Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 145
Filter
Add more filters

Publication year range
1.
Cell ; 183(2): 474-489.e17, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33035451

ABSTRACT

Mg2+ is the most abundant divalent cation in metazoans and an essential cofactor for ATP, nucleic acids, and countless metabolic enzymes. To understand how the spatio-temporal dynamics of intracellular Mg2+ (iMg2+) are integrated into cellular signaling, we implemented a comprehensive screen to discover regulators of iMg2+ dynamics. Lactate emerged as an activator of rapid release of Mg2+ from endoplasmic reticulum (ER) stores, which facilitates mitochondrial Mg2+ (mMg2+) uptake in multiple cell types. We demonstrate that this process is remarkably temperature sensitive and mediated through intracellular but not extracellular signals. The ER-mitochondrial Mg2+ dynamics is selectively stimulated by L-lactate. Further, we show that lactate-mediated mMg2+ entry is facilitated by Mrs2, and point mutations in the intermembrane space loop limits mMg2+ uptake. Intriguingly, suppression of mMg2+ surge alleviates inflammation-induced multi-organ failure. Together, these findings reveal that lactate mobilizes iMg2+ and links the mMg2+ transport machinery with major metabolic feedback circuits and mitochondrial bioenergetics.


Subject(s)
Endoplasmic Reticulum/metabolism , Lactic Acid/metabolism , Magnesium/metabolism , Animals , COS Cells , Calcium/metabolism , Calcium Signaling/physiology , Chlorocebus aethiops , Endoplasmic Reticulum/physiology , Female , HeLa Cells , Hep G2 Cells , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/metabolism
2.
Mol Cell ; 84(7): 1321-1337.e11, 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38513662

ABSTRACT

Intracellular Mg2+ (iMg2+) is bound with phosphometabolites, nucleic acids, and proteins in eukaryotes. Little is known about the intracellular compartmentalization and molecular details of Mg2+ transport into/from cellular organelles such as the endoplasmic reticulum (ER). We found that the ER is a major iMg2+ compartment refilled by a largely uncharacterized ER-localized protein, TMEM94. Conventional and AlphaFold2 predictions suggest that ERMA (TMEM94) is a multi-pass transmembrane protein with large cytosolic headpiece actuator, nucleotide, and phosphorylation domains, analogous to P-type ATPases. However, ERMA uniquely combines a P-type ATPase domain and a GMN motif for ERMg2+ uptake. Experiments reveal that a tyrosine residue is crucial for Mg2+ binding and activity in a mechanism conserved in both prokaryotic (mgtB and mgtA) and eukaryotic Mg2+ ATPases. Cardiac dysfunction by haploinsufficiency, abnormal Ca2+ cycling in mouse Erma+/- cardiomyocytes, and ERMA mRNA silencing in human iPSC-cardiomyocytes collectively define ERMA as an essential component of ERMg2+ uptake in eukaryotes.


Subject(s)
Adenosine Triphosphatases , P-type ATPases , Animals , Mice , Humans , Adenosine Triphosphatases/metabolism , Membrane Transport Proteins/metabolism , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Biological Transport , P-type ATPases/metabolism , Calcium/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases
3.
Cell ; 151(3): 630-44, 2012 Oct 26.
Article in English | MEDLINE | ID: mdl-23101630

ABSTRACT

Mitochondrial Ca(2+) (Ca(2+)(m)) uptake is mediated by an inner membrane Ca(2+) channel called the uniporter. Ca(2+) uptake is driven by the considerable voltage present across the inner membrane (ΔΨ(m)) generated by proton pumping by the respiratory chain. Mitochondrial matrix Ca(2+) concentration is maintained five to six orders of magnitude lower than its equilibrium level, but the molecular mechanisms for how this is achieved are not clear. Here, we demonstrate that the mitochondrial protein MICU1 is required to preserve normal [Ca(2+)](m) under basal conditions. In its absence, mitochondria become constitutively loaded with Ca(2+), triggering excessive reactive oxygen species generation and sensitivity to apoptotic stress. MICU1 interacts with the uniporter pore-forming subunit MCU and sets a Ca(2+) threshold for Ca(2+)(m) uptake without affecting the kinetic properties of MCU-mediated Ca(2+) uptake. Thus, MICU1 is a gatekeeper of MCU-mediated Ca(2+)(m) uptake that is essential to prevent [Ca(2+)](m) overload and associated stress.


