toxicity , Humans , Intracellular Membranes/metabolism , Lysosomes/metabolism , Neurons/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction
3.
Biochem Biophys Res Commun ; 592: 31-37, 2022 02 12.
Article En | MEDLINE | ID: mdl-35016149

Tributyltin (TBT) is an environmental pollutant that remains in marine sediments and is toxic to mammals. For example, TBT elicits neurotoxic and immunosuppressive effects on rats. However, it is not entirely understood how TBT causes toxicity. Autophagy plays a pivotal role in protein quality control and eliminates aggregated proteins and damaged organelles. We previously reported that TBT dephosphorylates mammalian target of rapamycin (mTOR), which may be involved in enhancement of autophagosome synthesis, in primary cultures of cortical neurons. Autophagosomes can accumulate due to enhancement of autophagosome synthesis or inhibition of autophagic degradation, and we did not clarify whether TBT alters autophagic flux. Here, we investigated the mechanism by which TBT causes accumulation of autophagosomes in SH-SY5Y cells. TBT inhibited autophagy without affecting autophagosome-lysosome fusion before it caused cell death. TBT dramatically decreased the acidity of lysosomes without affecting lysosomal membrane integrity. TBT decreased the mature protein level of cathepsin B, and this may be related to the decrease in lysosomal acidity. These results suggest that TBT inhibits autophagic degradation by decreasing lysosomal acidity. Autophagy impairment may be involved in the mechanism underlying neuronal death and/or T-cell-dependent thymus atrophy induced by TBT.


Autophagy , Lysosomes/metabolism , Trialkyltin Compounds/pharmacology , Autophagosomes/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , Hydrolysis , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Lysosomes/drug effects , Microtubule-Associated Proteins/metabolism , Sequestosome-1 Protein/metabolism
4.
J Cell Biochem ; 123(2): 155-160, 2022 02.
Article En | MEDLINE | ID: mdl-34668225

Drug repurposing is an attractive option for identifying new treatment strategies, in particular in extraordinary situations of urgent need such as the current coronavirus disease 2019 (Covid-19) pandemic. Recently, the World Health Organization announced testing of three drugs as potential Covid-19 therapeutics that are known for their dampening effect on the immune system. Thus, the underlying concept of selecting these drugs is to temper the potentially life-threatening overshooting of the immune system reacting to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection. This viewpoint discusses the possibility that the impact of these and other drugs on autophagy contributes to their therapeutic effect by hampering the SARS-CoV-2 life cycle.


Antiviral Agents/pharmacology , Artesunate/pharmacology , Autophagy/drug effects , COVID-19 Drug Treatment , Drug Repositioning , Imatinib Mesylate/pharmacology , Infliximab/pharmacology , Pandemics , SARS-CoV-2/drug effects , Antidepressive Agents/pharmacology , Antiviral Agents/therapeutic use , Artesunate/therapeutic use , Chloroquine/pharmacology , Drug Development , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/physiology , Endoplasmic Reticulum/virology , Endosomes/drug effects , Endosomes/virology , Humans , Hydroxychloroquine/pharmacology , Imatinib Mesylate/therapeutic use , Infliximab/therapeutic use , Intracellular Membranes/drug effects , Intracellular Membranes/physiology , Intracellular Membranes/virology , Ivermectin/pharmacology , Macrolides/pharmacology , Middle East Respiratory Syndrome Coronavirus/drug effects , Niclosamide/pharmacology , Niclosamide/therapeutic use , RNA, Viral/metabolism , SARS-CoV-2/physiology , Virus Replication
5.
Int J Mol Sci ; 22(20)2021 Oct 15.
Article En | MEDLINE | ID: mdl-34681775

Botulinum neurotoxin serotype A (BoNT/A) is the most potent protein toxin to humans. BoNT/A light chain (LC/A) cleavage of the membrane-bound SNAP-25 has been well-characterized, but how LC/A traffics to the plasma membrane to target SNAP-25 is unknown. Of the eight BoNT/A subtypes (A1-A8), LC/A3 has a unique short duration of action and low potency that correlate to the intracellular steady state of LC/A, where LC/A1 is associated with the plasma membrane and LC/A3 is present in the cytosol. Steady-state and live imaging of LC/A3-A1 chimeras identified a two-step process where the LC/A N terminus bound intracellular vesicles, which facilitated an internal α-helical-rich domain to mediate LC/A plasma membrane association. The propensity of LC/A variants for membrane association correlated with enhanced BoNT/A potency. Understanding the basis for light chain intracellular localization provides insight to mechanisms underlying BoNT/A potency, which can be extended to applications as a human therapy.


