Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Mol Pharmacol ; 100(4): 309-318, 2021 10.
Article in English | MEDLINE | ID: mdl-34312217

ABSTRACT

Microtubule-targeting agents (MTAs), including both microtubule stabilizers and destabilizers are highly effective chemotherapeutic drugs used in the treatment of solid tumors and hematologic malignancies. In addition to the shared ability of all MTAs to block cell cycle progression, growing evidence shows that different agents of this class can also have mechanistically distinct effects on nonmitotic microtubule-dependent cellular processes, including cellular signaling and transport. Herein, we test the biologic hypothesis that MTAs used in the treatment of triple-negative breast cancer (TNBC) can differentially affect innate immune signaling pathways independent of their antimitotic effects. Our data demonstrate that the microtubule destabilizer eribulin, but not the microtubule stabilizer paclitaxel, induces cGAS-STING-dependent expression of interferon-ß in both myeloid and TNBC cells. Activation of the cGAS-STING pathway by eribulin was further found to be mediated by the accumulation of cytoplasmic mitochondrial DNA. Together, these findings provide mechanistic insight into how eribulin can induce innate immune signaling independent of its antimitotic or cytotoxic effects. SIGNIFICANCE STATEMENT: Microtubule-targeting agents (MTAs) are often used in the treatment of breast cancer and have been used in combination with immune checkpoint inhibitors to improve efficacy. Although all clinically approved MTAs share an antimitotic mechanism of action, their distinct effects on interphase microtubules can promote differential downstream signaling consequences. This work shows that the microtubule destabilizer eribulin, but not the microtubule stabilizer paclitaxel, activates the cGAS-STING innate immune signaling pathway through the accumulation of mitochondrial DNA in the cytoplasm.


Subject(s)
Cytoplasm/metabolism , DNA, Mitochondrial/metabolism , Furans/pharmacology , Ketones/pharmacology , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Signal Transduction/drug effects , Animals , Cell Line, Tumor , Cytoplasm/drug effects , Humans , Immunity, Innate/drug effects , Immunity, Innate/physiology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Microtubules/drug effects , Microtubules/metabolism , Signal Transduction/physiology
2.
J Nat Prod ; 84(3): 750-761, 2021 03 26.
Article in English | MEDLINE | ID: mdl-33226219

ABSTRACT

A fundamental factor in natural product drug discovery programs is the necessity to identify the active component(s) from complex chemical mixtures. Whereas this has traditionally been accomplished using bioassay-guided fractionation, we questioned whether alternative techniques could supplement and, in some cases, even supplant this approach. We speculated that a combination of ligand-fishing methods and modern analytical tools (e.g., LC-MS and online natural product databases) offered a route to enhance natural product drug discovery. Herein, a candidate solution referred to as the lickety-split ligand-affinity-based molecular angling system (LLAMAS) is described. This approach utilizes an ultrafiltration-based LC-PDA-MS/MS-guided DNA-binding assay in combination with the (i) Global Natural Products Social Molecular Networking, (ii) Dictionary of Natural Products, and (iii) SciFinder platforms to identify DNA binders in complex chemical mixtures. LLAMAS was initially vetted in tests using known small-molecule DNA binders and then optimized to a 96-well plate-based format. A set of 332 plant samples used in traditional Chinese medicine was screened for DNA-binding activity with LLAMAS, resulting in the identification of seven DNA-binding molecules, including berberine (12), palmatine (13), coptisine (14), fangchinoline (15), tetrandrine (16), daurisoline (17), and dauricine (18). These results demonstrate that LLAMAS is an effective natural product discovery platform for the efficient identification and dereplication of DNA-binding molecules from complex mixtures.


