Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 115
Filter
Add more filters

Publication year range
1.
Biochemistry ; 63(8): 984-999, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38569593

ABSTRACT

Ferroptosis is a recently identified form of regulated cell death, characterized by excessive iron-dependent lipid peroxidation. Recent studies have demonstrated that protein disulfide isomerase (PDI) is an important mediator of chemically induced ferroptosis and also a new target for protection against ferroptosis-associated cell death. In the present study, we identified that 4-hydroxyestrone (4-OH-E1), a metabolic derivative of endogenous estrogen, is a potent small-molecule inhibitor of PDI, and can strongly protect against chemically induced ferroptotic cell death in the estrogen receptor-negative MDA-MB-231 human breast cancer cells. Pull-down and CETSA assays demonstrated that 4-OH-E1 can directly bind to PDI both in vitro and in intact cells. Computational modeling analysis revealed that 4-OH-E1 forms two hydrogen bonds with PDI His256, which is essential for its binding interaction and thus inhibition of PDI's catalytic activity. Additionally, PDI knockdown attenuates the protective effect of 4-OH-E1 as well as cystamine (a known PDI inhibitor) against chemically induced ferroptosis in human breast cancer cells. Importantly, inhibition of PDI by 4-OH-E1 and cystamine or PDI knockdown by siRNAs each markedly reduces iNOS activity and NO accumulation, which has recently been demonstrated to play an important role in erastin-induced ferroptosis. In conclusion, this study demonstrates that 4-OH-E1 is a novel inhibitor of PDI and can strongly inhibit ferroptosis in human breast cancer cells in an estrogen receptor-independent manner. The mechanistic understanding gained from the present study may also aid in understanding the estrogen receptor-independent cytoprotective actions of endogenous estrogen metabolites in many noncancer cell types.


Subject(s)
Breast Neoplasms , Hydroxyestrones , Piperazines , Protein Disulfide-Isomerases , Humans , Female , Protein Disulfide-Isomerases/chemistry , Breast Neoplasms/drug therapy , Cystamine , Cell Death , Estrogens , Receptors, Estrogen
2.
Cell Commun Signal ; 22(1): 431, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39243059

ABSTRACT

Recently we have shown that protein disulfide isomerase (PDI or PDIA1) is involved in mediating chemically-induced, glutathione (GSH) depletion-associated ferroptotic cell death through NOS activation (dimerization) and NO accumulation. The present study aims to determine the role of PDI in mediating chemically-induced hepatocyte injury in vitro and in vivo and whether PDI inhibitors can effectively protect against chemically-induced hepatocyte injury. We show that during the development of erastin-induced ferroptotic cell death, accumulation of cellular NO, ROS and lipid-ROS follows a sequential order, i.e., cellular NO accumulation first, followed by accumulation of cellular ROS, and lastly cellular lipid-ROS. Cellular NO, ROS and lipid-ROS each play a crucial role in mediating erastin-induced ferroptosis in cultured hepatocytes. In addition, it is shown that PDI is an important upstream mediator of erastin-induced ferroptosis through PDI-mediated conversion of NOS monomer to its dimer, which then leads to accumulation of cellular NO, ROS and lipid-ROS, and ultimately ferroptotic cell death. Genetic manipulation of PDI expression or pharmacological inhibition of PDI function each can effectively abrogate erastin-induced ferroptosis. Lastly, evidence is presented to show that PDI is also involved in mediating acetaminophen-induced liver injury in vivo using both wild-type C57BL/6J mice and hepatocyte-specific PDI conditional knockout (PDIfl/fl Alb-cre) mice. Together, our work demonstrates that PDI is an important upstream mediator of chemically-induced, GSH depletion-associated hepatocyte ferroptosis, and inhibition of PDI can effectively prevent this injury.


Subject(s)
Glutathione , Hepatocytes , Protein Disulfide-Isomerases , Reactive Oxygen Species , Protein Disulfide-Isomerases/metabolism , Protein Disulfide-Isomerases/genetics , Hepatocytes/metabolism , Hepatocytes/drug effects , Hepatocytes/pathology , Animals , Glutathione/metabolism , Reactive Oxygen Species/metabolism , Mice , Mice, Inbred C57BL , Piperazines/pharmacology , Ferroptosis/drug effects , Nitric Oxide/metabolism , Male , Humans
3.
J Chem Inf Model ; 63(11): 3230-3237, 2023 06 12.
Article in English | MEDLINE | ID: mdl-37235532

ABSTRACT

Protein-Protein binding affinity reflects the binding strength between the binding partners. The prediction of protein-protein binding affinity is important for elucidating protein functions and also for designing protein-based therapeutics. The geometric characteristics such as area (both interface and surface areas) in the structure of a protein-protein complex play an important role in determining protein-protein interactions and their binding affinity. Here, we present a free web server for academic use, AREA-AFFINITY, for prediction of protein-protein or antibody-protein antigen binding affinity based on interface and surface areas in the structure of a protein-protein complex. AREA-AFFINITY implements 60 effective area-based protein-protein affinity predictive models and 37 effective area-based models specific for antibody-protein antigen binding affinity prediction developed in our recent studies. These models take into consideration the roles of interface and surface areas in binding affinity by using areas classified according to different amino acid types with different biophysical nature. The models with the best performances integrate machine learning methods such as neural network or random forest. These newly developed models have superior or comparable performance compared to the commonly used existing methods. AREA-AFFINITY is available for free at: https://affinity.cuhk.edu.cn/.


Subject(s)
Machine Learning , Proteins , Protein Binding , Proteins/chemistry , Amino Acids/metabolism , Computers
4.
Acta Biochim Biophys Sin (Shanghai) ; 55(5): 853-865, 2023 May 30.
Article in English | MEDLINE | ID: mdl-37249331

ABSTRACT

Ferroptosis is a new form of nonapoptotic cell death closely associated with glutathione (GSH) peroxidase 4 inhibition and/or GSH depletion, resulting in the accumulation of cellular iron and lipid peroxides. The exact mechanism by which GSH depletion causes the accumulation of reactive oxygen species (ROS) and lipid-ROS and subsequent ferroptotic cell death in neuronal cells remains unclear. In the present study, using immortalized HT22 mouse hippocampal neuronal cells as a model, we show that nitric oxide (NO) accumulation via protein disulfide isomerase (PDI)-mediated neuronal nitric oxide synthase (nNOS) activation plays a critical role in chemically-induced ferroptosis. Mechanistically, we find that erastin-induced GSH depletion leads to activation of PDI, which then mediates ferroptosis by catalyzing nNOS dimerization, followed by accumulation of cellular NO, ROS and lipid ROS and ultimately ferroptotic cell death. Pharmacological inhibition of PDI enzymatic activity or selective PDI knockdown can effectively abrogate erastin-induced ferroptosis in HT22 cells. The results of this study reveal an important role of PDI in mediating chemically induced ferroptosis in a neuronal cell model, and PDI may serve as a potential drug target for protection against GSH depletion-associated ferroptotic neuronal cell death.


Subject(s)
Lipids , Mice , Animals , Reactive Oxygen Species/metabolism , Lipid Peroxidation , Cell Death
5.
Genes Chromosomes Cancer ; 61(4): 177-186, 2022 04.
Article in English | MEDLINE | ID: mdl-34687488

ABSTRACT

Anaplastic lymphoma kinase (ALK)-rearranged non-small cell lung cancer (NSCLC) respond well to ALK tyrosine kinase inhibitors (TKIs), and echinoderm microtubule-associated protein-like 4 (EML4)-ALK-rearranged NSCLC accounts for the majority of those patients. However, few studies have evaluated ALK-TKIs treatment for patients with huntingtin-interacting protein 1 (HIP1)-ALK fusions. This retrospective study evaluated the clinicopathological characteristics, genomic features, response to ALK-TKIs, and resistance mechanisms in 11 cases with HIP1-ALK fusions from five Chinese centers. Patients who received crizotinib at the Chinese centers had an objective response rate of 90% [9/10 cases, 95% confident index (CI): 54.1%-99.5%], median progression-free survival of 17.9 months (95% CI: 5.8-NA months), and median overall survival of 58.8 months (95% CI: 24.7-NA months). One patient who received first-line lorlatinib treatment achieved partial response for > 26.5 months. Despite the small sample size, HIP1-ALK (H21:A20) variant was the most common variant (four of 11 cases, 36.4%) and associated with better outcomes. Among the 11 cases, there were eight patients having available specimens for genetic testing before ALK-TKIs treatment and four patients undergoing biopsy after ALK-TKIs failure. The most common coexisting gene was TP53 among 11 patients and two of four patients after crizotinib failure harbored acquired ALK mutations (e.g., L1152V/Q1146K and L1196M). Brigatinib treatment appeared to be effective for a patient who failed crizotinib treatment because of the L1152V/Q1146K mutations, which might be related to increased binding affinity to these mutants. Although HIP1-ALK-rearranged NSCLC appears to initially respond well to ALK-TKIs, crizotinib resistance may be correlated with the AKAP9-BRAF fusion, ALK compound mutations (L1152V/Q1146K), and the ALK L1196M mutation. Larger studies are needed to evaluate the significance of HIP1-ALK-rearranged NSCLC.


Subject(s)
Anaplastic Lymphoma Kinase/genetics , Carcinoma, Non-Small-Cell Lung/genetics , DNA-Binding Proteins/genetics , Drug Resistance, Neoplasm , Gene Rearrangement , Lung Neoplasms/genetics , Oncogene Proteins, Fusion/genetics , Activin Receptors, Type II , Adult , Aged , Carcinoma, Non-Small-Cell Lung/pathology , Crizotinib/therapeutic use , Female , Humans , Immunoglobulin Fc Fragments , Lung Neoplasms/pathology , Male , Middle Aged , Protein Kinase Inhibitors/therapeutic use , Recombinant Fusion Proteins , Retrospective Studies , Survival Analysis
6.
Acta Pharmacol Sin ; 43(10): 2527-2541, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35347247

ABSTRACT

Oxidative stress is extensively involved in neurodegeneration. Clinical evidence shows that keeping the mind active through mentally-stimulating physical activities can effectively slow down the progression of neurodegeneration. With increased physical activities, more neurotransmitters would be released in the brain. In the present study, we investigated whether some of the released neurotransmitters might have a beneficial effect against oxidative neurodegeneration in vitro. Glutamate-induced, glutathione depletion-associated oxidative cytotoxicity in HT22 mouse hippocampal neuronal cells was used as an experimental model. We showed that norepinephrine (NE, 50 µM) or dopamine (DA, 50 µM) exerted potent protective effect against glutamate-induced cytotoxicity, but this effect was not observed when other neurotransmitters such as histamine, γ-aminobutyric acid, serotonin, glycine and acetylcholine were tested. In glutamate-treated HT22 cells, both NE and DA significantly suppressed glutathione depletion-associated mitochondrial dysfunction including mitochondrial superoxide accumulation, ATP depletion and mitochondrial AIF release. Moreover, both NE and DA inhibited glutathione depletion-associated MAPKs activation, p53 phosphorylation and GADD45α activation. Molecular docking analysis revealed that NE and DA could bind to protein disulfide isomerase (PDI). In biochemical enzymatic assay in vitro, NE and DA dose-dependently inhibited the reductive activity of PDI. We further revealed that the protective effect of NE and DA against glutamate-induced oxidative cytotoxicity was mediated through inhibition of PDI-catalyzed dimerization of the neuronal nitric oxide synthase. Collectively, the results of this study suggest that NE and DA may have a protective effect against oxidative neurodegeneration through inhibition of protein disulfide isomerase and the subsequent activation of the MAPKs‒p53‒GADD45α oxidative cascade.


Subject(s)
Cell Death , Dopamine , Neuroprotection , Norepinephrine , Protein Disulfide-Isomerases , Acetylcholine/pharmacology , Adenosine Triphosphate/metabolism , Animals , Cell Death/drug effects , Dopamine/pharmacology , Glutamic Acid/metabolism , Glutathione/metabolism , Glycine/pharmacology , Histamine/metabolism , Mice , Molecular Docking Simulation , Neuroprotection/drug effects , Neurotransmitter Agents , Nitric Oxide Synthase Type I/metabolism , Norepinephrine/pharmacology , Oxidative Stress , Protein Disulfide-Isomerases/drug effects , Protein Disulfide-Isomerases/metabolism , Serotonin/metabolism , Serotonin/pharmacology , Superoxides/metabolism , Superoxides/pharmacology , Tumor Suppressor Protein p53/metabolism , gamma-Aminobutyric Acid/metabolism
7.
Acta Biochim Biophys Sin (Shanghai) ; 54(4): 415-451, 2022 Apr 25.
Article in English | MEDLINE | ID: mdl-35607958

ABSTRACT

Hyperglycemia in diabetic patients is associated with abnormally-elevated cellular glucose levels. It is hypothesized that increased cellular glucose will lead to increased formation of endogenous methanol and/or formaldehyde, both of which are then metabolically converted to formic acid. These one-carbon metabolites are known to be present naturally in humans, and their levels are increased under diabetic conditions. Mechanistically, while formaldehyde is a cross-linking agent capable of causing extensive cytotoxicity, formic acid is an inhibitor of mitochondrial cytochrome oxidase, capable of inducing histotoxic hypoxia, ATP deficiency and cytotoxicity. Chronic increase in the production and accumulation of these toxic one-carbon metabolites in diabetic patients can drive the pathogenesis of ocular as well as other diabetic complications. This hypothesis is supported by a large body of experimental and clinical observations scattered in the literature. For instance, methanol is known to have organ- and species-selective toxicities, including the characteristic ocular lesions commonly seen in humans and non-human primates, but not in rodents. Similarly, some of the diabetic complications (such as ocular lesions) also have a characteristic species-selective pattern, closely resembling methanol intoxication. Moreover, while alcohol consumption or combined use of folic acid plus vitamin B is beneficial for mitigating acute methanol toxicity in humans, their use also improves the outcomes of diabetic complications. In addition, there is also a large body of evidence from biochemical and cellular studies. Together, there is considerable experimental support for the proposed hypothesis that increased metabolic formation of toxic one-carbon metabolites in diabetic patients contributes importantly to the development of various clinical complications.


Subject(s)
Diabetes Mellitus , Diabetic Retinopathy , Animals , Carbon , Diabetic Retinopathy/etiology , Formaldehyde , Formates , Glucose , Humans , Methanol/metabolism
8.
Acta Biochim Biophys Sin (Shanghai) ; 53(7): 848-869, 2021 Jul 05.
Article in English | MEDLINE | ID: mdl-33987639

ABSTRACT

Insulin receptor plays an important role in the regulation of energy metabolism. Dysfunction of insulin receptor (IR) can lead to many disease states, such as diabetes mellitus. Deciphering the complex dynamic structures of human IR and its mechanism of activation would greatly aid in understanding IR-mediated signaling pathways and also in designing new drugs (including nonpeptidal insulin analogs) to treat diabetes mellitus. Experimental evidence about IR structures has been gradually obtained by biologists over the past three decades. Based on available experimental structures of IR in different states, here we employ molecular modeling approach to construct the full-length IR structures in different states and model its structural and conformational changes during insulin-induced IR activation. Several key possible intermediate states are constructed based on structural alignment, rotation, and computational modeling. Based on the structures of the full-length IR in different states, it appears that there are two possible conformational transition pathways: one is symmetric and the other one is asymmetric. Structural changes and motions of different domains of the full-length IR along the pathways are analyzed. The role of insulin binding to IR in facilitating the conformational transition of the receptor is analyzed. Information and insights derived from our present structural modeling analyses may aid in understanding the complex dynamic, structural, and conformational changes during the process of IR activation.


Subject(s)
Insulin/chemistry , Models, Molecular , Receptor, Insulin/chemistry , Humans , Protein Structure, Quaternary
9.
Molecules ; 27(1)2021 Dec 30.
Article in English | MEDLINE | ID: mdl-35011450

ABSTRACT

In the kidney, prostaglandins formed by cyclooxygenase 1 and 2 (COX-1 and COX-2) play an important role in regulating renal blood flow. In the present study, we report our observations regarding a unique modulatory effect of renal microsomal preparation on COX-1/2-mediated formation of major prostaglandin (PG) products in vitro. We found that microsomes prepared from pig and rat kidneys had a dual stimulatory-inhibitory effect on the formation of certain PG products catalyzed by COX-1 and COX-2. At lower concentrations, kidney microsomes stimulated the formation of certain PG products, whereas at higher concentrations, their presence inhibited the formation. Presence of kidney microsomes consistently increased the Km values of the COX-1/2-mediated reactions, while the Vmax might be increased or decreased depending on stimulation or inhibition observed. Experimental evidence was presented to show that a protein component present in the pig kidney microsomes was primarily responsible for the activation of the enzyme-catalyzed arachidonic acid metabolism leading to the formation of certain PG products.


Subject(s)
Kidney/metabolism , Microsomes/metabolism , Prostaglandins/chemical synthesis , Animals , Arachidonic Acid/chemistry , Arachidonic Acid/metabolism , Catalysis , In Vitro Techniques , Kinetics , Prostaglandin-Endoperoxide Synthases/chemistry , Prostaglandin-Endoperoxide Synthases/metabolism , Rats , Swine
10.
Biochem Biophys Res Commun ; 491(4): 870-875, 2017 09 30.
Article in English | MEDLINE | ID: mdl-28709872

ABSTRACT

The insulin-like growth factor 1 receptor (IGF-1R) is a disulfide-linked heterotetramer containing two α-subunits and two ß-subunits. Earlier studies demonstrate that nitric oxide (NO) can adversely affect IGF-1 action in the central nervous system. It is known that NO can induce S-nitrosylation of the cysteine residues in proteins, thereby partly contributing to the regulation of protein function. In the present study, we sought to determine whether S-nitrosylation of the cysteine residues in IGF-1R is an important post-translational modification that regulates its response to IGF-1. Using cultured SH-SY5Y human neuroblastoma cells as an in vitro model, we found that treatment of cells with S-nitroso-cysteine (SNOC), a NO donor that can nitrosylate the cysteine residues in proteins, induces S-nitrosylation of the ß subunit of IGF-1R but not its α-subunit. IGF-1Rß S-nitrosylation by SNOC is coupled with increased dissociation of the IGF-1R protein complex. In addition, disruption of the IGF-1R function resulting from S-nitrosylation of the IGF-1Rß subunit is associated with disruption of the phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) signaling pathways. Further, we observed that SNOC-induced IGF-1Rß S-nitrosylation results in a dose-dependent inhibition of cell proliferation and survival. Together, these results suggest that elevated nitrosative stress may result in dysfunction of cellular IGF-1R signaling through S-nitrosylation of the cysteine residues in the IGF-1Rß subunit, thereby disrupting the downstream PI3K and MAPK signaling functions and ultimately resulting in inhibition of cell proliferation and survival.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Nitric Oxide/metabolism , Receptor, IGF Type 1/metabolism , Cell Proliferation/drug effects , Cysteine/analogs & derivatives , Cysteine/pharmacology , Dose-Response Relationship, Drug , Humans , S-Nitrosothiols/pharmacology , Structure-Activity Relationship , Tumor Cells, Cultured
11.
Chem Res Toxicol ; 30(7): 1448-1462, 2017 07 17.
Article in English | MEDLINE | ID: mdl-28616971

ABSTRACT

In animal models, estrogens are complete carcinogens in certain target sites. 4-Hydroxyestradiol (4-OH-E2), an endogenous metabolite of 17ß-estradiol (E2), is known to have prominent estrogenic activity plus potential genotoxicity and mutagenicity. We report here our finding that 4-OH-E2 does not induce pituitary tumors in ACI female rats, whereas E2 produces 100% pituitary tumor incidence. To probe the mechanism, we conducted a short-term animal experiment to compare the proliferative effect of 4-OH-E2 in several organs. We found that, whereas 4-OH-E2 had little ability to stimulate pituitary cell proliferation in ovariectomized female rats, it strongly stimulates cell proliferation in certain brain regions of these animals. Further, when we used in vitro cultured rat pituitary tumor cells as models, we found that 4-OH-E2 has similar efficacy as E2 in stimulating cell proliferation, but its potency is approximately 3 orders of magnitude lower than that of E2. Moreover, we found that the pituitary tumor cells have the ability to selectively metabolize 4-OH-E2 (but not E2) with ultrahigh efficiency. Additional analysis revealed that the rat pituitary expresses a membrane-bound catechol-O-methyltransferase that has an ultralow Km value (in nM range) for catechol estrogens. On the basis of these observations, it is concluded that rapid metabolic disposition of 4-OH-E2 through enzymatic O-methylation in rat anterior pituitary cells largely contributes to its apparent lack of cell proliferative and tumorigenic effects in this target site.


Subject(s)
Catechol O-Methyltransferase/metabolism , Estrogens, Catechol/pharmacology , Pituitary Gland, Anterior/drug effects , Pituitary Gland, Anterior/metabolism , Animals , Biocatalysis , Carcinogenesis/drug effects , Cell Proliferation/drug effects , Estrogens, Catechol/chemistry , Female , Humans , Methylation , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/enzymology , Rats , Rats, Inbred ACI , Tumor Cells, Cultured
12.
Biochem Biophys Res Commun ; 477(3): 495-502, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27317486

ABSTRACT

Glutathione depletion is a distinct cause underlying many forms of pathogenesis associated with oxidative stress and cytotoxicity. Earlier studies showed that glutamate-induced glutathione depletion in immortalized murine HT22 hippocampal neuronal cells leads to accumulation of reactive oxygen species (ROS) and ultimately cell death, but the precise mechanism underlying these processes is not clear. Here we show that during the induction of glutathione depletion, nitric oxide (NO) accumulation precedes ROS accumulation. While neuronal NO synthase (nNOS) in untreated HT22 cells exists mostly as a monomer, glutathione depletion results in increased formation of the dimer nNOS, accompanied by increases in the catalytic activity. We identified that nNOS dimerization is catalyzed by protein disulfide isomerase (PDI). Inhibition of PDI's isomerase activity effectively abrogates glutathione depletion-induced conversion of monomer nNOS into dimer nNOS, accumulation of NO and ROS, and cytotoxicity. Furthermore, we found that PDI is present in untreated cells in an inactive S-nitrosylated form, which becomes activated following glutathione depletion via S-denitrosylation. These results reveal a novel role for PDI in mediating glutathione depletion-induced oxidative cytotoxicity, as well as its role as a valuable therapeutic target for protection against oxidative cytotoxicity.


Subject(s)
Glutathione/metabolism , Protein Disulfide-Isomerases/metabolism , Animals , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism
13.
Am J Physiol Endocrinol Metab ; 308(5): E370-9, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25516546

ABSTRACT

Despite increased total food intake in healthy, late-stage pregnant women, their peak postprandial blood sugar levels are normally much lower than the levels seen in healthy nonpregnant women. In this study, we sought to determine whether estriol (E3), an endogenous estrogen predominantly produced during human pregnancy, contributes to the regulation of the postprandial blood glucose level in healthy normal rats. In vivo studies using rats showed that E3 blunted the speed and magnitude of the blood glucose rise following oral glucose administration, but it did not appear to affect the total amount of glucose absorbed. E3 also did not affect insulin secretion, but it significantly reduced the rate of intestinal glucose transport compared with vehicle-treated animals. Consistent with this finding, expression of the sodium-dependent glucose transporter 1 and 2 was significantly downregulated by E3 treatment in the brush-border membrane and basolateral membrane, respectively, of enterocytes. Most of the observed in vivo effects were noticeably stronger with E3 than with 17ß-estradiol. Using differentiated human Caco-2 enterocyte monolayer culture as an in vitro model, we confirmed that E3 at physiologically relevant concentrations could directly inhibit glucose uptake via suppression of glucose transporter 2 expression, whereas 17ß-estradiol did not have a similar effect. Collectively, these data showed that E3 can blunt the postprandial glycemic surge in rats through modulating the level of intestinal glucose transporters.


Subject(s)
Blood Glucose/drug effects , Estriol/pharmacology , Glucose Transporter Type 2/genetics , Intestines/drug effects , Postprandial Period/drug effects , Sodium-Glucose Transporter 1/genetics , Animals , Blood Glucose/metabolism , Caco-2 Cells , Estradiol/pharmacology , Gene Expression Regulation/drug effects , Glucose Transporter Type 2/metabolism , Humans , Insulin/blood , Intestinal Mucosa/metabolism , Male , Rats , Rats, Sprague-Dawley , Sodium-Glucose Transporter 1/metabolism
14.
Planta Med ; 81(10): 838-46, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26132849

ABSTRACT

The present study investigates the anticancer effect of ascorbate in MIA-PaCa-2 human pancreatic cancer cells using both in vitro and in vivo models, with a focus on assessing the role of oxidative stress and autophagy as important mechanistic elements in its anticancer actions. We showed that ascorbate suppresses the growth of human pancreatic cancer cells via the induction of oxidative stress and caspase-independent cell death. Ascorbate induces the formation of autophagosomes and the presence of autophagy inhibitors suppresses ascorbate-induced cell death. These data suggest that the induction of autophagosome formation contributes to ascorbate-induced pancreatic cancer cell death.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/metabolism , Ascorbic Acid/pharmacology , Autophagy/drug effects , Membrane Proteins/metabolism , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Beclin-1 , Cell Death/drug effects , Cell Line, Tumor/drug effects , Female , Humans , Membrane Proteins/genetics , Mice, Nude , Microtubule-Associated Proteins/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , RNA, Small Interfering , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
15.
Mol Carcinog ; 53(2): 125-37, 2014 Feb.
Article in English | MEDLINE | ID: mdl-22949227

ABSTRACT

Treatment of cancer cells with microtubule inhibitors causes mitotic arrest, which subsequently leads to cell death via activation of the intrinsic apoptotic pathway. Mitotically arrested cells typically display increased phosphorylation (i.e., inactivation) of two key anti-apoptotic proteins, Bcl-2 and Bcl-XL , but the mechanisms that regulate their phosphorylation as well as their role in apoptotic cell death following mitotic arrest are still poorly understood at present, which are the focus of this study. We recently showed that cyclin B1 and cell division cycle 2 (Cdc2) proteins are strongly up-regulated in human breast cancer cells following treatment with nocodazole (a prototypical microtubule inhibitor), and their up-regulation plays a critical role in the development of mitotic prometaphase arrest. In this study, we present evidence showing that the up-regulated cyclin B1/Cdc2 complex in nocodazole-treated human breast cancer cells is also responsible for the increased phosphorylation of Bcl-2 and Bcl-XL . However, only the increased phosphorylation of Bcl-XL , but not the phosphorylation of Bcl-2, contributes to subsequent activation of the intrinsic cell death pathway. In addition, evidence is presented to show that mitotic arrest deficient 2 (MAD2) is a key upstream mediator of the up-regulation of cyclin B1/Cdc2 as well as the subsequent increase in phosphorylationof Bcl-2 and Bcl-XL in nocodazole-treated cancer cells. Together, these results reveal that the up-regulated cyclin B1/Cdc2 complex not only mediates prometaphase arrest in nocodazole-treated cells, but also activates the subsequent intrinsic cell death pathway in these cells via increased phosphorylation of Bcl-XL .


Subject(s)
Apoptosis/genetics , Cell Cycle Checkpoints/genetics , Cyclin B1/genetics , Cyclin B/genetics , Mitosis/genetics , Phosphorylation/genetics , bcl-X Protein/genetics , Apoptosis/drug effects , CDC2 Protein Kinase , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinases , Humans , MCF-7 Cells , Mad2 Proteins/genetics , Mitosis/drug effects , Nocodazole/pharmacology , Phosphorylation/drug effects , Prometaphase/drug effects , Prometaphase/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Up-Regulation/drug effects
16.
Free Radic Biol Med ; 223: 473-492, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38992393

ABSTRACT

Ferroptosis is a form of iron-dependent regulated cell death which is different from apoptosis. Chemically-induced ferroptosis is characterized by an accumulation of lipid reactive oxygen species (ROS) in the cells. A number of earlier studies have suggested the involvement of mitochondrial ROS in ferroptosis, and the present study seeks to further investigate the role of mitochondrial ROS in the induction of chemically-induced ferroptotic cell death. We find that during erastin-induced, glutathione depletion-associated ferroptosis, mitochondrial ROS accumulation is an important late event, which likely is involved in the final execution of ferroptotic cell death. The mitochondrion-originated ROS is found to accumulate in large quantities inside the nuclei during the late phases of erastin-induced ferroptosis. Completion of the late-phase accumulation of mitochondrion-produced ROS inside the nucleus of a cell likely marks an irreversible point in the cell death process. Similarly, accumulation of large amounts of mitochondrion-produced ROS inside the nucleus is also observed in the late phases of RSL3-induced ferroptosis. The results of this study indicate that the mitochondrial ROS play an important role in the final steps of both erastin- and RSL3-induced ferroptotic cell death.


Subject(s)
Ferroptosis , Mitochondria , Piperazines , Reactive Oxygen Species , Ferroptosis/drug effects , Reactive Oxygen Species/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Humans , Piperazines/pharmacology , Glutathione/metabolism , Iron/metabolism , Cell Nucleus/metabolism , Lipid Peroxidation/drug effects , Animals , Apoptosis/drug effects , Carbolines
17.
Mol Neurobiol ; 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39354232

ABSTRACT

Ferroptosis, a regulated form of cell death characterized by excessive iron-dependent lipid peroxidation, can be readily induced in cultured cells by chemicals such as erastin and RSL3. Protein disulfide isomerase (PDI) has been identified as an upstream mediator of chemically induced ferroptosis and also a target for ferroptosis protection. In this study, we discovered that raloxifene (RAL), a selective estrogen receptor modulator known for its neuroprotective actions in humans, can effectively inhibit PDI function and provide robust protection against chemically induced ferroptosis in cultured HT22 neuronal cells. Specifically, RAL can bind directly to PDI both in vitro and in intact neuronal cells and inhibit its catalytic activity. Computational modeling analysis reveals that RAL can tightly bind to PDI through forming a hydrogen bond with its His256 residue, and biochemical analysis further shows that when PDI's His256 is mutated to Ala256, RAL loses its inhibition of PDI's catalytic activity. This inhibition of PDI by RAL significantly reduces the dimerization of both the inducible and neuronal nitric oxide synthases and the accumulation of nitric oxide, both of which have recently been shown to play a crucial role in mediating chemically induced ferroptosis through subsequent induction of ROS and lipid-ROS accumulation. In vivo behavioral analysis shows that mice treated with RAL are strongly protected against kainic acid-induced memory deficits and hippocampal neuronal damage. In conclusion, this study demonstrates that RAL is a potent inhibitor of PDI and can effectively prevent chemically induced ferroptosis in hippocampal neurons both in vitro and in vivo. These findings offer a novel estrogen receptor-independent mechanism for RAL's neuroprotective actions in animal models and humans.

18.
Biochim Biophys Acta ; 1823(8): 1306-15, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22580043

ABSTRACT

Earlier studies showed that 2-methoxyestradiol (2ME(2)), an endogenous nonpolar metabolite of estradiol-17ß, is a strong inducer of G(2)/M cell cycle arrest (based on analysis of cellular DNA content) in human cancer cell lines. The present study sought to investigate the molecular mechanism underlying 2ME(2)-induced cell cycle arrest. We found that 2ME(2) can selectively induce mitotic prometaphase arrest, but not G(2) phase arrest, in cultured MDA-MB-435s and MCF-7 human breast cancer cells. During the induction of prometaphase arrest, there is a time-dependent initial up-regulation of cyclin B1 and Cdc2 proteins, occurring around 12-24h. The strong initial up-regulation of cyclin B1 and Cdc2 matches in timing the 2ME(2)-induced prometaphase arrest. The 2ME(2)-induced prometaphase arrest is abrogated by selective knockdown of cyclin B1 and Cdc2, or by pre-treatment of cells with roscovitine, an inhibitor of cyclin-dependent kinases, or by co-treatment of cells with cycloheximide, a protein synthesis inhibitor that was found to suppress the early up-regulation of cyclin B1 and Cdc2. In addition, we provided evidence showing that MAD2 and JNK1 are important upstream mediators of 2ME(2)-induced up-regulation of cyclin B1 and Cdc2 as well as the subsequent induction of mitotic prometaphase arrest. In conclusion, treatment of human cancer cells with 2ME(2) causes up-regulation of cyclin B1 and Cdc2, which then mediate the induction of mitotic prometaphase arrest.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin B1/genetics , Cyclin B/genetics , Estradiol/analogs & derivatives , Prometaphase/drug effects , Up-Regulation/drug effects , 2-Methoxyestradiol , Breast Neoplasms , CDC2 Protein Kinase , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Nucleus Shape/drug effects , Cyclin B/metabolism , Cyclin B1/metabolism , Cyclin-Dependent Kinases , Estradiol/pharmacology , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Knockdown Techniques , Humans , Mad2 Proteins , Mitogen-Activated Protein Kinase 8/metabolism , RNA Interference , Repressor Proteins/metabolism
19.
J Cell Biochem ; 114(1): 192-203, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22903547

ABSTRACT

In a recent study, we showed that eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), two common omega-3 fatty acids, can cause ROS accumulation and subsequently induce caspase-8-dependent apoptosis in human breast cancer cells (Kang et al. [2010], PLoS ONE 5: e10296). In this study, we showed that the pancreas has a unique ability to accumulate EPA at a level markedly higher than several other tissues analyzed. Based on this finding, we sought to further investigate the anticancer actions of EPA and its analog DHA in human pancreatic cancer cells using both in vitro and in vivo models. EPA and DHA were found to induce ROS accumulation and caspase-8-dependent cell death in human pancreatic cancer cells (MIA-PaCa-2 and Capan-2) in vitro. Feeding animals with a diet supplemented with 5% fish oil, which contains high levels of EPA and DHA, also strongly suppresses the growth of MIA-PaCa-2 human pancreatic cancer xenografts in athymic nude mice, by inducing oxidative stress and cell death. In addition, we showed that EPA can concomitantly induce autophagy in these cancer cells, and the induction of autophagy diminishes its ability to induce apoptotic cell death. It is therefore suggested that combination of EPA with an autophagy inhibitor may be a useful strategy in increasing the therapeutic effectiveness in pancreatic cancer.


Subject(s)
Autophagy/drug effects , Caspase 8/metabolism , Docosahexaenoic Acids/therapeutic use , Eicosapentaenoic Acid/therapeutic use , Pancreatic Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Caspase 8/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Enzyme Activation/drug effects , Female , Gene Expression/drug effects , Humans , Mice , Mice, Nude , Oxidative Stress/drug effects , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , RNA, Small Interfering/genetics , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
20.
Biochem J ; 447(1): 115-23, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22747530

ABSTRACT

E(2) (17ß-oestradiol), a female sex hormone, has important biological functions in a woman's body. The pancreas, often considered a non-classical E(2)-targeting organ, is known to be functionally regulated by E(2), but little is known about how oestrogen actions are regulated in this organ. In the present study we report that PDIp (pancreas-specific protein disulfide isomerase), a protein-folding catalyst, can act as a major intracellular E(2) storage protein in a rat model to modulate the pancreatic tissue level, metabolism and action of E(2). The purified endogenous PDIp from both rat and human pancreatic tissues can bind E(2) with a K(d) value of approximately 150 nM. The endogenous PDIp-bound E(2) accounts for over 80% of the total protein-bound E(2) present in rat and human pancreatic tissues, and this binding protects E(2) from metabolic disposition and prolongs its duration of action. Importantly, we showed in ovariectomized female rats that the E(2) level in the pancreas reaches its highest level (9-fold increase over its basal level) at 24-48 h after a single injection of E(2), and even at 96 h its level is still approximately 5-fold higher. In contrast, the E(2) level in the uterus quickly returns to its basal level at 48 h after reaching its maximal level (approximately 2-fold increase) at 24 h. Taken together, these results show for the first time that PDIp is a predominant intracellular oestrogen storage protein in the pancreas, which offers novel mechanistic insights into the accumulation and action of oestrogen inside pancreatic cells.


Subject(s)
Estrogens/metabolism , Pancreas/metabolism , Protein Disulfide-Isomerases/metabolism , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Estradiol/metabolism , Female , Humans , Kinetics , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL