Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters










Publication year range
1.
Biomolecules ; 12(1)2022 01 12.
Article in English | MEDLINE | ID: mdl-35053271

ABSTRACT

The endohedral metallofullerenol Gd@C82(OH)22 has been identified as a possible antineoplastic agent that can inhibit both the growth and metastasis of cancer cells. Despite these potentially important effects, our understanding of the interactions between Gd@C82(OH)22 and biomacromolecules remains incomplete. Here, we study the interaction between Gd@C82(OH)22 and the human voltage-dependent anion channel 1 (hVDAC1), the most abundant porin embedded in the mitochondrial outer membrane (MOM), and a potential druggable target for novel anticancer therapeutics. Using in silico approaches, we observe that Gd@C82(OH)22 molecules can permeate and form stable interactions with the pore of hVDAC1. Further, this penetration can occur from either side of the MOM to elicit blockage of the pore. The binding between Gd@C82(OH)22 and hVDAC1 is largely driven by long-range electrostatic interactions. Analysis of the binding free energies indicates that it is thermodynamically more favorable for Gd@C82(OH)22 to bind to the hVDAC1 pore when it enters the channel from inside the membrane rather than from the cytoplasmic side of the protein. Multiple factors contribute to the preferential penetration, including the surface electrostatic landscape of hVDAC1 and the unique physicochemical properties of Gd@C82(OH)22. Our findings provide insights into the potential molecular interactions of macromolecular biological systems with the Gd@C82(OH)22 nanodrug.


Subject(s)
Antineoplastic Agents , Fullerenes , Neoplasms , Organometallic Compounds , Voltage-Dependent Anion Channel 1 , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Fullerenes/chemistry , Fullerenes/pharmacology , Gadolinium/chemistry , Gadolinium/pharmacology , Humans , Organometallic Compounds/pharmacology , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Voltage-Dependent Anion Channel 1/metabolism
2.
Oxid Med Cell Longev ; 2021: 7569168, 2021.
Article in English | MEDLINE | ID: mdl-34539973

ABSTRACT

Due to high energy and material metabolism requirements, mitochondria are frequently active in tumor cells. Our study found that the high energy metabolism status is positively correlated with the poor prognosis of patients with lung adenocarcinoma. We constructed a scoring system (mitoRiskscore) based on the gene expression of specific mitochondrial localized proteins through univariate and LASSO cox regression. It has been shown that high mitoRiskscore was correlated with a shorter survival time after surgery in patients with lung adenocarcinoma. Compared with the typical TNM grading system, the mitoRiskscore gene panel had higher prediction accuracy. A vast number of external verification results ensured its universality. Additionally, the mitoRiskscore could evaluate the metabolic pattern and chemotherapy sensitivity of the tumor samples. Lung adenocarcinoma with higher mitoRiskscore was more active in glycolysis, and oxidative phosphorylation expression of proliferation-related pathway genes was also significantly upregulated. In contrast, patients with low mitoRiskscore had similar metabolic patterns to normal tissues. In order to improve the accuracy of prediction ability and promote clinical usage, we developed a nomogram that combined mitoRiskscore and clinical prognostic factors to predict the 3-year, 5-year, and 10-year survival rates of patients. We also performed in vitro experiments to verify the function of the key genes in the mitoRiskscore panel. In conclusion, the mitoRiskscore scoring system may assist clinicians to judge the postoperative survival rate and chemotherapy of patients with lung adenocarcinoma.


Subject(s)
Adenocarcinoma/pathology , Lung Neoplasms/pathology , Mitochondrial Proteins/metabolism , A549 Cells , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Antineoplastic Agents/therapeutic use , Area Under Curve , Cell Proliferation , Databases, Genetic , Glycolysis/genetics , Humans , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Mitochondrial Proteins/genetics , Neoplasm Grading , Prognosis , RNA Interference , RNA, Small Interfering/metabolism , ROC Curve , Survival Rate , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Voltage-Dependent Anion Channel 1/genetics , Voltage-Dependent Anion Channel 1/metabolism
3.
Brain Res Bull ; 174: 131-140, 2021 09.
Article in English | MEDLINE | ID: mdl-34058286

