Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
Phytother Res ; 38(3): 1555-1573, 2024 Mar.
Article En | MEDLINE | ID: mdl-38281735

Anti-inflammatory and immune suppressive agents are required to moderate hyper-activation of lymphocytes under disease conditions or organ transplantation. However, selective disruption of mitochondrial redox has not been evaluated as a therapeutic strategy for suppression of T-cell-mediated pathologies. Using mitochondrial targeted curcumin (MitoC), we studied the effect of mitochondrial redox modulation on T-cell responses by flow cytometry, transmission electron microscopy, transcriptomics, and proteomics, and the role of Nrf2 was studied using Nrf2- /- mice. MitoC decreased mitochondrial TrxR activity, enhanced mitochondrial ROS (mROS) production, depleted mitochondrial glutathione, and suppressed activation-induced increase in mitochondrial biomass. This led to suppression of T-cell responses and metabolic reprogramming towards Treg differentiation. MitoC induced nuclear translocation and DNA binding of Nrf2, leading to upregulation of Nrf2-dependent genes and proteins. MitoC-mediated changes in mitochondrial redox and modulation of T-cell responses are abolished in Nrf2- /- mice. Restoration of mitochondrial thiols abrogated inhibition of T-cell responses. MitoC suppressed alloantigen-induced lymphoblast formation, inflammatory cytokines, morbidity, and mortality in acute graft-versus-host disease mice. Disruption of mitochondrial thiols but not mROS increase inculcates an Nrf2-dependent immune-suppressive disposition in T cells for the propitious treatment of graft-versus-host disease.


Curcumin , Curcumin/analogs & derivatives , Graft vs Host Disease , Animals , Mice , Curcumin/pharmacology , NF-E2-Related Factor 2/metabolism , T-Lymphocytes , Disease Models, Animal , Graft vs Host Disease/metabolism , Graft vs Host Disease/pathology , Sulfhydryl Compounds/metabolism , Sulfhydryl Compounds/pharmacology
2.
J Biosci ; 482023.
Article En | MEDLINE | ID: mdl-36971326

The present study primarily focuses on the efficacy of Malabaricone C (Mal C) as an anti-inflammatory agent. Mal C inhibited mitogen-induced T-cell proliferation and cytokine secretion. Mal C significantly reduced cellular thiols in lymphocytes. N-acetyl cysteine (NAC) restored cellular thiol levels and abrogated Mal C-mediated inhibition of T-cell proliferation and cytokine secretion. Physical interaction between Mal C and NAC was evinced from HPLC and spectral analysis. Mal C treatment significantly inhibited concanavalin A-induced phosphorylation of ERK/JNK and DNA binding of NF-κB. Administration of Mal C to mice suppressed T-cell proliferation and effector functions ex vivo. Mal C treatment did not alter the homeostatic proliferation of T-cells in vivo but completely abrogated acute graft-versus-host disease (GvHD)-associated morbidity and mortality. Our studies indicate probable use of Mal C for prophylaxis and treatment of immunological disorders caused due to hyper-activation of T-cells.


Myristica , Mice , Animals , Myristica/metabolism , Spices , Oxidation-Reduction , NF-kappa B/genetics , NF-kappa B/metabolism , Cytokines/genetics , Cytokines/metabolism , Anti-Inflammatory Agents/pharmacology
3.
Biochem J ; 476(2): 405-419, 2019 01 31.
Article En | MEDLINE | ID: mdl-30606737

