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










Publication year range
1.
Mol Cell Proteomics ; 22(5): 100543, 2023 05.
Article in English | MEDLINE | ID: mdl-37030595

ABSTRACT

Excitotoxicity, a neuronal death process in neurological disorders such as stroke, is initiated by the overstimulation of ionotropic glutamate receptors. Although dysregulation of proteolytic signaling networks is critical for excitotoxicity, the identity of affected proteins and mechanisms by which they induce neuronal cell death remain unclear. To address this, we used quantitative N-terminomics to identify proteins modified by proteolysis in neurons undergoing excitotoxic cell death. We found that most proteolytically processed proteins in excitotoxic neurons are likely substrates of calpains, including key synaptic regulatory proteins such as CRMP2, doublecortin-like kinase I, Src tyrosine kinase and calmodulin-dependent protein kinase IIß (CaMKIIß). Critically, calpain-catalyzed proteolytic processing of these proteins generates stable truncated fragments with altered activities that potentially contribute to neuronal death by perturbing synaptic organization and function. Blocking calpain-mediated proteolysis of one of these proteins, Src, protected against neuronal loss in a rat model of neurotoxicity. Extrapolation of our N-terminomic results led to the discovery that CaMKIIα, an isoform of CaMKIIß, undergoes differential processing in mouse brains under physiological conditions and during ischemic stroke. In summary, by identifying the neuronal proteins undergoing proteolysis during excitotoxicity, our findings offer new insights into excitotoxic neuronal death mechanisms and reveal potential neuroprotective targets for neurological disorders.


Subject(s)
Cell Death , Neurons , Synapses , Animals , Male , Mice , Rats , Calpain/metabolism , Cells, Cultured , Cysteine Proteinase Inhibitors/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neurons/pathology , Neurons/physiology , Neuroprotection , Proteome/analysis , Rats, Wistar , Stroke/pathology , Synapses/pathology , Synapses/physiology
2.
Cell Rep ; 41(12): 111862, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36543129

ABSTRACT

AMP-activated protein kinase (AMPK) is a master regulator of cellular energy homeostasis and a therapeutic target for metabolic diseases. Co/post-translational N-myristoylation of glycine-2 (Gly2) of the AMPK ß subunit has been suggested to regulate the distribution of the kinase between the cytosol and membranes through a "myristoyl switch" mechanism. However, the relevance of AMPK myristoylation for metabolic signaling in cells and in vivo is unclear. Here, we generated knockin mice with a Gly2-to-alanine point mutation of AMPKß1 (ß1-G2A). We demonstrate that non-myristoylated AMPKß1 has reduced stability but is associated with increased kinase activity and phosphorylation of the Thr172 activation site in the AMPK α subunit. Using proximity ligation assays, we show that loss of ß1 myristoylation impedes colocalization of the phosphatase PPM1A/B with AMPK in cells. Mice carrying the ß1-G2A mutation have improved metabolic health with reduced adiposity, hepatic lipid accumulation, and insulin resistance under conditions of high-fat diet-induced obesity.


Subject(s)
AMP-Activated Protein Kinases , Fatty Liver , Animals , Mice , Phosphorylation , AMP-Activated Protein Kinases/metabolism , Diet, High-Fat , Protein Processing, Post-Translational , Obesity , Myristic Acid/metabolism , Mice, Inbred C57BL , Protein Phosphatase 2C/metabolism
3.
Cell Rep ; 38(7): 110365, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35172150

ABSTRACT

AMP-activated protein kinase (AMPK) and mechanistic target of rapamycin complex 1 (mTORC1) are metabolic kinases that co-ordinate nutrient supply with cell growth. AMPK negatively regulates mTORC1, and mTORC1 reciprocally phosphorylates S345/7 in both AMPK α-isoforms. We report that genetic or torin1-induced loss of α2-S345 phosphorylation relieves suppression of AMPK signaling; however, the regulatory effect does not translate to α1-S347 in HEK293T or MEF cells. Dephosphorylation of α2-S345, but not α1-S347, transiently targets AMPK to lysosomes, a cellular site for activation by LKB1. By mass spectrometry, we find that α2-S345 is basally phosphorylated at 2.5-fold higher stoichiometry than α1-S347 in HEK293T cells and, unlike α1, phosphorylation is partially retained after prolonged mTORC1 inhibition. Loss of α2-S345 phosphorylation in endogenous AMPK fails to sustain growth of MEFs under amino acid starvation conditions. These findings uncover an α2-specific mechanism by which AMPK can be activated at lysosomes in the absence of changes in cellular energy.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Lysosomes/metabolism , AMP-Activated Protein Kinase Kinases/metabolism , Amino Acid Sequence , Animals , Enzyme Activation , Female , Glycogen Synthase Kinase 3/chemistry , Glycogen Synthase Kinase 3/metabolism , HEK293 Cells , HeLa Cells , Humans , Isoenzymes/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice, Inbred C57BL , Phosphorylation , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism
4.
J Biol Chem ; 295(48): 16239-16250, 2020 11 27.
Article in English | MEDLINE | ID: mdl-32913128

ABSTRACT

The calcium-calmodulin-dependent protein kinase kinase-2 (CaMKK2) is a key regulator of cellular and whole-body energy metabolism. It is known to be activated by increases in intracellular Ca2+, but the mechanisms by which it is inactivated are less clear. CaMKK2 inhibition protects against prostate cancer, hepatocellular carcinoma, and metabolic derangements induced by a high-fat diet; therefore, elucidating the intracellular mechanisms that inactivate CaMKK2 has important therapeutic implications. Here we show that stimulation of cAMP-dependent protein kinase A (PKA) signaling in cells inactivates CaMKK2 by phosphorylation of three conserved serine residues. PKA-dependent phosphorylation of Ser495 directly impairs calcium-calmodulin activation, whereas phosphorylation of Ser100 and Ser511 mediate recruitment of 14-3-3 adaptor proteins that hold CaMKK2 in the inactivated state by preventing dephosphorylation of phospho-Ser495 We also report the crystal structure of 14-3-3ζ bound to a synthetic diphosphorylated peptide that reveals how the canonical (Ser511) and noncanonical (Ser100) 14-3-3 consensus sites on CaMKK2 cooperate to bind 14-3-3 proteins. Our findings provide detailed molecular insights into how cAMP-PKA signaling inactivates CaMKK2 and reveals a pathway to inhibit CaMKK2 with potential for treating human diseases.


Subject(s)
14-3-3 Proteins/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Signal Transduction , 14-3-3 Proteins/genetics , Animals , COS Cells , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Cell Line, Tumor , Chlorocebus aethiops , Cyclic AMP-Dependent Protein Kinases/genetics , Enzyme Activation , Humans
5.
Nat Metab ; 2(9): 873-881, 2020 09.
Article in English | MEDLINE | ID: mdl-32719536

ABSTRACT

Long-chain fatty acids (LCFAs) play important roles in cellular energy metabolism, acting as both an important energy source and signalling molecules1. LCFA-CoA esters promote their own oxidation by acting as allosteric inhibitors of acetyl-CoA carboxylase, which reduces the production of malonyl-CoA and relieves inhibition of carnitine palmitoyl-transferase 1, thereby promoting LCFA-CoA transport into the mitochondria for ß-oxidation2-6. Here we report a new level of regulation wherein LCFA-CoA esters per se allosterically activate AMP-activated protein kinase (AMPK) ß1-containing isoforms to increase fatty acid oxidation through phosphorylation of acetyl-CoA carboxylase. Activation of AMPK by LCFA-CoA esters requires the allosteric drug and metabolite site formed between the α-subunit kinase domain and the ß-subunit. ß1 subunit mutations that inhibit AMPK activation by the small-molecule activator A769662, which binds to the allosteric drug and metabolite site, also inhibit activation by LCFA-CoAs. Thus, LCFA-CoA metabolites act as direct endogenous AMPK ß1-selective activators and promote LCFA oxidation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Acyl Coenzyme A/physiology , Allosteric Regulation/physiology , AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/genetics , Animals , Biphenyl Compounds , Catalytic Domain , Esters , Isoenzymes/chemistry , Isoenzymes/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Molecular , Mutation/genetics , Oxidation-Reduction , Palmitoyl Coenzyme A/metabolism , Phosphorylation , Pyrones/pharmacology , Thiophenes/pharmacology
6.
Bipolar Disord ; 22(8): 841-848, 2020 12.
Article in English | MEDLINE | ID: mdl-32216002