ABSTRACT

Neonatal hypoxic-ischemic encephalopathy (HIE), is a major cause of neurologic disorders in terms of neonates, with the unclear underlying mechanisms. In the study, triphenyl tetrazolium chloride (TTC) staining and Zea-longa score were performed to examine the neurologic damage in hypoxia and ischemia (HI) rats. The results showed that HI induced obviously infarct and serious neurologic impairment in neonatal rats. Then, protein chip was applied to detect the differential expression genes in cortex and hippocampus and found the brain-derived neurotrophic factor (BDNF) down-regulated both in cortex and hippocampus. Moreover, low expression of BDNF after HI in right cortex and hippocampus was validate by immunohistochemistry (IHC) and Western Blotting (WB). Afterwards, overexpressing and interfering HSV vector were produced, then verified by immunofluorescent staining and real-time quantitative polymerase chain reaction (qRT-PCR). The results of Tuj1 staining indicated that overexpression of BDNF could promote axonal regeneration and inhibit neuron swelling, whereas BDNF interference take an opposite effect after Oxygen glucose deprivation (OGD) injury. Finally, the interaction network among BDNF and associated proteins as examined by Genemania and confirmed by qRT-PCR. We found that the expression of VDAC1 was decreased and Stx1b was increased when BDNF overexpressing, which indicated that BDNF promoted neurite regrowth after OGD might be related to downregulation of VDAC1 and upregulation of Stx1b. Our results might provide novel strategy for the treatment of neurological defects induced by cerebral ischemia and hypoxia.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Cell Survival/drug effects , Genetic Therapy/methods , Hypoxia-Ischemia, Brain/therapy , Neurons/drug effects , Syntaxin 1/biosynthesis , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Animals , Animals, Newborn , Axons/drug effects , Brain-Derived Neurotrophic Factor/biosynthesis , Female , Glucose/deficiency , Nerve Regeneration/drug effects , Neurites , Pregnancy , Rats , Rats, Sprague-Dawley
4.
Biochem Biophys Res Commun ; 560: 52-58, 2021 06 30.
Article in English | MEDLINE | ID: mdl-33971568

ABSTRACT

Cisplatin is one of the most effective anti-cancer drugs, but its efficacy is limited by the development of resistance. Previous studies have shown that mitochondria play critical roles in cisplatin cytotoxicity, however, the exact mechanism of mitochondria involved in cisplatin sensitivity has not been clarified. In this study, cisplatin triggered mitochondrial oxidative stress and the decrease of mitochondria membrane potential in human cervical cancer cells. Then we screened a series of mitochondrial relevant inhibitors, including mitochondrial mPTP inhibitors DIDS and CsA, and mitochondrial respiratory complex inhibitors Rot and TTFA. Among these, only DIDS, as the inhibitor of mitochondrial outer membrane protein VDAC1, showed strong antagonism against cisplatin toxicity. DIDS mitigated cisplatin-induced MFN1-dependent mitochondrial fusion, mitochondrial dysfunction and oxidative damage. These findings demonstrated that VDAC1 may serve as a potential therapeutic target in the increase sensitivity of cisplatin, which provides an attractive pharmacological therapy to improve the effectiveness of chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Mitochondrial Dynamics/drug effects , Voltage-Dependent Anion Channel 1 , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , A549 Cells , Cell Survival/drug effects , HeLa Cells , Humans , MCF-7 Cells , Mitochondria/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects , Voltage-Dependent Anion Channel 1/antagonists & inhibitors
5.
Oxid Med Cell Longev ; 2020: 5950195, 2020.
Article in English | MEDLINE | ID: mdl-33082910

ABSTRACT

Endothelial injury plays a critical role in the pathogenesis of cardiovascular disorders and metabolic-associated vascular complications which are the leading cause of death worldwide. However, the mechanism underlying endothelial dysfunction is not completely understood. The study is aimed at investigating the role of tubulin polymerization-promoting protein family member 3 (TPPP3) in palmitic acid- (PA-) induced endothelial injury. The effect of TPPP3 on human umbilical vein endothelial cells (HUVECs) was determined by evaluating apoptosis, tube formation, and reactive oxygen species (ROS) production. TPPP3 silencing inhibited PA overload-induced apoptosis and production of ROS, along with the alteration of apoptosis-related key proteins such as BCL-2 and Bax. Mechanically, voltage-dependent anion channel 1 (VDAC1) was identified as a novel functional binding partner of TPPP3, and TPPP3 promoted VDAC1 protein stability and its activity. Further studies indicated that TPPP3 could promote apoptosis, ROS production, tube formation, and proapoptotic protein expression and reduce antiapoptotic protein expression through increasing VDAC1 expression under mildly elevated levels of PA. Collectively, these results demonstrated that TPPP3 could promote PA-induced oxidative damage in HUVECs via a VDAC1-dependent pathway, suggesting that TPPP3 might be considered as a potential therapeutic target in vascular disease.


Subject(s)
Cytoskeletal Proteins/metabolism , Reactive Oxygen Species/metabolism , Voltage-Dependent Anion Channel 1/metabolism , Apoptosis/drug effects , Cytoskeletal Proteins/antagonists & inhibitors , Cytoskeletal Proteins/genetics , Human Umbilical Vein Endothelial Cells , Humans , Neovascularization, Physiologic/drug effects , Oxidative Stress/drug effects , Palmitic Acid/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Up-Regulation/drug effects , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Voltage-Dependent Anion Channel 1/genetics , bcl-2-Associated X Protein/metabolism
6.
Biochem Pharmacol ; 177: 113995, 2020 07.
Article in English | MEDLINE | ID: mdl-32339494