Type II phosphatidylinositol 4-kinase ß (PtdIns 4-kinase II ß) is an enigma among the phosphatidylinositol 4-kinase family. The role of PtdIns 4-kinase II ß in MCF-7 cells was addressed with the help of short hairpin RNA (shRNA). PtdIns 4-kinase II ß shRNA transfection increased pan-caspase activity and induced apoptosis in cancerous MCF-7 cells. Non-cancerous MCF-10A cells were resistant to PtdIns 4-kinase II ß shRNA-induced apoptosis. Caspase 8 and 9 inhibitors rescued MCF-7 cells from apoptosis. Shotgun proteomic studies with Flag-tagged PtdIns 4-kinase II ß immunoprecipitates showed tumor suppressor prostate apoptosis response-4 (Par-4) as one of the interacting proteins in HEK293 cells. In reciprocal experiments, Par-4 antibodies co-precipitated PtdIns 4-kinase II ß from MCF-7 cells. Deletion of membrane localization motif (ΔCCPCC) or a mutation in ATP-binding region (D304A) of PtdIns 4-kinase II ß did not affect its interaction with Par-4. Pull-down assays with GST-PtdIns 4-kinase II ß-truncated mutants showed that the region between 101 and 215 amino acid residues is essential for interaction with Par-4. At molecular level, PtdIns 4-kinase II ß shRNA transfection increased Par-4 stability, its nuclear localization and inhibition of NF-κB binding to target DNA. Knocking down of Par-4 with siRNA (small interfering RNA) rescued MCF-7 cells from PtdIns 4-kinase II ß shRNA-induced apoptosis. These results suggest that PtdIns 4-kinase II ß may be a novel regulator of Par-4 through protein-protein interactions. These studies have potential implications in cancer therapy.


1-Phosphatidylinositol 4-Kinase/metabolism , Apoptosis Regulatory Proteins/metabolism , Apoptosis , Neoplasm Proteins/metabolism , Neoplasms/metabolism , 1-Phosphatidylinositol 4-Kinase/genetics , Amino Acid Motifs , Amino Acid Sequence , Apoptosis Regulatory Proteins/genetics , HEK293 Cells , Humans , MCF-7 Cells , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/pathology , Sequence Deletion
4.
Article En | MEDLINE | ID: mdl-29875077

The radiation-induced adaptive response (RI-AR) is a non-targeted effect which is outside the scope of the classical Linear-No-Threshold (LNT) dose-response paradigm. However, the mechanisms of the RI-AR are not well understood. We have studied the RI-AR in quiescent human peripheral blood mononuclear cells (PBMCs). PBMCs in G0 phase were 'primed' with a low dose (100 mGy gamma radiation) and then, after an 'adaptive window' of 4 h, 'challenged' with a high dose (2 Gy). A small (5.7%) increase in viability and a decrease in DNA strand breaks were seen in primed cells, compared to non-primed cells. This was consistent with lower levels of reactive oxygen species, higher mitochondrial membrane potential, and increased activity of antioxidant enzymes such as catalase, superoxide dismutase, thioredoxin reductase, and glutathione peroxidase, in the primed cells. Reduced oxidative stress in primed PBMCs correlated with greater nuclear translocation of the redox-sensitive transcription factors Nuclear factor kappa B (NF-κB) and Nuclear factor E2-related factor 2 (Nrf2). Distinct differences in responses were seen in PBMCs irradiated with low dose (100 mGy) and high dose (2 Gy). These findings provide insight into the mechanisms of radioadaptation in human cells.


Antioxidants/pharmacology , Gamma Rays , Gene Expression Regulation/drug effects , Leukocytes, Mononuclear/drug effects , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Oxidative Stress/drug effects , Adult , Catalase , DNA Damage , Gene Expression Regulation/radiation effects , Glutathione Peroxidase/metabolism , Healthy Volunteers , Humans , Leukocytes, Mononuclear/radiation effects , Oxidation-Reduction , Oxidative Stress/radiation effects , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Young Adult
5.
Anticancer Res ; 36(12): 6409-6417, 2016 12.
Article En | MEDLINE | ID: mdl-27919962

