Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 48
1.
Breast Cancer Res ; 22(1): 130, 2020 11 25.
Article En | MEDLINE | ID: mdl-33239070

BACKGROUND: Chemotherapy is the standard treatment for breast cancer; however, the response to chemotherapy is disappointingly low. Here, we investigated the alternative therapeutic efficacy of novel combination treatment with necroptosis-inducing small molecules to overcome chemotherapeutic resistance in tyrosine aminoacyl-tRNA synthetase (YARS)-positive breast cancer. METHODS: Pre-chemotherapeutic needle biopsy of 143 invasive ductal carcinomas undergoing the same chemotherapeutic regimen was subjected to proteomic analysis. Four different machine learning algorithms were employed to determine signature protein combinations. Immunoreactive markers were selected using three common candidate proteins from the machine-learning algorithms and verified by immunohistochemistry using 123 cases of independent needle biopsy FFPE samples. The regulation of chemotherapeutic response and necroptotic cell death was assessed using lentiviral YARS overexpression and depletion 3D spheroid formation assay, viability assays, LDH release assay, flow cytometry analysis, and transmission electron microscopy. The ROS-induced metabolic dysregulation and phosphorylation of necrosome complex by YARS were assessed using oxygen consumption rate analysis, flow cytometry analysis, and 3D cell viability assay. The therapeutic roles of SMAC mimetics (LCL161) and a pan-BCL2 inhibitor (ABT-263) were determined by 3D cell viability assay and flow cytometry analysis. Additional biologic process and protein-protein interaction pathway analysis were performed using Gene Ontology annotation and Cytoscape databases. RESULTS: YARS was selected as a potential biomarker by proteomics-based machine-learning algorithms and was exclusively associated with good response to chemotherapy by subsequent immunohistochemical validation. In 3D spheroid models of breast cancer cell lines, YARS overexpression significantly improved chemotherapy response via phosphorylation of the necrosome complex. YARS-induced necroptosis sequentially mediated mitochondrial dysfunction through the overproduction of ROS in breast cancer cell lines. Combination treatment with necroptosis-inducing small molecules, including a SMAC mimetic (LCL161) and a pan-BCL2 inhibitor (ABT-263), showed therapeutic efficacy in YARS-overexpressing breast cancer cells. CONCLUSIONS: Our results indicate that, before chemotherapy, an initial screening of YARS protein expression should be performed, and YARS-positive breast cancer patients might consider the combined treatment with LCL161 and ABT-263; this could be a novel stepwise clinical approach to apply new targeted therapy in breast cancer patients in the future.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/therapy , Carcinoma, Ductal, Breast/therapy , Neoadjuvant Therapy/methods , Tyrosine-tRNA Ligase/analysis , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis Regulatory Proteins/agonists , Apoptosis Regulatory Proteins/metabolism , Biopsy , Breast/pathology , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Cell Line, Tumor , Clinical Decision-Making/methods , Drug Synergism , Female , Humans , Mastectomy , Mitochondrial Proteins/agonists , Mitochondrial Proteins/metabolism , Necroptosis/drug effects , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Thiazoles/pharmacology , Thiazoles/therapeutic use , Tyrosine-tRNA Ligase/metabolism
2.
Neuropharmacology ; 181: 108326, 2020 12 15.
Article En | MEDLINE | ID: mdl-32966847

We have previously shown that sphingosine kinase 2 (SPK2) interacts with Bcl-2 via its BH3 domain, activating autophagy by inducing the dissociation of Beclin-1/Bcl-2 complexes, and that a TAT-SPK2 peptide containing the BH3 domain of SPK2 protects neurons against ischemic injury. The goals of the present study were to establish the functional significance of these findings, by testing whether TAT-SPK2 was effective in a mouse model of ischemic stroke, and to explore potential underlying mechanisms. Mice were administered with TAT-SPK2 by intraperitoneal injection before or after transient middle cerebral artery occlusion (tMCAO). Infarct volume, neurological deficit and brain water content were assessed 24 h after reperfusion. Mitophagy inhibitor Mdivi-1 and BNIP3 siRNAs were used to examine the involvement of BNIP3-dependent mitophagy in the neuroprotection of TAT-SPK2. Mitophagy was quantified by immunoblotting, immunofluorescence and electron microscopy. The interaction between TAT-SPK2 and Bcl-2, Bcl-2 and BNIP3 was detected by co-immunoprecipitation. In the tMCAO model, pre-treatment with TAT-SPK2 significantly reduced infarct volume, improved neurological function and decreased brain edema. Neuroprotection by TAT-SPK2 was still seen when the peptide was administered 3 h after reperfusion. TAT-SPK2 also significantly improved functional recovery and reduced long-term brain atrophy of the ischemic hemisphere 30 days after administration. Our studies further showed that TAT-SPK2 directly binds to Bcl-2 and disrupts Bcl-2/Beclin-1 or Bcl-2/BNIP3 complexes to induce mitophagy. These results suggest that TAT-SPK2 protects neurons against ischemia reperfusion injury by activating BNIP3-mediated mitophagy. Agents exploiting this molecular mechanism are potential candidates for the treatment of ischemic stroke.


Gene Products, tat/pharmacology , Membrane Proteins/agonists , Mitochondrial Proteins/agonists , Mitophagy/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Peptides/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , Reperfusion Injury/prevention & control , Animals , Autophagy , Beclin-1 , Brain Edema/prevention & control , Infarction, Middle Cerebral Artery/drug therapy , Ischemic Stroke/drug therapy , Male , Mice , Mice, Inbred ICR , Mitochondria/drug effects , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , RNA, Small Interfering/pharmacology
3.
J Immunol ; 203(12): 3407-3415, 2019 12 15.
Article En | MEDLINE | ID: mdl-31694910

Inflammatory bowel disease (IBD) is a complex autoimmune disease with dysfunction in pattern-recognition responses, including within the NLR family. Nucleotide-binding oligomerization domain, leucine rich repeat containing X1 (NLRX1) is a unique NLR with regulatory and anti-inflammatory functions resulting in protection from IBD in mouse models. NX-13 is an orally active, gut-restricted novel drug candidate that selectively targets and activates the NLRX1 pathway locally in the gut. In vitro and in vivo efficacy of NLRX1 activation by NX-13 was examined. Oral treatment with NX-13 alleviates disease severity, colonic leukocytic infiltration, and cytokine markers of inflammation in three mouse models of IBD (dextran sulfate sodium, Mdr1a-/-, and CD45RBhi adoptive transfer). Treatment of naive CD4+ T cells with NX-13 in vitro decreases differentiation into Th1 and Th17 subsets with increased oxidative phosphorylation and decreased NF-κB activation and reactive oxygen species. With stimulation by PMA/ionomycin, TNF-α, or H2O2, PBMCs from ulcerative colitis patients treated with NX-13 had decreased NF-κB activity, TNF-α+ and IFN-γ+ CD4+ T cells and overall production of IL-6, MCP1, and IL-8. NX-13 activates NLRX1 to mediate a resistance to both inflammatory signaling and oxidative stress in mouse models and human primary cells from ulcerative colitis patients with effects on NF-κB activity and oxidative phosphorylation. NX-13 is a promising oral, gut-restricted NLRX1 agonist for treating IBD.


Benzene Derivatives/therapeutic use , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Mitochondrial Proteins/metabolism , Pyridines/therapeutic use , Signal Transduction/drug effects , Administration, Oral , Animals , Benzene Derivatives/administration & dosage , Benzene Derivatives/pharmacology , Cell Differentiation/drug effects , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Humans , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/agonists , Oxidative Phosphorylation/drug effects , Oxidative Stress/drug effects , Pyridines/administration & dosage , Pyridines/pharmacology , Reactive Oxygen Species/metabolism
4.
Br J Haematol ; 185(2): 219-231, 2019 04.
Article En | MEDLINE | ID: mdl-30836448

Evasion of apoptosis has been identified as one of the essential hallmarks of cancer. Inhibitor of apoptosis proteins (IAPs) are implicated in a host of myeloid malignancies, providing the rationale for strategies aimed at neutralizing IAPs to lower the cancer cell apoptosis threshold. Modes of IAP antagonism may include down-regulating IAP expression, up-regulating endogenous pro-apoptotic proteins, such as tumour necrosis factor-α or Fas ligand, or directly antagonizing IAP activity against caspases. Direct targeting of IAPs using mimetics of the second mitochondria-derived activator of caspase (SMAC) protein has shown therapeutic promise by sensitizing the effect of chemotherapy on malignant cells. In pre-clinical studies, SMAC mimetics have demonstrated broad synergistic activity with a wide range of therapeutics, including cytotoxic chemotherapy, receptor tyrosine kinase inhibitors, agents targeting death receptors and alternative mechanisms of cell death, such as necroptosis or autophagy and immune check point blockade. SMAC mimetics represent a novel approach for further investigation in patients with high-risk, chemo-refractory blood cancers, as single agents or in thoughtfully selected combinations. In this review, we discuss the development and therapeutic rationale of small molecule SMAC mimetics, with an emphasis on agents in clinical development for myeloid malignancies.


Antimetabolites, Antineoplastic/therapeutic use , Apoptosis Regulatory Proteins/agonists , Leukemia, Myeloid/drug therapy , Mitochondrial Proteins/agonists , Myelodysplastic Syndromes/drug therapy , Peptidomimetics/therapeutic use , Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/metabolism , Humans , Leukemia, Myeloid/metabolism , Molecular Targeted Therapy/methods , Myelodysplastic Syndromes/metabolism , Peptidomimetics/pharmacology
5.
Science ; 360(6386): 336-341, 2018 04 20.
Article En | MEDLINE | ID: mdl-29674596

Mitofusins (MFNs) promote fusion-mediated mitochondrial content exchange and subcellular trafficking. Mutations in Mfn2 cause neurodegenerative Charcot-Marie-Tooth disease type 2A (CMT2A). We showed that MFN2 activity can be determined by Met376 and His380 interactions with Asp725 and Leu727 and controlled by PINK1 kinase-mediated phosphorylation of adjacent MFN2 Ser378 Small-molecule mimics of the peptide-peptide interface of MFN2 disrupted this interaction, allosterically activating MFN2 and promoting mitochondrial fusion. These first-in-class mitofusin agonists overcame dominant mitochondrial defects provoked in cultured neurons by CMT2A mutants MFN2 Arg94→Gln94 and MFN2 Thr105→Met105, as demonstrated by amelioration of mitochondrial dysmotility, fragmentation, depolarization, and clumping. A mitofusin agonist normalized axonal mitochondrial trafficking within sciatic nerves of MFN2 Thr105→Met105 mice, promising a therapeutic approach for CMT2A and other untreatable diseases of impaired neuronal mitochondrial dynamism and/or trafficking.