Botulinum Toxins, Type A/metabolism , Cell Membrane/metabolism , Intracellular Membranes/metabolism , Animals , Botulinum Toxins, Type A/pharmacokinetics , Cell Membrane/drug effects , Female , Humans , Intracellular Membranes/drug effects , Mice , Mice, Inbred ICR , Protein Binding , Synaptosomal-Associated Protein 25/metabolism , Tumor Cells, Cultured
6.
Commun Biol ; 4(1): 828, 2021 07 01.
Article En | MEDLINE | ID: mdl-34211117

The heterotrimeric Sec61 complex is a major site for the biogenesis of transmembrane proteins (TMPs), accepting nascent TMP precursors that are targeted to the endoplasmic reticulum (ER) by the signal recognition particle (SRP). Unlike most single-spanning membrane proteins, the integration of type III TMPs is completely resistant to small molecule inhibitors of the Sec61 translocon. Using siRNA-mediated depletion of specific ER components, in combination with the potent Sec61 inhibitor ipomoeassin F (Ipom-F), we show that type III TMPs utilise a distinct pathway for membrane integration at the ER. Hence, following SRP-mediated delivery to the ER, type III TMPs can uniquely access the membrane insertase activity of the ER membrane complex (EMC) via a mechanism that is facilitated by the Sec61 translocon. This alternative EMC-mediated insertion pathway allows type III TMPs to bypass the Ipom-F-mediated blockade of membrane integration that is seen with obligate Sec61 clients.


Endoplasmic Reticulum/metabolism , Intracellular Membranes/metabolism , Membrane Proteins/metabolism , Protein Biosynthesis , SEC Translocation Channels/metabolism , Animals , Endoplasmic Reticulum/drug effects , Glycoconjugates/pharmacology , HeLa Cells , Humans , Immunoblotting , Intracellular Membranes/drug effects , Models, Biological , Protein Transport/drug effects , RNA Interference , SEC Translocation Channels/genetics , Signal Recognition Particle/metabolism
7.
Mol Nutr Food Res ; 65(15): e2100157, 2021 08.
Article En | MEDLINE | ID: mdl-34061446

SCOPE: The muscle loss during aging results from the blunt of protein synthesis and poses threat to the elderly health. This study aims to investigate whether betaine affects muscle loss by improving protein synthesis. METHODS AND RESULTS: Male C57BL/6J mice are raised from age 12 or 15 months. Mice are fed with AIN-93M diet without or with 2% w/v betaine in distilled water as control group or betaine intervention group (Bet), respectively. Betaine supplementation to mice demonstrates better body composition, grip strength, and motor function. Muscle morphology upregulates expression of myogenic regulate factors, and elevates myosin heavy chain and also improves in Bet group. Betaine promotes muscle protein synthesis via tethering mammalian target of rapamycin complex1 protein kinase (mTORC1) on the lysosomal membrane thereby activating mTORC1 signaling. All these effects aforementioned are time-dependent (p < 0.05). Ultrahigh-performance liquid chromatography results show that betaine increases S-adenosyl-l-methionine (SAM) via methionine cycle. SAM sensor-Samtor-overexpression in C2C12 cells could displace mTORC1 from lysosome thereby inhibiting the mTORC1 signaling. Addition of betaine attenuates this inhibition by increasing SAM level and then disrupting interaction of Samtor complex. CONCLUSIONS: These observations indicate that betaine could promisingly promote protein synthesis to delay age-related muscle loss.


Betaine/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/metabolism , Methyltransferases/antagonists & inhibitors , Muscle, Skeletal/drug effects , Muscle, Skeletal/physiopathology , S-Adenosylmethionine/metabolism , Aging/drug effects , Aging/pathology , Animals , Gene Expression Regulation/drug effects , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Male , Methionine/metabolism , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Protein Biosynthesis/drug effects , Signal Transduction/drug effects
8.
Adv Drug Deliv Rev ; 175: 113820, 2021 08.
Article En | MEDLINE | ID: mdl-34087327

The interactions between inorganic-based nanomaterials (NMs) and biological membranes are among the most important phenomena for developing NM-based therapeutics and resolving nanotoxicology. Herein, we introduce the structural and functional effects of inorganic-based NMs on biological membranes, mainly the plasma membrane and the endomembrane system, with an emphasis on the interface, which involves highly complex networks between NMs and biomolecules (such as membrane proteins and lipids). Significant efforts have been devoted to categorizing and analyzing the interaction mechanisms in terms of the physicochemical characteristics and biological effects of NMs, which can directly or indirectly influence the effects of NMs on membranes. Importantly, we summarize that the biological membranes act as platforms and thereby mediate NMs-immune system contacts. In this overview, the existing challenges and potential applications in the areas are addressed. A strong understanding of the discussed concepts will promote therapeutic NM designs for drug delivery systems by leveraging the NMs-membrane interactions and their functions.