ABSTRACT

OBJECTIVES: Loss-of-function mutations in the gene encoding the calcium-calmodulin (Ca2+ -CaM)-dependent protein kinase kinase-2 (CaMKK2) enzyme are linked to bipolar disorder. Recently, a de novo arginine to cysteine (R311C) mutation in CaMKK2 was identified from a whole exome sequencing study of bipolar patients and their unaffected parents. The aim of the present study was to determine the functional consequences of the R311C mutation on CaMKK2 activity and regulation by Ca2+ -CaM. METHODS: The effects of the R311C mutation on CaMKK2 activity and Ca2+ -CaM activation were examined using a radiolabeled adenosine triphosphate (ATP) kinase assay. We performed immunoblot analysis to determine whether the R311C mutation impacts threonine-85 (T85) autophosphorylation, an activating phosphorylation site on CaMKK2 that has also been implicated in bipolar disorder. We also expressed the R311C mutant in CaMKK2 knockout HAP1 cells and used immunoblot analysis and an MTS reduction assay to study its effects on Ca2+ -dependent downstream signaling and cell viability, respectively. RESULTS: The R311C mutation maps to the conserved HRD motif within the catalytic loop of CaMKK2 and caused a marked reduction in kinase activity and Ca2+ -CaM activation. The R311C mutation virtually abolished T85 autophosphorylation in response to Ca2+ -CaM and exerted a dominant-negative effect in cells as it impaired the ability of wild-type CaMKK2 to initiate downstream signaling and maintain cell viability. CONCLUSIONS: The highly disruptive, loss-of-function impact of the de novo R311C mutation in human CaMKK2 provides a compelling functional rationale for being considered a potential rare monogenic cause of bipolar disorder.


Subject(s)
Bipolar Disorder/genetics , Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Calcium/metabolism , Calmodulin/metabolism , Bipolar Disorder/diagnosis , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Calmodulin/genetics , Genetic Variation , Humans , Mutation , Phosphorylation , Signal Transduction/physiology
7.
Cell Commun Signal ; 18(1): 25, 2020 02 13.
Article in English | MEDLINE | ID: mdl-32054489

ABSTRACT

BACKGROUND: Eukaryotic elongation factor-2 kinase (eEF2K) is a Ca 2+ /calmodulin (CaM)-dependent protein kinase that inhibits protein synthesis. However, the role of eEF2K in cancer development was reported paradoxically and remains to be elucidated. METHODS: Herein, A549 cells with eEF2K depletion or overexpression by stably transfected lentivirus plasmids were used in vitro and in vivo study. MTT and colony assays were used to detect cell proliferation and growth. Extracellular glucose and lactate concentration were measured using test kit. Immunoblot and co-immunoprecipitation assays were used to examine the molecular biology changes and molecular interaction in these cells. LC-MS/MS analysis and [γ- 32 P] ATP kinase assay were used to identify combining protein and phosphorylation site. Nude mice was utilized to study the correlation of eEF2K and tumor growth in vivo. RESULTS: We demonstrated that eEF2K inhibited lung cancer cells proliferation and affected the inhibitory effects of EGFR inhibitor gefitinib. Mechanistically, we showed that eEF2K formed a complex with PKM2 and STAT3, thereby phosphorylated PKM2 at T129, leading to reduced dimerization of PKM2. Subsequently, PKM2 impeded STAT3 phosphorylation and STAT3-dependent c-Myc expression. eEF2K depletion promoted the nuclear translocation of PKM2 and increased aerobic glycolysis reflected by increased lactate secretion and glucose. CONCLUSIONS: Our findings define a novel mechanism underlying the regulation of cancer cell proliferation by eEF2K independent of its role in protein synthesis, disclosing the diverse roles of eEF2K in cell biology, which lays foundation for the development of new anticancer therapeutic strategies.


Subject(s)
Carrier Proteins/metabolism , Elongation Factor 2 Kinase/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Membrane Proteins/metabolism , STAT3 Transcription Factor/metabolism , Thyroid Hormones/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Gefitinib/pharmacology , HEK293 Cells , Humans , Mice, Nude , Phosphorylation/drug effects , Phosphothreonine/metabolism , Protein Multimerization/drug effects , Proto-Oncogene Proteins c-myc/metabolism , Tumor Burden/drug effects , Thyroid Hormone-Binding Proteins
8.
Nat Metab ; 2(1): 41-49, 2020 01.
Article in English | MEDLINE | ID: mdl-31993556