Charcot-Marie-Tooth Disease/drug therapy , Drug Design , Mitochondria/drug effects , Mitochondrial Diseases/drug therapy , Mitochondrial Proteins/agonists , Oligopeptides/pharmacology , Small Molecule Libraries/pharmacology , Amino Acid Substitution , Animals , Arginine/genetics , Axons/drug effects , Axons/physiology , Charcot-Marie-Tooth Disease/genetics , Disease Models, Animal , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Glutamine/genetics , Humans , Methionine/genetics , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oligopeptides/chemistry , Oligopeptides/therapeutic use , Phosphorylation , Protein Kinases/metabolism , Sciatic Nerve/drug effects , Sciatic Nerve/physiopathology , Small Molecule Libraries/therapeutic use , Threonine/genetics
6.
Toxicol Lett ; 289: 1-13, 2018 Jun 01.
Article En | MEDLINE | ID: mdl-29501571

We performed a multiple 'omics study by integrating data on epigenomic, transcriptomic, and proteomic perturbations associated with mitochondrial dysfunction in primary human hepatocytes caused by the liver toxicant valproic acid (VPA), to deeper understand downstream events following epigenetic alterations in the mitochondrial genome. Furthermore, we investigated persistence of cross-omics changes after terminating drug treatment. Upon transient methylation changes of mitochondrial genes during VPA-treatment, increasing complexities of gene-interaction networks across time were demonstrated, which normalized during washout. Furthermore, co-expression between genes and their corresponding proteins increased across time. Additionally, in relation to persistently decreased ATP production, we observed decreased expression of mitochondrial complex I and III-V genes. Persistent transcripts and proteins were related to citric acid cycle and ß-oxidation. In particular, we identified a potential novel mitochondrial-nuclear signaling axis, MT-CO2-FN1-MYC-CPT1. In summary, this cross-omics study revealed dynamic responses of the mitochondrial epigenome to an impulse toxicant challenge resulting in persistent mitochondrial dysfunctioning. Moreover, this approach allowed for discriminating between the toxic effect of VPA and adaptation.


Anticonvulsants/adverse effects , DNA, Mitochondrial/drug effects , Hepatocytes/drug effects , Mitochondria, Liver/drug effects , Mitochondrial Proteins/metabolism , Models, Biological , Valproic Acid/adverse effects , Adenosine Triphosphate/metabolism , Cells, Cultured , DNA Methylation/drug effects , DNA, Mitochondrial/metabolism , Electron Transport Complex I/antagonists & inhibitors , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex III/antagonists & inhibitors , Electron Transport Complex III/genetics , Electron Transport Complex III/metabolism , Electron Transport Complex IV/antagonists & inhibitors , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Epigenomics , Gene Expression Profiling , Hepatocytes/enzymology , Hepatocytes/metabolism , Humans , Kinetics , Mitochondria, Liver/enzymology , Mitochondria, Liver/metabolism , Mitochondrial Proteins/agonists , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Proteomics
7.
Int J Mol Sci ; 19(2)2018 Jan 25.
Article En | MEDLINE | ID: mdl-29370072

Potassium channel openers (KCOs) have been shown to play a role in cytoprotection through the activation of mitochondrial potassium channels. Recently, in several reports, a number of data has been described as off-target actions for KCOs. In the present study, we investigated the effects of BKCa channel openers CGS7181, CGS7184, NS1619, and NS004 in neuronal cells. For the purpose of this research, we used a rat brain, the mouse hippocampal HT22 cells, and the human astrocytoma U-87 MG cell line. We showed that CGS7184 activated the mitochondrial BKCa (mitoBKCa) channel in single-channel recordings performed on astrocytoma mitoplasts. Moreover, when applied to the rat brain homogenate or isolated rat brain mitochondria, CGS7184 increased the oxygen consumption rate, and can thus be considered a potentially cytoprotective agent. However, experiments on intact neuronal HT22 cells revealed that both CGS7181 and CGS7184 induced HT22 cell death in a concentration- and time-dependent manner. By contrast, we did not observe cell death when NS1619 or NS004 was applied. CGS7184 toxicity was not abolished by BKCa channel inhibitors, suggesting that the observed effects were independent of a BKCa-type channel activity. CGS7184 treatment resulted in an increase of cytoplasmic Ca2+ concentration that likely involved efflux from internal calcium stores and the activation of calpains (calcium-dependent proteases). The cytotoxic effect of the channel opener was partially reversed by a calpain inhibitor. Our data show that KCOs under study not only activate mitoBKCa channels from brain tissue, but also induce cell death when used in cellular models.