Subject(s)
Biological Products/chemistry , DNA/chemistry , Drug Discovery/methods , Chromatography, Liquid , Tandem Mass Spectrometry , Ultrafiltration
3.
J Nat Prod ; 83(10): 3080-3092, 2020 10 23.
Article in English | MEDLINE | ID: mdl-33021790

ABSTRACT

There are no targeted therapies available for triple-negative breast cancers (TNBCs) in part because they represent a heterogeneous group of tumors with diverse oncogenic drivers. Our goal is to identify targeted therapies for subtypes of these cancers using a mechanism-blind screen of natural product extract libraries. An extract from Desmanthodium guatemalense was 4-fold more potent for cytotoxicity against MDA-MB-231 cells, which represent the mesenchymal stem-like (MSL) subtype, as compared to cells of other TNBC subtypes. Bioassay-guided fractionation led to the isolation of six polyacetylenes, and subsequent investigations of plant sources known to produce polyacetylenes yielded six additional structurally related compounds. A subset of these compounds retained selective cytotoxic effects in MSL subtype cells. Studies suggest that these selective effects do not appear to be due to PPARγ agonist activities that have previously been reported for polyacetylenes. A CRISPR-Cas9-mediated gene knockout screen was employed to identify the mechanism of selective cytotoxic activity of the most potent and selective compound, dehydrofalcarinol (1a). This genomic screen identified HSD17B11, the gene encoding the enzyme 17ß-hydroxysteroid dehydrogenase type 11, as a mediator of the selective cytotoxic effects of 1a in MDA-MB-231 cells that express high levels of this protein. The Project Achilles cancer dependency database further identified a subset of Ewing sarcoma cell lines as highly dependent on HSD17B11 expression, and it was found these were also highly sensitive to 1a. This report demonstrates the value of CRISPR-Cas9 genome-wide screens to identify the mechanisms underlying the selective activities of natural products.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , CRISPR-Cas Systems , Gene Knockout Techniques/methods , Neoplastic Stem Cells/drug effects , Triple Negative Breast Neoplasms/drug therapy , 17-Hydroxysteroid Dehydrogenases/drug effects , 17-Hydroxysteroid Dehydrogenases/genetics , Aldehyde Oxidoreductases/drug effects , Aldehyde Oxidoreductases/genetics , Cell Line, Tumor , Female , Humans , Molecular Structure , PPAR gamma/agonists , RNA, Small Interfering/pharmacology
4.
J Nat Prod ; 83(3): 584-592, 2020 03 27.
Article in English | MEDLINE | ID: mdl-32105068

ABSTRACT

An extract prepared from the fruit of Choerospondias axillaris exhibited differential cytotoxic effects when tested in a panel of pediatric cancer cell lines [Ewing sarcoma (A-673), rhabdomyosarcoma (SJCRH30), medulloblastoma (D283), and hepatoblastoma (Hep293TT)]. Bioassay-guided fractionation led to the purification of five new hydroquinone-based metabolites, choerosponols A-E (1-5), bearing unsaturated hydrocarbon chains. The structures of the natural products were determined using a combination of 1D and 2D NMR, HRESIMS, ECD spectroscopy, and Mosher ester analyses. The purified compounds were evaluated for their antiproliferative and cytotoxic activities, revealing that 1, which contains a benzofuran moiety, exhibited over 50-fold selective antiproliferative activity against Ewing sarcoma and medulloblastoma cells with growth inhibitory (GI50) values of 0.19 and 0.07 µM, respectively. The effects of 1 were evaluated in a larger panel of cancer cell lines, and these data were used in turn to interrogate the Project Achilles cancer dependency database, leading to the identification of the MCT1 transporter as a functional target of 1. These data highlight the utility of publicly available cancer dependency databases such as Project Achilles to facilitate the identification of the mechanisms of action of compounds with selective activities among cancer cell lines, which can be a major challenge in natural products drug discovery.


Subject(s)
Anacardiaceae/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Plant Extracts/pharmacology , Cell Line, Tumor , Fruit/chemistry , Humans , Molecular Structure , Phytochemicals/pharmacology , Vietnam
5.
Mol Carcinog ; 58(1): 102-112, 2019 01.
Article in English | MEDLINE | ID: mdl-30302860