Membranes/drug effects , Nanostructures , Animals , Cell Membrane/drug effects , Cell Membrane/metabolism , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Membrane Lipids/metabolism , Membrane Proteins/metabolism , Membranes/metabolism
9.
Nat Commun ; 12(1): 2616, 2021 05 10.
Article En | MEDLINE | ID: mdl-33972548

FUN14 domain-containing protein 1 (FUNDC1) is an integral mitochondrial outer-membrane protein, and mediates the formation of mitochondria-associated endoplasmic reticulum membranes (MAMs). This study aims to determine the contributions of FUNDC1-mediated MAMs to angiogenesis in vitro and in vivo. In cultured endothelial cells, VEGF significantly increases the formation of MAMs and MAM-related proteins, including FUNDC1. Endothelial cell-specific deletion of FUNDC1, which disrupts MAM formation in endothelial cells, lowers VEGFR2 expression and reduces tube formation, spheroid-sprouting, and functional blood vessel formation in vitro and in vivo. Conversely, increased MAM formation using MAM linkers mimics the effects of VEGF and promotes endothelial angiogenesis. Mechanistically, increased MAMs formation led to increased levels of Ca2+ in cytosol, promoted the phosphorylation of serum response factor (SRF) and enhanced the binding of SRF to VEGFR2 promoter, resulting in increased VEGFR2 production, with consequent angiogenesis. Moreover, blocking FUNDC1-related MAM formation with a cell-penetrating inhibitory peptide significantly suppresses the expressions of downstream angiogenic genes and inhibits tumor angiogenesis. We conclude that decreased MAMs formation by silencing FUNDC1 can inhibit angiogenesis by decreasing VEGFR2 expression, and targeting FUNDC1-dependent MAMs might be a promising approach for treating human disorders characterized by defective angiogenesis.


Endoplasmic Reticulum/metabolism , Endothelial Cells/metabolism , Intracellular Membranes/metabolism , Membrane Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Physiologic/genetics , Animals , Calcium/metabolism , Cytoplasm/metabolism , Endoplasmic Reticulum/ultrastructure , Gene Silencing , Human Umbilical Vein Endothelial Cells , Humans , Intracellular Membranes/drug effects , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Mitochondria/ultrastructure , Mitochondrial Proteins/genetics , Neovascularization, Physiologic/drug effects , Phosphorylation , RNA, Small Interfering , Retina/metabolism , Serum Response Factor/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vascular Endothelial Growth Factor Receptor-2/pharmacology , Xenograft Model Antitumor Assays
10.
Sci Rep ; 11(1): 10609, 2021 05 19.
Article En | MEDLINE | ID: mdl-34011952

In cancer cells only, TLR3 acquires death receptor properties by efficiently triggering the extrinsic pathway of apoptosis with Caspase-8 as apical protease. Here, we demonstrate that in the absence of Caspase-8, activation of TLR3 can trigger a form of programmed cell death, which is distinct from classical apoptosis. When TLR3 was activated in the Caspase-8 negative neuroblastoma cell line SH-SY5Y, cell death was accompanied by lysosomal permeabilization. Despite caspases being activated, lysosomal permeabilization as well as cell death were not affected by blocking caspase-activity, positioning lysosomal membrane permeabilization (LMP) upstream of caspase activation. Taken together, our data suggest that LMP with its deadly consequences represents a "default" death mechanism in cancer cells, when Caspase-8 is absent and apoptosis cannot be induced.


Apoptosis , Caspase 8/metabolism , Neuroblastoma/metabolism , Neuroblastoma/pathology , Toll-Like Receptor 3/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Enzyme Activation/drug effects , Humans , Interferon Type I/pharmacology , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Necroptosis/drug effects , Permeability/drug effects , Poly I-C/pharmacology
11.
Article En | MEDLINE | ID: mdl-33945875