ABSTRACT

Central to cellular metabolism and cell proliferation are highly conserved signalling pathways controlled by mammalian target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK)1,2, dysregulation of which are implicated in pathogenesis of major human diseases such as cancer and type 2 diabetes. AMPK pathways leading to reduced cell proliferation are well established and, in part, act through inhibition of TOR complex-1 (TORC1) activity. Here we demonstrate reciprocal regulation, specifically that TORC1 directly down-regulates AMPK signalling by phosphorylating the evolutionarily conserved residue Ser367 in the fission yeast AMPK catalytic subunit Ssp2, and AMPK α1Ser347/α2Ser345 in the mammalian homologs, which is associated with reduced phosphorylation of activation loop Thr172. Genetic or pharmacological inhibition of TORC1 signalling led to AMPK activation in the absence of increased AMP:ATP ratios; under nutrient stress conditions this was associated with growth limitation in both yeast and human cell cultures. Our findings reveal fundamental, bi-directional regulation between two major metabolic signalling networks and uncover new opportunity for cancer treatment strategies aimed at suppressing cell proliferation in the nutrient-poor tumor microenvironment.


Subject(s)
Adenylate Kinase/antagonists & inhibitors , Cell Proliferation/physiology , Mechanistic Target of Rapamycin Complex 1/physiology , Nutrients/metabolism , Stress, Physiological , Adenylate Kinase/chemistry , Adenylate Kinase/metabolism , Catalytic Domain , Diabetes Mellitus, Type 2/metabolism , Down-Regulation , Enzyme Activation , Humans , Mechanistic Target of Rapamycin Complex 1/drug effects , Neoplasms/metabolism , Phosphorylation , Schizosaccharomyces/metabolism , Schizosaccharomyces pombe Proteins/metabolism , Signal Transduction/physiology
9.
J Neurochem ; 153(1): 10-32, 2020 04.
Article in English | MEDLINE | ID: mdl-31630412

ABSTRACT

Perception of our environment entirely depends on the close interaction between the central and peripheral nervous system. In order to communicate each other, both systems must develop in parallel and in coordination. During development, axonal projections from the CNS as well as the PNS must extend over large distances to reach their appropriate target cells. To do so, they read and follow a series of axon guidance molecules. Interestingly, while these molecules play critical roles in guiding developing axons, they have also been shown to be critical in other major neurodevelopmental processes, such as the migration of cortical progenitors. Currently, a major hurdle for brain repair after injury or neurodegeneration is the absence of axonal regeneration in the mammalian CNS. By contrasts, PNS axons can regenerate. Many hypotheses have been put forward to explain this paradox but recent studies suggest that hacking neurodevelopmental mechanisms may be the key to promote CNS regeneration. Here we provide a seminar report written by trainees attending the second Flagship school held in Alpbach, Austria in September 2018 organized by the International Society for Neurochemistry (ISN) together with the Journal of Neurochemistry (JCN). This advanced school has brought together leaders in the fields of neurodevelopment and regeneration in order to discuss major keystones and future challenges in these respective fields.


Subject(s)
Axon Guidance/physiology , Axons/physiology , Brain/ultrastructure , Animals , Axons/ultrastructure , Brain/growth & development , Brain/physiology , Humans , Nerve Regeneration , Optic Chiasm/growth & development , Peripheral Nervous System/growth & development , Peripheral Nervous System/physiology , Spinal Cord/growth & development , Spinal Cord/physiology , Spinal Cord/ultrastructure
10.
Cell Death Dis ; 10(3): 213, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30824683

ABSTRACT

Excitotoxicity, caused by overstimulation or dysregulation of ionotropic glutamate receptors (iGluRs), is a pathological process directing neuronal death in many neurological disorders. The aberrantly stimulated iGluRs direct massive influx of calcium ions into the affected neurons, leading to changes in expression and phosphorylation of specific proteins to modulate their functions and direct their participation in the signalling pathways that induce excitotoxic neuronal death. To define these pathways, we used quantitative proteomic approaches to identify these neuronal proteins (referred to as the changed proteins) and determine how their expression and/or phosphorylation dynamically changed in association with excitotoxic cell death. Our data, available in ProteomeXchange with identifier PXD008353, identified over 100 changed proteins exhibiting significant alterations in abundance and/or phosphorylation levels at different time points (5-240 min) in neurons after glutamate overstimulation. Bioinformatic analyses predicted that many of them are components of signalling networks directing defective neuronal morphology and functions. Among them, the well-known neuronal survival regulators including mitogen-activated protein kinases Erk1/2, glycogen synthase kinase 3 (GSK3) and microtubule-associated protein (Tau), were selected for validation by biochemical approaches, which confirmed the findings of the proteomic analysis. Bioinformatic analysis predicted Protein Kinase B (Akt), c-Jun kinase (JNK), cyclin-dependent protein kinase 5 (Cdk5), MAP kinase kinase (MEK), Casein kinase 2 (CK2), Rho-activated protein kinase (Rock) and Serum/glucocorticoid-regulated kinase 1 (SGK1) as the potential upstream kinases phosphorylating some of the changed proteins. Further biochemical investigation confirmed the predictions of sustained changes of the activation states of neuronal Akt and CK2 in excitotoxicity. Thus, future investigation to define the signalling pathways directing the dynamic alterations in abundance and phosphorylation of the identified changed neuronal proteins will help elucidate the molecular mechanism of neuronal death in excitotoxicity.


Subject(s)
Glutamic Acid/toxicity , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Signal Transduction/drug effects , Animals , Casein Kinase II/chemistry , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cell Death , Cell Survival , Cells, Cultured , Chromatography, Liquid , Computational Biology , Glutamic Acid/metabolism , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Mice , Mitogen-Activated Protein Kinase 1/chemistry , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/chemistry , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Nerve Tissue Proteins/chemistry , Neurons/cytology , Neurons/pathology , Phosphorylation , Proteomics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, trkA/genetics , Receptor, trkA/metabolism , Signal Transduction/genetics , Software , Tandem Mass Spectrometry , tau Proteins/chemistry , tau Proteins/genetics , tau Proteins/metabolism
11.
J Biol Chem ; 293(23): 8874-8885, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29695504

ABSTRACT

Inhibition of the metabolic regulator AMP-activated protein kinase (AMPK) is increasingly being investigated for its therapeutic potential in diseases where AMPK hyperactivity results in poor prognoses, as in established cancers and neurodegeneration. However, AMPK-inhibitory tool compounds are largely limited to compound C, which has a poor selectivity profile. Here we identify the pyrimidine derivative SBI-0206965 as a direct AMPK inhibitor. SBI-0206965 inhibits AMPK with 40-fold greater potency and markedly lower kinase promiscuity than compound C and inhibits cellular AMPK signaling. Biochemical characterization reveals that SBI-0206965 is a mixed-type inhibitor. A co-crystal structure of the AMPK kinase domain/SBI-0206965 complex shows that the drug occupies a pocket that partially overlaps the ATP active site in a type IIb inhibitor manner. SBI-0206965 has utility as a tool compound for investigating physiological roles for AMPK and provides fresh impetus to small-molecule AMPK inhibitor therapeutic development.


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , Benzamides/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/metabolism , Animals , Benzamides/chemistry , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans , Molecular Docking Simulation , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemistry
12.
Cell Chem Biol ; 25(6): 728-737.e9, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29657085

ABSTRACT

The AMP-activated protein kinase (AMPK) αßγ heterotrimer regulates cellular energy homeostasis with tissue-specific isoform distribution. Small-molecule activation of skeletal muscle α2ß2 AMPK complexes may prove a valuable treatment strategy for type 2 diabetes and insulin resistance. Herein, we report the small-molecule SC4 is a potent, direct AMPK activator that preferentially activates α2 complexes and stimulates skeletal muscle glucose uptake. In parallel with the term secretagog, we propose "importagog" to define a substance that induces or augments cellular uptake of another substance. Three-dimensional structures of the glucose importagog SC4 bound to activated α2ß2γ1 and α2ß1γ1 complexes reveal binding determinants, in particular a key interaction between the SC4 imidazopyridine 4'-nitrogen and ß2-Asp111, which provide a design paradigm for ß2-AMPK therapeutics. The α2ß2γ1/SC4 structure reveals an interaction between a ß2 N-terminal α helix and the α2 autoinhibitory domain. Our results provide a structure-function guide to accelerate development of potent, but importantly tissue-specific, ß2-AMPK therapeutics.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Benzoates/pharmacology , Glucose/metabolism , Muscle, Skeletal/drug effects , Pyridines/pharmacology , Small Molecule Libraries/pharmacology , Animals , Benzoates/chemical synthesis , Benzoates/chemistry , COS Cells , Cell Line , Chlorocebus aethiops , Crystallography, X-Ray , Enzyme Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Molecular Structure , Muscle, Skeletal/metabolism , Pyridines/chemical synthesis , Pyridines/chemistry , Rats , Rats, Wistar , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry
13.
Cell Death Discov ; 4: 39, 2018 Dec.
Article in English | MEDLINE | ID: mdl-29531836