ABSTRACT

Despite effective surgical methods for non-melanoma skin cancer (NMSC), patients suffer from tissue damage, scarring, or even disfigurement; thus, there is a need for chemopreventive approaches. Because of the complex interplay between glucocorticoids (GCs), inflammation, and cancer, we sought to determine the role of 11ß-hydroxysteroid dehydrogenase 1 and 2 (11ßHSD1 and 2) in regulating GCs during skin cancer development and progression. 11ßHSDs modulate the activation of GCs in a tissue-specific manner and have been reported to play a role in development and progression of other types of cancer, but their role has not yet been reported in NMSC. Here, we found a significant upregulation of 11ßHSD2 protein in skin cancer cells when compared to normal skin cells, suggesting a role for this enzyme in the multifactorial process of skin cancer development. In addition, inhibition of 11ßHSD2 with siRNA resulted in significant reduction in colony formation in vitro. Finally, our in vivo study elucidated that inhibition of 11ßHSD2 with pharmacological inhibitor, Glycyrrhetinic acid (GA) could significantly diminish tumorigenesis in a well-studied in vivo mouse model of NMSC. Overall, these studies highlight for the first time a potential novel role for 11ßHSD2 in NMSC development and may allow for new GC treatment approaches capable of avoiding deactivation by the enzyme. If 11ßHSD2 can be inhibited as we have done here, or circumvented using modified GCs, this may lead to more efficacious outcomes for NMSC patients by preventing deactivation of the GC and minimizing resistance.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenases/antagonists & inhibitors , Anti-Inflammatory Agents/pharmacology , Gene Expression Regulation, Enzymologic/drug effects , Glucocorticoids/pharmacology , Glycyrrhetinic Acid/pharmacology , Skin Neoplasms/prevention & control , Animals , Apoptosis , Cell Proliferation , Female , Humans , Mice , Skin Neoplasms/enzymology , Skin Neoplasms/pathology , Tumor Cells, Cultured
6.
Mol Carcinog ; 57(12): 1698-1706, 2018 12.
Article in English | MEDLINE | ID: mdl-30129681

ABSTRACT

The phytonutrient ursolic acid (UA), present in apples, rosemary, and other plant sources, has anti-cancer properties in a number of systems, including skin cancers. However, few reports have examined upstream mechanisms by which UA may prevent or treat cancer. Recent reports have indicated UA induces death of cancer cell lines via AMP-activated protein kinase (AMPK), an energy-sensing kinase which possesses both pro-metabolic and anti-cancer effects. Other studies have shown UA activates peroxisome proliferator activated receptor α (PPARα) and the glucocorticoid receptor (GR). Here, we found the cytotoxic effect of UA in skin carcinoma cells required AMPK activation. In addition, two inhibitors of PPARα partially reversed the cytotoxic effects of UA, suggesting its effects are at least partially mediated through this receptor. Finally, inhibition of the GR did not reverse the effects of UA nor did this compound bind the GR under the conditions of experiments performed. Overall, studies elucidating the anti-cancer effects of UA may allow for the development of more potent analogues utilizing similar mechanisms. These studies may also reveal the mediators of any possible side effects or resistance mechanisms to UA therapy.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , PPAR alpha/metabolism , Skin Neoplasms/metabolism , Triterpenes/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Signal Transduction/drug effects , Skin Neoplasms/drug therapy , Ursolic Acid
7.
Biomacromolecules ; 19(11): 4270-4276, 2018 11 12.
Article in English | MEDLINE | ID: mdl-30231201

ABSTRACT

Cationic polymers have emerged as appealing nonviral gene vectors for decades, which, however, suffer from the paradox between low molecular weight and high transfection efficacy. Low molecular weight cationic polymers (LCPs) are well cell tolerated but are perplexed by orders-of-magnitude less efficacy compared to their macromolecular counterparts. The deficiency mainly lies in weak DNA binding of polymers and difficulty in endosomal escape of formulated polyplexes. Herein, we demonstrate that, through zinc (Zn) coordinated modification of LCPs, the high transfection efficiency and low molecular weight (thus low cytotoxicity) can be achieved simultaneously. The Zn coordinated ligand shows a high affinity to phosphate components and therefore will largely benefit the DNA packaging and endosomal membrane destabilization, addressing the defects of LCPs in gene delivery. Zn coordinative functionalization of LCPs breaks up the "efficacy-toxicity" paradox and provides great promise for the development of clinically efficient and safe nonviral gene vectors.


Subject(s)
Cations/chemistry , Colonic Neoplasms/therapy , Erythrocytes/metabolism , Mesenchymal Stem Cells/metabolism , Polymers/administration & dosage , Transfection/methods , Zinc/chemistry , Animals , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Erythrocytes/cytology , Female , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors/administration & dosage , Humans , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred BALB C , Molecular Weight , Polymers/chemistry , Sheep , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Mol Carcinog ; 53(9): 753-63, 2014 Sep.
Article in English | MEDLINE | ID: mdl-23625588

ABSTRACT

Glucocorticoids (GCs) are well-known anti-inflammatory compounds, but they also inhibit cell proliferation depending on cell type. Similarly, peroxisome proliferator-activated receptors (PPARα, PPARδ, and PPARγ) also possess anti-proliferation properties beyond their canonical roles as metabolic mediators. In the present study, we investigated the potential additive or synergistic inhibitory effects on cancer cell proliferation by simultaneous application of fenofibrate and budesonide, agonists for PPARα and glucocorticoid receptor, respectively. We observed differential effects on cell proliferation in A549 and SK-MES-1 lung cancer cells by budesonide and fenofibrate. Fenofibrate inhibited cell proliferation in both TP53 wild type and deficient lung cancer cells. The anti-proliferation effect of budesonide in TP53 wild type A549 cells was abolished in SK-MES-1 cells that do not have wild type TP53 protein. An additive effect against cell proliferation by budesonide and fenofibrate combination was observed only in TP53 wild type A549 cancer cells. Analysis of cell cycle distribution and cyclin profile indicated that the inhibition of cell proliferation was associated with G1 cell cycle arrest. The suppression of NF-κB activity and ERK signaling may contribute to the inhibition of cell proliferation by budesonide and or fenofibrate. The additive inhibitory effect on cell proliferation by budesonide and fenofibrate combination suggests that the same or greater therapeutic effect could be achieved with reduced dosage and side effects when the two compounds are applied simultaneously.


Subject(s)
Adenocarcinoma/drug therapy , Budesonide/pharmacology , Cell Proliferation/drug effects , Fenofibrate/pharmacology , Lung Neoplasms/drug therapy , PPAR alpha/agonists , Receptors, Glucocorticoid/agonists , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Anti-Inflammatory Agents/pharmacology , Blotting, Western , Cell Cycle/drug effects , Flow Cytometry , Humans , Hypolipidemic Agents/pharmacology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , NF-kappa B/genetics , NF-kappa B/metabolism , PPAR alpha/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Glucocorticoid/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
9.
Muscle Nerve ; 44(6): 947-56, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22102466

ABSTRACT

INTRODUCTION: Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease. We sought to determine whether peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) would have a beneficial effect on this disease. METHODS: PGC-1α transgenic mice were crossed with SOD1 mutant G93A DL mice. RESULTS: We observed a moderate but non-significant increase in average lifespan in PGC-1α/G93A DL mice, as compared with G93A DL mice (292 ± 3 days vs. 274 ± 7 days). Although the onset of ALS was not altered, progression of the disease was significantly slower (≈34% increase in duration) in the PGC-1α/G93A DL mice. These mice also exhibited markedly improved performance on the rotarod test, and the improved motor activity was associated with a decreased loss of motor neurons and less degeneration of neuromuscular junctions. CONCLUSION: A sustained level of excitatory amino acid transporter protein 2 (EAAT2) in astrocytes of the PGC-1α/G93A DL mice may contribute to neuronal protection.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Disease Models, Animal , Disease Progression , Neurons/metabolism , Trans-Activators/genetics , Alanine/genetics , Amino Acid Substitution/genetics , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Female , Glycine/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Transcription Factors
10.
Cancers (Basel) ; 13(11)2021 Jun 03.
Article in English | MEDLINE | ID: mdl-34205051

ABSTRACT

Extracellular vesicles play a central role in intercellular communication and contribute to cancer progression, including the epithelial-to-mesenchymal transition (EMT). Microtubule targeting agents (MTAs) including eribulin and paclitaxel continue to provide significant value in cancer therapy and their abilities to inhibit oncogenic signaling pathways, including eribulin's capacity to reverse EMT are being revealed. Because microtubules are involved in the intracellular trafficking required for the formation and cargo loading of small extracellular vesicles (sEVs), we investigated whether MTA-mediated disruption of microtubule-dependent transport would impact sEV release and their cargo. Eribulin and paclitaxel caused an intracellular accumulation of CD63, a tetraspanin component of sEVs, in late/multivesicular endosomes of triple-negative breast cancer cells, consistent with the disruption of endosomal sorting and exosome cargo loading in these cells. While the concentrations of sEVs released from MTA-treated cells were not significantly altered, levels of CD63 and the CD63-associated cargos, ILK and ß-integrin, were reduced in sEVs isolated from eribulin-treated HCC1937 cells as compared to vehicle or paclitaxel-treated cells. These results show that eribulin can reduce specific sEV cargos, including ILK, a major transducer of EMT in the tumor microenvironment, which may contribute to eribulin's ability to reverse EMT to promote anticancer efficacy.

11.
RSC Adv ; 10(56): 34203-34214, 2020.
Article in English | MEDLINE | ID: mdl-32944226

ABSTRACT

The structures of several aquaglyceroporins have been resolved to atomic resolution showing two or more glycerols bound inside a channel and confirming a glycerol-facilitator's affinity for its substrate glycerol. However, the kinetics data of glycerol transport experiments all point to unsaturated transport that is characteristic of low substrate affinity in terms of the Michaelis-Menten kinetics. In this article, we present an in silico-in vitro research focused on AQP3, one of the human aquaglyceroporins that is natively expressed in the abundantly available erythrocytes. We conducted 2.1 µs in silico simulations of AQP3 embedded in a model erythrocyte membrane with intracellular-extracellular asymmetries in leaflet lipid compositions and compartment salt ions. From the equilibrium molecular dynamics (MD) simulations, we elucidated the mechanism of glycerol transport at high substrate concentrations. From the steered MD simulations, we computed the Gibbs free-energy profile throughout the AQP3 channel. From the free-energy profile, we quantified the kinetics of glycerol transport that is unsaturated due to glycerol-glycerol interactions mediated by AQP3 resulting in the concerted movement of two glycerol molecules for the transport of one glycerol molecule across the cell membrane. We conducted in vitro experiments on glycerol uptake into human erythrocytes for a wide range of substrate concentrations and various temperatures. The experimental data quantitatively validated our theoretical-computational conclusions on the unsaturated glycerol transport through AQP3 that has high affinity for glycerol.

12.
RSC Adv ; 10(36): 21283-21291, 2020.
Article in English | MEDLINE | ID: mdl-32612811

ABSTRACT

Modelling water and membrane lipids is an essential element in the computational research of biophysical/biochemical processes such as water transport across the cell membrane. In this study, we examined the accuracies of two popular water models, TIP3P and TIP4P, in the molecular dynamics simulations of erythrocyte aquaporins (AQP1 and AQP3). We modelled the erythrocyte membrane as an asymmetric lipid bilayer with appropriate lipid compositions of its inner and outer leaflet, in comparison with a symmetric lipid bilayer of a single lipid type. We computed the AQP1/3 permeabilities with the transition state theory with full correction for recrossing events. We also conducted cell swelling assays for water transport across the erythrocyte membrane. The experimental results agree with the TIP3P water-erythrocyte membrane model, in confirmation of the expected accuracy of the erythrocyte membrane model, the TIP3P water model, and the CHARMM parameters for water-protein interactions.

13.
Biochim Biophys Acta Biomembr ; 1861(4): 768-775, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30659792

ABSTRACT

For its fundamental relevance, transport of water and glycerol across the erythrocyte membrane has long been investigated before and after the discovery of aquaporins (AQPs), the membrane proteins responsible for water and glycerol transport. AQP1 is abundantly expressed in the human erythrocyte for maintaining its hydrohomeostasis where AQP3 is also expressed (at a level ~30-folds lower than AQP1) facilitating glycerol transport. This research is focused on two of the remaining questions: How permeable is AQP3 to water? What is the glycerol-AQP3 affinity under near-physiological conditions? Through atomistic modelling and large-scale simulations, we found that AQP3 is two to three times more permeable to water than AQP1 and that the glycerol-AQP3 affinity is approximately 500/M. Using these computed values along with the data from the latest literature on AQP1 and on erythrocyte proteomics, we estimated the water and glycerol transport rates across the membrane of an entire erythrocyte. We used these rates to predict the time courses of erythrocyte swelling-shrinking in response to inward and outward osmotic gradients. Experimentally, we monitored the time course of human erythrocytes when subject to an osmotic or glycerol gradient with light scattering in a stopped-flow spectrometer. We observed close agreement between the experimentally measured and the computationally predicted time courses of erythrocytes, which corroborated our computational conclusions on the AQP3 water-permeability and the glycerol-AQP3 affinity.