Subject(s)
Calcium-Binding Proteins/metabolism , Calcium/metabolism , Cation Transport Proteins/metabolism , Cell Survival , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Apoptosis , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/genetics , Cation Transport Proteins/chemistry , Cation Transport Proteins/genetics , Gene Knockdown Techniques , HeLa Cells , Humans , Mitochondrial Membrane Transport Proteins/chemistry , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membranes/metabolism
4.
Mol Cell ; 65(6): 1014-1028.e7, 2017 Mar 16.
Article in English | MEDLINE | ID: mdl-28262504

ABSTRACT

Ca2+ dynamics and oxidative signaling are fundamental mechanisms for mitochondrial bioenergetics and cell function. The MCU complex is the major pathway by which these signals are integrated in mitochondria. Whether and how these coactive elements interact with MCU have not been established. As an approach toward understanding the regulation of MCU channel by oxidative milieu, we adapted inflammatory and hypoxia models. We identified the conserved cysteine 97 (Cys-97) to be the only reactive thiol in human MCU that undergoes S-glutathionylation. Furthermore, biochemical, structural, and superresolution imaging analysis revealed that MCU oxidation promotes MCU higher order oligomer formation. Both oxidation and mutation of MCU Cys-97 exhibited persistent MCU channel activity with higher [Ca2+]m uptake rate, elevated mROS, and enhanced [Ca2+]m overload-induced cell death. In contrast, these effects were largely independent of MCU interaction with its regulators. These findings reveal a distinct functional role for Cys-97 in ROS sensing and regulation of MCU activity.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling , Calcium/metabolism , Endothelial Cells/metabolism , Ion Channel Gating , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Reactive Oxygen Species/metabolism , Animals , COS Cells , Calcium Channels/chemistry , Calcium Channels/genetics , Calcium Signaling/drug effects , Cell Death , Cell Hypoxia , Chlorocebus aethiops , Cysteine , Endothelial Cells/drug effects , Endothelial Cells/pathology , Energy Metabolism , Glutathione/metabolism , HEK293 Cells , HeLa Cells , Humans , Ion Channel Gating/drug effects , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondria/drug effects , Mitochondria/pathology , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/pathology , Mutation , Oxidation-Reduction , Protein Multimerization , Protein Processing, Post-Translational , Protein Structure, Quaternary , Structure-Activity Relationship , Thrombin/pharmacology , Time Factors , Transfection
5.
Hum Mol Genet ; 31(3): 376-385, 2022 02 03.
Article in English | MEDLINE | ID: mdl-34494107

ABSTRACT

Calcium signaling via mitochondrial calcium uniporter (MCU) complex coordinates mitochondrial bioenergetics with cellular energy demands. Emerging studies show that the stability and activity of the pore-forming subunit of the complex, MCU, is dependent on the mitochondrial phospholipid, cardiolipin (CL), but how this impacts calcium-dependent mitochondrial bioenergetics in CL-deficiency disorder like Barth syndrome (BTHS) is not known. Here we utilized multiple models of BTHS including yeast, mouse muscle cell line, as well as BTHS patient cells and cardiac tissue to show that CL is required for the abundance and stability of the MCU-complex regulatory subunit MICU1. Interestingly, the reduction in MICU1 abundance in BTHS mitochondria is independent of MCU. Unlike MCU and MICU1/MICU2, other subunit and associated factor of the uniporter complex, EMRE and MCUR1, respectively, are not affected in BTHS models. Consistent with the decrease in MICU1 levels, we show that the kinetics of MICU1-dependent mitochondrial calcium uptake is perturbed and acute stimulation of mitochondrial calcium signaling in BTHS myoblasts fails to activate pyruvate dehydrogenase, which in turn impairs the generation of reducing equivalents and blunts mitochondrial bioenergetics. Taken together, our findings suggest that defects in mitochondrial calcium signaling could contribute to cardiac and skeletal muscle pathologies observed in BTHS patients.


Subject(s)
Barth Syndrome , Calcium , Animals , Barth Syndrome/genetics , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Signaling , Calcium-Binding Proteins/metabolism , Humans , Mice , Mitochondrial Membrane Transport Proteins/metabolism , Saccharomyces cerevisiae/metabolism
6.
Nat Rev Mol Cell Biol ; 13(9): 549-65, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22914293

ABSTRACT

Stromal interaction molecule (STIM) proteins function in cells as dynamic coordinators of cellular calcium (Ca(2+)) signals. Spanning the endoplasmic reticulum (ER) membrane, they sense tiny changes in the levels of Ca(2+) stored within the ER lumen. As ER Ca(2+) is released to generate primary Ca(2+) signals, STIM proteins undergo an intricate activation reaction and rapidly translocate into junctions formed between the ER and the plasma membrane. There, STIM proteins tether and activate the highly Ca(2+)-selective Orai channels to mediate finely controlled Ca(2+) signals and to homeostatically balance cellular Ca(2+). Details are emerging on the remarkable organization within these STIM-induced junctional microdomains and the identification of new regulators and alternative target proteins for STIM.