ABSTRACT

Few discoveries have influenced drug discovery programs more than the finding that mitochondrial membranes undergo swings in permeability in response to cellular perturbations. The conductor of these permeability changes is the aptly named mitochondrial permeability transition pore which, although not yet precisely defined, is comprised of several integral proteins that differentially act to regulate the flux of ions, proteins and metabolic byproducts during the course of cellular physiological functions but also pathophysiological insults. Pursuit of the pore's exact identity remains a topic of keen interest, but decades of research have unearthed provocative functions for the integral proteins leading to their evaluation to develop novel therapeutics for a wide range of clinical indications. Chief amongst these targeted, integral proteins have been the Voltage Dependent Anion Channel (VDAC) and the F1FO ATP synthase. Research associated with the roles and ligands of VDAC has been extensive and we will expand upon 3 examples of ligand:VDAC interactions for consideration of drug discovery projects: Tubulin:VDAC1, Hexokinase I/II:VDAC1 and olesoxime:VDAC1. The discoveries that cyclosporine blocks mitochondrial permeability transition via binding to cyclophilin D, and that cyclophilin D is an important component of F1FO ATP synthase, has heightened interest in the F1FO ATP synthase as a focal point for drug discovery, and we will discuss 2 plausible campaigns associated with disease indications. To date no drug has emerged from prospective targeting these integral proteins; however, continued exploration such as the approaches suggested in this Commentary will increase the likelihood of providing important therapeutics for severely unmet medical needs.


Subject(s)
Amyotrophic Lateral Sclerosis/drug therapy , Cholestenones/therapeutic use , Cyclosporine/therapeutic use , Mitochondria/drug effects , Mitochondrial Membrane Transport Proteins/genetics , Voltage-Dependent Anion Channel 1/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , Cyclophilins/genetics , Cyclophilins/metabolism , Gene Expression Regulation , Hexokinase/genetics , Hexokinase/metabolism , Humans , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Proton-Translocating ATPases/antagonists & inhibitors , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/pathology , Permeability/drug effects , Protein Binding , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Tubulin/genetics , Tubulin/metabolism , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Voltage-Dependent Anion Channel 1/metabolism
7.
Int J Biol Sci ; 16(9): 1536-1550, 2020.
Article in English | MEDLINE | ID: mdl-32226300

ABSTRACT

Non-small cell lung cancer (NSCLC) with epidermal growth factor receptor (EGFR) wild-type is intrinsic resistance to EGFR-tyrosine kinase inhibitors (TKIs). In this study, we assessed whether the combination of bisdemethoxycurcumin (BDMC) and icotinib could surmount primary EGFR-TKI resistance in NSCLC cells and investigated its molecular mechanism. Results showed that the combination of BDMC and icotinib produced potently synergistic growth inhibitory effect on primary EGFR-TKI-resistant NSCLC cell lines H460 (EGFR wild-type and K-ras mutation) and H1781 (EGFR wild-type and Her2 mutation). Compared with BDMC or icotinib alone, the two drug combination induced more significant apoptosis and autophagy via suppressing EGFR activity and interaction of Sp1 and HDCA1/HDCA2, which was accompanied by accumulation of reactive oxygen species (ROS), induction of DNA damage, and inhibition of cell migration and invasion. ROS inhibitor (NAC) and autophagy inhibitors (CQ or 3-MA) partially reversed BDMC plus icotinib-induced growth inhibitory effect on the NSCLC cells. Meanwhile, co-treatment with NAC attenuated the two drug combination-induced autophagy, apoptosis, DNA damage and decrease of cell migration and invasion ability. Also, 3-MA or CQ can abate the combination treatment-induced apoptosis and DNA damage, suggesting that there is crosstalk between different signaling pathways in the effect produced by the combination treatment. Our data indicate that BMDC has the potential to improve the treatment of primary EGFR-TKI resistant NISCLC that cannot be controlled with single-target agent, such as icotinib.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Crown Ethers/therapeutic use , Diarylheptanoids/therapeutic use , Lung Neoplasms/drug therapy , Quinazolines/therapeutic use , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/drug effects , DNA Damage , Drug Resistance, Neoplasm/drug effects , Drug Synergism , ErbB Receptors/antagonists & inhibitors , Histone Deacetylase Inhibitors/therapeutic use , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Reactive Oxygen Species/metabolism , Sp Transcription Factors/antagonists & inhibitors , Voltage-Dependent Anion Channel 1/antagonists & inhibitors
8.
Oncogene ; 39(15): 3056-3074, 2020 04.
Article in English | MEDLINE | ID: mdl-32066881

ABSTRACT

The Bcl-xL apoptosis inhibitor plays a major role in vertebrate development. In addition to its effect on apoptosis, Bcl-xL is also involved in cell migration and mitochondrial metabolism. These effects may favour the onset and dissemination of metastasis. However, the underlying molecular mechanisms remain to be fully understood. Here we focus on the control of cell migration by Bcl-xL in the context of breast cancer cells. We show that Bcl-xL silencing led to migration defects in Hs578T and MDA-MB231 cells. These defects were rescued by re-expressing mitochondria-addressed, but not endoplasmic reticulum-addressed, Bcl-xL. The use of BH3 mimetics, such as ABT-737 and WEHI-539 confirmed that the effect of Bcl-xL on migration did not depend on interactions with BH3-containing death accelerators such as Bax or BH3-only proteins. In contrast, the use of a BH4 peptide that disrupts the Bcl-xL/VDAC1 complex supports that Bcl-xL by acting on VDAC1 permeability contributes to cell migration through the promotion of reactive oxygen species production by the electron transport chain. Collectively our data highlight the key role of Bcl-xL at the interface between cell metabolism, cell death, and cell migration, thus exposing the VDAC1/Bcl-xL interaction as a promising target for anti-tumour therapy in the context of metastatic breast cancer.


Subject(s)
Breast Neoplasms/pathology , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , bcl-X Protein/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Biphenyl Compounds/pharmacology , Biphenyl Compounds/therapeutic use , Breast/pathology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Disease Models, Animal , Female , Gene Knockdown Techniques , Humans , Lymphatic Metastasis/pathology , Mitochondria/drug effects , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Nitrophenols/pharmacology , Nitrophenols/therapeutic use , Piperazines/pharmacology , Piperazines/therapeutic use , Protein Binding/drug effects , RNA, Small Interfering/metabolism , Sulfonamides/pharmacology , Sulfonamides/therapeutic use , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Voltage-Dependent Anion Channel 1/metabolism , Xenograft Model Antitumor Assays , Zebrafish , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/genetics
9.
Biofactors ; 46(1): 83-93, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31618500

ABSTRACT

MicroRNAs have been reported to be implicated in myocardial ischemia/reperfusion (I/R) injury. The purpose of this study was to investigate the effect of miR-183-5p on I/R injury. Overexpression of miR-183-5p by agomiR transfection alleviated cardiac dysfunction and significantly reduced the infarct size in rats with myocardial I/R. MiR-183-5p also alleviated myocardial apoptosis with reduced apoptotic cells and lower levels of apoptosis associated proteins. in vitro experiments were conducted on rat H9c2 cells treated with anoxia/reoxygenation (A/R). Annexin V/propidium iodide (PI) staining and flow cytometry reported that the ratio of apoptotic cells decreased by miR-183-5p transfection before A/R treatment. Moreover, according to binding sequence prediction and Dual luciferase reporter assay, we explored that voltage-dependent anion channel 1 (VDAC1), which aggravates myocardial injury and apoptosis reported in our former research, was a target of miR-183-5p. In conclusion, miR-183-5p can efficiently attenuate I/R injury and miR-183-5p may exert its effect through repressing VDAC1 expression.


Subject(s)
MicroRNAs/metabolism , Myocardial Reperfusion Injury/prevention & control , Protective Agents/metabolism , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Animals , Apoptosis/genetics , Disease Models, Animal , Male , MicroRNAs/genetics , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Rats , Rats, Wistar , Voltage-Dependent Anion Channel 1/genetics
10.
Cell Mol Life Sci ; 77(18): 3611-3626, 2020 Sep.
Article in English | MEDLINE | ID: mdl-31760463

ABSTRACT

An intrinsically disordered neuronal protein α-synuclein (αSyn) is known to cause mitochondrial dysfunction, contributing to loss of dopaminergic neurons in Parkinson's disease. Through yet poorly defined mechanisms, αSyn crosses mitochondrial outer membrane and targets respiratory complexes leading to bioenergetics defects. Here, using neuronally differentiated human cells overexpressing wild-type αSyn, we show that the major metabolite channel of the outer membrane, the voltage-dependent anion channel (VDAC), is a pathway for αSyn translocation into the mitochondria. Importantly, the neuroprotective cholesterol-like synthetic compound olesoxime inhibits this translocation. By applying complementary electrophysiological and biophysical approaches, we provide mechanistic insights into the interplay between αSyn, VDAC, and olesoxime. Our data suggest that olesoxime interacts with VDAC ß-barrel at the lipid-protein interface thus hindering αSyn translocation through the VDAC pore and affecting VDAC voltage gating. We propose that targeting αSyn translocation through VDAC could represent a key mechanism for the development of new neuroprotective strategies.


Subject(s)
Cholestenones/pharmacology , Mitochondria/drug effects , Protective Agents/pharmacology , Voltage-Dependent Anion Channel 1/metabolism , alpha-Synuclein/metabolism , Apoptosis , Cell Line, Tumor , Cell Survival/drug effects , Humans , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Protein Binding , Protein Transport/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Reactive Oxygen Species/metabolism , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Voltage-Dependent Anion Channel 1/genetics , alpha-Synuclein/genetics
11.
Cancer Res ; 79(24): 6215-6226, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31582380

ABSTRACT

The regulators of mitochondrial cell death in cancer have remained elusive, hampering the development of new therapies. Here, we showed that protein isoforms of mitochondrial fission factor (MFF1 and MFF2), a molecule that controls mitochondrial size and shape, that is, mitochondrial dynamics, were overexpressed in patients with non-small cell lung cancer and formed homo- and heterodimeric complexes with the voltage-dependent anion channel-1 (VDAC1), a key regulator of mitochondrial outer membrane permeability. MFF inserted into the interior hole of the VDAC1 ring using Arg225, Arg236, and Gln241 as key contact sites. A cell-permeable MFF Ser223-Leu243 d-enantiomeric peptidomimetic disrupted the MFF-VDAC1 complex, acutely depolarized mitochondria, and triggered cell death in heterogeneous tumor types, including drug-resistant melanoma, but had no effect on normal cells. In preclinical models, treatment with the MFF peptidomimetic was well-tolerated and demonstrated anticancer activity in patient-derived xenografts, primary breast and lung adenocarcinoma 3D organoids, and glioblastoma neurospheres. These data identify the MFF-VDAC1 complex as a novel regulator of mitochondrial cell death and an actionable therapeutic target in cancer. SIGNIFICANCE: These findings describe mitochondrial fission regulation using a peptidomimetic agent that disturbs the MFF-VDAC complex and displays anticancer activity in multiple tumor models.See related commentary by Rao, p. 6074.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Membrane Proteins/metabolism , Mitochondria/pathology , Mitochondrial Dynamics/drug effects , Mitochondrial Proteins/metabolism , Animals , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Humans , Lung Neoplasms/drug therapy , Male , Membrane Proteins/antagonists & inhibitors , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/pathology , Mitochondrial Proteins/antagonists & inhibitors , Permeability/drug effects , Protein Multimerization/drug effects , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Voltage-Dependent Anion Channel 1/metabolism , Xenograft Model Antitumor Assays
12.
Bioorg Chem ; 91: 103146, 2019 10.
Article in English | MEDLINE | ID: mdl-31377389

ABSTRACT

Warburg hypothesized that the energy consumption of cancer cells is different than the normal cells. When compared to normal conditions, cancer cells do not undergo tricarboxylic acid (TCA) cycle therefore resulting in more lactate in the cells. Glycolysis pathway is a way of cancer cells to provide energy. The first step in glycolysis is the phosphorylation of glucose to glucose-6-phosphate. This reaction is catalyzed by the hexokinase-II enzyme (HK-II) which is known to be overexpressed in tumor cells. The feeding of cancer cells can be prevented by inhibiting the hexokinase-II enzyme in the first step of aerobic glycolysis. In literature, Methyl Jasmonate (MJ) is known as a Hexokinase-II inhibitor since it disposes VDAC and HK-II interaction on mitochondrial membrane. In our study, we aimed to increase the activity by synthesizing the novel MJ analogues with appropriate modifications. Here we report Hexokinase-2 enzyme and cell viability study results in different cancer cells. Based on the three different cancer cell lines we investigated, our novel MJ analogues proved to be more potent than the original molecule. Thus this research may provide more efficacious/novel HK-II inhibitors and may shed light to develop new anti-cancer agents.


Subject(s)
Acetates/chemistry , Acetates/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Cyclopentanes/chemistry , Cyclopentanes/pharmacology , Hexokinase/antagonists & inhibitors , Neoplasms/drug therapy , Oxylipins/chemistry , Oxylipins/pharmacology , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Glucose/metabolism , Glycolysis , Hexokinase/metabolism , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasms/pathology , Phosphorylation , Tumor Cells, Cultured , Voltage-Dependent Anion Channel 1/metabolism
13.
J Leukoc Biol ; 105(5): 935-946, 2019 05.
Article in English | MEDLINE | ID: mdl-30698851

ABSTRACT

Acute lymphoblastic leukemia (ALL) comprises a heterogeneous group of hematologic malignancies, arising from diverse genetic alterations in the early lymphocyte development. T-cell subtype of ALL (T-ALL) accounts for about 15% and 25% of ALL in children and adults, respectively. Being less frequent among ALL subtypes, T-ALL represents a high-risk factor for poor prognosis due to its aggressiveness and resistance to common antileukemic drugs. Mitochondria were widely explored recently as a target for anticancer treatment because they are involved in a metabolic reprogramming of a cancer cell and play key roles in reactive oxygen species generation, Ca2+ signaling, and cell death induction. Accordingly, a new class of anticancer compounds named mitocans has been developed, which target mitochondria at distinct crucial points to promote their dysfunction and subsequent cell death. The present review analyses the role of mitochondria in malignant reprogramming and emerging therapeutic strategies targeting mitochondria as an "Achilles' heel" in T-ALL, with an emphasis on BH3 mimetics, sequestering pro-survival BCL proteins and voltage-dependent anion channel (VDAC)1-directed drugs, which promote the suppression of aerobic glycolysis, VDAC1 closure, mitochondrial Ca2+ overload, stoppage of the oxidative phosphorylation, oxidative stress, and release of proapoptotic factors.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis Regulatory Proteins/genetics , Gene Expression Regulation, Leukemic , Mitochondria/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Voltage-Dependent Anion Channel 1/genetics , Adult , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis Regulatory Proteins/agonists , Apoptosis Regulatory Proteins/metabolism , Calcium/metabolism , Calcium Signaling , Cell Transformation, Neoplastic , Child , Glycolysis/drug effects , Glycolysis/genetics , Humans , Mitochondria/metabolism , Molecular Targeted Therapy/methods , Oxidative Phosphorylation/drug effects , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Prognosis , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Voltage-Dependent Anion Channel 1/metabolism
14.
Biochem Biophys Res Commun ; 505(4): 1203-1210, 2018 11 10.
Article in English | MEDLINE | ID: mdl-30327144

ABSTRACT

Sulindac sulfone is a metabolite of sulindac, a non-steroidal anti-inflammatory drug (NSAID), without anti-inflammatory ability. However, sulindac sulfone has been reported to significantly reduce polyps in patients with colorectal adenomatous polyposis in clinical trials. Thus, sulindac sulfone is expected to be useful for the chemoprevention of neoplasia with few side effects related to anti-inflammatory ability. To date, the molecular targets of sulindac sulfone have not yet fully investigated. Therefore, in order to newly identify sulindac sulfone-binding proteins, we generated sulindac sulfone-fixed FG beads and purified sulindac sulfone-binding proteins from human colon cancer HT-29 cells. we identified mitochondrial outer membrane proteins voltage-dependent anion channel (VDAC) 1 and VDAC2 as novel molecular targets of sulindac sulfone, and sulindac sulfone directly bound to both VDAC1 and VDAC2. Double knockdown of VDAC1 and VDAC2 by siRNA inhibited growth and arrested the cell cycle at G1 phase in HT-29 cells. Depletion of VDAC1 and VDAC2 also inhibited the mTORC1 pathway with a reduction in cyclin D1. Interestingly, these effects were consistent with those of sulindac sulfone against human colon cancer cells, suggesting that sulindac sulfone negatively regulates the function of VDAC1 and VDAC2. In the present study, our data suggested that VDAC1 and VDAC2 are direct targets of sulindac sulfone which suppresses the mTORC1 pathway and induces G1 arrest.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/pharmacology , Colonic Neoplasms/metabolism , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Sulindac/analogs & derivatives , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Voltage-Dependent Anion Channel 2/antagonists & inhibitors , Adenosine Triphosphate/biosynthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Cell Cycle Checkpoints , Colonic Neoplasms/pathology , HT29 Cells , Humans , Sulindac/chemistry , Sulindac/metabolism , Sulindac/pharmacology , Voltage-Dependent Anion Channel 1/metabolism , Voltage-Dependent Anion Channel 2/metabolism
15.
BMC Nephrol ; 19(1): 140, 2018 06 15.
Article in English | MEDLINE | ID: mdl-29907098

ABSTRACT

BACKGROUND: The mechanism of podocyte apoptosis is not fully understood. In addition, the role of the inositol 1,4,5-triphosphate receptor (IP3R)/glucose-regulated protein 75 (Grp75)/voltage-dependent anion channel 1 (VDAC1)/mitochondrial calcium uniporter (MCU) calcium regulation axis, which is located at sites of endoplasmic reticulum (ER) mitochondria coupling, in the mechanism of podocyte apoptosis is unclear. This study aimed to understand the roles of this axis in podocyte apoptosis and explore potential targets for podocyte protection. METHODS: The expression of IP3R, Grp75, VDAC1, and MCU and mitochondrial Ca2+ were analyzed during Adriamycin- or angiotensin II-induced apoptosis in cultured mouse podocytes. The interaction between IP3R, Grp75, and VDAC1 was investigated using co-immunoprecipitation experiments. The effects of IP3R, Grp75, and MCU agonists and antagonists on mitochondrial Ca2+ and apoptosis were investigated in cultured podocytes. The podocyte-protective effects of an MCU inhibitor were further investigated in rats with Adriamycin-induced nephropathy. RESULTS: Increased expression of IP3R, Grp75, VDAC1 and MCU, enhanced interaction among the IP3R-Grp75-VDAC1 complex, mitochondrial Ca2+ overload, and increased active caspase-3 levels were confirmed during Adriamycin- or angiotensin II-induced mouse podocyte apoptosis. Agonists of this axis facilitated mitochondrial Ca2+ overload and podocyte apoptosis, whereas specific antagonists against IP3R, Grp75, or MCU prevented mitochondrial Ca2+ overload and podocyte apoptosis. A specific MCU inhibitor prevented Adriamycin-induced proteinuria and podocyte foot process effacement in rats. CONCLUSIONS: This study identified a novel pathway in which the IP3R-Grp75-VDAC1-MCU calcium regulation axis mediated podocyte apoptosis by facilitating mitochondrial Ca2+ overload. Antagonists that inhibit Ca2+ transfer from ER to mitochondria protected mouse podocytes from apoptosis. An MCU inhibitor protected podocytes and decreased proteinuria in rats with Adriamycin-induced nephropathy. Therefore, antagonists to this pathway have promise as novel podocyte-protective drugs.


Subject(s)
Calcium/physiology , Doxorubicin/toxicity , Kidney Diseases/metabolism , Macrocyclic Compounds/pharmacology , Oxazoles/pharmacology , Podocytes/metabolism , Proteinuria/metabolism , Adenosylhomocysteinase/antagonists & inhibitors , Adenosylhomocysteinase/biosynthesis , Animals , Antibiotics, Antineoplastic/toxicity , Apoptosis/drug effects , Apoptosis/physiology , Calcium Channels/biosynthesis , Cells, Cultured , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/biosynthesis , Kidney Diseases/chemically induced , Kidney Diseases/drug therapy , Macrocyclic Compounds/therapeutic use , Male , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/biosynthesis , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Oxazoles/therapeutic use , Podocytes/drug effects , Proteinuria/drug therapy , Rats , Rats, Sprague-Dawley , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Voltage-Dependent Anion Channel 1/biosynthesis
16.
Neurosci Bull ; 33(6): 711-721, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29196874

ABSTRACT

Voltage-dependent anion channel 1 (VDAC1) is thought to contribute to the progression of tumor development. However, whether VDAC1 contributes to bone cancer pain remains unknown. In this study, we found that the expression of VDAC1 was upregulated in the L2-5 segments of the spinal dorsal horn at 2 and 3 weeks after injection of tumor cells into the tibial cavity. Intrathecal injection of a VDAC1 inhibitor significantly reversed the pain hypersensitivity and reduced the over-expression of Toll-like receptor 4 (TLR4). Intrathecal injection of minocycline, an inhibitor of microglia, also attenuated the pain hypersensitivity of rat models of bone cancer pain. These results suggest that VDAC1 plays a significant role in the development of complicated cancer pain, possibly by regulating the expression of TLR4.


Subject(s)
Bone Neoplasms/metabolism , Cancer Pain/metabolism , Hyperalgesia/metabolism , Microglia/metabolism , Spinal Cord Dorsal Horn/metabolism , Toll-Like Receptor 4/metabolism , Voltage-Dependent Anion Channel 1/metabolism , Animals , Cancer Pain/drug therapy , Disease Models, Animal , Female , Hyperalgesia/drug therapy , Lumbar Vertebrae , Microglia/drug effects , Rats , Rats, Sprague-Dawley , Spinal Cord Dorsal Horn/drug effects , Toll-Like Receptor 4/drug effects , Up-Regulation , Voltage-Dependent Anion Channel 1/antagonists & inhibitors
17.
Sci Rep ; 7(1): 14749, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29116138

ABSTRACT

Peripheral vascular occlusive disease (PVOD) is a common manifestation of atherosclerosis, and it has a high rate of morbidity. Therapeutic angiogenesis would re-establish blood perfusion and rescue ischemic tissue. Vascular endothelial growth factor (VEGF) induces angiogenesis and can potentially be used to treat ischemic diseases, yet in clinical trials VEGF has not fulfilled its full potential with side effects. Whether amino acids promote angiogenesis and the molecular mechanisms are largely unknown. Here we showed that (1) Glycine significantly promoted angiogenesis both in vitro and in vivo and effectively protected mitochondrial function. (2) Activation of glycine transporter 1(GlyT1) induced by VEGF led to an increase in intracellular glycine. (3) Glycine directly bounded to voltage dependent anion channel 1 (VDAC1) on the mitochondrial outer membrane and inhibited its opening. These original results highlight glycine as a necessary mediator in VEGF signalling via the GlyT1-glycine-mTOR-VDAC1 axis pathway. Therefore, the findings in this study are of significance providing new mechanistic insights into angiogenesis and providing better understanding of glycine function in angiogenesis, which may provide valuable information for development of novel therapeutic targets for the treatment of angiogenic vascular disorders.


Subject(s)
Glycine/metabolism , Neovascularization, Pathologic/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Animals , Glycine Plasma Membrane Transport Proteins/metabolism , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism
18.
Neuro Oncol ; 19(7): 951-964, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28339833

ABSTRACT

BACKGROUND: Glioblastoma (GBM), an aggressive brain tumor with frequent relapses and a high mortality, still awaits an effective treatment. Like many cancers, GBM cells acquire oncogenic properties, including metabolic reprogramming, vital for growth. As such, tumor metabolism is an emerging avenue for cancer therapy. One relevant target is the voltage-dependent anion channel 1 (VDAC1), a mitochondrial protein controlling cell energy and metabolic homeostasis. METHODS: We used VDAC1-specific short interfering (si)RNA (si-VDAC1) to treat GBM cell lines and subcutaneous or intracranial-orthotopic GBM xenograft mouse models. Tumors were monitored using MRI, immunohistochemistry, immunoblotting, immunofluorescence, quantitative real-time PCR, transcription factor expression, and DNA microarray analyses. RESULTS: Silencing VDAC1 expression using si-VDAC1 in 9 glioblastoma-related cell lines, including patient-derived cells, led to marked decreases in VDAC1 levels and cell growth. Using si-VDAC1 in subcutaneous or intracranial-orthotopic GBM models inhibited tumor growth and reversed oncogenic properties, such as reprogrammed metabolism, stemness, angiogenesis, epithelial-mesenchymal transition, and invasiveness. In cells in culture, si-VDAC1 inhibits cancer neurosphere formation and, in tumors, targeted cancer stem cells, leading to their differentiation into neuronal-like cells. These VDAC1 depletion-mediated effects involved alterations in transcription factors regulating signaling pathways associated with cancer hallmarks. CONCLUSION: VDAC1 offers a target for GBM treatment, allowing for attacks on the interplay between metabolism and oncogenic signaling networks, leading to tumor cell differentiation into neuron- and astrocyte-like cells. Simultaneously attacking all of these processes, VDAC1 depletion overcame GBM heterogeneity and can replace several anticancer drugs that separately target angiogenesis, proliferation, or metabolism.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Animals , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Cell Line, Tumor , Cell Proliferation , Gene Expression , Glioblastoma/genetics , Glioblastoma/therapy , Humans , Male , Mice, Nude , RNA, Small Interfering/administration & dosage , Transcriptome , Xenograft Model Antitumor Assays
19.
J Biol Chem ; 291(48): 24986-25003, 2016 Nov 25.
Article in English | MEDLINE | ID: mdl-27738100

ABSTRACT

Apoptosis is thought to play a critical role in several pathological processes, such as neurodegenerative diseases (i.e. Parkinson's and Alzheimer's diseases) and various cardiovascular diseases. Despite the fact that apoptotic mechanisms are well defined, there is still no substantial therapeutic strategy to stop or even slow this process. Thus, there is an unmet need for therapeutic agents that are able to block or slow apoptosis in neurodegenerative and cardiovascular diseases. The outer mitochondrial membrane protein voltage-dependent anion channel 1 (VDAC1) is a convergence point for a variety of cell survival and death signals, including apoptosis. Recently, we demonstrated that VDAC1 oligomerization is involved in mitochondrion-mediated apoptosis. Thus, VDAC1 oligomerization represents a prime target for agents designed to modulate apoptosis. Here, high-throughput compound screening and medicinal chemistry were employed to develop compounds that directly interact with VDAC1 and prevent VDAC1 oligomerization, concomitant with an inhibition of apoptosis as induced by various means and in various cell lines. The compounds protected against apoptosis-associated mitochondrial dysfunction, restoring dissipated mitochondrial membrane potential, and thus cell energy and metabolism, decreasing reactive oxidative species production, and preventing detachment of hexokinase bound to mitochondria and disruption of intracellular Ca2+ levels. Thus, this study describes novel drug candidates with a defined mechanism of action that involves inhibition of VDAC1 oligomerization, apoptosis, and mitochondrial dysfunction. The compounds VBIT-3 and VBIT-4 offer a therapeutic strategy for treating different diseases associated with enhanced apoptosis and point to VDAC1 as a promising target for therapeutic intervention.


Subject(s)
Apoptosis/drug effects , Mitochondria/metabolism , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Animals , Apoptosis/genetics , Calcium Signaling/drug effects , Calcium Signaling/genetics , Energy Metabolism/drug effects , Energy Metabolism/genetics , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Knockout , Mitochondria/genetics , Protein Multimerization/drug effects , Protein Multimerization/genetics , Rats , Voltage-Dependent Anion Channel 1/genetics , Voltage-Dependent Anion Channel 1/metabolism
20.
PLoS One ; 11(8): e0159529, 2016.
Article in English | MEDLINE | ID: mdl-27479079

ABSTRACT

To fully understand the glycolytic behavior of cancer cells, it is important to recognize how it is linked to pH dynamics. Here, we evaluated the acute effects of mild acidification and alkalization on cancer cell glucose uptake and glycolytic flux and investigated the role of hexokinase (HK). Cancer cells exposed to buffers with graded pH were measured for 18F-fluorodeoxyglucose (FDG) uptake, lactate production and HK activity. Subcellular localization of HK protein was assessed by western blots and confocal microscopy. The interior of T47D breast cancer cells was mildly alkalized to pH 7.5 by a buffer pH of 7.8, and this was accompanied by rapid increases of FDG uptake and lactate extrusion. This shift toward glycolytic flux led to the prompt recovery of a reversed pH gradient. In contrast, mild acidification rapidly reduced cellular FDG uptake and lactate production. Mild acidification decreased and mild alkalization increased mitochondrial HK translocation and enzyme activity. Cells transfected with specific siRNA against HK-1, HK-2 and voltage-dependent anion channel (VDAC)1 displayed significant attenuation of pH-induced changes in FDG uptake. Confocal microscopy showed increased co-localization of HK-1 and HK-2 with VDAC1 by alkaline treatment. In isolated mitochondria, acidic pH increased and alkaline pH decreased release of free HK-1 and HK-2 from the mitochondrial pellet into the supernatant. Furthermore, experiments using purified proteins showed that alkaline pH promoted co-immunoprecipitation of HK with VDAC protein. These findings demonstrate that mild alkalization is sufficient to acutely trigger cancer cell glycolytic flux through enhanced activity of HK by promoting its mitochondrial translocation and VDAC binding. This process might serve as a mechanism through which cancer cells trigger the Warburg effect to maintain a dysregulated pH.


Subject(s)
Hexokinase/metabolism , Voltage-Dependent Anion Channel 1/metabolism , Cell Line, Tumor , Fluorodeoxyglucose F18/metabolism , Glycolysis , Hexokinase/antagonists & inhibitors , Hexokinase/genetics , Humans , Hydrogen-Ion Concentration , Immunoprecipitation , Lactic Acid/metabolism , Microscopy, Confocal , Mitochondria/metabolism , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Voltage-Dependent Anion Channel 1/antagonists & inhibitors , Voltage-Dependent Anion Channel 1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...