The biogenesis of peroxisomes in relation to the trafficking of proteins to peroxisomes has been extensively examined. However, the supply of phospholipids, which is needed to generate peroxisomal membranes in mammals, remains unclear. Therefore, we herein investigated metabolic alterations induced by clofibric acid, a peroxisome proliferator, in the synthesis of phospholipids, particularly phosphatidylethanolamine (PE) molecular species, and their relationship with the biogenesis of peroxisomal membranes. The subcutaneous administration of clofibric acid to rats at a relatively low dose (130 mg/kg) once a day time-dependently and gradually increased the integrated perimeter of peroxisomes per 100 µm2 hepatocyte cytoplasm (PA). A strong correlation was observed between the content (µmol/mg DNA) of PE containing arachidonic acid (20:4) and PA (r2 = 0.9168). Moreover, the content of PE containing octadecenoic acid (18:1) positively correlated with PA (r2 = 0.8094). The treatment with clofibric acid markedly accelerated the formation of 16:0-20:4 PE by increasing the production of 20:4 and the activity of acyl chain remodeling of pre-existing PE molecular species. Increases in the acyl chain remodeling of PE by clofibric acid were mainly linked to the up-regulated expression of the Lpcat3 gene. On the other hand, clofibric acid markedly increased the formation of palmitic acid (16:0)-18:1 PE through de novo synthesis. These results suggest that the enhanced formation of particular PE molecular species is related to increases in the mass of peroxisomal membranes in peroxisome proliferation in the liver.


Arachidonic Acid/biosynthesis , Arachidonic Acid/chemistry , Clofibric Acid/pharmacology , Intracellular Membranes/drug effects , Liver/cytology , Peroxisomes/drug effects , Phosphatidylethanolamines/chemistry , Animals , Intracellular Membranes/metabolism , Male , Peroxisomes/metabolism , Rats , Rats, Wistar
12.
Int J Mol Sci ; 22(5)2021 Feb 25.
Article En | MEDLINE | ID: mdl-33668885

Lysosomal membrane permeabilization (LMP) has been proposed to precede nanoparticle-induced macrophage injury and NLRP3 inflammasome activation; however, the underlying mechanism(s) of LMP is unknown. We propose that nanoparticle-induced lysosomal hyperpolarization triggers LMP. In this study, a rapid non-invasive method was used to measure changes in lysosomal membrane potential of murine alveolar macrophages (AM) in response to a series of nanoparticles (ZnO, TiO2, and CeO2). Crystalline SiO2 (micron-sized) was used as a positive control. Changes in cytosolic potassium were measured using Asante potassium green 2. The results demonstrated that ZnO or SiO2 hyperpolarized the lysosomal membrane and decreased cytosolic potassium, suggesting increased lysosome permeability to potassium. Time-course experiments revealed that lysosomal hyperpolarization was an early event leading to LMP, NLRP3 activation, and cell death. In contrast, TiO2- or valinomycin-treated AM did not cause LMP unless high doses led to lysosomal hyperpolarization. Neither lysosomal hyperpolarization nor LMP was observed in CeO2-treated AM. These results suggested that a threshold of lysosomal membrane potential must be exceeded to cause LMP. Furthermore, inhibition of lysosomal hyperpolarization with Bafilomycin A1 blocked LMP and NLRP3 activation, suggesting a causal relation between lysosomal hyperpolarization and LMP.


Intracellular Membranes/physiology , Lysosomes/metabolism , Membrane Potentials/physiology , Nanoparticles/toxicity , Animals , Cell Death/drug effects , Cytosol/metabolism , Female , Hydrogen-Ion Concentration , Inflammasomes/metabolism , Intracellular Membranes/drug effects , Lysosomes/drug effects , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/metabolism , Male , Membrane Potentials/drug effects , Mice, Inbred C57BL , Models, Biological , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Permeability , Potassium/metabolism , Reproducibility of Results , Silicon Dioxide/toxicity
13.
Int J Mol Sci ; 22(2)2021 Jan 11.
Article En | MEDLINE | ID: mdl-33440911

Changes in zinc content and dysregulated zinc homeostatic mechanisms have been recognized in several solid malignancies such as prostate cancer, breast cancer, or pancreatic cancer. Moreover, it has been shown that zinc serum and/or tissue levels are altered in melanoma with varying effects on melanoma development and biology. This study was conducted to explore the effects of acute increases of intracellular zinc in a set of melanoma tissue explants obtained from clinical samples. Measurements of their zinc content showed an extant heterogeneity in total and free intracellular zinc pools associated with varying biological behavior of individual cells, e.g., autophagy levels and propensity to cell death. Use of zinc pyrithione elevated intracellular zinc in a short time frame which resulted in marked changes in mitochondrial activity and lysosomes. These alterations were accompanied by significantly enhanced autophagy flux and subsequent cell demise in the absence of typical apoptotic cell death markers. The present results show for the first time that acutely increased intracellular zinc in melanoma cells specifically enhances their autophagic activity via mitochondria and lysosomes which leads to autophagic cell death. While biologically relevant, this discovery may contribute to our understanding and exploration of zinc in relation to autophagy as a means of controlling melanoma growth and survival.


Autophagy , Lysosomes/metabolism , Melanoma/metabolism , Mitochondria/metabolism , Zinc/metabolism , Apoptosis , Autophagy/drug effects , Cell Death , Cell Line, Tumor , Cell Proliferation , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Intracellular Space/metabolism , Melanocytes/drug effects , Melanocytes/metabolism , Mitochondria/drug effects , Time Factors , Zinc/pharmacology
14.
Cell Biol Int ; 45(3): 633-641, 2021 Mar.
Article En | MEDLINE | ID: mdl-33247607

Carbon tetrachloride (CCl4 ) causes hepatotoxicity in mammals, with its hepatocytic metabolism producing radicals that attack the intracellular membrane system and destabilize intracellular vesicle transport. Inhibition of intracellular transport causes lipid droplet retention and abnormal protein distribution. The intracellular transport of synthesized lipids and proteins from the endoplasmic reticulum (ER) to the Golgi apparatus is performed by coat complex II (COPII) vesicle transport, but how CCl4 inhibits COPII vesicle transport has not been elucidated. COPII vesicle formation on the ER membrane is initiated by the recruitment of Sar1 protein from the cytoplasm to the ER membrane, followed by that of the COPII coat constituent proteins (Sec23, Sec24, Sec13, and Sec31). In this study, we evaluated the effect of CCl4 on COPII vesicle formation using the RLC-16 rat hepatocyte cell line. Our results showed that CCl4 suppressed ER-Golgi transport in RLC-16 cells. Using a reconstituted system of rat liver tissue-derived cytoplasm and RLC-16 cell-derived ER membranes, CCl4 treatment inhibited the recruitment of Sar1 and Sec13 from the cytosolic fraction to ER membranes. CCl4 -induced changes in the ER membrane accordingly inhibited the accumulation of COPII vesicle-coated constituent proteins on the ER membrane, as well as the formation of COPII vesicles, which suppressed lipid and protein transport between the ER and Golgi apparatus. Our data suggest that CCl4 inhibits ER-Golgi intracellular transport by inhibiting COPII vesicle formation on the ER membrane in hepatocytes.


COP-Coated Vesicles/metabolism , Carbon Tetrachloride/toxicity , Endoplasmic Reticulum/metabolism , Golgi Apparatus/metabolism , Hepatocytes/metabolism , Intracellular Membranes/metabolism , Animals , COP-Coated Vesicles/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cytosol/drug effects , Cytosol/metabolism , Endoplasmic Reticulum/drug effects , Golgi Apparatus/drug effects , Hepatocytes/drug effects , Intracellular Membranes/drug effects , Male , Protein Transport/drug effects , Rats, Sprague-Dawley
15.
Oxid Med Cell Longev ; 2020: 6569728, 2020.
Article En | MEDLINE | ID: mdl-33149811

Mitochondrial oxidative stress and dysfunction play an important role of atrial remodeling and atrial fibrillation (AF) in diabetes mellitus. Endoplasmic reticulum (ER) stress has been linked to both physiological and pathological states including diabetes. The aim of this project is to explore the roles of ER stress in hyperglycemia-induced mitochondrial dysfunction and cell death of atrial cardiomyocytes. High glucose upregulated ER stress, mitochondrial oxidative stress, and mitochondria-associated ER membrane (MAM)- enriched proteins (such as glucose-regulated protein 75 (GRP75) and mitofusin-2 (Mfn2)) of primary cardiomyocytes in vitro. Sodium phenylbutyrate (4-PBA) prevented the above changes. Silencing of Mfn2 in HL-1 cells decreased the Ca2+ transfer from ER to mitochondria under ER stress conditions, which were induced by the ER stress agonist, tunicamycin (TM). Electron microscopy data suggested that Mfn2 siRNA significantly disrupted ER-mitochondria tethering in ER stress-injured HL-1 cells. Mfn2 silencing attenuated mitochondrial oxidative stress and Ca2+ overload, increased mitochondrial membrane potential and mitochondrial oxygen consumption, and protected cells from TM-induced apoptosis. In summary, Mfn2 plays an important role in high glucose-induced ER stress in atrial cardiomyocytes, and Mfn2 silencing prevents mitochondrial Ca2+ overload-mediated mitochondrial dysfunction, thereby decreasing ER stress-mediated cardiomyocyte cell death.


Down-Regulation , Endoplasmic Reticulum Stress , GTP Phosphohydrolases/genetics , Heart Atria/pathology , Hyperglycemia/pathology , Mitochondria, Heart/metabolism , Mitochondrial Proteins/genetics , Myocytes, Cardiac/pathology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers/metabolism , Calcium/metabolism , Cell Death/drug effects , Cell Line , Down-Regulation/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/drug effects , GTP Phosphohydrolases/metabolism , Gene Silencing , Glucose/toxicity , Intracellular Membranes/drug effects , Intracellular Membranes/metabolism , Mitochondria, Heart/ultrastructure , Mitochondrial Proteins/metabolism , Models, Biological , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Oxidative Stress/drug effects , RNA, Small Interfering/metabolism , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Tunicamycin/pharmacology , Up-Regulation/drug effects
16.
Int J Oncol ; 57(6): 1280-1292, 2020 12.
Article En | MEDLINE | ID: mdl-33173988

The proton pump inhibitor lansoprazole (LPZ) inhibits the growth of several cancer cell lines, including A549 and CAL 27. We previously reported that macrolide antibiotics such as azithromycin (AZM) and clarithromycin (CAM) potently inhibit autophagic flux and that combining AZM or CAM with the epidermal growth factor receptor inhibitors enhanced their antitumor effect against various cancer cells. In the present study, we conducted the combination treatment with LPZ and macrolide antibiotics against A549 and CAL 27 cells and evaluated cytotoxicity and morphological changes using cell proliferation and viability assays, flow cytometric analysis, immunoblotting, and morphological assessment. Combination therapy with LPZ and AZM greatly enhanced LPZ­induced cell death, whereas treatment with AZM alone exhibited negligible cytotoxicity. The observed cytotoxic effect was not mediated through apoptosis or necroptosis. Transmission electron microscopy of A549 cells treated with the LPZ + AZM combination revealed morphological changes associated with necrosis and accumulated autolysosomes with undigested contents. Furthermore, the A549 cell line with ATG5 knockout exhibited complete inhibition of autophagosome formation, which did not affect LPZ + AZM treatment­induced cytotoxicity, thus excluding the involvement of autophagy­dependent cell death in LPZ + AZM treatment­induced cell death. A549 cells treated with LPZ + AZM combination therapy retained the endosomal Alexa­dextran for extended duration as compared to untreated control cells, thus indicating impairment of lysosomal digestion. Notably, lysosomal galectin­3 puncta expression induced due to lysosomal membrane permeabilization was increased in cells treated with LPZ + AZM combination as compared to the treatment by either agent alone. Collectively, the present results revealed AZM­induced autolysosome accumulation, potentiated LPZ­mediated necrosis, and lysosomal membrane permeabilization, thus suggesting the potential clinical application of LPZ + AZM combination therapy for cancer treatment.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Azithromycin/pharmacology , Lansoprazole/pharmacology , Lysosomes/drug effects , Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/metabolism , Azithromycin/therapeutic use , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Drug Synergism , Gene Knockout Techniques , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/ultrastructure , Lansoprazole/therapeutic use , Lysosomes/pathology , Lysosomes/ultrastructure , Microscopy, Electron, Transmission , Neoplasms/pathology , Permeability/drug effects
17.
Int J Mol Sci ; 21(9)2020 Apr 29.
Article En | MEDLINE | ID: mdl-32365555

O-methyl-serine dodecylamine hydrochloride (MSDH) is a detergent that accumulates selectively in lysosomes, a so-called lysosomotropic detergent, with unexpected chemical properties. At physiological pH, it spontaneously forms vesicles, which disassemble into small aggregates (probably micelles) below pH 6.4. In this study, we characterize the interaction between MSDH and liposomes at different pH and correlate the findings to toxicity in human fibroblasts. We find that the effect of MSDH on lipid membranes is highly pH-dependent. At neutral pH, the partitioning of MSDH into the liposome membrane is immediate and causes the leakage of small fluorophores, unless the ratio between MSDH and lipids is kept low. At pH 5, the partitioning of MSDH into the membrane is kinetically impeded since MSDH is charged and a high ratio between MSDH and the lipids is required to permeabilize the membrane. When transferred to cell culture conditions, the ratio between MSDH and plasma membrane lipids must therefore be low, at physiological pH, to maintain plasma membrane integrity. Transmission electron microscopy suggests that MSDH vesicles are taken up by endocytosis. As the pH of the endosomal compartment progressively drops, MSDH vesicles disassemble, leading to a high concentration of increasingly charged MSDH in small aggregates inside the lysosomes. At sufficiently high MSDH concentrations, the lysosome is permeabilized, the proteolytic content released to the cytosol and apoptotic cell death is induced.


Amides/chemistry , Amides/pharmacology , Detergents/chemistry , Detergents/pharmacology , Lipid Bilayers/adverse effects , Lysosomes/drug effects , Serine/analogs & derivatives , Cell Membrane Permeability/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Endocytosis/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Humans , Hydrogen-Ion Concentration , Intracellular Membranes/drug effects , Intracellular Membranes/ultrastructure , Lipid Bilayers/chemistry , Lipids/chemistry , Serine/chemistry , Serine/pharmacology
18.
Ecotoxicol Environ Saf ; 197: 110618, 2020 Jul 01.
Article En | MEDLINE | ID: mdl-32302861

Benzotriazole (BTR) is a common corrosion inhibitor used to protect copper (Cu) and Cu alloys. To reveal the combined subacute toxicity of BTR and Cu at environmental levels on terrestrial animals, the activity of antioxidative enzymes and the glutathione levels in earthworms (Eisenia fetida) of the single or co-exposure treatments were determined. The activity of both antioxidant enzymes and non-enzymatic antioxidants was affected by BTR in earthworms. Moreover, the analyses of lysosomal neutral red retention time and total antioxidant capacity indicated a detoxification effect of BTR on Cu-induced impairments of the antioxidant defense capacity in earthworms. The apoptotic rate of coelomocytes in earthworms of the co-exposure treatment was lower than that in earthworms treated with Cu only, indicating that BTR alleviates Cu mediated lysosomal membrane damage and antioxidant defense system responses in earthworms.


Copper/toxicity , Oligochaeta/drug effects , Soil Pollutants/toxicity , Triazoles/toxicity , Animals , Antioxidants/metabolism , Apoptosis , Drug Interactions , Glutathione/metabolism , Intracellular Membranes/drug effects , Lysosomes/drug effects , Oligochaeta/enzymology , Oligochaeta/metabolism
19.
Autophagy ; 16(6): 1044-1060, 2020 06.
Article En | MEDLINE | ID: mdl-31517566

Macroautophagy/autophagy can enable cancer cells to withstand cellular stress and maintain bioenergetic homeostasis by sequestering cellular components into newly formed double-membrane vesicles destined for lysosomal degradation, potentially affecting the efficacy of anti-cancer treatments. Using 13C-labeled choline and 13C-magnetic resonance spectroscopy and western blotting, we show increased de novo choline phospholipid (ChoPL) production and activation of PCYT1A (phosphate cytidylyltransferase 1, choline, alpha), the rate-limiting enzyme of phosphatidylcholine (PtdCho) synthesis, during autophagy. We also discovered that the loss of PCYT1A activity results in compromised autophagosome formation and maintenance in autophagic cells. Direct tracing of ChoPLs with fluorescence and immunogold labeling imaging revealed the incorporation of newly synthesized ChoPLs into autophagosomal membranes, endoplasmic reticulum (ER) and mitochondria during anticancer drug-induced autophagy. Significant increase in the colocalization of fluorescence signals from the newly synthesized ChoPLs and mCherry-MAP1LC3/LC3 (microtubule-associated protein 1 light chain 3) was also found on autophagosomes accumulating in cells treated with autophagy-modulating compounds. Interestingly, cells undergoing active autophagy had an altered ChoPL profile, with longer and more unsaturated fatty acid/alcohol chains detected. Our data suggest that de novo synthesis may be required to increase autophagosomal ChoPL content and alter its composition, together with replacing phospholipids consumed from other organelles during autophagosome formation and turnover. This addiction to de novo ChoPL synthesis and the critical role of PCYT1A may lead to development of agents targeting autophagy-induced drug resistance. In addition, fluorescence imaging of choline phospholipids could provide a useful way to visualize autophagosomes in cells and tissues. ABBREVIATIONS: AKT: AKT serine/threonine kinase; BAX: BCL2 associated X, apoptosis regulator; BECN1: beclin 1; ChoPL: choline phospholipid; CHKA: choline kinase alpha; CHPT1: choline phosphotransferase 1; CTCF: corrected total cell fluorescence; CTP: cytidine-5'-triphosphate; DCA: dichloroacetate; DMEM: dulbeccos modified Eagles medium; DMSO: dimethyl sulfoxide; EDTA: ethylenediaminetetraacetic acid; ER: endoplasmic reticulum; GDPD5: glycerophosphodiester phosphodiesterase domain containing 5; GFP: green fluorescent protein; GPC: glycerophosphorylcholine; HBSS: hanks balances salt solution; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; LPCAT1: lysophosphatidylcholine acyltransferase 1; LysoPtdCho: lysophosphatidylcholine; MRS: magnetic resonance spectroscopy; MTORC1: mechanistic target of rapamycin kinase complex 1; PCho: phosphocholine; PCYT: choline phosphate cytidylyltransferase; PLA2: phospholipase A2; PLB: phospholipase B; PLC: phospholipase C; PLD: phospholipase D; PCYT1A: phosphate cytidylyltransferase 1, choline, alpha; PI3K: phosphoinositide-3-kinase; pMAFs: pancreatic mouse adult fibroblasts; PNPLA6: patatin like phospholipase domain containing 6; Pro-Cho: propargylcholine; Pro-ChoPLs: propargylcholine phospholipids; PtdCho: phosphatidylcholine; PtdEth: phosphatidylethanolamine; PtdIns3P: phosphatidylinositol-3-phosphate; RPS6: ribosomal protein S6; SCD: stearoyl-CoA desaturase; SEM: standard error of the mean; SM: sphingomyelin; SMPD1/SMase: sphingomyelin phosphodiesterase 1, acid lysosomal; SGMS: sphingomyelin synthase; WT: wild-type.