Indoles/pharmacology , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Membrane Transport Modulators/pharmacology , Mitochondrial Proteins/metabolism , Animals , Apoptosis/drug effects , Calcium/metabolism , Calpain/metabolism , Cell Line, Tumor , Humans , Indoles/toxicity , Large-Conductance Calcium-Activated Potassium Channels/agonists , Male , Membrane Transport Modulators/toxicity , Mitochondrial Proteins/agonists , Rats , Rats, Wistar
8.
Cell Chem Biol ; 25(3): 268-278.e4, 2018 03 15.
Article En | MEDLINE | ID: mdl-29290623

Mitochondria are dynamic organelles that produce most of the cellular ATP, and are involved in many other cellular functions such as Ca2+ signaling, differentiation, apoptosis, cell cycle, and cell growth. One key process of mitochondrial dynamics is mitochondrial fusion, which is catalyzed by mitofusins (MFN1 and MFN2) and OPA1. The outer mitochondrial membrane protein MFN2 plays a relevant role in the maintenance of mitochondrial metabolism, insulin signaling, and mutations that cause neurodegenerative disorders. Therefore, modulation of proteins involved in mitochondrial dynamics has emerged as a potential pharmacological strategy. Here, we report the identification of small molecules by high-throughput screen that promote mitochondrial elongation in an MFN1/MFN2-dependent manner. Detailed analysis of their mode of action reveals a previously unknown connection between pyrimidine metabolism and mitochondrial dynamics. Our data indicate a link between pyrimidine biosynthesis and mitochondrial dynamics, which maintains cell survival under stress conditions characterized by loss of pyrimidine synthesis.


Mitochondria/metabolism , Mitochondrial Dynamics/drug effects , Pyrimidines/metabolism , Small Molecule Libraries/pharmacology , Animals , Apoptosis/drug effects , Cell Line , Dihydroorotate Dehydrogenase , Doxorubicin/pharmacology , Electron Transport Complex III/antagonists & inhibitors , Electron Transport Complex III/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Humans , Leflunomide/pharmacology , Mice , Mitochondria/drug effects , Mitochondrial Membrane Transport Proteins/agonists , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proteins/agonists , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Porins/genetics , Porins/metabolism , Pyrimidines/biosynthesis , RNA, Messenger/metabolism , Transcriptome/drug effects
9.
Stem Cells ; 35(7): 1655-1662, 2017 07.
Article En | MEDLINE | ID: mdl-28544378

High attrition rates and loss of capital plague the drug discovery process. This is particularly evident for mitochondrial disease that typically involves neurological manifestations and is caused by nuclear or mitochondrial DNA defects. This group of heterogeneous disorders is difficult to target because of the variability of the symptoms among individual patients and the lack of viable modeling systems. The use of induced pluripotent stem cells (iPSCs) might significantly improve the search for effective therapies for mitochondrial disease. iPSCs can be used to generate patient-specific neural cell models in which innovative compounds can be identified or validated. Here we discuss the promises and challenges of iPSC-based drug discovery for mitochondrial disease with a specific focus on neurological conditions. We anticipate that a proper use of the potent iPSC technology will provide critical support for the development of innovative therapies against these untreatable and detrimental disorders. Stem Cells 2017;35:1655-1662.


Drug Discovery/methods , Induced Pluripotent Stem Cells/drug effects , Mitochondria/drug effects , Mitochondrial Diseases/drug therapy , Neurons/drug effects , Neuroprotective Agents/pharmacology , Cell Differentiation , DNA, Mitochondrial/genetics , Gene Expression , High-Throughput Screening Assays , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Mitochondria/metabolism , Mitochondria/pathology , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondrial Diseases/pathology , Mitochondrial Proteins/agonists , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Models, Biological , Mutation , Neurons/metabolism , Neurons/pathology , Organoids/drug effects , Organoids/metabolism , Organoids/pathology , Precision Medicine
10.
J Steroid Biochem Mol Biol ; 172: 29-35, 2017 09.
Article En | MEDLINE | ID: mdl-28549691

Investigating differentially expressed proteins in a milieu rich in cholesterol oxidation products, we found via mass spectrometry-based proteomics that surface levels of heat shock protein 60 (HSP60) were upregulated on monocytic cells in the presence of 27-hydroxycholesterol (27OHChol). The elevated levels of cytoplasmic membrane HSP60 were verified via Western blot analysis and visualized by confocal microscopy. Treatment with 27OHChol also resulted in increased levels of cellular HSP60 without altering its transcription. Cholesterol, however, did not affect cell-surface levels and cellular amount of HSP60. GSK 2033, an LXR antagonist, inhibited expression of live X receptor α, but not of HSP60, induced by 27OHChol. Treatment with 27OHChol also resulted in increased release of HSP60 from monocytic cells, but the release was significantly reduced by inhibitors of endoplasmic reticulum-Golgi protein trafficking, brefeldin A and monensin. Results of the current study indicate that 27OHChol upregulates not only cell-surface and cellular levels of HSP60 but also its release from monocytic cells, thereby contributing to activation of the immune system.


Chaperonin 60/genetics , Hydroxycholesterols/pharmacokinetics , Mitochondrial Proteins/genetics , Monocytes/drug effects , Amino Acid Sequence , Animals , Brefeldin A/pharmacology , Cell Line , Cell Membrane/drug effects , Cell Membrane/immunology , Cell Membrane/metabolism , Chaperonin 60/agonists , Chaperonin 60/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/metabolism , Gene Expression Regulation , Golgi Apparatus/drug effects , Golgi Apparatus/immunology , Golgi Apparatus/metabolism , Humans , Hydroxycholesterols/metabolism , Immunity, Cellular , Liver X Receptors/antagonists & inhibitors , Liver X Receptors/genetics , Liver X Receptors/metabolism , Mitochondrial Proteins/agonists , Mitochondrial Proteins/metabolism , Monensin/pharmacology , Monocytes/cytology , Monocytes/immunology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Sulfonamides/pharmacology , Transcription, Genetic
11.
J Biol Chem ; 292(24): 10306-10315, 2017 06 16.
Article En | MEDLINE | ID: mdl-28446607

Juvenile hormone (JH) controls many biological activities in insects, including development, metamorphosis, and reproduction. In the Aedes aegypti mosquito, a vector of dengue, yellow fever, chikungunya, and zika viruses, the metabolic tissue (the fat body, which is an analogue of the vertebrate liver) produces yolk proteins for developing oocytes. JH is important for the fat body to acquire competence for yolk protein production. However, the molecular mechanisms of how JH promotes mosquito reproduction are not completely understood. In this study we show that stimulation of the JH receptor methoprene-tolerant (Met) activates expression of genes encoding the regulator of ribosome synthesis 1 (RRS1) and six ribosomal proteins (two ribosomal large subunit proteins, two ribosomal small subunit proteins, and two mitochondrial ribosomal proteins). Moreover, RNAi-mediated depletion of RRS1 decreased biosynthesis of the ribosomal protein L32 (RpL32). Depletion of Met, RRS1, or RpL32 led to retardation of ovarian growth and reduced mosquito fecundity, which may at least in part have resulted from decreased vitellogenin protein production in the fat body. In summary, our results indicate that JH is critical for inducing the expression of ribosomal protein genes and demonstrate that RRS1 mediates the JH signal to enhance both ribosomal biogenesis and vitellogenesis.


Aedes/metabolism , Insect Proteins/agonists , Juvenile Hormones/metabolism , Organelle Biogenesis , Ribosomal Proteins/agonists , Ribosomes/metabolism , Vitellogenesis , Aedes/growth & development , Animals , Fat Body/growth & development , Fat Body/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Insect Proteins/antagonists & inhibitors , Insect Proteins/genetics , Insect Proteins/metabolism , Insecticide Resistance , Mitochondrial Proteins/agonists , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Organ Culture Techniques , Ovary/growth & development , Ovary/metabolism , Polyribosomes/metabolism , RNA Interference , Ribosomal Proteins/antagonists & inhibitors , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Signal Transduction , Vitellogenins/antagonists & inhibitors , Vitellogenins/genetics , Vitellogenins/metabolism
12.
J Biol Chem ; 292(15): 6095-6107, 2017 04 14.
Article En | MEDLINE | ID: mdl-28196863

The first step in glutamine catabolism is catalysis by the mitochondrial enzyme glutaminase, with a specific isoform, glutaminase C (GAC), being highly expressed in cancer cells. GAC activation requires the formation of homotetramers, promoted by anionic allosteric activators such as inorganic phosphate. This leads to the proper orientation of a flexible loop proximal to the dimer-dimer interface that is essential for catalysis (i.e. the "activation loop"). A major class of allosteric inhibitors of GAC, with the prototype being bis-2-(5-phenylacetamido-1,2,4-thiadiazol-2-yl)ethyl sulfide (BPTES) and the related molecule CB-839, binds to the activation loop and induces the formation of an inactive tetramer (two inhibitors bound per active tetramer). Here we describe a direct readout for monitoring the dynamics of the activation loop of GAC in response to these allosteric inhibitors, as well as allosteric activators, through the substitution of phenylalanine at position 327 with tryptophan (F327W). The tryptophan fluorescence of the GAC(F327W) mutant undergoes a marked quenching upon the binding of BPTES or CB-839, yielding titration profiles that make it possible to measure the binding affinities of these inhibitors for the enzyme. Allosteric activators like phosphate induce the opposite effect (i.e. fluorescence enhancement). These results describe direct readouts for the binding of the BPTES class of allosteric inhibitors as well as for inorganic phosphate and related activators of GAC, which should facilitate screening for additional modulators of this important metabolic enzyme.


Benzeneacetamides/chemistry , Enzyme Activators/chemistry , Enzyme Inhibitors/chemistry , Glutaminase/antagonists & inhibitors , Glutaminase/chemistry , Mitochondrial Proteins/agonists , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/chemistry , Sulfides/chemistry , Thiadiazoles/chemistry , Allosteric Regulation , Amino Acid Substitution , Animals , Glutaminase/genetics , Mice , Mitochondrial Proteins/genetics , Mutation, Missense , Protein Structure, Secondary , Spectrometry, Fluorescence
13.
Biochem Biophys Res Commun ; 478(2): 838-44, 2016 09 16.
Article En | MEDLINE | ID: mdl-27510639

Ferroptosis is a form of non-apoptotic cell death originally identified in cancer cells. However, the key regulator of ferroptosis in mitochondria remains unknown. Here, we show that CDGSH iron sulfur domain 1 (CISD1, also termed mitoNEET), an iron-containing outer mitochondrial membrane protein, negatively regulates ferroptotic cancer cell death. The classical ferroptosis inducer erastin promotes CISD1 expression in an iron-dependent manner in human hepatocellular carcinoma cells (e.g., HepG2 and Hep3B). Genetic inhibition of CISD1 increased iron-mediated intramitochondrial lipid peroxidation, which contributes to erastin-induced ferroptosis. In contrast, stabilization of the iron sulfur cluster of CISD1 by pioglitazone inhibits mitochondrial iron uptake, lipid peroxidation, and subsequent ferroptosis. These findings indicate a novel role of CISD1 in protecting against mitochondrial injury in ferroptosis.


Apoptosis Regulatory Proteins/genetics , Apoptosis/genetics , Gene Expression Regulation, Neoplastic , Iron/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Apoptosis/drug effects , Apoptosis Regulatory Proteins/agonists , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/metabolism , Deferoxamine/pharmacology , Hep G2 Cells , Humans , Iron Chelating Agents/pharmacology , Lipid Peroxidation/drug effects , Mitochondria/drug effects , Mitochondrial Proteins/agonists , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/metabolism , Oxidative Stress , Pioglitazone , Piperazines/antagonists & inhibitors , Piperazines/pharmacology , Thiazolidinediones/pharmacology
14.
Oncotarget ; 7(35): 56253-56265, 2016 Aug 30.
Article En | MEDLINE | ID: mdl-27494845

Bcl2 and IAP families are anti-apoptotic proteins deregulated in multiple myeloma (MM) cells. Pharmacological inhibition of each of these families has shown significant activity only in subgroups of MM patients. Here, we have examined a broad-spectrum Bcl2 family inhibitor Obatoclax (OBX) in combination with a Smac mimetic LCL161 in MM cell lines and patient cells. LCL161/OBX combination induced synergistic cytotoxicity and anti-proliferative effects on a broad range of human MM cell lines. The cytotoxicity was mediated through inhibition of the IAPs, activation of caspases and up regulation of the pro-apoptotic proteins Bid, Bim, Puma and Noxa by the drug combination. In addition, we observed that OBX caused ER stress and activated the Unfolded Protein Response (UPR) leading to drug resistance. LCL161, however inhibited spliced Xbp-1, a pro-survival factor. In addition, we observed that OBX increased GRP78 localization to the cell surface, which then induced PI3K dependent Akt activation and resistance to cell death. LCL161 was able to block OBX induced Akt activation contributing to synergistic cell death. Our results support clinical evaluation of this combination strategy in relapsed refractory MM patients.


Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Endoplasmic Reticulum Stress/drug effects , Multiple Myeloma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Pyrroles/pharmacology , Thiazoles/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis Regulatory Proteins , Caspases/metabolism , Cell Line, Tumor , Down-Regulation , Drug Resistance, Neoplasm , Drug Synergism , Endoplasmic Reticulum Chaperone BiP , Heat-Shock Proteins/metabolism , Humans , Indoles , Intracellular Signaling Peptides and Proteins/agonists , Mitochondrial Proteins/agonists , Multiple Myeloma/pathology , Neoplasm Recurrence, Local/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrroles/therapeutic use , Thiazoles/therapeutic use , Unfolded Protein Response/drug effects , Up-Regulation , X-Box Binding Protein 1/metabolism
15.
Toxicol Lett ; 258: 227-236, 2016 Sep 06.
Article En | MEDLINE | ID: mdl-27392435

Application of cisplatin (DDP) for treating lung cancer is restricted due to its toxicity and lung cancer's drug resistance. In this study, we examined the effect of Jinfukang (JFK), an effective herbal medicine against lung cancer, on DDP-induced cytotoxicity in lung cancer cells. Morphologically, we observed that JFK increases DDP-induced pro-apoptosis in A549 cells in a synergistic manner. Transcriptome profiling analysis indicated that the combination of JFK and DDP regulates genes involved in apoptosis-related signaling pathways. Moreover, we found that the combination of JFK and DDP produces synergistic pro-apoptosis effect in other lung cancer cell lines, such as NCI-H1975, NCI-H1650, and NCI-H2228. Particularly, we demonstrated that AIFM2 is activated by the combined treatment of JFK and DDP and partially mediates the synergistic pro-apoptosis effect. Collectively, this study not only offered the first evidence that JFK promotes DDP-induced cytotoxicity, and activation of AIFM2 enhances apoptosis of human lung cancer cells undergoing toxicological stress, but also provided a novel insight for improving cytotoxicity by combining JFK with DDP to treat lung cancer cells.


Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis Regulatory Proteins/agonists , Apoptosis/drug effects , Drugs, Chinese Herbal/pharmacology , Lung Neoplasms/drug therapy , Mitochondrial Proteins/agonists , Neoplasm Proteins/agonists , A549 Cells , Adenocarcinoma, Bronchiolo-Alveolar/drug therapy , Adenocarcinoma, Bronchiolo-Alveolar/metabolism , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Cisplatin/pharmacology , Drug Resistance, Neoplasm , Drug Synergism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks/drug effects , Humans , Lung Neoplasms/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA Interference , RNA, Small Interfering
16.
Cell Mol Life Sci ; 73(15): 2871-96, 2016 08.
Article En | MEDLINE | ID: mdl-27007507

Sirtuins are an evolutionary conserved family of NAD(+)-dependent protein lysine deacylases. Mammals have seven Sirtuin isoforms, Sirt1-7. They contribute to regulation of metabolism, stress responses, and aging processes, and are considered therapeutic targets for metabolic and aging-related diseases. While initial studies were focused on Sirt1 and 2, recent progress on the mitochondrial Sirtuins Sirt3, 4, and 5 has stimulated research and drug development for these isoforms. Here we review the roles of Sirtuins in regulating mitochondrial functions, with a focus on the mitochondrially located isoforms, and on their contributions to disease pathologies. We further summarize the compounds available for modulating the activity of these Sirtuins, again with a focus on mitochondrial isoforms, and we describe recent results important for the further improvement of compounds. This overview illustrates the potential of mitochondrial Sirtuins as drug targets and summarizes the status, progress, and challenges in developing small molecule compounds modulating their activity.


Drug Discovery , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Sirtuins/metabolism , Small Molecule Libraries/pharmacology , Animals , Drug Discovery/methods , Humans , Mitochondria/pathology , Mitochondrial Proteins/agonists , Mitochondrial Proteins/analysis , Mitochondrial Proteins/antagonists & inhibitors , Models, Molecular , Molecular Targeted Therapy/methods , Protein Isoforms/analysis , Protein Isoforms/metabolism , Sirtuins/analysis , Sirtuins/antagonists & inhibitors , Small Molecule Libraries/chemistry
17.
J Biol Chem ; 291(9): 4523-36, 2016 Feb 26.
Article En | MEDLINE | ID: mdl-26733201

Inhibiting class I histone deacetylases (HDACs) increases energy expenditure, reduces adiposity, and improves insulin sensitivity in obese mice. However, the precise mechanism is poorly understood. Here, we demonstrate that HDAC1 is a negative regulator of the brown adipocyte thermogenic program. The Hdac1 level is lower in mouse brown fat (BAT) than white fat, is suppressed in mouse BAT during cold exposure or ß3-adrenergic stimulation, and is down-regulated during brown adipocyte differentiation. Remarkably, overexpressing Hdac1 profoundly blocks, whereas deleting Hdac1 significantly enhances, ß-adrenergic activation-induced BAT-specific gene expression in brown adipocytes. ß-Adrenergic activation in brown adipocytes results in a dissociation of HDAC1 from promoters of BAT-specific genes, including uncoupling protein 1 (Ucp1) and peroxisome proliferator-activated receptor γ co-activator 1α (Pgc1α), leading to increased acetylation of histone H3 lysine 27 (H3K27), an epigenetic mark of gene activation. This is followed by dissociation of the polycomb repressive complexes, including the H3K27 methyltransferase enhancer of zeste homologue (EZH2), suppressor of zeste 12 (SUZ12), and ring finger protein 2 (RNF2) from (and concomitant recruitment of H3K27 demethylase ubiquitously transcribed tetratricopeptide repeat on chromosome X (UTX) to) Ucp1 and Pgc1α promoters, leading to decreased H3K27 trimethylation, a histone transcriptional repression mark. Thus, HDAC1 negatively regulates the brown adipocyte thermogenic program, and inhibiting Hdac1 promotes BAT-specific gene expression through a coordinated control of increased acetylation and decreased methylation of H3K27, thereby switching the transcriptional repressive state to the active state at the promoters of Ucp1 and Pgc1α. Targeting HDAC1 may be beneficial in prevention and treatment of obesity by enhancing BAT thermogenesis.


Adipocytes, Brown/metabolism , Histone Deacetylase 1/metabolism , Histones/metabolism , Ion Channels/metabolism , Mitochondrial Proteins/metabolism , Protein Processing, Post-Translational , Thermogenesis , Transcription Factors/metabolism , Acetylation/drug effects , Adipocytes, Brown/drug effects , Adipocytes, Brown/enzymology , Adrenergic beta-3 Receptor Agonists/pharmacology , Animals , Cell Line, Transformed , Enhancer of Zeste Homolog 2 Protein , Gene Expression Regulation/drug effects , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/genetics , Humans , Ion Channels/agonists , Ion Channels/antagonists & inhibitors , Ion Channels/genetics , Lysine/metabolism , Methylation/drug effects , Mice, Inbred Strains , Mitochondrial Proteins/agonists , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Polycomb Repressive Complex 1/agonists , Polycomb Repressive Complex 1/antagonists & inhibitors , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Polycomb Repressive Complex 2/agonists , Polycomb Repressive Complex 2/antagonists & inhibitors , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism , Promoter Regions, Genetic/drug effects , Protein Processing, Post-Translational/drug effects , RNA Interference , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Thermogenesis/drug effects , Transcription Factors/agonists , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Uncoupling Protein 1
18.
Oncotarget ; 6(32): 33382-96, 2015 Oct 20.
Article En | MEDLINE | ID: mdl-26450902

Recent research on non-thermal plasma (NTP, an ionized gas) has identified it as a novel cancer therapeutic tool. However, the molecular mechanism remains unclear. In this study, we demonstrated NTP induced cell death of head and neck cancer (HNC) through the AKT ubiquitin-proteasome system. NTP increased the gene expression of mitochondrial E3 ubiquitin protein ligase 1 (MUL1), an E3 ligase for AKT, and NTP-induced HNC cell death was prevented by MUL1 siRNA. We also showed that MUL1 inhibited the level of AKT and p-AKT and MUL1 expression was increased by NTP-induced ROS. Furthermore, we optimized and manufactured a new type of NTP, a liquid type of NTP (LTP). In syngeneic and xenograft in vivo tumor models, LTP inhibited tumor progression by increasing the MUL1 level and reducing p-AKT levels, indicating that LTP also has an anti-cancer effect through the same mechanism as that of NTP. Taken together, our results suggest that NTP and LTP have great potential for HNC therapy.


Carcinoma, Squamous Cell/metabolism , Head and Neck Neoplasms/metabolism , Mitochondrial Proteins/agonists , Oncogene Protein v-akt/metabolism , Plasma Gases/pharmacology , Proteolysis/drug effects , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Enzyme Activation/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , RNA, Small Interfering/pharmacology , Squamous Cell Carcinoma of Head and Neck , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Xenograft Model Antitumor Assays
19.
Bull Exp Biol Med ; 159(5): 601-3, 2015 Sep.
Article En | MEDLINE | ID: mdl-26459485

Long-term food restriction (3 weeks, 60% of normal consumption of control animals) was followed by an increase in DNA and protein content in the intercapsular brown fat of mice. As the animals were kept under thermoneutral conditions, these changes are thought to be a result of food restriction.


Adipose Tissue, Brown/metabolism , Body Temperature Regulation/physiology , DNA/biosynthesis , Food Deprivation/physiology , Ion Channels/biosynthesis , Mitochondrial Proteins/biosynthesis , Animals , Body Weight , DNA/agonists , Homeostasis/physiology , Ion Channels/agonists , Male , Mice , Mice, Inbred ICR , Mitochondrial Proteins/agonists , Oxygen Consumption/physiology , Temperature , Uncoupling Protein 1
20.
Br J Nutr ; 114(11): 1774-83, 2015 Dec 14.
Article En | MEDLINE | ID: mdl-26395755

Obesity is one of the major health problems throughout the world. The present study investigated the preventive effect of epilactose--a rare non-digestible disaccharide--on obesity and metabolic disorders in mice fed high-fat (HF) diets. Feeding with HF diets increased body weight gain, fat pad weight and adipocyte size in mice (P<0·01), and these increases were effectively prevented by the use of supplemental epilactose without influencing food intake (P<0·01). Caecal pools of SCFA such as acetic and propionic acids in mice fed epilactose were higher compared with mice not receiving epilactose. Supplemental epilactose increased the expression of uncoupling protein (UCP)-1, which enhances energy expenditure, to 2-fold in the gastrocnemius muscle (P=0·04) and to 1·3-fold in the brown adipose tissue (P=0·02) in mice fed HF diets. Feeding HF diets induced pro-inflammatory macrophage infiltration into white adipose tissue, as indicated by the increased expression of monocyte chemotactic protein-1, TNF-α and F4/80, and these increases were attenuated by supplemental epilactose. In differentiated myogenic-like C2C12 cells, propionic acid, but not acetic or n-butyric acids, directly enhanced UCP-1 expression by approximately 2-fold (P<0·01). Taken together, these findings indicate that the epilactose-mediated increase in UCP-1 in the skeletal muscle and brown adipose tissue can enhance whole-body energy expenditure, leading to effective prevention of obesity and metabolic disorders in mice fed HF diets. It is suggested that propionic acid--a bacterial metabolite--acts as a mediator to induce UCP-1 expression in skeletal muscles.


Anti-Obesity Agents/therapeutic use , Disaccharides/therapeutic use , Ion Channels/agonists , Mitochondrial Proteins/agonists , Muscle, Skeletal/metabolism , Obesity/prevention & control , Prebiotics , Up-Regulation , Adipose Tissue, Brown/immunology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anti-Obesity Agents/metabolism , Cell Line , Diet, High-Fat/adverse effects , Disaccharides/metabolism , Energy Metabolism , Fermentation , Gastrointestinal Microbiome , Ion Channels/genetics , Ion Channels/metabolism , Macrophage Activation , Male , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Muscle, Skeletal/immunology , Obesity/immunology , Obesity/metabolism , Obesity/microbiology , Propionates/metabolism , Random Allocation , Uncoupling Protein 1
...