Subject(s)
Calcium Signaling , Calcium/metabolism , Cell Adhesion Molecules/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Animals , Cell Membrane/metabolism , Humans , Intercellular Junctions/metabolism , Models, Biological , Stromal Interaction Molecule 1 , Stromal Interaction Molecule 2
7.
Nature ; 545(7652): 93-97, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28445457

ABSTRACT

Mitochondrial calcium (mCa2+) has a central role in both metabolic regulation and cell death signalling, however its role in homeostatic function and disease is controversial. Slc8b1 encodes the mitochondrial Na+/Ca2+ exchanger (NCLX), which is proposed to be the primary mechanism for mCa2+ extrusion in excitable cells. Here we show that tamoxifen-induced deletion of Slc8b1 in adult mouse hearts causes sudden death, with less than 13% of affected mice surviving after 14 days. Lethality correlated with severe myocardial dysfunction and fulminant heart failure. Mechanistically, cardiac pathology was attributed to mCa2+ overload driving increased generation of superoxide and necrotic cell death, which was rescued by genetic inhibition of mitochondrial permeability transition pore activation. Corroborating these findings, overexpression of NCLX in the mouse heart by conditional transgenesis had the beneficial effect of augmenting mCa2+ clearance, preventing permeability transition and protecting against ischaemia-induced cardiomyocyte necrosis and heart failure. These results demonstrate the essential nature of mCa2+ efflux in cellular function and suggest that augmenting mCa2+ efflux may be a viable therapeutic strategy in disease.


Subject(s)
Calcium/metabolism , Homeostasis , Mitochondria/metabolism , Sodium-Calcium Exchanger/metabolism , Animals , Cell Survival , Death, Sudden , Female , Gene Deletion , HeLa Cells , Heart Failure/metabolism , Heart Failure/pathology , Humans , Male , Mice , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Necrosis , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Sodium-Calcium Exchanger/genetics , Superoxides/metabolism , Tamoxifen/pharmacology , Ventricular Remodeling
8.
Mol Cell ; 60(1): 47-62, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26387735

ABSTRACT

Mitochondrial permeability transition is a phenomenon in which the mitochondrial permeability transition pore (PTP) abruptly opens, resulting in mitochondrial membrane potential (ΔΨm) dissipation, loss of ATP production, and cell death. Several genetic candidates have been proposed to form the PTP complex, however, the core component is unknown. We identified a necessary and conserved role for spastic paraplegia 7 (SPG7) in Ca(2+)- and ROS-induced PTP opening using RNAi-based screening. Loss of SPG7 resulted in higher mitochondrial Ca(2+) retention, similar to cyclophilin D (CypD, PPIF) knockdown with sustained ΔΨm during both Ca(2+) and ROS stress. Biochemical analyses revealed that the PTP is a heterooligomeric complex composed of VDAC, SPG7, and CypD. Silencing or disruption of SPG7-CypD binding prevented Ca(2+)- and ROS-induced ΔΨm depolarization and cell death. This study identifies an ubiquitously expressed IMM integral protein, SPG7, as a core component of the PTP at the OMM and IMM contact site.


Subject(s)
Cyclophilins/metabolism , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Mitochondria/metabolism , Voltage-Dependent Anion Channel 1/metabolism , ATPases Associated with Diverse Cellular Activities , Binding Sites , Calcium/metabolism , Cell Death , Cyclophilins/chemistry , HEK293 Cells , HeLa Cells , Humans , Membrane Potential, Mitochondrial , Metalloendopeptidases/chemistry , Mitochondrial Membranes/metabolism , RNA Interference , Reactive Oxygen Species/metabolism
9.
Proc Natl Acad Sci U S A ; 117(28): 16383-16390, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32601238

ABSTRACT

Calcium uptake by the mitochondrial calcium uniporter coordinates cytosolic signaling events with mitochondrial bioenergetics. During the past decade all protein components of the mitochondrial calcium uniporter have been identified, including MCU, the pore-forming subunit. However, the specific lipid requirements, if any, for the function and formation of this channel complex are currently not known. Here we utilize yeast, which lacks the mitochondrial calcium uniporter, as a model system to address this problem. We use heterologous expression to functionally reconstitute human uniporter machinery both in wild-type yeast as well as in mutants defective in the biosynthesis of phosphatidylethanolamine, phosphatidylcholine, or cardiolipin (CL). We uncover a specific requirement of CL for in vivo reconstituted MCU stability and activity. The CL requirement of MCU is evolutionarily conserved with loss of CL triggering rapid turnover of MCU homologs and impaired calcium transport. Furthermore, we observe reduced abundance and activity of endogenous MCU in mammalian cellular models of Barth syndrome, which is characterized by a partial loss of CL. MCU abundance is also decreased in the cardiac tissue of Barth syndrome patients. Our work raises the hypothesis that impaired mitochondrial calcium transport contributes to the pathogenesis of Barth syndrome, and more generally, showcases the utility of yeast phospholipid mutants in dissecting the phospholipid requirements of ion channel complexes.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Mitochondria/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/metabolism , Animals , Barth Syndrome/genetics , Barth Syndrome/metabolism , Biological Transport , Calcium Channels/chemistry , Calcium Channels/genetics , Cardiolipins/genetics , Cardiolipins/metabolism , Humans , Mice , Mitochondria/chemistry , Mitochondria/genetics , Myoblasts/metabolism , Phospholipids , Protein Stability , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics
10.
EMBO Rep ; 21(10): e48483, 2020 10 05.
Article in English | MEDLINE | ID: mdl-32851774