Subject(s)
Aquaporin 3/chemistry , Erythrocyte Membrane/chemistry , Glycerol/chemistry , Aquaporin 3/metabolism , Cell Membrane Permeability , Erythrocyte Membrane/metabolism , Glycerol/metabolism , Humans
14.
Physiol Genomics ; 34(1): 112-26, 2008 Jun 12.
Article in English | MEDLINE | ID: mdl-18445702

ABSTRACT

How higher organisms respond to elevated oxidative stress in vivo is poorly understood. Therefore, we measured oxidative stress parameters and gene expression alterations (Affymetrix arrays) in the liver caused by elevated reactive oxygen species induced in vivo by diquat or by genetic ablation of the major antioxidant enzymes CuZn-superoxide dismutase (Sod1) and glutathione peroxidase-1 (Gpx1). Diquat (50 mg/kg) treatment resulted in a significant increase in oxidative damage within 3-6 h in wild-type mice without any lethality. In contrast, treatment of Sod1(-/-) or Gpx1(-/-) mice with a similar concentration of diquat resulted in a significant increase in oxidative damage within an hour of treatment and was lethal, i.e., these mice are extremely sensitive to the oxidative stress generated by diquat. The expression response to elevated oxidative stress in vivo does not involve an upregulation of classic antioxidant genes, although long-term oxidative stress in Sod1(-/-) mice leads to a significant upregulation of thiol antioxidants (e.g., Mt1, Srxn1, Gclc, Txnrd1), which appears to be mediated by the redox-sensitive transcription factor Nrf2. The main finding of our study is that the common response to elevated oxidative stress with diquat treatment in wild-type, Gpx1(-/-), and Sod1(-/-) mice and in untreated Sod1(-/-) mice is an upregulation of p53 target genes (p21, Gdf15, Plk3, Atf3, Trp53inp1, Ddit4, Gadd45a, Btg2, Ndrg1). A retrospective comparison with previous studies shows that induction of these p53 target genes is a conserved expression response to oxidative stress, in vivo and in vitro, in different species and different cells/organs.


Subject(s)
Gene Expression Profiling , Oxidative Stress/genetics , Animals , Antioxidants/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , DNA/metabolism , Diquat/toxicity , Gene Expression Regulation/drug effects , Glutathione Peroxidase/deficiency , Lipid Peroxidation/drug effects , Liver Diseases/enzymology , Liver Diseases/genetics , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Oligonucleotide Array Sequence Analysis , Oxidative Stress/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Superoxide Dismutase/deficiency , Tumor Suppressor Protein p53/metabolism , Glutathione Peroxidase GPX1
15.
ACS Chem Neurosci ; 9(11): 2815-2823, 2018 11 21.
Article in English | MEDLINE | ID: mdl-29865792

ABSTRACT

Fourteen glucose transporters (GLUTs) play essential roles in human physiology by facilitating glucose diffusion across the cell membrane. Due to its central role in the energy metabolism of the central nervous system, GLUT3 has been thoroughly investigated. However, the Gibbs free-energy gradient (what drives the facilitated diffusion of glucose) has not been mapped out along the transport path. Some fundamental questions remain. Here we present a molecular dynamics study of GLUT3 embedded in a lipid bilayer to quantify the free-energy profile along the entire transport path of attracting a ß-d-glucose from the interstitium to the inside of GLUT3 and, from there, releasing it to the cytoplasm by Arrhenius thermal activation. From the free-energy profile, we elucidate the unique Michaelis-Menten characteristics of GLUT3, low KM and high VMAX, specifically suitable for neurons' high and constant demand of energy from their low-glucose environments. We compute GLUT3's binding free energy for ß-d-glucose to be -4.6 kcal/mol in agreement with the experimental value of -4.4 kcal/mol ( KM = 1.4 mM). We also compute the hydration energy of ß-d-glucose, -18.0 kcal/mol vs the experimental data, -17.8 kcal/mol. In this, we establish a dynamics-based connection from GLUT3's crystal structure to its cellular thermodynamics with quantitative accuracy. We predict equal Arrhenius barriers for glucose uptake and efflux through GLUT3 to be tested in future experiments.


Subject(s)
Energy Metabolism , Glucose Transporter Type 3/metabolism , Glucose/metabolism , Lipid Bilayers/metabolism , Thermodynamics , Facilitated Diffusion , Humans , Molecular Dynamics Simulation
16.
ACS Macro Lett ; 7(7): 868-874, 2018 Jul 17.
Article in English | MEDLINE | ID: mdl-35650761

ABSTRACT

Amines have been extensively involved in vector design thus far, however, their clinical translation has been impeded by several obstacles: cytotoxicity, polyplex serum instability and low efficacy in vivo. In pursuit of functional groups to substitute amines in vector design to address these disadvantages is of great significance. Herein, we report well-tailored noncationic copolymers that contain hydrophilic, hydrophobic, and zinc coordinative moieties through reversible addition-fragmentation chain transfer (RAFT) polymerization for efficient and safe gene delivery. These polymers are capable of condensing DNA, enabling the formation of uncharged polyplexes. Especially, the zinc coordinative ligand can simultaneously benefit strong DNA binding, robust cellular uptake, efficacious endosomal destabilization, low cytotoxicity, and avoidance of serum protein adsorption. The coordinative module holds great promise to substitute amines and inspires the development of next-generation gene vectors. More importantly, the coordinative copolymers illuminate the possibility and potential of noncationic gene delivery systems for clinical applications.

17.
Ann N Y Acad Sci ; 1100: 264-79, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17460188

ABSTRACT

Peroxisome proliferation activator receptor (PPAR) gamma-coactivator 1alpha (PGC-1alpha), a transcription coactivator, functions as a master regulator of a wide array of metabolic and physiological processes and is an essential factor in the process of mitochondrial biogenesis. Transfection of NIH 3T3 fibroblasts with a mouse cDNA for PGC-1alpha led to the induction of markers of mitochondrial biogenesis, that is, mitochondrial transcription factor A (mtTFA), cytochrome c, and mitochondrial DNA (mtDNA). Mitochondrial biogenesis-associated net protein synthesis appears to be accomplished by a reduction in the rate of mitochondrial protein degradation with little or no change in the rate of protein synthesis. Overexpression of PGC-1alpha did not adversely affect cellular proliferation. Cellular ATP levels were increased in the transfected cells and they were more resistant to oxidative stress than the control nontransfected 3T3 cells. This resistance to oxidative stress was manifested by both an improved viability and the maintenance of mitochondrial membrane potential in the transfected cells when exposed to t-butyl hydroperoxide (t-BOOH). It therefore appears that PGC-1alpha overexpression stimulates mitochondrial biogenesis in 3T3 cells making them more resistant to oxidative stressors.


Subject(s)
Fibroblasts/metabolism , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Trans-Activators/physiology , 3T3 Cells , Adenosine Triphosphate/metabolism , Animals , Cell Proliferation , Cell Survival , DNA, Mitochondrial/metabolism , Gene Expression Regulation , Mice , Oxidative Stress , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Transcription Factors , Transfection , tert-Butylhydroperoxide/pharmacology
18.
Adv Physiol Educ ; 30(4): 145-51, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17108241

ABSTRACT

Peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1alpha is a member of a family of transcription coactivators that plays a central role in the regulation of cellular energy metabolism. It is strongly induced by cold exposure, linking this environmental stimulus to adaptive thermogenesis. PGC-1alpha stimulates mitochondrial biogenesis and promotes the remodeling of muscle tissue to a fiber-type composition that is metabolically more oxidative and less glycolytic in nature, and it participates in the regulation of both carbohydrate and lipid metabolism. It is highly likely that PGC-1alpha is intimately involved in disorders such as obesity, diabetes, and cardiomyopathy. In particular, its regulatory function in lipid metabolism makes it an inviting target for pharmacological intervention in the treatment of obesity and Type 2 diabetes.


