Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.888
Filter
1.
Exp Neurol ; 367: 114470, 2023 09.
Article in English | MEDLINE | ID: mdl-37327964

ABSTRACT

Clinical evidence indicates that major depression is a common comorbidity of chronic pain, including neuropathic pain; however, the cellular basis for chronic pain-mediated major depression remains unclear. Mitochondrial dysfunction induces neuroinflammation and has been implicated in various neurological diseases, including depression. Nevertheless, the relationship between mitochondrial dysfunction and anxiodepressive-like behaviors in the neuropathic pain state remains unclear. The current study examined whether hippocampal mitochondrial dysfunction and downstream neuroinflammation are involved in anxiodepressive-like behaviors in mice with neuropathic pain, which was induced by partial sciatic nerve ligation (PSNL). At 8 weeks after surgery, there was decreased levels of mitochondrial damage-associated molecular patterns, such as cytochrome c and mitochondrial transcription factor A, and increased level of cytosolic mitochondrial DNA in the contralateral hippocampus, suggesting the development of mitochondrial dysfunction. Type I interferon (IFN) mRNA expression in the hippocampus was also increased at 8 weeks after PSNL surgery. The restoration of mitochondrial function by curcumin blocked the increased cytosolic mitochondrial DNA and type I IFN expression in PSNL mice and improved anxiodepressive-like behaviors. Blockade of type I IFN signaling by anti-IFN alpha/beta receptor 1 antibody also improved anxiodepressive-like behaviors in PSNL mice. Together, these findings suggest that neuropathic pain induces hippocampal mitochondrial dysfunction followed by neuroinflammation, which may contribute to anxiodepressive-behaviors in the neuropathic pain state. Improving mitochondrial dysfunction and inhibiting type I IFN signaling in the hippocampus might be a novel approach to reducing comorbidities associated with neuropathic pain, such as depression and anxiety.


Subject(s)
Anxiety , Depression , Interferon Type I , Mitochondria , Neuralgia , Animals , Male , Mice , Anxiety/complications , Anxiety/drug therapy , Anxiety/metabolism , Chronic Pain/complications , Chronic Pain/metabolism , Chronic Pain/pathology , Chronic Pain/psychology , Curcumin/pharmacology , Curcumin/therapeutic use , Cytosol/drug effects , Cytosol/metabolism , Depression/complications , Depression/drug therapy , Depression/metabolism , DNA, Mitochondrial/metabolism , Frontal Lobe/metabolism , Frontal Lobe/pathology , Hippocampus/drug effects , Hippocampus/immunology , Hippocampus/metabolism , Hippocampus/pathology , Interferon Type I/antagonists & inhibitors , Interferon Type I/genetics , Interferon Type I/metabolism , Microglia/drug effects , Microglia/immunology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Neuralgia/complications , Neuralgia/metabolism , Neuralgia/pathology , Neuralgia/psychology , Neuroinflammatory Diseases/complications , Sciatic Nerve/surgery
2.
Mol Pharmacol ; 101(3): 132-143, 2022 03.
Article in English | MEDLINE | ID: mdl-34969832

ABSTRACT

Calcium- and voltage-gated K+ channels of large conductance (BKs) are expressed in the cell membranes of all excitable tissues. Currents mediated by BK channel-forming slo1 homotetramers are consistently inhibited by increases in membrane cholesterol (CLR). The molecular mechanisms leading to this CLR action, however, remain unknown. Slo1 channels are activated by increases in calcium (Ca2+) nearby Ca2+-recognition sites in the slo1 cytosolic tail: one high-affinity and one low-affinity site locate to the regulator of conductance for K+ (RCK) 1 domain, whereas another high-affinity site locates within the RCK2 domain. Here, we first evaluated the crosstalking between Ca2+ and CLR on the function of slo1 (cbv1 isoform) channels reconstituted into planar lipid bilayers. CLR robustly reduced channel open probability while barely decreasing unitary current amplitude, with CLR maximal effects being observed at 10-30 µM internal Ca2+ CLR actions were not only modulated by internal Ca2+ levels but also disappeared in absence of this divalent. Moreover, in absence of Ca2+, BK channel-activating concentrations of magnesium (10 mM) did not support CLR action. Next, we evaluated CLR actions on channels where the different Ca2+-sensing sites present in the slo1 cytosolic domain became nonfunctional via mutagenesis. CLR still reduced the activity of low-affinity Ca2+ (RCK1:E379A, E404A) mutants. In contrast, CLR became inefficacious when both high-affinity Ca2+ sites were mutated (RCK1:D367A,D372A and RCK2:D899N,D900N,D901N,D902N,D903N), yet still was able to decrease the activity of each high-affinity site mutant. Therefore, BK channel inhibition by CLR selectively requires optimal levels of Ca2+ being recognized by either of the slo1 high-affinity Ca2+-sensing sites. SIGNIFICANCE STATEMENT: Results reveal that inhibition of calcium/voltage-gated K+ channel of large conductance (BK) (slo1) channels by membrane cholesterol requires a physiologically range of internal calcium (Ca2+) and is selectively linked to the two high-affinity Ca2+-sensing sites located in the cytosolic tail domain, which underscores that Ca2+ and cholesterol actions are allosterically coupled to the channel gate. Cholesterol modification of BK channel activity likely contributes to disruption of normal physiology by common health conditions that are triggered by disruption of cholesterol homeostasis.