ABSTRACT

MICU1 is a mitochondrial inner membrane protein that inhibits mitochondrial calcium entry; elevated MICU1 expression is characteristic of many cancers, including ovarian cancer. MICU1 induces both glycolysis and chemoresistance and is associated with poor clinical outcomes. However, there are currently no available interventions to normalize aberrant MICU1 expression. Here, we demonstrate that microRNA-195-5p (miR-195) directly targets the 3' UTR of the MICU1 mRNA and represses MICU1 expression. Additionally, miR-195 is under-expressed in ovarian cancer cell lines, and restoring miR-195 expression reestablishes native MICU1 levels and the associated phenotypes. Stable expression of miR-195 in a human xenograft model of ovarian cancer significantly reduces tumor growth, increases tumor doubling times, and enhances overall survival. In conclusion, miR-195 controls MICU1 levels in ovarian cancer and could be exploited to normalize aberrant MICU1 expression, thus reversing both glycolysis and chemoresistance and consequently improving patient outcomes.


Subject(s)
Cation Transport Proteins , MicroRNAs , Ovarian Neoplasms , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Glycolysis/genetics , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Ovarian Neoplasms/genetics
11.
Am J Physiol Cell Physiol ; 320(4): C465-C482, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33296287

ABSTRACT

Calcium (Ca2+) signaling is critical for cell function and cell survival. Mitochondria play a major role in regulating the intracellular Ca2+ concentration ([Ca2+]i). Mitochondrial Ca2+ uptake is an important determinant of cell fate and governs respiration, mitophagy/autophagy, and the mitochondrial pathway of apoptosis. Mitochondrial Ca2+ uptake occurs via the mitochondrial Ca2+ uniporter (MCU) complex. This review summarizes the present knowledge on the function of MCU complex, regulation of MCU channel, and the role of MCU in Ca2+ homeostasis and human disease pathogenesis. The channel core consists of four MCU subunits and essential MCU regulators (EMRE). Regulatory proteins that interact with them include mitochondrial Ca2+ uptake 1/2 (MICU1/2), MCU dominant-negative ß-subunit (MCUb), MCU regulator 1 (MCUR1), and solute carrier 25A23 (SLC25A23). In addition to these proteins, cardiolipin, a mitochondrial membrane-specific phospholipid, has been shown to interact with the channel core. The dynamic interplay between the core and regulatory proteins modulates MCU channel activity after sensing local changes in [Ca2+]i, reactive oxygen species, and other environmental factors. Here, we highlight the structural details of the human MCU heteromeric assemblies and their known roles in regulating mitochondrial Ca2+ homeostasis. MCU dysfunction has been shown to alter mitochondrial Ca2+ dynamics, in turn eliciting cell apoptosis. Changes in mitochondrial Ca2+ uptake have been implicated in pathological conditions affecting multiple organs, including the heart, skeletal muscle, and brain. However, our structural and functional knowledge of this vital protein complex remains incomplete, and understanding the precise role for MCU-mediated mitochondrial Ca2+ signaling in disease requires further research efforts.


Subject(s)
Calcium Channels/metabolism , Calcium Signaling , Energy Metabolism , Mitochondria/metabolism , Animals , Apoptosis , Calcium Channels/chemistry , Calcium Channels/drug effects , Calcium Channels/genetics , Calcium Signaling/drug effects , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Energy Metabolism/drug effects , Gene Expression Regulation , Humans , Membrane Potential, Mitochondrial , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/pathology , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Molecular Targeted Therapy , Muscular Diseases/drug therapy , Muscular Diseases/genetics , Muscular Diseases/metabolism , Muscular Diseases/pathology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Protein Conformation , Reactive Oxygen Species/metabolism , Structure-Activity Relationship
12.
J Biol Chem ; 295(21): 7249-7260, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32277051

ABSTRACT

Exposure to chronic hyperglycemia because of diabetes mellitus can lead to development and progression of diabetic kidney disease (DKD). We recently reported that reduced superoxide production is associated with mitochondrial dysfunction in the kidneys of mouse models of type 1 DKD. We also demonstrated that humans with DKD have significantly reduced levels of mitochondrion-derived metabolites in their urine. Here we examined renal superoxide production in a type 2 diabetes animal model, the db/db mouse, and the role of a mitochondrial protectant, MTP-131 (also called elamipretide, SS-31, or Bendavia) in restoring renal superoxide production and ameliorating DKD. We found that 18-week-old db/db mice have reduced renal and cardiac superoxide levels, as measured by dihydroethidium oxidation, and increased levels of albuminuria, mesangial matrix accumulation, and urinary H2O2 Administration of MTP-131 significantly inhibited increases in albuminuria, urinary H2O2, and mesangial matrix accumulation in db/db mice and fully preserved levels of renal superoxide production in these mice. MTP-131 also reduced total renal lysocardiolipin and major lysocardiolipin subspecies and preserved lysocardiolipin acyltransferase 1 expression in db/db mice. These results indicate that, in type 2 diabetes, DKD is associated with reduced renal and cardiac superoxide levels and that MTP-131 protects against DKD and preserves physiological superoxide levels, possibly by regulating cardiolipin remodeling.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Diabetic Nephropathies , Mitochondria , Oligopeptides/pharmacology , Superoxides/metabolism , Animals , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/genetics , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Humans , Male , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology
13.
J Mol Cell Cardiol ; 127: 232-245, 2019 02.
Article in English | MEDLINE | ID: mdl-30611795