BACKGROUND: Ascorbic acid is proposed to have antitumor potential against certain cancer types but has the limitation of requiring high doses for treating cancer. Ascorbyl stearate (ASC-S) is a fatty acid ester derivative of ascorbic acid with comparable potent apoptotic activity. The present study was aimed at understanding the pathway involved in apoptotic activity of ASC-S in cervical cancer cells. MATERIALS AND METHODS: The effect of ASC-S on reactive oxygen species (ROS), and mitochondrial membrane potential (MMP) was studied in HeLa cells. Furthermore, the dose-dependent effect of ASC-S on release of cytochrome c, pro-caspase-9, caspase-3, BH3 interacting-domain death agonist (BID), truncated BH3 interacting-domain death agonist (t-BID), FAS ligand (FASL) and transcription factors nuclear factor-kappa B (NF-ĸB), nuclear factor of activated T-cells (NFAT) and activator protein-1 (AP1) were studied in HeLa cells. RESULTS: Treatment of HeLa cells with ASC-S significantly increased the MMP. The modulation of MMP resulted in cleavage of BID, expression of FAS, cleavage of pro-caspase-9 and release of cytochrome c into cytosol. In addition, ASC-S treatment resulted in deregulation of transcription factors NF-ĸB, NFAT and AP1, which play an important role in the development of inflammation and cancer. CONCLUSION: Our data, for the first time, suggest that ASC-S has an apoptotic effect against HeLa cells by inducing change in mitochondrial membrane permeability, cytochrome c release and subsequent activation of caspase-3 and NF-ĸB.


Apoptosis/drug effects , Ascorbic Acid/analogs & derivatives , Mitochondria/metabolism , Ascorbic Acid/pharmacology , Dose-Response Relationship, Drug , HeLa Cells , Humans , Matrix Metalloproteinases/metabolism , Membrane Potential, Mitochondrial , Reactive Oxygen Species/metabolism , Transcription Factors/metabolism
6.
Biochem Pharmacol ; 108: 75-89, 2016 May 15.
Article En | MEDLINE | ID: mdl-27019135

NF-κB is a crucial mediator of inflammatory and immune responses and a number of phytochemicals that can suppress this immune-regulatory transcription factor are known to have promising anti-inflammatory potential. However, we report that inducer of pro-inflammatory transcription factor NF-κB functions as an anti-inflammatory agent. Our findings reveal that a plant derived flavonoid baicalein could suppress mitogen induced T cell activation, proliferation and cytokine secretion. Treatment of CD4+ T cells with baicalein prior to transfer in to lymphopenic allogenic host significantly suppressed graft versus host disease. Interestingly, addition of baicalein to murine splenic lymphocytes induced DNA binding of NF-κB but did not suppress Concanavalin A induced NF-κB. Since baicalein did not inhibit NF-κB binding to DNA, we hypothesized that baicalein may be suppressing NF-κB trans-activation. Thioredoxin system is implicated in the regulation of NF-κB trans-activation potential and therefore inhibition of thioredoxin system may be responsible for suppression of NF-κB dependent genes. Baicalein not only inhibited TrxR activity in cell free system but also suppressed mitogen induced thioredoxin activity in the nuclear compartment of lymphocytes. Similar to baicalein, pharmacological inhibitors of thioredoxin system also could suppress mitogen induced T cell proliferation without inhibiting DNA binding of NF-κB. Further, activation of cellular thioredoxin system by the use of pharmacological activator or over-expression of thioredoxin could abrogate the anti-inflammatory action of baicalein. We propose a novel strategy using baicalein to limit NF-κB dependent inflammatory responses via inhibition of thioredoxin system.


Anti-Inflammatory Agents/pharmacology , Flavanones/pharmacology , NF-kappa B/metabolism , T-Lymphocytes/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Concanavalin A/pharmacology , Cytokines/metabolism , DNA/metabolism , Graft vs Host Disease/pathology , Graft vs Host Disease/prevention & control , Lymphopenia/pathology , Lymphopenia/therapy , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitogens/pharmacology , NF-kappa B/genetics , Protein Binding , Spleen/cytology , Spleen/drug effects , Spleen/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Thioredoxins/antagonists & inhibitors , Transcriptional Activation
7.
Free Radic Biol Med ; 85: 56-70, 2015 Aug.
Article En | MEDLINE | ID: mdl-25872101

Hematopoietic stem cells and progenitor cells (HSPC) are low in abundance and exhibit high radiosensitivity and their ability to divide dramatically decreases following exposure to ionizing radiation. Our earlier studies have shown antiapoptotic, immune-stimulatory, and antioxidant effects of chlorophyllin, a constituent of the over the counter drug derifil. Here we describe the beneficial effects of chlorophyllin against radiation-induced hematopoietic syndrome. Chlorophyllin administration significantly enhanced the abundance of HSPC in vivo. It induced a transient cell cycle arrest in lineage-negative cells in the bone marrow. However, the chlorophyllin-treated mice exposed to whole body irradiation (WBI) had a significantly higher proportion of actively dividing HSPC in the bone marrow as compared to only WBI-exposed mice. It significantly increased the number of colony forming units (CFUs) by bone marrow cells in vitro and spleen CFUs in irradiated mice in vivo. Pharmacokinetic study showed that chlorophyllin had a serum half-life of 141.8 min in mice. Chlorophyllin upregulated antiapoptotic genes and antioxidant machinery via activation of prosurvival transcription factors Nrf-2 and NF-κB and increased the survival and recovery of bone marrow cells in mice exposed to WBI. Chlorophyllin stimulated granulocyte production in bone marrow and increased the abundance of peripheral blood neutrophils by enhancing serum levels of granulocyte-colony stimulation factor (GCSF). Most importantly, prophylactic treatment of mice with chlorophyllin significantly abrogated radiation-induced mortality. Chlorophyllin mitigates radiation-induced hematopoietic syndrome by increasing the abundance of hematopoietic stem cells, enhancing granulopoiesis, and stimulating prosurvival pathways in bone marrow cells and lymphocytes.


Antioxidants/pharmacology , Chlorophyllides/pharmacology , Hematopoiesis/drug effects , Hematopoiesis/radiation effects , Hematopoietic Stem Cells/drug effects , Radiation Injuries/prevention & control , Animals , Male , Mice , Mice, Inbred BALB C
8.
Biochem Pharmacol ; 88(1): 95-105, 2014 Mar 01.
Article En | MEDLINE | ID: mdl-24406247

Low levels of oxidative stress have been shown to activate Nrf-2, an important anti-inflammatory transcription factor, by us and also by several other investigators. Earlier we showed that pro-oxidants protect normal lymphocytes against radiation injury by activating Nrf-2. In the present study, we have investigated the effect of oxidative stress on immune responses and delineated the underlying mechanism. Hydrogen peroxide, tert-butylhydroquinone and 1,4-naphthoquinone (NQ) inhibited mitogen induced proliferation of lymphocytes. NQ also inhibited mitogen (Concanavalin A) induced cytokine secretion by murine T cells and lipopolysaccharide induced release of cytokines, nitric oxide and cyclooxygenase-2 expression by macrophages. NQ modulated cellular redox by decreasing GSH/GSSG ratio and the immunosuppressive effects of NQ were significantly abrogated by thiol containing antioxidants and not by non-thiol antioxidants. This redox perturbation led to activation of Nrf-2 pathway and inhibition of NF-κB. NQ treatment increased total protein S-thiolation, induced glutathionylation of KEAP-1 protein and decreased IKKß levels in lymphocytes. Molecular docking studies revealed that NQ can disrupt KEAP-1/Nrf-2 interaction by directly blocking the binding site of Nrf-2 in the KEAP-1 protein. Further, inhibitors of Nrf-2 and HO-1 abrogated the anti-inflammatory effects of NQ. T cells isolated from spleen and gut associated lymphoid tissue of NQ administered mice also showed suppression of NF-κB activation and were hyporesponsive to mitogenic stimulation. These results demonstrate that pro-oxidants modulate inflammatory and immune responses via oxidative stress mediated KEAP-1 glutathionylation and IKKß degradation.


Adaptor Proteins, Signal Transducing/metabolism , Anti-Inflammatory Agents/pharmacology , Cytokines/metabolism , Cytoskeletal Proteins/metabolism , Glutathione/metabolism , NF-E2-Related Factor 2/metabolism , Naphthoquinones/pharmacology , Oxidative Stress/drug effects , Animals , Cell Line , Cell Proliferation/drug effects , Cytokines/immunology , Kelch-Like ECH-Associated Protein 1 , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Mice , Mice, Inbred BALB C , Molecular Docking Simulation , Nitric Oxide/metabolism , Oxidative Stress/immunology
9.
Inflammation ; 37(2): 542-54, 2014 Apr.
Article En | MEDLINE | ID: mdl-24234154

Plumbagin has been reported to modulate cellular redox status and suppress NF-κB. In the present study, we investigated the effect of plumbagin on lipopolysaccharide (LPS)-induced endotoxic shock, oxidative stress and inflammatory parameters in vitro and in vivo. Plumbagin inhibited LPS-induced nitric oxide, TNF-α, IL-6 and prostaglandin-E2 production in a concentration-dependent manner in RAW 264.7 cells without inducing any cell death. Plumbagin modulated cellular redox status in RAW cells. Plumbagin treatment significantly reduced MAPkinase and NF-κB activation in macrophages. Plumbagin prevented mice from endotoxic shock-associated mortality and decreased serum levels of pro-inflammatory markers. Plumbagin administration ameliorated LPS-induced oxidative stress in peritoneal macrophages and splenocytes. Plumbagin also attenuated endotoxic shock-associated changes in liver and lung histopathology and decreased the activation of ERK and NF-κB in liver. These findings demonstrate the efficacy of plumbagin in preventing LPS-induced endotoxemia and also provide mechanistic insights into the anti-inflammatory effects of plumbagin.


Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Inflammation Mediators/metabolism , NF-kappa B/metabolism , Naphthoquinones/pharmacology , Oxidative Stress/drug effects , Shock, Septic/prevention & control , Animals , Cell Line , Dinoprostone/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Down-Regulation , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/metabolism , Interleukin-6/metabolism , Lipopolysaccharides , Liver/drug effects , Liver/immunology , Liver/metabolism , Lung/drug effects , Lung/immunology , Lung/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Nitric Oxide/metabolism , Shock, Septic/blood , Shock, Septic/chemically induced , Shock, Septic/immunology , Signal Transduction/drug effects , Spleen/drug effects , Spleen/immunology , Spleen/metabolism , Time Factors , Tumor Necrosis Factor-alpha/metabolism
13.
Free Radic Biol Med ; 53(7): 1421-30, 2012 Oct 01.
Article En | MEDLINE | ID: mdl-22917978

Schisandrin B (SB), a dibenzocyclooctadiene derivative isolated from Schisandra chinensis and used commonly in traditional Chinese medicine for the treatment of hepatitis and myocardial disorders, has been recently shown to modulate cellular redox balance. Since we have shown that cellular redox plays an important role in the modulation of immune responses, the present studies were undertaken to study the effects of SB on activation and effector functions of lymphocytes. SB altered the redox status of lymphocytes by enhancing the basal reactive oxygen species levels and altering the GSH/GSSG ratio in lymphocytes. It also induced nuclear translocation of redox sensitive transcription factor Nrf2 and increased the transcription of its dependent genes. SB inhibited mitogen-induced proliferation and cytokine secretion by lymphocytes. SB also significantly inhibited mitogen-induced upregulation of T cell costimulatory molecules and activation markers. It was observed that SB inhibited mitogen-induced phosphorylation of c-Raf, MEK, ERK, JNK, and p38. It suppressed IκBα degradation and nuclear translocation of NF-κB in activated lymphocytes. Anti-inflammatory effects of SB were significantly abrogated by the inhibitors of Nrf2 and HO-1, suggesting the involvement of this pathway. Similar anti-inflammatory effects of SB on lymphocyte proliferation and cytokine secretion were also observed in vivo. To our knowledge, this is the first report showing that the anti-inflammatory effects of SB are mediated via modulation of Nrf2 and NF-κB in lymphocytes.


Anti-Inflammatory Agents/pharmacology , Lignans/pharmacology , Lymphocytes/drug effects , NF-E2-Related Factor 2/genetics , NF-kappa B/genetics , Polycyclic Compounds/pharmacology , Animals , Concanavalin A/pharmacology , Cyclooctanes/pharmacology , Cytokines/biosynthesis , Cytokines/immunology , Gene Expression Regulation , Heme Oxygenase-1/genetics , Heme Oxygenase-1/immunology , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , Lymphocyte Activation , Lymphocytes/cytology , Lymphocytes/immunology , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Kinases/immunology , NF-E2-Related Factor 2/immunology , NF-kappa B/immunology , Oxidation-Reduction , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/immunology , Signal Transduction , Spleen/cytology , Spleen/drug effects , Spleen/immunology
14.
J Biol Chem ; 286(9): 7339-47, 2011 Mar 04.
Article En | MEDLINE | ID: mdl-21156795

Doxorubicin is one of the most effective molecules used in the treatment of various tumors. Contradictory reports often open windows to understand the role of p53 tumor suppressor in doxorubicin-mediated cell death. In this report, we provide evidences that doxorubicin induced more cell death in p53-negative tumor cells. Several cells, having p53 basal expression, showed increase in p53 DNA binding upon doxorubicin treatment. Doxorubicin induced cell death in p53-positive cells through expression of p53-dependent genes and activation of caspases and caspase-mediated cleavage of cellular proteins. Surprisingly, in p53-negative cells, doxorubicin-mediated cell death was more aggressive (faster and intense). Doxorubicin increased the amount of Fas ligand (FasL) by enhancing activator protein (AP) 1 DNA binding in both p53-positive and p53-negative cells, but the basal expression of Fas was higher in p53-negative cells. Anti-FasL antibody considerably protected doxorubicin-mediated cell death in both types of cells. Activation of caspases was faster in p53-negative cells upon doxorubicin treatment. In contrast, the basal expression of Ras oncoprotein was higher in p53-positive cells, which might increase the basal expression of Fas in these cells. Overexpression of Ras decreased the amount of Fas in p53-negative cells, thereby decreasing doxorubicin-mediated aggressive cell death. Overall, this study will help to understand the much studied chemotherapeutic drug, doxorubicin-mediated cell signaling cascade, that leads to cell death in p53-positive and -negative cells. High basal expression of Fas might be an important determinant in doxorubicin-mediated cell death in p53-negative cells.


Apoptosis/drug effects , Apoptosis/physiology , Doxorubicin/pharmacology , Tumor Suppressor Protein p53/metabolism , ras Proteins/metabolism , Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms , Caspases/metabolism , Drug Resistance, Neoplasm/physiology , Fas Ligand Protein/genetics , Fas Ligand Protein/metabolism , Female , HCT116 Cells , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Transcription Factor AP-1/metabolism , U937 Cells
15.
J Biol Chem ; 286(6): 4690-702, 2011 Feb 11.
Article En | MEDLINE | ID: mdl-21127062

Considering the role of retinoids in regulation of more than 500 genes involved in cell cycle and growth arrest, a detailed understanding of the mechanism and its regulation is useful for therapy. The extract of the medicinal plant Neem (Azadirachta indica) is used against several ailments especially for anti-inflammatory, anti-itching, spermicidal, anticancer, and insecticidal activities. In this report we prove the detailed mechanism on the regulation of retinoic acid-mediated cell signaling by azadirachtin, active components of neem extract. Azadirachtin repressed all trans-retinoic acid (ATRA)-mediated nuclear transcription factor κB (NF-κB) activation, not the DNA binding but the NF-κB-dependent gene expression. It did not inhibit IκBα degradation, IκBα kinase activity, or p65 phosphorylation and its nuclear translocation but inhibited NF-κB-dependent reporter gene expression. Azadirachtin inhibited TRAF6-mediated, but not TRAF2-mediated NF-κB activation. It inhibited ATRA-induced Sp1 and CREB (cAMP-response element-binding protein) DNA binding. Azadirachtin inhibited ATRA binding with retinoid receptors, which is supported by biochemical and in silico evidences. Azadirachtin showed strong interaction with retinoid receptors. It suppressed ATRA-mediated removal of retinoid receptors, bound with DNA by inhibiting ATRA binding to its receptors. Overall, our data suggest that azadirachtin interacts with retinoic acid receptors and suppresses ATRA binding, inhibits falling off the receptors, and activates transcription factors like CREB, Sp1, NF-κB, etc. Thus, azadirachtin exerts anti-inflammatory and anti-metastatic responses by a novel pathway that would be beneficial for further anti-inflammatory and anti-cancer therapies.


Antineoplastic Agents/pharmacology , Azadirachta/chemistry , Cell Nucleus/metabolism , Insecticides/pharmacology , Limonins/pharmacology , Receptors, Retinoic Acid/metabolism , Tretinoin/pharmacology , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/physiology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Cell Nucleus/genetics , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Humans , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Insecticides/chemistry , Limonins/chemistry , NF-KappaB Inhibitor alpha , Phosphorylation/drug effects , Phosphorylation/physiology , Receptors, Retinoic Acid/genetics , Sp1 Transcription Factor/genetics , Sp1 Transcription Factor/metabolism , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , U937 Cells
16.
Breast Cancer Res Treat ; 120(3): 671-83, 2010 Apr.
Article En | MEDLINE | ID: mdl-19649704

Doxorubicin is one of the most effective molecules used in the treatment of various tumors. Contradictory reports often open windows to understand the doxorubicin-mediated signaling to exert its apoptosis effect. In this report, we provide evidences that doxorubicin induced biphasic induction of nuclear factor kappaB (NF-kappaB) of immediate activation followed by decrease in the amount of RelA (p65) subunit possibly by inducing the activity of proteasome, but not proteases. Further induction of NF-kappaB was observed through interleukin 8 (IL-8), expressed by doxorubicin treatment. Increased amount of IL-8 induced apoptosis via increase in the releases of intracellular Ca(2+), activation of calcineurin, nuclear translocation of nuclear factor activated T cell (NF-AT), and NF-AT-dependent FasL expression. Anti-IL-8 or -FasL antibody, dominant negative TRAF6 (TRAF6-DN), or TRAF6 binding peptide (TRAF6-BP) inhibited doxorubicin-mediated late phase induction of NF-kappaB and diminished cell death. Thus, our study clearly demonstrated that doxorubicin-mediated cell death is obtained through expression of IL-8. IL-8-mediated calcification is required for enhancement of doxorubicin-mediated cell death. Overall, this study will help to understand the much studied chemotherapeutic drug, doxorubicin-mediated cell signaling cascade to exert its effect during chemotherapy.


Adenocarcinoma/pathology , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Breast Neoplasms/pathology , Doxorubicin/pharmacology , Fas Ligand Protein/physiology , Interleukin-8/physiology , NF-kappa B/physiology , Neoplasm Proteins/physiology , Calcineurin/metabolism , Calcium Signaling/drug effects , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Fas Ligand Protein/biosynthesis , Fas Ligand Protein/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter , Humans , NFATC Transcription Factors/physiology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/physiology , Transcription Factor RelA/biosynthesis , Transcription Factor RelA/genetics , bcl-2-Associated X Protein/biosynthesis , bcl-2-Associated X Protein/genetics
17.
J Biol Chem ; 285(8): 5888-95, 2010 Feb 19.
Article En | MEDLINE | ID: mdl-20018848

The role of azadirachtin, an active component of a medicinal plant Neem (Azadirachta indica), on TNF-induced cell signaling in human cell lines was investigated. Azadirachtin blocks TNF-induced activation of nuclear factor kappaB (NF-kappaB) and also expression of NF-kappaB-dependent genes such as adhesion molecules and cyclooxygenase 2. Azadirachtin inhibits the inhibitory subunit of NF-kappaB (IkappaB alpha) phosphorylation and thereby its degradation and RelA (p65) nuclear translocation. It blocks IkappaB alpha kinase (IKK) activity ex vivo, but not in vitro. Surprisingly, azadirachtin blocks NF-kappaB DNA binding activity in transfected cells with TNF receptor-associated factor (TRAF)2, TNF receptor-associated death domain (TRADD), IKK, or p65, but not with TNFR, suggesting its effect is at the TNFR level. Azadirachtin blocks binding of TNF, but not IL-1, IL-4, IL-8, or TNF-related apoptosis-inducing ligand (TRAIL) with its respective receptors. Anti-TNFR antibody or TNF protects azadirachtin-mediated down-regulation of TNFRs. Further, in silico data suggest that azadirachtin strongly binds in the TNF binding site of TNFR. Overall, our data suggest that azadirachtin modulates cell surface TNFRs thereby decreasing TNF-induced biological responses. Thus, azadirachtin exerts an anti-inflammatory response by a novel pathway, which may be beneficial for anti-inflammatory therapy.


Anti-Inflammatory Agents/pharmacology , Limonins/pharmacology , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Active Transport, Cell Nucleus/drug effects , Binding Sites , Cell Nucleus/metabolism , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Gene Expression Regulation/drug effects , HeLa Cells , Humans , I-kappa B Kinase/metabolism , Insecticides/pharmacology , Phosphorylation/drug effects , Protein Structure, Tertiary , TNF Receptor-Associated Death Domain Protein/metabolism , TNF Receptor-Associated Factor 2/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Transcription Factor RelA/metabolism , U937 Cells
18.
J Cell Biochem ; 107(2): 203-13, 2009 May 15.
Article En | MEDLINE | ID: mdl-19242952

Doxorubicin is one of the most effective agents used in the treatment of various tumors. Its use is restricted by the development of resistance to apoptosis, the mechanism of which is not fully understood. Nuclear transcription factor kappaB (NF-kappaB) has been shown both to block apoptosis and to promote cell proliferation, and hence has been considered as an important target for anticancer drug development. We found that in wild type and Dox-revertant MCF-7 cells, Doxorubicin induced NF-kappaB was transient and Dox-resistant cells showed high basal activity of NF-kappaB and expression of genes dependent on it. Moreover, in resistant cells Doxorubicin was unable to induce apoptosis as detected by assays for reactive oxygen intermediates generation, lipid peroxidation, cytotoxicity, PARP degradation and Bcl-2 expression. High basal expressions of multi-drug resistant protein and transglutaminase were found in Dox-resistant cells and inhibition of NF-kappaB decreased those amounts and also sensitized these cells by Doxorubicin. These observations collectively suggest that high NF-kappaB activity confers resistance to Doxorubicin and its inhibition potentiates apoptosis. This study indicates that NF-kappaB plays an important role in chemoresistance and establishes the fact that inhibition of NF-kappaB will be a novel approach in chemotherapy.


Antibiotics, Antineoplastic/pharmacology , Apoptosis/physiology , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/physiology , NF-kappa B/metabolism , Cell Line , Flow Cytometry , Free Radicals/metabolism , Gene Expression Regulation , Humans , Immunohistochemistry , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology
...