Subject(s)
Calcium/metabolism , Cholesterol/metabolism , Cytosol/metabolism , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/antagonists & inhibitors , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Animals , Calcium Channel Blockers/pharmacology , Cytosol/drug effects , HEK293 Cells , Humans , Large-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Protein Structure, Secondary , Rats
3.
Int J Mol Sci ; 22(23)2021 Nov 23.
Article in English | MEDLINE | ID: mdl-34884427

ABSTRACT

Little is known about the effect of lead on the activity of the vacuolar K+ channels. Here, the patch-clamp technique was used to compare the impact of lead (PbCl2) on the slow-activating (SV) and fast-activating (FV) vacuolar channels. It was revealed that, under symmetrical 100-mM K+, the macroscopic currents of the SV channels exhibited a typical slow activation and a strong outward rectification of the steady-state currents, while the macroscopic currents of the FV channels displayed instantaneous currents, which, at the positive potentials, were about three-fold greater compared to the one at the negative potentials. When PbCl2 was added to the bath solution at a final concentration of 100 µM, it decreased the macroscopic outward currents of both channels but did not change the inward currents. The single-channel recordings demonstrated that cytosolic lead causes this macroscopic effect by a decrease of the single-channel conductance and decreases the channel open probability. We propose that cytosolic lead reduces the current flowing through the SV and FV channels, which causes a decrease of the K+ fluxes from the cytosol to the vacuole. This finding may, at least in part, explain the mechanism by which cytosolic Pb2+ reduces the growth of plant cells.


Subject(s)
Beta vulgaris/growth & development , Lead/pharmacology , Potassium Channels/metabolism , Vacuoles/metabolism , Beta vulgaris/drug effects , Beta vulgaris/metabolism , Cytosol/drug effects , Cytosol/metabolism , Gene Expression Regulation, Plant/drug effects , Patch-Clamp Techniques , Plant Proteins/drug effects , Plant Proteins/metabolism , Potassium Channels/drug effects , Vacuoles/drug effects
4.
Cells ; 10(11)2021 10 30.
Article in English | MEDLINE | ID: mdl-34831186

ABSTRACT

Macrophage stimulation by pathogen-associated molecular patterns (PAMPs) like lipopolysaccharide (LPS) or lipoteichoic acid (LTA) drives a proinflammatory phenotype and induces a metabolic reprogramming to sustain the cell's function. Nevertheless, the relationship between metabolic shifts and gene expression remains poorly explored. In this context, the metabolic enzyme ATP citrate lyase (ACLY), the producer of citrate-derived acetyl-coenzyme A (CoA), plays a critical role in supporting a proinflammatory response. Through immunocytochemistry and cytosol-nucleus fractionation, we found a short-term ACLY nuclear translocation. Protein immunoprecipitation unveiled the role of nuclear ACLY in NF-κB acetylation and in turn its full activation in human PBMC-derived macrophages. Notably, sepsis in the early hyperinflammatory phase triggers ACLY-mediated NF-κB acetylation. The ACLY/NF-κB axis increases the expression levels of proinflammatory genes, including SLC25A1-which encodes the mitochondrial citrate carrier-and ACLY, thus promoting the existence of a proinflammatory loop involving SLC25A1 and ACLY genes.


Subject(s)
ATP Citrate (pro-S)-Lyase/metabolism , Cell Nucleus/metabolism , Gene Expression Regulation , Inflammation/genetics , Macrophages/metabolism , NF-kappa B/metabolism , ATP Citrate (pro-S)-Lyase/genetics , Acetylation/drug effects , Aged , Cell Nucleus/drug effects , Cytosol/drug effects , Cytosol/metabolism , Female , Gene Expression Regulation/drug effects , Humans , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Male , Middle Aged , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Organic Anion Transporters/genetics , Organic Anion Transporters/metabolism , Sepsis/genetics , Teichoic Acids/pharmacology , Up-Regulation/genetics , Young Adult
5.
Cells ; 10(11)2021 11 05.
Article in English | MEDLINE | ID: mdl-34831263

ABSTRACT

Both, the decreased L-type Ca2+ current (ICa,L) density and increased spontaneous Ca2+ release from the sarcoplasmic reticulum (SR), have been associated with atrial fibrillation (AF). In this study, we tested the hypothesis that remodeling of 3',5'-cyclic adenosine monophosphate (cAMP)-dependent protein kinase A (PKA) signaling is linked to these compartment-specific changes (up- or down-regulation) in Ca2+-handling. Perforated patch-clamp experiments were performed in atrial myocytes from 53 patients with AF and 104 patients in sinus rhythm (Ctl). A significantly higher frequency of transient inward currents (ITI) activated by spontaneous Ca2+ release was confirmed in myocytes from AF patients. Next, inhibition of PKA by H-89 promoted a stronger effect on the ITI frequency in these myocytes compared to myocytes from Ctl patients (7.6-fold vs. 2.5-fold reduction), while the ß-agonist isoproterenol (ISO) caused a greater increase in Ctl patients (5.5-fold vs. 2.1-fold). ICa,L density was larger in myocytes from Ctl patients at baseline (p < 0.05). However, the effect of ISO on ICa,L density was only slightly stronger in AF than in Ctl myocytes (3.6-fold vs. 2.7-fold). Interestingly, a significant reduction of ICa,L and Ca2+ sparks was observed upon Ca2+/Calmodulin-dependent protein kinase II inhibition by KN-93, but this inhibition had no effect on ITI. Fluorescence resonance energy transfer (FRET) experiments showed that although AF promoted cytosolic desensitization to ß-adrenergic stimulation, ISO increased cAMP to similar levels in both groups of patients in the L-type Ca2+ channel and ryanodine receptor compartments. Basal cAMP signaling also showed compartment-specific regulation by phosphodiesterases in atrial myocytes from 44 Ctl and 43 AF patients. Our results suggest that AF is associated with opposite changes in compartmentalized PKA/cAMP-dependent regulation of ICa,L (down-regulation) and ITI (up-regulation).


Subject(s)
Atrial Fibrillation/metabolism , Calcium Signaling , Cyclic AMP/metabolism , Adrenergic beta-Antagonists/pharmacology , Aged , Animals , Calcium/metabolism , Calcium Channels, L-Type/metabolism , Calcium Signaling/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Carvedilol/pharmacology , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Cytosol/drug effects , Cytosol/metabolism , Female , Humans , Male , Middle Aged , Receptors, Adrenergic, beta/metabolism , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism
6.
PLoS Negl Trop Dis ; 15(11): e0009951, 2021 11.
Article in English | MEDLINE | ID: mdl-34780470

ABSTRACT

With current drug treatments failing due to toxicity, low efficacy and resistance; leishmaniasis is a major global health challenge that desperately needs new validated drug targets. Inspired by activity of the natural chalcone 2',6'-dihydroxy-4'-methoxychalcone (DMC), the nitro-analogue, 3-nitro-2',4',6'- trimethoxychalcone (NAT22, 1c) was identified as potent broad spectrum antileishmanial drug lead. Structural modification provided an alkyne containing chemical probe that labelled a protein within the parasite that was confirmed as cytosolic tryparedoxin peroxidase (cTXNPx). Crucially, labelling is observed in both promastigote and intramacrophage amastigote life forms, with no evidence of host macrophage toxicity. Incubation of the chalcone in the parasite leads to ROS accumulation and parasite death. Deletion of cTXNPx, by CRISPR-Cas9, dramatically impacts upon the parasite phenotype and reduces the antileishmanial activity of the chalcone analogue. Molecular docking studies with a homology model of in-silico cTXNPx suggest that the chalcone is able to bind in the putative active site hindering access to the crucial cysteine residue. Collectively, this work identifies cTXNPx as an important target for antileishmanial chalcones.


Subject(s)
Antiprotozoal Agents/therapeutic use , Chalcone/metabolism , Chalcone/pharmacology , Cytosol/drug effects , Leishmania/drug effects , Peroxidases/antagonists & inhibitors , Protozoan Proteins/antagonists & inhibitors , Animals , Antiprotozoal Agents/administration & dosage , Antiprotozoal Agents/pharmacology , Cells, Cultured , Chalcone/administration & dosage , Chalcone/analogs & derivatives , Cytosol/enzymology , Cytosol/parasitology , Drug Discovery , Humans , Leishmania/classification , Leishmaniasis/drug therapy , Leishmaniasis/parasitology , Macrophages/drug effects , Macrophages/parasitology , Mice , Mice, Inbred BALB C , Molecular Docking Simulation , Peroxidases/metabolism , Protozoan Proteins/metabolism
7.
Biochem Biophys Res Commun ; 585: 191-195, 2021 12 31.
Article in English | MEDLINE | ID: mdl-34813979