ABSTRACT

Sepsis is the overwhelming systemic immune response to infection, which can result in multiple organ dysfunction and septic shock. Myocardial dysfunction during sepsis is associated with advanced disease and significantly increased in-hospital mortality. Our group has shown that energetic failure and excess reactive oxygen species (ROS) generation constitute major components of myocardial dysfunction in sepsis. Because ROS production is central to cellular metabolic health, we tested if the synthetic anti-oxidant lignan secoisolariciresinol diglucoside (SDG; LGM2605) would alleviate septic cardiac dysfunction and investigated the underlying mechanism. Using the cecal ligation and puncture (CLP) mouse model of peritonitis-induced sepsis, we observed impairment of cardiac function beginning at 4 h post-CLP surgery. Treatment of mice with LGM2605 (100 mg/kg body weight, i.p.) 6 h post-CLP surgery reduced cardiac ROS accumulation and restored cardiac function. Assessment of mitochondrial respiration (Seahorse XF) in primary cardiomyocytes obtained from adult C57BL/6 mice that had undergone CLP and treatment with LGM2605 showed restored basal and maximal respiration, as well as preserved oxygen consumption rate (OCR) associated with spare capacity. Further analyses aiming to identify the cellular mechanisms that may account for improved cardiac function showed that LGM2605 restored mitochondria abundance, increased mitochondrial calcium uptake and preserved mitochondrial membrane potential. In addition to protecting against cardiac dysfunction, daily treatment with LGM2605 and antibiotic ertapenem (70 mg/kg) protected against CLP-associated mortality and reversed hypothermia when compared against mice receiving ertapenem and saline. Therefore, treatment of septic mice with LGM2605 emerges as a novel pharmacological approach that reduces cardiac ROS accumulation, protects cardiac mitochondrial function, alleviates cardiac dysfunction, and improves survival.


Subject(s)
Butylene Glycols/chemical synthesis , Butylene Glycols/therapeutic use , Cardiomyopathies/complications , Cardiomyopathies/drug therapy , Glucosides/chemical synthesis , Glucosides/therapeutic use , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Sepsis/complications , Sepsis/drug therapy , Animals , Antioxidants/metabolism , Autophagy/drug effects , Biomarkers/metabolism , Butylene Glycols/chemistry , Butylene Glycols/pharmacology , Calcium/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/physiopathology , Cecum/pathology , Cell Line , Cytokines/blood , Disease Models, Animal , Gene Expression Regulation/drug effects , Glucosides/chemistry , Glucosides/pharmacology , Humans , Inflammation Mediators/metabolism , Ligation , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred C57BL , Mitochondria, Heart/drug effects , Myocardium/metabolism , Myocytes, Cardiac/drug effects , NF-kappa B/metabolism , Organelle Biogenesis , Oxidative Stress/drug effects , Oxygen Consumption/drug effects , Punctures , Sepsis/genetics , Sepsis/physiopathology
14.
J Cell Physiol ; 234(9): 15048-15060, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30637731

ABSTRACT

The mechanisms by which Trpm2 channels enhance mitochondrial bioenergetics and protect against oxidative stress-induced cardiac injury remain unclear. Here, the role of proline-rich tyrosine kinase 2 (Pyk2) in Trpm2 signaling is explored. Activation of Trpm2 in adult myocytes with H2 O2 resulted in 10- to 21-fold increases in Pyk2 phosphorylation in wild-type (WT) myocytes which was significantly lower (~40%) in Trpm2 knockout (KO) myocytes. Pyk2 phosphorylation was inhibited (~54%) by the Trpm2 blocker clotrimazole. Buffering Trpm2-mediated Ca2+ increase with 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (BAPTA) resulted in significantly reduced pPyk2 in WT but not in KO myocytes, indicating Ca2+ influx through activated Trpm2 channels phosphorylated Pyk2. Part of phosphorylated Pyk2 translocated from cytosol to mitochondria which has been previously shown to augment mitochondrial Ca2+ uptake and enhance adenosine triphosphate generation. Although Trpm2-mediated Ca2+ influx phosphorylated Ca2+ -calmodulin kinase II (CaMKII), the CaMKII inhibitor KN93 did not significantly affect Pyk2 phosphorylation in H2 O2 -treated WT myocytes. After ischemia/reperfusion (I/R), Pyk2 phosphorylation and its downstream prosurvival signaling molecules (pERK1/2 and pAkt) were significantly lower in KO-I/R when compared with WT-I/R hearts. After hypoxia/reoxygenation, mitochondrial membrane potential was lower and superoxide level was higher in KO myocytes, and were restored to WT values by the mitochondria-targeted superoxide scavenger MitoTempo. Our results suggested that Ca2+ influx via tonically activated Trpm2 phosphorylated Pyk2, part of which translocated to mitochondria, resulting in better mitochondrial bioenergetics to maintain cardiac health. After I/R, Pyk2 activated prosurvival signaling molecules and prevented excessive increases in reactive oxygen species, thereby affording protection from I/R injury.

15.
J Cell Physiol ; 234(4): 4432-4444, 2019 04.
Article in English | MEDLINE | ID: mdl-30256393

ABSTRACT

The pathophysiology of human immunodeficiency virus (HIV)-associated cardiomyopathy remains uncertain. We used HIV-1 transgenic (Tg26) mice to explore mechanisms by which HIV-related proteins impacted on myocyte function. Compared to adult ventricular myocytes isolated from nontransgenic (wild type [WT]) littermates, Tg26 myocytes had similar mitochondrial membrane potential (ΔΨ m ) under normoxic conditions but lower Δ Ψ m after hypoxia/reoxygenation (H/R). In addition, Δ Ψ m in Tg26 myocytes failed to recover after Ca 2+ challenge. Functionally, mitochondrial Ca 2+ uptake was severely impaired in Tg26 myocytes. Basal and maximal oxygen consumption rates (OCR) were lower in normoxic Tg26 myocytes, and further reduced after H/R. Complex I subunit and ATP levels were lower in Tg26 hearts. Post-H/R, mitochondrial superoxide (O 2•- ) levels were higher in Tg26 compared to WT myocytes. Overexpression of B-cell lymphoma 2-associated athanogene 3 (BAG3) reduced O 2•- levels in hypoxic WT and Tg26 myocytes back to normal. Under normoxic conditions, single myocyte contraction dynamics were similar between WT and Tg26 myocytes. Post-H/R and in the presence of isoproterenol, myocyte contraction amplitudes were lower in Tg26 myocytes. BAG3 overexpression restored Tg26 myocyte contraction amplitudes to those measured in WT myocytes post-H/R. Coimmunoprecipitation experiments demonstrated physical association of BAG3 and the HIV protein Tat. We conclude: (a) Under basal conditions, mitochondrial Ca 2+ uptake, OCR, and ATP levels were lower in Tg26 myocytes; (b) post-H/R, Δ Ψ m was lower, mitochondrial O 2•- levels were higher, and contraction amplitudes were reduced in Tg26 myocytes; and (c) BAG3 overexpression decreased O 2•- levels and restored contraction amplitudes to normal in Tg26 myocytes post-H/R in the presence of isoproterenol.


Subject(s)
Cardiomyopathies/metabolism , Energy Metabolism , HIV Infections/complications , HIV-1/genetics , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/physiopathology , Cardiomyopathies/virology , Cell Hypoxia , Cells, Cultured , Disease Models, Animal , HIV Infections/virology , Membrane Potential, Mitochondrial , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria, Heart/virology , Myocardial Contraction , Myocytes, Cardiac/virology , Oxidation-Reduction , Oxidative Stress , Oxygen Consumption , Reactive Oxygen Species/metabolism , Signal Transduction , Ventricular Function, Left
16.
Am J Physiol Lung Cell Mol Physiol ; 317(6): L791-L804, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31313618

ABSTRACT

Pulmonary emphysema is characterized by alveolar type II (ATII) cell death, destruction of alveolar wall septa, and irreversible airflow limitation. Cigarette smoke induces oxidative stress and is the main risk factor for this disease development. ATII cells isolated from nonsmokers, smokers, and patients with emphysema were used for this study. ATII cell apoptosis in individuals with this disease was detected. DJ-1 and S100A8 have cytoprotective functions against oxidative stress-induced cell injury. Reduced DJ-1 and S100A8 interaction was found in ATII cells in patients with emphysema. The molecular function of S100A8 was determined by an analysis of the oxidation status of its cysteine residues using chemoselective probes. Decreased S100A8 sulfination was observed in emphysema patients. In addition, its lower levels correlated with higher cell apoptosis induced by cigarette smoke extract in vitro. Cysteine at position 106 within DJ-1 is a central redox-sensitive residue. DJ-1 C106A mutant construct abolished the cytoprotective activity of DJ-1 against cell injury induced by cigarette smoke extract. Furthermore, a molecular and complementary relationship between DJ-1 and S100A8 was detected using gain- and loss-of-function studies. DJ-1 knockdown sensitized cells to apoptosis induced by cigarette smoke extract, and S100A8 overexpression provided cytoprotection in the absence of DJ-1. DJ-1 knockout mice were more susceptible to ATII cell apoptosis induced by cigarette smoke compared with wild-type mice. Our results indicate that the impairment of DJ-1 and S100A8 function may contribute to cigarette smoke-induced ATII cell injury and emphysema pathogenesis.