Antineoplastic Agents/pharmacology , Autophagosomes/enzymology , Autophagosomes/metabolism , Choline-Phosphate Cytidylyltransferase/metabolism , Furans/pharmacology , Macroautophagy , Phosphatidylcholines/biosynthesis , Pyridines/pharmacology , Pyrimidines/pharmacology , Animals , Autophagosomes/drug effects , Autophagosomes/ultrastructure , CHO Cells , Cell Line, Tumor , Choline/metabolism , Choline-Phosphate Cytidylyltransferase/genetics , Cricetulus , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Knockout Techniques , Humans , Intracellular Membranes/drug effects , Intracellular Membranes/enzymology , Intracellular Membranes/metabolism , Macroautophagy/drug effects , Magnetic Resonance Spectroscopy , Mass Spectrometry , Metabolomics , Mice , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/metabolism , Phosphoinositide-3 Kinase Inhibitors , Vacuoles/drug effects , Vacuoles/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
20.
Mycotoxin Res ; 36(1): 23-30, 2020 Feb.
Article En | MEDLINE | ID: mdl-31264166

The mycotoxin enniatin B1 (ENN B1) is widely present in grain-based feed and food products. In the present study, we have investigated how this lipophilic and ionophoric molecule can affect the lysosomal stability and chaperone-mediated autophagy (CMA) in wild-type (WT) and in lysosome-associated membrane proteins (LAMP)-1/2 double-deficient (DD) mouse embryonic fibroblasts (MEF). The cell viability and lysosomal pH were assessed using the Neutral Red (NR) cytotoxicity assay and the LysoSensor® Yellow/Blue DND-160, respectively. Changes in the expression of the CMA-related components LAMP-2 and the chaperones heat shock cognate (hsc) 70 and heat shock protein (hsp) 90 were determined in cytosolic extracts by immunoblotting. In the NR assay, LAMP-1/2 DD MEF cells were significantly less sensitive to ENN B1 than WT MEF cells after 24 h exposure to ENN B1 at levels of 2.5-10 µmol/L. Exposure to ENN B1 at concentrations below the half maximal effective concentration (EC50) (1.5-1.7 µmol/L) increased the lysosomal pH in WT MEF, but not in LAMP-1/2 DD cells, suggesting that lysosomal LAMP-2 is an early target of ENN B1-induced lysosomal alkalization and cytotoxicity in MEF cells. Additionally, cytosolic hsp90 and LAMP-2 levels slightly increased after exposure for 4 h, indicating lysosomal membrane permeabilization (LMP). In summary, it appeared that ENN B1 can destabilize the LAMP-2 complex in the lysosomal membrane at concentrations close to the EC50, resulting in the alkalinization of lysosomes, partial LMP, and thereby leakage of CMA-associated components into the cytosol.


Depsipeptides/toxicity , Intracellular Membranes/drug effects , Lysosomes/pathology , Mycotoxins/toxicity , Permeability/drug effects , Animals , Chaperone-Mediated Autophagy/drug effects , Fibroblasts , Gene Deletion , HSC70 Heat-Shock Proteins/drug effects , HSC70 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/drug effects , HSP90 Heat-Shock Proteins/metabolism , Hydrogen-Ion Concentration/drug effects , Lysosomal-Associated Membrane Protein 2/drug effects , Lysosomal-Associated Membrane Protein 2/genetics , Lysosomal-Associated Membrane Protein 2/metabolism , Mice , Molecular Chaperones/drug effects , Molecular Chaperones/metabolism
...