ABSTRACT

Human induced pluripotent stem cells (iPSCs) are a valuable tool for studying the cardiac developmental process in vitro, and cardiomyocytes derived from iPSCs are a putative cell source for personalized medicine. Changes in mitochondrial morphology have been shown to occur during cellular reprogramming and pluripotent stem cell differentiation. However, the relationships between mitochondrial dynamics and cardiac mesoderm commitment of iPSCs remain unclear. Here we demonstrate that changes in mitochondrial morphology from a small granular fragmented phenotype in pluripotent stem cells to a filamentous reticular elongated network in differentiated cardiomyocytes are required for cardiac mesodermal differentiation. Genetic and pharmacological inhibition of the mitochondrial fission protein, Drp1, by either small interfering RNA or Mdivi-1, respectively, increased cardiac mesoderm gene expression in iPSCs. Treatment of iPSCs with Mdivi-1 during embryoid body formation significantly increased the percentage of beating embryoid bodies and expression of cardiac-specific genes. Furthermore, Drp1 gene silencing was accompanied by increased mitochondrial respiration and decreased aerobic glycolysis. Our findings demonstrate that shifting the balance of mitochondrial morphology toward fusion by inhibition of Drp1 promoted cardiac differentiation of human iPSCs with a metabolic shift from glycolysis towards oxidative phosphorylation. These findings suggest that Drp1 may represent a new molecular target for future development of strategies to promote the differentiation of human iPSCs into cardiac lineages for patient-specific cardiac regenerative medicine.

14.
Stem Cells Dev ; 26(24): 1771-1780, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29054138

ABSTRACT

Cardiac stem cell (CSC) therapy is a promising approach to treat ischemic heart disease. However, the poor survival of transplanted stem cells in the ischemic myocardium has been a major impediment in achieving an effective cell-based therapy against myocardial infarction. Inhibiting mitochondrial fission has been shown to promote survival of several cell types. However, the role of mitochondrial morphology in survival of human CSC remains unknown. In this study, we investigated whether mitochondrial division inhibitor-1 (Mdivi-1), an inhibitor of mitochondrial fission protein dynamin-related protein-1 (Drp1), can improve survival of a novel population of human W8B2+ CSCs in hydrogen peroxide (H2O2)-induced oxidative stress and simulated ischemia-reperfusion injury models. Mdivi-1 significantly reduced H2O2-induced cell death in a dose-dependent manner. This cytoprotective effect was accompanied by an increased proportion of cells with tubular mitochondria, but independent of mitochondrial membrane potential recovery and reduction of mitochondrial superoxide production. In simulated ischemia-reperfusion injury model, Mdivi-1 given as a pretreatment or throughout ischemia-reperfusion injury significantly reduced cell death. However, the cytoprotective effect of Mdivi-1 was not observed when given at reperfusion. Moreover, the cytoprotective effect of Mdivi-1 in the simulated ischemia-reperfusion injury model was not accompanied by changes in mitochondrial morphology, mitochondrial membrane potential, or mitochondrial reactive oxygen species production. Mdivi-1 also did not affect mitochondrial bioenergetics of intact W8B2+ CSCs. Taken together, these experiments demonstrated that Mdivi-1 treatment of human W8B2+ CSCs enhances their survival and can be employed to improve therapeutic efficacy of CSCs for ischemic heart disease.


Subject(s)
Antigens, Surface/metabolism , Oxidative Stress/drug effects , Protective Agents/pharmacology , Quinazolinones/pharmacology , Reperfusion Injury/drug therapy , Stem Cells/drug effects , Apoptosis/drug effects , Cell Death/drug effects , Cells, Cultured , Humans , Hydrogen Peroxide/pharmacology , Membrane Potential, Mitochondrial/drug effects , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Dynamics/drug effects , Reactive Oxygen Species/metabolism , Reperfusion Injury/metabolism , Stem Cells/metabolism
15.
Nat Commun ; 8(1): 571, 2017 09 18.
Article in English | MEDLINE | ID: mdl-28924239

ABSTRACT

AMP-activated protein kinase (AMPK) is a metabolic stress-sensing enzyme responsible for maintaining cellular energy homeostasis. Activation of AMPK by salicylate and the thienopyridone A-769662 is critically dependent on phosphorylation of Ser108 in the ß1 regulatory subunit. Here, we show a possible role for Ser108 phosphorylation in cell cycle regulation and promotion of pro-survival pathways in response to energy stress. We identify the autophagy initiator Unc-51-like kinase 1 (ULK1) as a ß1-Ser108 kinase in cells. Cellular ß1-Ser108 phosphorylation by ULK1 was dependent on AMPK ß-subunit myristoylation, metabolic stress associated with elevated AMP/ATP ratio, and the intrinsic energy sensing capacity of AMPK; features consistent with an AMP-induced myristoyl switch mechanism. We further demonstrate cellular AMPK signaling independent of activation loop Thr172 phosphorylation, providing potential insight into physiological roles for Ser108 phosphorylation. These findings uncover new mechanisms by which AMPK could potentially maintain cellular energy homeostasis independently of Thr172 phosphorylation.AMPK is involved in sensing of metabolic stress. The authors show that the autophagy initiator ULK1 phosphorylates ß1-Ser108 on the regulatory ß1-subunit, sensitizing AMPK to allosteric drugs, and activates signaling pathways that appear independent of Thr172 phosphorylation in the kinase activation loop.


Subject(s)
AMP-Activated Protein Kinases/physiology , Autophagy-Related Protein-1 Homolog/physiology , Stress, Physiological , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Autophagy/physiology , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Biphenyl Compounds , COS Cells , Chlorocebus aethiops , HEK293 Cells , Homeostasis , Humans , Models, Biological , Phosphorylation , Pyrones/pharmacology , Salicylates/pharmacology , Thiophenes/pharmacology
16.
Sci Rep ; 7: 43264, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28230171

ABSTRACT

The Ca2+-calmodulin dependent protein kinase kinase-2 (CaMKK2) is a key regulator of neuronal function and whole-body energy metabolism. Elevated CaMKK2 activity is strongly associated with prostate and hepatic cancers, whereas reduced CaMKK2 activity has been linked to schizophrenia and bipolar disease in humans. Here we report the functional effects of nine rare-variant point mutations that were detected in large-scale human genetic studies and cancer tissues, all of which occur close to two regulatory phosphorylation sites and the catalytic site on human CaMKK2. Four mutations (G87R, R139W, R142W and E268K) cause a marked decrease in Ca2+-independent autonomous activity, however S137L and P138S mutants displayed increased autonomous and Ca2+-CaM stimulated activities. Furthermore, the G87R mutant is defective in Thr85-autophosphorylation dependent autonomous activity, whereas the A329T mutation rendered CaMKK2 virtually insensitive to Ca2+-CaM stimulation. The G87R and R139W mutants behave as dominant-negative inhibitors of CaMKK2 signaling in cells as they block phosphorylation of the downstream substrate AMP-activated protein kinase (AMPK) in response to ionomycin. Our study provides insight into functionally disruptive, rare-variant mutations in human CaMKK2, which have the potential to influence risk and burden of disease associated with aberrant CaMKK2 activity in human populations carrying these variants.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Kinase/genetics , Calcium-Calmodulin-Dependent Protein Kinase Kinase/metabolism , Calcium/metabolism , Calmodulin/metabolism , Gene Expression Regulation , Genetic Variation , Humans , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation, Missense , Phosphorylation
17.
Pharmacol Ther ; 160: 159-79, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26899498

ABSTRACT

Excitotoxicity, a pathological process caused by over-stimulation of ionotropic glutamate receptors, is a major cause of neuronal loss in acute and chronic neurological conditions such as ischaemic stroke, Alzheimer's and Huntington's diseases. Effective neuroprotective drugs to reduce excitotoxic neuronal loss in patients suffering from these neurological conditions are urgently needed. One avenue to achieve this goal is to clearly define the intracellular events mediating the neurotoxic signals originating from the over-stimulated glutamate receptors in neurons. In this review, we first focus on the key cellular events directing neuronal death but not involved in normal physiological processes in the neurotoxic signalling pathways. These events, referred to as pathologically activated events, are potential targets for the development of neuroprotectant therapeutics. Inhibitors blocking some of the known pathologically activated cellular events have been proven to be effective in reducing stroke-induced brain damage in animal models. Notable examples are inhibitors suppressing the ion channel activity of neurotoxic glutamate receptors and those disrupting interactions of specific cellular proteins occurring only in neurons undergoing excitotoxic cell death. Among them, Tat-NR2B9c and memantine are clinically effective in reducing brain damage caused by some acute and chronic neurological conditions. Our second focus is evaluation of the suitability of the other inhibitors for use as neuroprotective therapeutics. We also discuss the experimental approaches suitable for bridging our knowledge gap in our current understanding of the excitotoxic signalling mechanism in neurons and discovery of new pathologically activated cellular events as potential targets for neuroprotection.


Subject(s)
Cell Death/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Stroke/drug therapy , Animals , Humans , Memantine/pharmacology , Memantine/therapeutic use , Peptides/pharmacology , Peptides/therapeutic use , Signal Transduction/drug effects
18.
Brain Res ; 1594: 1-14, 2015 Jan 12.
Article in English | MEDLINE | ID: mdl-25451123

ABSTRACT

BACKGROUND: Src-family kinases (SFKs) are involved in neuronal survival and their aberrant regulation contributes to neuronal death. However, how they control neuronal survival and death remains unclear. OBJECTIVE: To define the effect of inhibition of Src activity and expression on neuronal survival. RESULTS: In agreement with our previous findings, we demonstrated that Src was cleaved by calpain to form a 52-kDa truncated fragment in neurons undergoing excitotoxic cell death, and expression of the recombinant truncated Src fragment induced neuronal death. The data confirm that the neurotoxic signaling pathways are intact in the neurons we used for our study. To define the functional role of neuronal SFKs, we treated these neurons with SFK inhibitors and discovered that the treatment induced cell death, suggesting that the catalytic activity of one or more of the neuronal SFKs is critical to neuronal survival. Using small hairpin RNAs that suppress Src expression, we demonstrated that Src is indispensable to neuronal survival. Additionally, we found that neuronal death induced by expression of the neurotoxic truncated Src mutant, treatment of SFK inhibitors or knock-down of Src expression caused inhibition of the neuroprotective protein kinases Erk1/2, or Akt. CONCLUSIONS: Src is critical to both neuronal survival and death. Intact Src sustains neuronal survival. However, in the excitotoxic condition, calpain cleavage of Src generates a neurotoxic truncated Src fragment. Both intact Src and the neurotoxic truncated Src fragment exert their biological actions by controlling the activities of neuroprotective protein kinases.


Subject(s)
Neurons/enzymology , Signal Transduction/physiology , src-Family Kinases/metabolism , Animals , Blotting, Western , Calpain/metabolism , Cell Survival , Fluorescent Antibody Technique , Mice , Mice, Inbred C57BL , Peptide Fragments/metabolism
20.
Int J Food Microbiol ; 142(3): 375-8, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20685000

ABSTRACT

Cronobacter spp. formerly known as Enterobacter sakazakii is an occasional contaminant of powdered infant formula (PIF). This pathogen has been associated with out-breaks of a rare form of infant meningitis, necrotizing enterocolitis (NEC), bacteremia and neonate deaths. The organism is ranked by the International Commission for Microbiological Specifications for Foods (ICMSF) as a 'Severe hazard for restricted populations, life threatening or substantial chronic sequelae or long duration'. Present study aimed to isolate Cronobacter spp. from PIF and clinical samples, such as blood, stool and CSF collected from 93 neonates and child patients, age ranged from 0 to 24months. We did not detect Cronobacter spp. in any of these samples. Later 32 PIF samples collected from retail markets in Bangladesh were tested for the presence of Cronobacter spp. Of these only one was found to be contaminated with Cronobacter sp. This is the first case of Cronobacter contaminated PIF found in Bangladesh to be reported. The organism was successfully identified based on its typical culture characteristics, producing blue-green colonies on chromogenic DFI agar and also by a standardized conventional PCR assay targeting the alpha glucosidase and 16S rRNA gene sequence of Cronobacter sp. The 16S rRNA gene was partially sequenced to provide for the phylogenetic analysis of this isolate (DA01) and found to cluster with some other Cronobacter isolates in the phylogram.


Subject(s)
Cronobacter sakazakii/isolation & purification , Food Contamination/analysis , Infant Food/microbiology , Infant Formula , Bangladesh , Base Sequence , Cluster Analysis , Consumer Product Safety , Cronobacter sakazakii/classification , Feces/microbiology , Food Microbiology , Humans , Infant , Infant Food/analysis , Infant, Newborn , Molecular Sequence Data , Phylogeny , Polymerase Chain Reaction , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics
SELECTION OF CITATIONS
SEARCH DETAIL