Subject(s)
Alveolar Epithelial Cells/pathology , Apoptosis , Calgranulin A/metabolism , Protein Deglycase DJ-1/metabolism , Pulmonary Alveoli/pathology , Pulmonary Emphysema/pathology , Aged , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Animals , Calgranulin A/genetics , Cytoprotection , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Oxidative Stress , Protein Deglycase DJ-1/genetics , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , Pulmonary Emphysema/genetics , Pulmonary Emphysema/metabolism , Smoke/adverse effects
17.
Am J Physiol Lung Cell Mol Physiol ; 317(4): L475-L485, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31313616

ABSTRACT

The alveolus participates in gas exchange, which can be impaired by environmental factors and toxins. There is an increase in using electronic cigarettes (e-cigarettes); however, their effect on human primary alveolar epithelial cells is unknown. Human lungs were obtained from nonsmoker organ donors to isolate alveolar type II (ATII) cells. ATII cells produce and secrete pulmonary surfactant and restore the epithelium after damage, and mitochondrial function is important for their metabolism. Our data indicate that human ATII cell exposure to e-cigarette aerosol increased IL-8 levels and induced DNA damage and apoptosis. We also studied the cytoprotective effect of DJ-1 against ATII cell injury. DJ-1 knockdown in human primary ATII cells sensitized cells to mitochondrial dysfunction as detected by high mitochondrial superoxide production, decreased mitochondrial membrane potential, and calcium elevation. DJ-1 knockout (KO) mice were more susceptible to ATII cell apoptosis and lung injury induced by e-cigarette aerosol compared with wild-type mice. Regulation of the oxidative phosphorylation (OXPHOS) is important for mitochondrial function and protection against oxidative stress. Major subunits of the OXPHOS system are encoded by both nuclear and mitochondrial DNA. We found dysregulation of OXPHOS complexes in DJ-1 KO mice after exposure to e-cigarette aerosol, which could disrupt the nuclear/mitochondrial stoichiometry, resulting in mitochondrial dysfunction. Together, our results indicate that DJ-1 deficiency sensitizes ATII cells to damage induced by e-cigarette aerosol leading to lung injury.


Subject(s)
Alveolar Epithelial Cells/drug effects , Electronic Nicotine Delivery Systems , Interleukin-8/genetics , Nicotine/pharmacology , Protein Deglycase DJ-1/genetics , Aerosols , Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Calcium/metabolism , DNA Damage , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Interleukin-8/metabolism , Membrane Potential, Mitochondrial , Mice , Mice, Knockout , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects , Primary Cell Culture , Protein Deglycase DJ-1/deficiency , Protein Deglycase DJ-1/metabolism , Pulmonary Alveoli/cytology , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , Superoxides/metabolism
18.
Blood ; 130(5): 567-580, 2017 08 03.
Article in English | MEDLINE | ID: mdl-28500171

ABSTRACT

Platelet-derived microparticles (PMPs) are associated with enhancement of metastasis and poor cancer outcomes. Circulating PMPs transfer platelet microRNAs (miRNAs) to vascular cells. Solid tumor vasculature is highly permeable, allowing the possibility of PMP-tumor cell interaction. Here, we show that PMPs infiltrate solid tumors in humans and mice and transfer platelet-derived RNA, including miRNAs, to tumor cells in vivo and in vitro, resulting in tumor cell apoptosis. MiR-24 was a major species in this transfer. PMP transfusion inhibited growth of both lung and colon carcinoma ectopic tumors, whereas blockade of miR-24 in tumor cells accelerated tumor growth in vivo, and prevented tumor growth inhibition by PMPs. Conversely, Par4-deleted mice, which had reduced circulating microparticles (MPs), supported accelerated tumor growth which was halted by PMP transfusion. PMP targeting was associated with tumor cell apoptosis in vivo. We identified direct RNA targets of platelet-derived miR-24 in tumor cells, which included mitochondrial mt-Nd2, and Snora75, a noncoding small nucleolar RNA. These RNAs were suppressed in PMP-treated tumor cells, resulting in mitochondrial dysfunction and growth inhibition, in an miR-24-dependent manner. Thus, platelet-derived miRNAs transfer in vivo to tumor cells in solid tumors via infiltrating MPs, regulate tumor cell gene expression, and modulate tumor progression. These findings provide novel insight into mechanisms of horizontal RNA transfer and add multiple layers to the regulatory roles of miRNAs and PMPs in tumor progression. Plasma MP-mediated transfer of regulatory RNAs and modulation of gene expression may be a common feature with important outcomes in contexts of enhanced vascular permeability.