ABSTRACT

Impairment of pancreatic ß cells is a principal driver of the development of diabetes. Restoring normal insulin release from the ß cells depends on the ATP produced by the intracellular mitochondria. In maintaining mitochondrial function, the tumor suppressor p53 has emerged as a novel regulator of metabolic homeostasis and participates in adaptations to nutritional changes. In this study, we used orotic acid, an intermediate in the pathway for de novo synthesis of the pyrimidine nucleotide, to reduce genotoxicity. Administration of orotic acid reduced p53 activation of MIN6 ß cells and subsequently reduced ß cell death in the db/db mouse. Orotic acid intake helped to maintain the islet size, number of ß cells, and protected insulin secretion in the db/db mouse. In conclusion, orotic acid treatment maintained ß cell function and reduced cell death, and may therefore, be a future therapeutic strategy for the prevention and treatment of diabetes.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Disease Models, Animal , Insulin-Secreting Cells/drug effects , Orotic Acid/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Blood Glucose/metabolism , Body Weight/drug effects , Cell Line, Tumor , Cytosol/drug effects , Cytosol/metabolism , Diabetes Mellitus, Type 2/blood , Humans , Insulin Secretion/drug effects , Insulin-Secreting Cells/metabolism , Male , Mice, Inbred C57BL , Orotic Acid/administration & dosage , Orotic Acid/blood , Protective Agents/administration & dosage , Protective Agents/pharmacology
8.
Biomolecules ; 11(10)2021 10 11.
Article in English | MEDLINE | ID: mdl-34680130

ABSTRACT

Although the causes of hepatotoxicity among alcohol-abusing HIV patients are multifactorial, alcohol remains the least explored "second hit" for HIV-related hepatotoxicity. Here, we investigated whether metabolically derived acetaldehyde impairs lysosomes to enhance HIV-induced hepatotoxicity. We exposed Cytochrome P450 2E1 (CYP2E1)-expressing Huh 7.5 (also known as RLW) cells to an acetaldehyde-generating system (AGS) for 24 h. We then infected (or not) the cells with HIV-1ADA then exposed them again to AGS for another 48 h. Lysosome damage was assessed by galectin 3/LAMP1 co-localization and cathepsin leakage. Expression of lysosome biogenesis-transcription factor, TFEB, was measured by its protein levels and by in situ immunofluorescence. Exposure of cells to both AGS + HIV caused the greatest amount of lysosome leakage and its impaired lysosomal biogenesis, leading to intrinsic apoptosis. Furthermore, the movement of TFEB from cytosol to the nucleus via microtubules was impaired by AGS exposure. The latter impairment appeared to occur by acetylation of α-tubulin. Moreover, ZKSCAN3, a repressor of lysosome gene activation by TFEB, was amplified by AGS. Both these changes contributed to AGS-elicited disruption of lysosome biogenesis. Our findings indicate that metabolically generated acetaldehyde damages lysosomes and likely prevents their repair and restoration, thereby exacerbating HIV-induced hepatotoxicity.


Subject(s)
Ethanol/toxicity , HIV Infections/pathology , Liver/pathology , Liver/virology , Lysosomes/metabolism , Organelle Biogenesis , Acetaldehyde/metabolism , Acetylcysteine/pharmacology , Apoptosis/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cathepsins/metabolism , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cytosol/drug effects , Cytosol/metabolism , Humans , Liver/drug effects , Lysosomes/drug effects , Models, Biological , Oxidative Stress/drug effects , Proteasome Endopeptidase Complex/metabolism , Transcription Factors/metabolism
9.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34493661

ABSTRACT

Regulation of apoptosis is tightly linked with the targeting of numerous Bcl-2 proteins to the mitochondrial outer membrane (MOM), where their activation or inhibition dictates cell death or survival. According to the traditional view of apoptotic regulation, BH3-effector proteins are indispensable for the cytosol-to-MOM targeting and activation of proapoptotic and antiapoptotic members of the Bcl-2 protein family. This view is challenged by recent studies showing that these processes can occur in cells lacking BH3 effectors by as yet to be determined mechanism(s). Here, we exploit a model membrane system that recapitulates key features of MOM to demonstrate that the proapoptotic Bcl-2 protein BAX and antiapoptotic Bcl-xL have an inherent ability to interact with membranes in the absence of BH3 effectors, but only in the presence of cellular concentrations of Mg2+/Ca2+ Under these conditions, BAX and Bcl-xL are selectively targeted to membranes, refolded, and activated in the presence of anionic lipids especially the mitochondrial-specific lipid cardiolipin. These results provide a mechanistic explanation for the mitochondrial targeting and activation of Bcl-2 proteins in cells lacking BH3 effectors. At cytosolic Mg2+ levels, the BH3-independent activation of BAX could provide localized amplification of apoptotic signaling at regions enriched in cardiolipin (e.g., contact sites between MOM and mitochondrial inner membrane). Increases in MOM cardiolipin, as well as cytosolic [Ca2+] during apoptosis could further contribute to its MOM targeting and activity. Meanwhile, the BH3-independent targeting and activation of Bcl-xL to the MOM is expected to counter the action of proapoptotic BAX, thereby preventing premature commitment to apoptosis.


Subject(s)
Cardiolipins/pharmacology , Cell Membrane Permeability , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism , Cytosol/drug effects , Cytosol/metabolism , Humans , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membranes/drug effects , bcl-2-Associated X Protein/genetics , bcl-X Protein/genetics
10.
Cells ; 10(7)2021 07 05.
Article in English | MEDLINE | ID: mdl-34359868

ABSTRACT

Hsp90ß is a major chaperone involved in numerous cellular processes. Hundreds of client proteins depend on Hsp90ß for proper folding and/or activity. Regulation of Hsp90ß is critical to coordinate its tasks and is mediated by several post-translational modifications. Here, we focus on two phosphorylation sites located in the charged linker region of human Hsp90ß, Ser226 and Ser255, which have been frequently reported but whose function remains unclear. Targeted measurements by mass spectrometry indicated that intracellular Hsp90ß is highly phosphorylated on both sites (>90%). The level of phosphorylation was unaffected by various stresses (e.g., heat shock, inhibition with drugs) that impact Hsp90ß activity. Mutating the two serines to alanines increased the amount of proteins interacting with Hsp90ß globally and increased the sensitivity to tryptic cleavage in the C-terminal domain. Further investigation revealed that phosphorylation on Ser255 and to a lesser extent on Ser226 is decreased in the conditioned medium of cultured K562 cells, and that a non-phosphorylatable double alanine mutant was secreted more efficiently than the wild type. Overall, our results show that phosphorylation events in the charged linker regulate both the interactions of Hsp90ß and its secretion, through changes in the conformation of the chaperone.


Subject(s)
HSP90 Heat-Shock Proteins/metabolism , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Culture Media, Conditioned/pharmacology , Cytosol/drug effects , Cytosol/metabolism , HEK293 Cells , HSP90 Heat-Shock Proteins/chemistry , Heat-Shock Response/drug effects , Humans , K562 Cells , Mutant Proteins/metabolism , Mutation/genetics , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Binding/drug effects , Protein Domains , Proteolysis/drug effects , Stress, Physiological/drug effects
11.
Oxid Med Cell Longev ; 2021: 5572129, 2021.
Article in English | MEDLINE | ID: mdl-34394828

ABSTRACT

Prion diseases are caused by PrPsc accumulation in the brain, which triggers dysfunctional mitochondrial injury and reactive oxygen species (ROS) generation in neurons. Recent studies on prion diseases suggest that endoplasmic reticulum (ER) stress induced by misfolding proteins such as misfolded prion protein results in activation of calcineurin. Calcineurin is a calcium-related protein phosphatase of type 2B that exists in copious quantities in the brain and acts as a critical nodal component in the control of cellular functions. To investigate the relationship between calcineurin and intracellular ROS, we assessed the alteration of CaN and ROS induced by prion peptide (PrP) 106-126. Human prion peptide increased mitochondrial ROS by activating calcineurin, and the inhibition of calcineurin activity protected mitochondrial function and neuronal apoptosis in neuronal cells. These results suggest that calcineurin plays a pivotal role in neuronal apoptosis by mediating mitochondrial injury and ROS in prion diseases.


Subject(s)
Calcineurin/metabolism , Mitochondria/drug effects , Peptides/pharmacology , Prion Proteins/chemistry , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cytosol/drug effects , Cytosol/metabolism , Humans , Mitochondria/metabolism , Peptides/chemical synthesis , Tacrolimus/pharmacology , Up-Regulation/drug effects
12.
RNA ; 27(10): 1241-1256, 2021 10.
Article in English | MEDLINE | ID: mdl-34244458

ABSTRACT

Stress granules (SGs) are membraneless organelles composed of mRNAs and RNA binding proteins which undergo assembly in response to stress-induced inactivation of translation initiation. In general, SG recruitment is limited to a subpopulation of a given mRNA species and RNA-seq analyses of purified SGs revealed that signal sequence-encoding (i.e., endoplasmic reticulum [ER]-targeted) transcripts are significantly underrepresented, consistent with prior reports that ER localization can protect mRNAs from SG recruitment. Using translational profiling, cell fractionation, and single molecule mRNA imaging, we examined SG biogenesis following activation of the unfolded protein response (UPR) by 1,4-dithiothreitol (DTT) and report that gene-specific subsets of cytosolic and ER-targeted mRNAs can be recruited into SGs. Furthermore, we demonstrate that SGs form in close proximity to or directly associated with the ER membrane. ER-associated SG assembly was also observed during arsenite stress, suggesting broad roles for the ER in SG biogenesis. Recruitment of a given mRNA into SGs required stress-induced translational repression, though translational inhibition was not solely predictive of an mRNA's propensity for SG recruitment. SG formation was prevented by the transcriptional inhibitors actinomycin D or triptolide, suggesting a functional link between gene transcriptional state and SG biogenesis. Collectively these data demonstrate that ER-targeted and cytosolic mRNAs can be recruited into ER-associated SGs and this recruitment is sensitive to transcriptional inhibition. We propose that newly transcribed mRNAs exported under conditions of suppressed translation initiation are primary SG substrates, with the ER serving as the central subcellular site of SG formation.


Subject(s)
Cytoplasmic Granules/genetics , Endoplasmic Reticulum/genetics , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Unfolded Protein Response , Biomarkers/metabolism , Cytoplasmic Granules/drug effects , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/ultrastructure , Cytosol/drug effects , Cytosol/metabolism , Dactinomycin/pharmacology , Diterpenes/pharmacology , Dithiothreitol/pharmacology , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Epoxy Compounds/pharmacology , Gene Expression , HeLa Cells , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Organelle Biogenesis , Peptide Chain Initiation, Translational/drug effects , Phenanthrenes/pharmacology , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Single Molecule Imaging , Stress, Physiological/drug effects , Transcription, Genetic/drug effects , beta 2-Microglobulin/genetics , beta 2-Microglobulin/metabolism
13.
Cells ; 10(7)2021 06 23.
Article in English | MEDLINE | ID: mdl-34201461

ABSTRACT

Cholinergic innervation in the pancreas controls both the release of digestive enzymes to support the intestinal digestion and absorption, as well as insulin release to promote nutrient use in the cells of the body. The effects of muscarinic receptor stimulation are described in detail for endocrine beta cells and exocrine acinar cells separately. Here we describe morphological and functional criteria to separate these two cell types in situ in tissue slices and simultaneously measure their response to ACh stimulation on cytosolic Ca2+ oscillations [Ca2+]c in stimulatory glucose conditions. Our results show that both cell types respond to glucose directly in the concentration range compatible with the glucose transporters they express. The physiological ACh concentration increases the frequency of glucose stimulated [Ca2+]c oscillations in both cell types and synchronizes [Ca2+]c oscillations in acinar cells. The supraphysiological ACh concentration further increases the oscillation frequency on the level of individual beta cells, inhibits the synchronization between these cells, and abolishes oscillatory activity in acinar cells. We discuss possible mechanisms leading to the observed phenomena.


Subject(s)
Acetylcholine/pharmacology , Acinar Cells/metabolism , Calcium Signaling , Cytosol/metabolism , Insulin-Secreting Cells/metabolism , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Cytosol/drug effects , Female , Glucose/pharmacology , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Male , Mice, Inbred C57BL , Receptors, Muscarinic/metabolism , Time Factors
14.
FASEB J ; 35(7): e21712, 2021 07.
Article in English | MEDLINE | ID: mdl-34110637

ABSTRACT

Palmitic acid (PA) is a saturated fatty acid whose high consumption has been largely associated with the development of different metabolic alterations, such as insulin resistance, metabolic syndrome, and type 2 diabetes. Particularly in the brain, insulin signaling disruption has been linked to cognitive decline and is considered a risk factor for Alzheimer's disease. Cumulative evidence has demonstrated the participation of PA in the molecular cascade underlying cellular insulin resistance in peripheral tissues, but its role in the development of neuronal insulin resistance and the mechanisms involved are not fully understood. It has generally been accepted that the brain does not utilize fatty acids as a primary energy source, but recent evidence shows that neurons possess the machinery for fatty acid ß-oxidation. However, it is still unclear under what conditions neurons use fatty acids as energy substrates and the implications of their oxidative metabolism in modifying insulin-stimulated effects. In the present work, we have found that neurons differentiated from human neuroblastoma MSN exposed to high but nontoxic concentrations of PA generate ATP through mitochondrial metabolism, which is associated with an increase in the cytosolic Ca2+ and diminished insulin signaling in neurons. These findings reveal a novel mechanism by which saturated fatty acids produce Ca2+ entry and insulin resistance that may play a causal role in increasing neuronal vulnerability associated with metabolic diseases.


Subject(s)
Calcium/metabolism , Energy Metabolism/drug effects , Insulin Resistance/physiology , Neurons/drug effects , Palmitic Acid/pharmacology , Adenosine Triphosphate/metabolism , Cell Line, Tumor , Cytosol/drug effects , Cytosol/metabolism , Fatty Acids/pharmacology , Humans , Insulin/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Neuroblastoma/metabolism , Neurons/metabolism , Signal Transduction/drug effects
15.
Transl Res ; 236: 133-146, 2021 10.
Article in English | MEDLINE | ID: mdl-33905948

ABSTRACT

Hypertrophic scars are the most common post-burn complications characterized by fibroblast proliferation and excessive extracellular matrix deposition. The intermediate-conductance Ca2+-activated K+ channel (KCa3.1) mediates fibroblast activation, resulting in several fibrotic diseases; however, this channel's role in the formation of post-burn hypertrophic skin scars remains unknown. Herein, we investigated the role of KCa3.1 and the therapeutic potential of TRAM-34, a selective inhibitor of KCa3.1, in hypertrophic skin scar formation following burn injury. Cytosolic Ca2+ levels, the expression of KCa3.1 and hypertrophic markers, and the proliferation of skin fibroblasts obtained directly from patients with third-degree burns who consequently developed post-burn hypertrophic scars were assessed. The anti-fibrotic effect of KCa3.1 inhibition by TRAM-34 was evaluated in vitro (fibroblasts) and in vivo (mouse burn models). Fibroblasts from burn wounds exhibited remarkably higher levels of cytosolic Ca2+ than normal cells. KCa3.1 expression was markedly higher in the membrane fraction but lower in the cytosolic fraction of burn wound fibroblasts than in normal cells. Selective inhibition of KCa3.1 by TRAM-34 markedly reduced not only the proliferation of burn wound fibroblasts but also the expression of hypertrophic markers in these cells. Anti-scarring molecular, histological, and visual effects of TRAM-34 were confirmed in murine burn models. Altered subcellular expression of KCa3.1 is a novel mechanism underlying the cellular response to burn injury. Our results suggest that selective inhibition of KCa3.1 by TRAM-34 has therapeutic potential against post-burn hypertrophic scar formation.


Subject(s)
Burns/drug therapy , Burns/metabolism , Cicatrix, Hypertrophic/drug therapy , Cicatrix, Hypertrophic/etiology , Intermediate-Conductance Calcium-Activated Potassium Channels/metabolism , Pyrazoles/therapeutic use , Adolescent , Adult , Aged , Animals , Biomarkers/metabolism , Burns/genetics , Burns/pathology , Calcium/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cicatrix, Hypertrophic/genetics , Cytosol/drug effects , Cytosol/metabolism , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Male , Mice , Middle Aged , Pyrazoles/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Young Adult
16.
Am J Physiol Lung Cell Mol Physiol ; 321(1): L91-L101, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33908264

ABSTRACT

During agonist stimulation of airway smooth muscle (ASM), agonists such as ACh induce a transient increase in cytosolic Ca2+ concentration ([Ca2+]cyt), which leads to a contractile response [excitation-contraction (E-C) coupling]. Previously, the sensitivity of the contractile response of ASM to elevated [Ca2+]cyt (Ca2+ sensitivity) was assessed as the ratio of maximum force to maximum [Ca2+]cyt. However, this static assessment of Ca2+ sensitivity overlooks the dynamic nature of E-C coupling in ASM. In this study, we simultaneously measured [Ca2+]cyt and isometric force responses to three concentrations of ACh (1, 2.6, and 10 µM). Both maximum [Ca2+]cyt and maximum force responses were ACh concentration dependent, but force increased disproportionately, thereby increasing static Ca2+ sensitivity. The dynamic properties of E-C coupling were assessed in several ways. The temporal delay between the onset of ACh-induced [Ca2+]cyt and onset force responses was not affected by ACh concentration. The rates of rise of the ACh-induced [Ca2+]cyt and force responses increased with increasing ACh concentration. The integral of the phase-loop plot of [Ca2+]cyt and force from onset to steady state also increased with increasing ACh concentration, whereas the rate of relaxation remained unchanged. Although these results suggest an ACh concentration-dependent increase in the rate of cross-bridge recruitment and in the rate of rise of [Ca2+]cyt, the extent of regulatory myosin light-chain (rMLC20) phosphorylation was not dependent on ACh concentration. We conclude that the dynamic properties of [Ca2+]cyt and force responses in ASM are dependent on ACh concentration but reflect more than changes in the extent of rMLC20 phosphorylation.


Subject(s)
Calcium/metabolism , Cholinergic Agents/pharmacology , Cytosol/metabolism , Muscle Contraction , Muscle, Smooth/metabolism , Respiratory System/metabolism , Animals , Cytosol/drug effects , Female , Male , Muscle, Smooth/drug effects , Respiratory System/drug effects , Swine
17.
Oxid Med Cell Longev ; 2021: 8877691, 2021.
Article in English | MEDLINE | ID: mdl-33628390

ABSTRACT

Hydrogen sulfide (H2S) is naturally synthesized in a wide range of mammalian tissues. Whether H2S is involved in the regulation of erythrocyte functions remains unknown. Using mice with a genetic deficiency in a H2S natural synthesis enzyme cystathionine-γ-lyase (CSE) and high-throughput metabolomic profiling, we found that levels of erythrocyte 2,3-bisphosphoglycerate (2,3-BPG), an erythroid-specific metabolite negatively regulating hemoglobin- (Hb-) oxygen (O2) binding affinity, were increased in CSE knockout (Cse -/-) mice under normoxia. Consistently, the 50% oxygen saturation (P50) value was increased in erythrocytes of Cse -/- mice. These effects were reversed by treatment with H2S donor GYY4137. In the models of cultured mouse and human erythrocytes, we found that H2S directly acts on erythrocytes to decrease 2,3-BPG production, thereby enhancing Hb-O2 binding affinity. Mouse genetic studies showed that H2S produced by peripheral tissues has a tonic inhibitory effect on 2,3-BPG production and consequently maintains Hb-O2 binding affinity in erythrocytes. We further revealed that H2S promotes Hb release from the membrane to the cytosol and consequently enhances bisphosphoglycerate mutase (BPGM) anchoring to the membrane. These processes might be associated with S-sulfhydration of Hb. Moreover, hypoxia decreased the circulatory H2S level and increased the erythrocyte 2,3-BPG content in mice, which could be reversed by GYY4137 treatment. Altogether, our study revealed a novel signaling pathway that regulates oxygen-carrying capacity in erythrocytes and highlights a previously unrecognized role of H2S in erythrocyte 2,3-BPG production.


Subject(s)
2,3-Diphosphoglycerate/metabolism , Erythrocytes/metabolism , Hemoglobins/metabolism , Hydrogen Sulfide/pharmacology , Oxygen/metabolism , Animals , Bisphosphoglycerate Mutase/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Cells, Cultured , Cytosol/drug effects , Cytosol/metabolism , Erythrocytes/drug effects , Humans , Hypoxia/metabolism , Mice, Inbred C57BL , Models, Biological , Protein Transport/drug effects , Sulfates/metabolism
18.
Toxicology ; 453: 152726, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33617915

ABSTRACT

Cadmium (Cd) is a ubiquitous environmental and occupational pollutant that is considered as a high-risk factor for neurodegenerative diseases. However, the mechanism underlying Cd-induced neurotoxicity has not been fully elucidated. Abnormal mitochondrial distribution and excessive mitochondrial fission are increasingly implicated in various neurological pathologies. Herein, by exposing primary cortical neurons to Cd (10 and 100 µM) for various times (0, 6, 12, and 24 h), we observed that the rapid motility of the mitochondria in neurons progressively slowed. Many more mitochondria were transported and distributed to the somas of Cd-treated neurons. Coupled with abnormal mitochondrial distribution, Cd exposure triggered excessive mitochondrial fragmentation, followed by mitochondrial membrane potential loss and neuronal damage. However, BAPTA-AM, a chelator of cytosolic calcium ([Ca2+]c), significantly attenuated Cd-induced abnormal mitochondrial distribution and excessive mitochondrial fission, which protected against Cd-induced mitochondrial damage and neuronal toxicity. In contrast to the increase in [Ca2+]c, Cd exposure had no effect on the level of mitochondrial calcium ([Ca2+]m). Inhibiting [Ca2+]m uptake, either by ruthenium 360 (Ru360) or by knock-out of mitochondrial calcium uniporter (MCU), failed to alleviate Cd-induced mitochondrial damage and neuronal toxicity. Additionally, in MCU knock-out neurons, BAPTA-AM effectively prevented Cd-induced abnormal mitochondrial distribution and excessive mitochondrial fission. Taken together, Cd exposure disrupts mitochondrial distribution and activates excessive mitochondrial fission by elevating [Ca2+]c independent of MCU-mediated mitochondrial calcium uptake, thereby leading to neurotoxicity. Chelating overloaded [Ca2+]c is a promising strategy to prevent the neurotoxicity of Cd.


Subject(s)
Cadmium/toxicity , Calcium Channels/deficiency , Calcium/metabolism , Cytosol/metabolism , Mitochondria/metabolism , Mitochondrial Dynamics/physiology , Animals , Animals, Newborn , Calcium Channels/genetics , Cells, Cultured , Cytosol/drug effects , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondrial Dynamics/drug effects , Rats , Rats, Sprague-Dawley
19.
Nat Commun ; 12(1): 290, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33436590

ABSTRACT

The environmental contaminant 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) causes hepatic toxicity associated with prominent lipid accumulation in humans. Here, the authors report that the lysosomal copper transporter SLC46A3 is induced by TCDD and underlies the hepatic lipid accumulation in mice, potentially via effects on mitochondrial function. SLC46A3 was localized to the lysosome where it modulated intracellular copper levels. Forced expression of hepatic SLC46A3 resulted in decreased mitochondrial membrane potential and abnormal mitochondria morphology consistent with lower copper levels. SLC46A3 expression increased hepatic lipid accumulation similar to the known effects of TCDD exposure in mice and humans. The TCDD-induced hepatic triglyceride accumulation was significantly decreased in Slc46a3-/- mice and was more pronounced when these mice were fed a high-fat diet, as compared to wild-type mice. These data are consistent with a model where lysosomal SLC46A3 induction by TCDD leads to cytosolic copper deficiency resulting in mitochondrial dysfunction leading to lower lipid catabolism, thus linking copper status to mitochondrial function, lipid metabolism and TCDD-induced liver toxicity.


Subject(s)
Copper Transport Proteins/metabolism , Copper/metabolism , Cytosol/metabolism , Homeostasis , Lysosomes/metabolism , Proton-Coupled Folate Transporter/metabolism , Animals , Copper Transport Proteins/genetics , Cytosol/drug effects , Green Fluorescent Proteins/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Homeostasis/drug effects , Ions , Liver/metabolism , Lysosomes/drug effects , Male , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Polychlorinated Dibenzodioxins/toxicity , Proton-Coupled Folate Transporter/genetics , Receptors, Aryl Hydrocarbon/metabolism , Substrate Specificity/drug effects , Superoxide Dismutase/metabolism , Triglycerides/metabolism
20.
Cell Biol Int ; 45(3): 633-641, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33247607

ABSTRACT

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.


Subject(s)
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
SELECTION OF CITATIONS
SEARCH DETAIL
...