Subject(s)
Energy Metabolism/physiology , Heat-Shock Proteins/physiology , Transcription Factors/physiology , Adaptation, Physiological/physiology , Diabetes Mellitus, Type 2/etiology , Glucose/metabolism , Heart/growth & development , Heat-Shock Proteins/metabolism , Humans , Muscle Fibers, Skeletal/physiology , Muscle, Skeletal/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Thermogenesis/physiology , Transcription Factors/metabolism
19.
Mol Cancer Ther ; 3(11): 1385-96, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15542777

ABSTRACT

Irofulven (hydroxymethylacylfulvene) is a novel antitumor drug, which acts by alkylating cellular macromolecular targets. The drug is a potent inducer of apoptosis in various types of tumor cells, whereas it is nonapoptotic in normal cells. This study defined molecular responses to irofulven involving mitochondrial dysfunction and leading to death of prostate tumor LNCaP-Pro5 cells. Irofulven caused early (2-5 hours) translocation of the proapoptotic Bax from cytosol to mitochondria followed by the dissipation of mitochondrial membrane potential and cytochrome c release at 4 to 12 hours. These effects preceded caspase activation and during the first 6 hours were not affected by caspase inhibitors. Processing of caspase-9 initiated the caspase cascade at approximately 6 hours and progressed over time. The activation of the caspase cascade provided a positive feedback loop that enhanced Bcl-2-independent translocation and cytochrome c release. General and specific caspase inhibitors abrogated irofulven-induced apoptotic DNA fragmentation with the following order of potency: pan-caspase > or = caspase-9 > caspase-8/6 > caspase-2 > caspase-3/7 > caspase-1/4. Abrogation of caspase-mediated DNA fragmentation failed to salvage irofulven-treated cells from growth inhibition and loss of viability, demonstrating a substantial contribution of a caspase-independent cell death. Monobromobimane, an inhibitor of alternative caspase-independent apoptotic pathway that is mediated by mitochondrial permeability transition, antagonized both apoptosis, measured as phosphatidylserine externalization, and cytotoxicity of irofulven. Collectively, the results indicate that irofulven-induced signaling is integrated at the level of mitochondrial dysfunction. The induction of both caspase-dependent and caspase-independent death pathways is consistent with pleiotropic effects of irofulven, which include targeting of cellular DNA and proteins.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Enzyme Inhibitors/pharmacology , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/pathology , Sesquiterpenes/pharmacology , Bridged Bicyclo Compounds/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cytochromes c/metabolism , DNA Fragmentation/drug effects , Enzyme Inhibitors/chemistry , Humans , Male , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Structure , Prostatic Neoplasms/metabolism , Protein Transport/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Sesquiterpenes/chemistry , Signal Transduction/drug effects , bcl-2-Associated X Protein
20.
Melanoma Res ; 25(2): 103-12, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25647735

ABSTRACT

Malignant melanoma is associated with a 5-year survival rate of less than 20% once metastasized. Malignant melanoma cells exhibit increased levels of autophagy, a process of intracellular digestion that allows cells to survive various stresses including chemotherapies, resulting in reduced patient survival. Autophagy can be inhibited by chemicals like chloroquine (CQ), which prevents fusion of autophagosomes to lysosomes, resulting in autophagosome accumulation in most systems. Here, we describe how tested CQ to see whether it could sensitize B16F10 metastatic mouse melanoma cells to the anticancer activities of the natural compounds ursolic acid (UA) and resveratrol (RES). CQ with UA or RES strongly and synergistically reduced the viability of B16F10 mouse melanoma and A375 human melanoma cells. Surprisingly, flow cytometry of acridine orange-stained cells showed that UA or RES in combination with CQ significantly reduced autophagosome levels. Western blotting analysis revealed that CQ plus UA or RES paradoxically increased LC3II, indicative of autophagosome accumulation. In addition, CQ plus RES synergistically decreased the levels of both autophagy initiator beclin-1 and autophagy supporter p62. These results indicate that CQ with UA or RES strongly and synergistically reduces the viability of B16F10 and A375 melanoma cells. However, studies on B16F10 cells have shown that the synergistic effect was not mediated by inhibition of autophagy induced by UA or RES. These compounds are well-tolerated in humans, and CQ has shown promise as an adjuvant therapy. These combinations may be valuable treatment strategies for melanoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Chloroquine/pharmacology , Melanoma, Experimental/drug therapy , Skin Neoplasms/drug therapy , Stilbenes/pharmacology , Triterpenes/pharmacology , Animals , Autophagy/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Drug Synergism , Humans , Melanoma, Experimental/pathology , Mice , Resveratrol , Signal Transduction/drug effects , Skin Neoplasms/pathology , Ursolic Acid
SELECTION OF CITATIONS
SEARCH DETAIL