Subject(s)
Blood Platelets/metabolism , Cell-Derived Microparticles/metabolism , Colonic Neoplasms/metabolism , Lung Neoplasms/metabolism , MicroRNAs/metabolism , Animals , Cell-Derived Microparticles/transplantation , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Mice , NADH Dehydrogenase/genetics , NADH Dehydrogenase/metabolism , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Receptors, Proteinase-Activated
19.
Am J Physiol Cell Physiol ; 315(4): C571-C586, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30020827

ABSTRACT

Transient receptor potential melastatin channel subfamily member 2 (TRPM2) has an essential function in cell survival and is highly expressed in many cancers. Inhibition of TRPM2 in neuroblastoma by depletion with CRISPR technology or expression of dominant negative TRPM2-S has been shown to significantly reduce cell viability. Here, the role of proline-rich tyrosine kinase 2 (Pyk2) in TRPM2 modulation of neuroblastoma viability was explored. In TRPM2-depleted cells, phosphorylation and expression of Pyk2 and cAMP-responsive element-binding protein (CREB), a downstream target, were significantly reduced after application of the chemotherapeutic agent doxorubicin. Overexpression of wild-type Pyk2 rescued cell viability. Reduction of Pyk2 expression with shRNA decreased cell viability and CREB phosphorylation and expression, demonstrating Pyk2 modulates CREB activation. TRPM2 depletion impaired phosphorylation of Src, an activator of Pyk2, and this may be a mechanism to reduce Pyk2 phosphorylation. TRPM2 inhibition was previously demonstrated to decrease mitochondrial function. Here, CREB, Pyk2, and phosphorylated Src were reduced in mitochondria of TRPM2-depleted cells, consistent with their role in modulating expression and activation of mitochondrial proteins. Phosphorylated Src and phosphorylated and total CREB were reduced in TRPM2-depleted nuclei. Expression and function of mitochondrial calcium uniporter (MCU), a target of phosphorylated Pyk2 and CREB, were significantly reduced. Wild-type TRPM2 but not Ca2+-impermeable mutant E960D reconstituted phosphorylation and expression of Pyk2 and CREB in TRPM2-depleted cells exposed to doxorubicin. Results demonstrate that TRPM2 expression protects the viability of neuroblastoma through Src, Pyk2, CREB, and MCU activation, which play key roles in maintaining mitochondrial function and cellular bioenergetics.


Subject(s)
Calcium Channels/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Focal Adhesion Kinase 2/genetics , Neuroblastoma/drug therapy , TRPM Cation Channels/genetics , Calcium Signaling/genetics , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mitochondria/genetics , Neuroblastoma/genetics , Neuroblastoma/pathology , Phosphorylation , Reactive Oxygen Species/metabolism , src-Family Kinases/genetics
20.
J Cell Physiol ; 233(2): 748-758, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28493473

ABSTRACT

Cardiovascular disease remains a leading cause of morbidity and mortality in HIV-positive patients, even in those whose viral loads are well controlled with antiretroviral therapy. However, the underlying molecular events responsible for the development of cardiac disease in the setting of HIV remain unknown. The HIV-encoded Tat protein plays a critical role in the activation of HIV gene expression and profoundly impacts homeostasis in both HIV-infected cells and uninfected cells that have taken up released Tat via a bystander effect. Since cardiomyocyte function, including excitation-contraction coupling, greatly depends on energy provided by the mitochondria, in this study, we performed a series of experiments to assess the impact of Tat on mitochondrial function and bioenergetics pathways in a primary cell culture model derived from neonatal rat ventricular cardiomyocytes (NRVCs). Our results show that the presence of Tat in cardiomyocytes is accompanied by a decrease in oxidative phosphorylation, a decline in the levels of ATP, and an accumulation of reactive oxygen species (ROS). Tat impairs the uptake of mitochondrial Ca2+ ([Ca2+ ]m ) and the electrophysiological activity of cardiomyocytes. Tat also affects the protein clearance pathway and autophagy in cardiomyocytes under stress due to hypoxia-reoxygenation conditions. A reduction in the level of ubiquitin along with dysregulated degradation of autophagy proteins including SQSTM1/p62 and a reduction of LC3 II were detected in cardiomyocytes harboring Tat. These results suggest that, by targeting mitochondria and protein quality control, Tat significantly impacts bioenergetics and autophagy resulting in dysregulation of cardiomyocyte health and homeostasis.


Subject(s)
Energy Metabolism , HIV-1/metabolism , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , tat Gene Products, Human Immunodeficiency Virus/metabolism , Adenosine Triphosphate/metabolism , Animals , Apoptosis , Autophagy , Calcium/metabolism , Calcium Channels/metabolism , Cell Hypoxia , Cells, Cultured , Host-Pathogen Interactions , Membrane Potentials , Microtubule-Associated Proteins/metabolism , Mitochondria, Heart/virology , Mitophagy , Myocytes, Cardiac/virology , Oxidative Phosphorylation , Primary Cell Culture , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Sequestosome-1 Protein/metabolism , Signal Transduction , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL