Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 42
1.
Neuroreport ; 32(17): 1379-1387, 2021 12 08.
Article En | MEDLINE | ID: mdl-34718250

OBJECTIVES: Paeoniflorin, an active component of Radix Paeoniae Alba, has a neuroprotective effect in Parkinson's animal models. However, its mechanism of action remains to be determined. METHODS: In this study, we hypothesized that the neuroprotective effect of paeoniflorin occurs through the α-synuclein/protein kinase C δ subtype (PKC-δ) signaling pathway. We tested our hypothesis in the 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP)-induced mouse model of Parkinson's disease. We evaluated the effects of paeoniflorin on the expression levels of signal components of the α-synuclein/PKC-δ pathway, cellular apoptosis and motor performance. RESULTS: Our results demonstrated that paeoniflorin restored the motor performance impairment caused by MPTP, inhibited apoptosis, and protected the ultrastructure of neurons. Paeoniflorin treatment also resulted in the dose-dependent upregulation of an antiapoptotic protein, B-cell lymphoma-2, at the mRNA and protein levels, similar to the effects of the positive control, selegiline. In contrast, paeoniflorin treatment downregulated the expression of pro-apoptotic proteins BCL2-Associated X2, α-synuclein, and PKC-δ at the mRNA and protein levels, as well as the level of the activated form of nuclear factor kappa B (p-NF-κB p65). CONCLUSIONS: Thus, our results showed that paeoniflorin exerts its neuroprotective effect by regulating the α-synuclein/PKC-δ signaling pathway to reduce neuronal apoptosis.


Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Glucosides/pharmacology , Monoterpenes/pharmacology , Parkinsonian Disorders/metabolism , Protein Kinase C-delta/drug effects , Substantia Nigra/drug effects , alpha-Synuclein/drug effects , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Annexin A5/drug effects , Annexin A5/metabolism , Antiparkinson Agents/pharmacology , Disease Models, Animal , Mice , Microscopy, Electron, Transmission , Neurotoxins , Parkinsonian Disorders/pathology , Parkinsonian Disorders/physiopathology , Protein Kinase C-delta/metabolism , Rotarod Performance Test , Selegiline/pharmacology , Substantia Nigra/metabolism , Substantia Nigra/pathology , alpha-Synuclein/metabolism
2.
Neurotox Res ; 38(2): 385-397, 2020 Aug.
Article En | MEDLINE | ID: mdl-32378056

Tramadol is a synthetic analogue of codeine that is often prescribed for the treatment of mild to moderate pains. It has a number of side effects including emotional instability and anxiety. In this study, we focus on the structural and functional changes of prefrontal cortex under chronic exposure to tramadol. At the cellular level, the amounts of ROS and annexin V in PC12 cells were evidently increased upon exposure to tramadol (at a concentration of 600 µM for 48 h). To this end, the rats were daily treated with tramadol at doses of 50 mg/kg for 3 weeks. Our findings reveal that tramadol provokes atrophy and apoptosis by the induction of apoptotic markers such as Caspase 3 and 8, pro-inflammatory markers, and downregulation of GDNF. Moreover, it triggers microgliosis and astrogliosis along with neuronal death in the prefrontal cortex. Behavioral disturbance and cognitive impairment are other side effects of tramadol. Overall, our results indicate tramadol-induced neurodegeneration in the prefrontal cortex mainly through activation of neuroinflammatory response.


Apoptosis/drug effects , Neurons/drug effects , Prefrontal Cortex/drug effects , Tramadol/toxicity , Animals , Annexin A5/drug effects , Annexin A5/metabolism , Astrocytes/drug effects , Astrocytes/pathology , Atrophy , Caspase 3/drug effects , Caspase 3/metabolism , Caspase 8/drug effects , Caspase 8/metabolism , Cell Death , Glial Cell Line-Derived Neurotrophic Factor/drug effects , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Gliosis/chemically induced , Gliosis/metabolism , Gliosis/pathology , Male , Microglia/drug effects , Microglia/pathology , Neurons/metabolism , Neurons/pathology , PC12 Cells , Prefrontal Cortex/metabolism , Prefrontal Cortex/pathology , Rats , Reactive Oxygen Species/metabolism
3.
Cancer ; 123(9): 1662-1673, 2017 05 01.
Article En | MEDLINE | ID: mdl-28055106

BACKGROUND: Nucleophosmin 1 (NPM1) is a nucleocytoplasmic shuttling protein mainly localized in the nucleolus. NPM1 is frequently mutated in acute myeloid leukemia (AML). NPM1c oligomerizes with wild-type nucleophosmin 1 (wt-NPM1), and this leads to its continuous cytoplasmic delocalization and contributes to leukemogenesis. Recent studies have shown that Cytoplasmic NPM1 (NPM1c) degradation leads to growth arrest and apoptosis of NPM1c AML cells and corrects wt-NPM1 normal nucleolar localization. METHODS: AML cells expressing wt-NPM1 or NPM1c or transfected with wt-NPM1 or NPM1c as well as wt-NPM1 and NPM1c AML xenograft mice were used. Cell growth was assessed with trypan blue or a CellTiter 96 proliferation kit. The cell cycle was studied with a propidium iodide (PI) assay. Caspase-mediated intrinsic apoptosis was assessed with annexin V/PI, the mitochondrial membrane potential, and poly(adenosine diphosphate ribose) polymerase cleavage. The expression of NPM1, p53, phosphorylated p53, and p21 was analyzed via immunoblotting. Localization was performed with confocal microscopy. The leukemia burden was evaluated by flow cytometry with an anti-human CD45 antibody. RESULTS: The imidazoquinoxaline 1-(3-methoxyphenyl)-N-methylimidazo[1,2-a]quinoxalin-4-amine (EAPB0503) induced selective proteasome-mediated degradation of NPM1c, restored wt-NPM1 nucleolar localization in NPM1c AML cells, and thus yielded selective growth arrest and apoptosis. Introducing NPM1c to cells normally harboring wt-NPM1 sensitized them to EAPB0503 and led to their growth arrest. Moreover, EAPB0503 selectively reduced the leukemia burden in NPM1c AML xenograft mice. CONCLUSIONS: These findings further reinforce the idea of targeting the NPM1c oncoprotein to eradicate leukemic cells and warrant a broader preclinical evaluation and then a clinical evaluation of this promising drug. Cancer 2017;123:1662-1673. © 2017 American Cancer Society.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Leukemia, Myeloid, Acute/drug therapy , Mutant Proteins/drug effects , Nuclear Proteins/drug effects , Quinoxalines/pharmacology , Animals , Annexin A5/drug effects , Annexin A5/metabolism , Cell Line, Tumor , Cell Nucleolus/metabolism , Cyclin-Dependent Kinase Inhibitor p21/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytoplasm/metabolism , Flow Cytometry , Humans , Immunoblotting , Leukemia, Myeloid, Acute/genetics , Mice , Microscopy, Confocal , Mutant Proteins/metabolism , Mutation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleophosmin , Phosphorylation/drug effects , Poly(ADP-ribose) Polymerases/drug effects , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
4.
Asian Pac J Cancer Prev ; 13(4): 1395-9, 2012.
Article En | MEDLINE | ID: mdl-22799338

BACKGROUND: Histone deacetylase (HDAC) inhibitors have been reported to induce cell growth arrest, apoptosis and differentiation of tumor cells. The present study aimed to examine the effects of trichostatin A (TSA), one such inhibitor, on the cell cycle, apoptosis and invasiveness of osteosarcoma cells. METHODS: MG- 63 cells were treated with TSA at various concentrations. Then, cell growth and apoptosis were determined by 3-(4, 5-dimethyl-2-thiazolyl)-2H-tetrazolium bromide (MTT) and TUNEL assays, respectively; cell cycling was assessed by flow cytometry; invasion assays were performed with the transwell Boyden Chamber system. RESULTS: MTT assays revealed that TSA significantly inhibited the growth of MG-63 cells in a concentration and time dependent manner. TSA treated cells demonstrated morphological changes indicative of apoptosis and TUNEL assays revealed increased apoptosis of MG-63 cells after TSA treatment. Flow cytometry showed that TSA arrested the cell cycle in G1/G2 phase and annexin V positive apoptotic cells increased markedly. In addition, the invasiveness of MG-63 cells was inhibited by TSA in a concentration dependent manner. CONCLUSION: Our findings demonstrate that TSA inhibits the proliferation, induces apoptosis and inhibits invasiveness of osteosarcoma cells in vitro. HDAC inhibitors may thus have promise to become new therapeutic agents against osteosarcoma.


Apoptosis/drug effects , Cell Proliferation/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Osteosarcoma/pathology , Annexin A5/drug effects , Annexin A5/metabolism , Cell Line, Tumor , Humans , Neoplasm Invasiveness , Osteosarcoma/drug therapy , Osteosarcoma/metabolism
5.
Oral Dis ; 18(3): 280-4, 2012 Apr.
Article En | MEDLINE | ID: mdl-22093110

OBJECTIVE: To determine the effects of fluorides on endothelial functioning. MATERIALS AND METHODS: We analyzed expressions of adhesion molecules, ICAM-1 and ICAM-3, and annexin V, on the surface of human umbilical vein endothelial cells (HUVECs) exposed to various concentrations of NaF and SnF(2) . We compared the effects of fluoride-induced changes with those obtained when stimulating HUVECs with TNF-α and verified whether N-acetyl cysteine (NAC), well-known antioxidant, can prevent both fluoride- and TNF-α-induced alterations. RESULTS: The expressions of annexin V and ICAM-1 increased significantly after adding NaF (5.0 or 7.5mM) or Sn(2) F (0.5 or 0.75mM) to the culture medium. Pre-incubating HUVECs with NAC prevented the effects induced by 5.0 mM of NaF and 0.5 mM of Sn(2) F. Only the highest concentration of NaF (7.5mM) triggered the expression of ICAM-3. The expressions of all three molecules increased significantly upon stimulating the cultures with TNF-α (20ng ml(-1) ); these changes were not reversed by pre-incubation with NAC. CONCLUSIONS: Fluorides induce oxidative stress, resulting in apoptosis and activation of HUVECs, manifested by an elevated expression of ICAM-1. The oxidative stress resulting from a stimulation by the highest NaF concentration triggers ICAM-3 expression on the HUVECs' surface.


Apoptosis/drug effects , Cariostatic Agents/pharmacology , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Fluorides/pharmacology , Acetylcysteine/pharmacology , Annexin A5/drug effects , Antigens, CD/drug effects , Antioxidants/pharmacology , Cell Adhesion Molecules/drug effects , Cell Culture Techniques , Cells, Cultured , Endothelium, Vascular/cytology , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Humans , Intercellular Adhesion Molecule-1/drug effects , Oxidative Stress/drug effects , Sodium Fluoride/pharmacology , Tin Fluorides/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Umbilical Veins/cytology
6.
Rev Med Interne ; 31(12): 854-7, 2010 Dec.
Article Fr | MEDLINE | ID: mdl-20888088

Studies have shown a protective effect of hydroxychloroquine on thrombosis in systemic lupus erythematosus patients. Recent in vitro studies have demonstrated that this molecule was able to restore the anticoagulant action of annexin A5, which is reduced in presence of antiphospholipid antibodies. Hydroxychloroquine use may be a new approach of the prevention of thrombosis in the antiphospholipid syndrome, which remains to be validated by well-conducted clinical trials.


Antiphospholipid Syndrome/drug therapy , Antirheumatic Agents/therapeutic use , Hydroxychloroquine/therapeutic use , Thrombosis/prevention & control , Annexin A5/blood , Annexin A5/drug effects , Antiphospholipid Syndrome/blood , Antiphospholipid Syndrome/immunology , Biomarkers/blood , Enzyme Inhibitors/blood , Evidence-Based Medicine , Humans , Lupus Erythematosus, Systemic/blood , Lupus Erythematosus, Systemic/drug therapy , Thrombosis/immunology , Treatment Outcome
7.
Scand J Rheumatol ; 39(1): 77-83, 2010.
Article En | MEDLINE | ID: mdl-20132075

OBJECTIVES: Anti-phospholipid antibodies (aPL), including anti-cardiolipin antibodies (aCL), are risk factors for cardiovascular disease (CVD) in the general population and in patients with the anti-phospholipid syndrome (APS; Hughes syndrome). APS may be primary but is also common in patients with systemic lupus erythematosus (SLE). The anti-coagulant protein annexin A5 (ANXA5) is implicated in CVD by interfering with phospholipids and aPL. METHODS: ANXA5 binding to human umbilical venous endothelial cells (HUVECs) was determined by flow cytometry. RESULTS: When cells were cultured in serum from APS patients with a high aPL titre (aPL-S), binding of ANXA5 to HUVECs was reduced. Monoclonal immunoglobulin (Ig)G aPL against cardiolipin (mAb-CL) dose-dependently reduced ANXA5 binding to endothelium. Preincubation of intravenous (IV)Ig at therapeutically relevant doses with aPL-S and mAb-aCL restored ANXA5 binding to comparable levels when normal healthy serum (NHS) was used. By contrast, IVIg per se had the capacity to reduce ANXA5 binding to endothelium when added to NHS (but not to aPL-S). CONCLUSIONS: Decreased ANXA5 binding to endothelium, mediated by aPL, is a novel mechanism of atherothrombosis that can be countered by IVIg in vitro. IVIg per se could, to a lesser degree, cause decreased ANXA5 binding in NHS, which raises the possibility that some antibodies in IVIg can be involved in a side-effect reported in IVIg treatment, namely atherothrombosis and CVD. Increasing ANXA5 binding, either by addition of ANXA5 or by use of neutralizing antibodies in IVIg, represents a possible therapeutic strategy that deserves further study.


Annexin A5/drug effects , Annexin A5/metabolism , Antibodies, Anticardiolipin/blood , Cardiovascular Diseases/physiopathology , Immunoglobulins, Intravenous/pharmacology , Annexin A5/immunology , Antibodies, Anticardiolipin/metabolism , Antiphospholipid Syndrome/blood , Antiphospholipid Syndrome/immunology , Binding Sites/drug effects , Binding Sites/physiology , Case-Control Studies , Cells, Cultured , Endothelial Cells/immunology , Endothelial Cells/metabolism , Female , Flow Cytometry , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunoglobulins, Intravenous/immunology , Male , Probability , Reference Values , Umbilical Veins/cytology
8.
Biochim Biophys Acta ; 1800(4): 439-47, 2010 Apr.
Article En | MEDLINE | ID: mdl-20064588

BACKGROUND: Plagiochin E (PLE) is an antifungal active macrocyclic bis(bibenzyl) isolated from liverwort Marchantia polymorpha L. To elucidate the mechanism of action, previous studies revealed that the antifungal effect of PLE was associated with the accumulation of ROS, an important regulator of apoptosis in Candida albicans. The present study was designed to find whether PLE caused apoptosis in C. albicans. METHODS: We assayed the cell cycle by flow cytometry using PI staining, observed the ultrastructure by transmission electron microscopy, studied the nuclear fragmentation by DAPI staining, and investigated the exposure of phosphatidylserine at the outer layer of the cytoplasmic membrane by the FITC-annexin V staining. The effect of PLE on expression of CDC28, CLB2, and CLB4 was determined by RT-PCR. Besides, the activity of metacaspase was detected by FITC-VAD-FMK staining, and the release of cytochrome c from mitochondria was also determined. Furthermore, the effect of antioxidant L-cysteine on PLE-induced apoptosis in C. albicans was also investigated. RESULTS: Cells treated with PLE showed typical markers of apoptosis: G(2)/M cell cycle arrest, chromatin condensation, nuclear fragmentation, and phosphatidylserine exposure. The expression of CDC28, CLB2, and CLB4 was down-regulated by PLE, which may contribute to PLE-induced G(2)/M cell cycle arrest. Besides, PLE promoted the cytochrome c release and activated the metacaspase, which resulted in the yeast apoptosis. The addition of L-cysteine prevented PLE-induced nuclear fragmentation, phosphatidylserine exposure, and metacaspase activation, indicating the ROS was an important mediator of PLE-induced apoptosis. CONCLUSIONS: PLE induced apoptosis in C. albicans through a metacaspase-dependent apoptotic pathway. GENERAL SIGNIFICANCE: In this study, we reported for the first time that PLE induced apoptosis in C. albicans through activating the metacaspase. These results would conduce to elucidate its underlying antifungal mechanism.


Antifungal Agents/pharmacology , Bridged-Ring Compounds/pharmacology , Candida albicans/drug effects , Macrocyclic Compounds/pharmacology , Stilbenes/pharmacology , Annexin A5/drug effects , Annexin A5/metabolism , Antifungal Agents/isolation & purification , Apoptosis/drug effects , Bridged-Ring Compounds/chemistry , Bridged-Ring Compounds/isolation & purification , Candida albicans/cytology , Candida albicans/enzymology , Candida albicans/genetics , Caspase Inhibitors , Cell Cycle/drug effects , DNA Primers , Flow Cytometry , Macrocyclic Compounds/chemistry , Macrocyclic Compounds/isolation & purification , Marchantia/chemistry , Models, Molecular , Molecular Conformation , Protease Inhibitors/pharmacology , RNA, Fungal/genetics , RNA, Fungal/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Stilbenes/chemistry , Stilbenes/isolation & purification
9.
J Oral Pathol Med ; 38(10): 792-800, 2009 Nov.
Article En | MEDLINE | ID: mdl-19656269

BACKGROUND: Although N(1)-guanyl-1,7,-diamineoheptane (GC7), an inhibitor of deoxyhypusine synthase, has been shown to inhibit cell growth, the mechanism of its action is not completely understood. In this study, we investigated the mechanisms of the effects of GC7 on cell growth, differentiation and apoptosis in relation to adenosine monophosphate-activated protein kinase (AMPK) activation, as AMPK is known to be a possible target for cancer treatment. METHODS: The effects of GC7 on the growth of immortalized human oral keratinocytes (IHOK) and primary oral cancer cells (HN4), was investigated using MTT assay, Western blotting, cell cycle analysis, DNA fragmentation and expression of apoptotic pathway proteins. RESULTS: N(1)-guanyl-1,7,-diamineoheptane inhibited cell proliferation in a time- and dose-dependent manner in IHOK and HN4 cells. GC7 treatment decreased the expression of differentiation markers, such as involucrin, CK13 and CK19. The major mechanism of growth inhibition by GC7 treatment was induction of apoptosis, which is supported by sub-G(1) phase arrest, annexin V-FITC staining and DNA fragmentation analysis. GC7 treatment increased the cytosolic level of cytochrome c and resulted in caspase-3 activation. GC7 treatment also resulted in a strong activation of AMPK. Furthermore, specific AMPK activator blocked the GC7-induced growth inhibition effect, as well as apoptosis. CONCLUSION: These results demonstrate that GC7 blocks immortalized and malignant keratinocyte cell proliferation and differentiation by inducing apoptosis through the mitochondrial and AMPK pathways. On the basis of these observations, we propose that a strategy combining GC7 and AMPK inhibition could be developed into a novel chemotherapeutic modality in oral cancer.


AMP-Activated Protein Kinases/drug effects , Apoptosis/drug effects , Carcinoma/pathology , Enzyme Inhibitors/pharmacology , Guanine/analogs & derivatives , Keratinocytes/drug effects , Mitochondria/drug effects , Mouth Mucosa/drug effects , Mouth Neoplasms/pathology , Oxidoreductases Acting on CH-NH Group Donors/antagonists & inhibitors , Annexin A5/drug effects , Caspase 3/drug effects , Cell Cycle Proteins/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cytochromes c/drug effects , DNA Fragmentation , Dose-Response Relationship, Drug , Enzyme Activation , Enzyme Inhibitors/administration & dosage , G1 Phase/drug effects , Guanine/administration & dosage , Guanine/pharmacology , Humans , Keratin-13/antagonists & inhibitors , Keratin-19/antagonists & inhibitors , Mouth Mucosa/pathology , Protein Precursors/antagonists & inhibitors
10.
Brain Res ; 1283: 155-66, 2009 Aug 04.
Article En | MEDLINE | ID: mdl-19505441

Neuroblastoma is a pediatric extracranial tumor and a major cause of death in children under age 2. Conventional therapy shows inefficacy in most cases and thus development of new therapeutic strategies is urgently needed. We explored the efficacy of combination of the small molecule Bcl-2 inhibitor HA14-1 (HA) and the isoflavonoid genistein (GST) in human malignant neuroblastoma SK-N-BE2 and SH-SY5Y cells. Combination of 10 microM HA and 250 microM GST was optimal for SK-N-BE2 cells and combination of 5 microM HA and 100 microM GST was optimal for SH-SY5Y cells for induction of apoptosis. Phase-contrast microscopy and Wright staining showed morphological features of apoptosis. Cell cycle analysis and Annexin V-FITC/PI binding assay showed that combination of HA and GST was more effective in inducing apoptosis in both cell lines than either HA or GST alone. Western blotting showed that combination of HA and GST caused upregulation of Bax and down regulation of Bcl-2 resulting in increased Bax:Bcl-2 ratio and mitochondrial release of cytochrome c, Smac, and AIF. Down regulation of survival factors such as NF-kappaB, N-Myc, and survivin promoted apoptosis. Activation of caspase-8, calpain, and caspase-3 occurred in course of apoptosis. Increased calpain and caspase-3 activities were confirmed in the degradation of alpha-spectrin to 145 kD spectrin break down product (SBDP) and 120 kD SBDP, respectively. Thus, combination of HA and GST could serve as a promising therapeutic strategy for increasing apoptosis in different human malignant neuroblastoma cells.


Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis Regulatory Proteins/antagonists & inhibitors , Benzopyrans/pharmacology , Cysteine Endopeptidases/drug effects , Genistein/pharmacology , Neuroblastoma/drug therapy , Nitriles/pharmacology , Annexin A5/drug effects , Annexin A5/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis/physiology , Apoptosis Regulatory Proteins/metabolism , Benzopyrans/therapeutic use , Cell Cycle/drug effects , Cell Cycle/physiology , Cysteine Endopeptidases/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Genistein/therapeutic use , Humans , Neuroblastoma/metabolism , Neuroblastoma/physiopathology , Nitriles/therapeutic use , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
11.
Pediatr Allergy Immunol ; 20(4): 392-8, 2009 Jun.
Article En | MEDLINE | ID: mdl-18713315

Decreased severity of graft-versus-host disease after mismatched umbilical cord blood (UCB) transplantation may be attributed in part to the increased propensity to apoptosis of UCB T cells following activation. Interleukin (IL)-15, a pleiotropic cytokine that is essential for T-cell proliferation and survival, may serve as promising immunomodulative therapy post-CB transplantation for its anti-apoptotic effect. This study aimed to determine the kinetics of Fas or tumor necrosis factor-alpha receptor (TNFR) mediated caspase-3 expression and apoptosis of anti-CD3/anti-CD28 activated UCB T cells in the influence of IL-15. Activated caspase-3 expression was analyzed by Western blotting and the percentage of apoptotic cells was determined by annexin-V/propidium iodide (PI) flow cytometric staining. Significant expression of Fas and TNFR2 was detected on anti-CD3/anti-CD28 pre-activated UCB T cells. These cells were susceptible to anti-Fas but not TNF-alpha-induced apoptosis. Kinetic study shows that caspase-3 expression became evident at 6th-8th h following anti-Fas stimulation, while early apoptotic cells with annexin-V(+)/PI(-) expression appeared at 12th-16th h. IL-15, though successful in decreasing apoptosis in pre-activated UCB T cells, failed to completely prevent Fas-mediated caspase-3 expression and apoptosis of CB T cells. The pre-activated UCB and adult peripheral blood T cells behaved similarly with regard to death receptor expression, caspase-3 expression and apoptosis upon Fas-engagement. Although IL-15 promotes overall activated UCB T-cell survival, it did not particularly prevent Fas-mediated activation-induced cell death.


Apoptosis/immunology , Fetal Blood/immunology , T-Lymphocytes/immunology , fas Receptor/immunology , Annexin A5/drug effects , Annexin A5/immunology , Antibodies/immunology , CD28 Antigens/immunology , CD3 Complex/immunology , Caspase 3/drug effects , Caspase 3/metabolism , Female , Humans , Infant, Newborn , Interleukin-15/pharmacology , Lymphocyte Activation , Male , T-Lymphocytes/drug effects , Tumor Necrosis Factor-alpha/pharmacology
12.
Clin Cancer Res ; 14(10): 3113-23, 2008 May 15.
Article En | MEDLINE | ID: mdl-18483379

PURPOSE: To evaluate the potential utility of histone deacetylase inhibitors (HDACi) for treatment of retinoblastoma (RB). EXPERIMENTAL DESIGN: Growth-inhibitory effects of HDACi [trichostatin A (TSA), suberoylanilide hydroxamic acid (SAHA), or MS-275] were assessed in human and transgenic murine RB cells. Effects of TSA and MS-275 were also assessed in combination with standard therapeutic agents for RB. Proapoptotic effects of MS-275 and TSA were evaluated by caspase-3/7 activity, Annexin V translocation, and/or Bim expression analyses. Effects of MS-275 on cell cycle distribution and reactive oxygen species levels were determined by flow cytometry. Retinal tissue morphology was evaluated in mice after local administration of MS-275. Analysis of retinal acetyl-histone levels was used to assess MS-275 delivery after systemic administration. Therapeutic effects of MS-275 were determined in transgenic mouse and rat ocular xenograft models of RB after i.p. injection of 20 mg/kg every other day for 21 or 13 days, respectively. RESULTS: TSA, SAHA, and MS-275 dose dependently reduced RB cell survival. TSA and MS-275 showed additive growth-inhibitory effects in combination with carboplatin, etoposide, or vincristine. TSA and MS-275 increased caspase-3/7 activity. MS-275 increased Annexin V membrane translocation and induced G1 arrest. Cytotoxicity of MS-275 was dependent on increased reactive oxygen species levels and was reversed by antioxidant pretreatment. Intraocular administration of 1 microL of 10 micromol/L MS-275 did not alter ocular tissue morphology. Increased acetyl-histone levels confirmed MS-275 delivery to retinal tissue after systemic administration. MS-275 significantly reduced tumor burden in both mouse and rat models of RB. CONCLUSIONS: HDACi should be considered for clinical trials in children with RB.


Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Retinal Neoplasms/drug therapy , Retinoblastoma/drug therapy , Animals , Annexin A5/drug effects , Apoptosis/drug effects , Benzamides/pharmacology , Blotting, Western , Caspase 3/drug effects , Caspase 7/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Flow Cytometry , Humans , Hydroxamic Acids/pharmacology , In Vitro Techniques , Mice , Mice, Transgenic , Polymerase Chain Reaction , Pyridines/pharmacology , Rats , Reactive Oxygen Species/metabolism , Vorinostat , Xenograft Model Antitumor Assays
13.
Toxicol Appl Pharmacol ; 230(1): 57-66, 2008 Jul 01.
Article En | MEDLINE | ID: mdl-18377941

Arsenic contamination in drinking water is one of the biggest natural calamities, which has become an imperative threat to human health throughout the world. Abbreviation of erythrocyte lifespan leading to the development of anemia is a common sequel in arsenic exposed population. This study was undertaken to explore the mechanism of cell death in human erythrocytes during chronic arsenic exposure. Results revealed transformation of smooth discoid red cells into evaginated echinocytic form in the exposed individuals. Further distortion converted reversible echinocytes to irreversible spheroechinocytes. Arsenic toxicity increased membrane microviscosity along with an elevation of cholesterol/phospholipid ratio, which hampered the flexibility of red cell membrane and made them less deformable. Significant increase in the binding of merocyanine 540 with erythrocyte membrane due to arsenic exposure indicated disruption of lipid packing in the outer leaflet of the cell membrane resulting from altered transbilayer phospholipid asymmetry. Arsenic induced eryptosis was characterized by cell shrinkage and exposure of phosphatidylserine at the cell surface. Furthermore, metabolic starvation with depletion of cellular ATP triggered apoptotic removal of erythrocytes from circulation. Significant decrease in reduced glutathione content indicating defective antioxidant capacity was coupled with enhancement of malondialdehyde and protein carbonyl levels, which pointed to oxidative damage to erythrocyte membrane. Arsenic toxicity intervened into red cell membrane integrity eventually leading to membrane destabilization and hemoglobin release. The study depicted the involvement of both erythrophagocytosis and hemolysis in the destruction of human erythrocytes during chronic arsenic exposure.


Annexin A5/drug effects , Arsenic/adverse effects , Erythrocyte Membrane/drug effects , Hemolysis/drug effects , Phosphatidylserines/metabolism , Water Pollutants, Chemical/adverse effects , Adult , Anemia/etiology , Annexin A5/metabolism , Arsenic/analysis , Arsenic/urine , Erythrocyte Membrane/metabolism , Female , Humans , India , Male , Water Pollutants, Chemical/analysis , Water Pollutants, Chemical/urine
14.
Blood ; 111(4): 1951-61, 2008 Feb 15.
Article En | MEDLINE | ID: mdl-18024794

Adhesion of leukemic cells to vascular cells may confer resistance to chemotherapeutic agents. We hypothesized that disruption of leukemic cell cytoskeletal stability and interference with vascular cell interactions would promote leukemic cell death. We demonstrate that low and nontoxic doses of microtubule-destabilizing agent combretastatin-A4-phosphate (CA4P) inhibit leukemic cell proliferation in vitro and induce mitotic arrest and cell death. Treatment of acute myeloid leukemias (AMLs) with CA4P leads to disruption of mitochondrial membrane potential, release of proapoptotic mitochondrial membrane proteins, and DNA fragmentation, resulting in cell death in part through a caspase-dependent manner. Furthermore, CA4P increases intracellular reactive oxygen species (ROS), and antioxidant treatment imparts partial protection from cell death, suggesting that ROS accumulation contributes to CA4P-induced cytotoxicity in AML. In vivo, CA4P inhibited proliferation and circulation of leukemic cells and diminished the extent of perivascular leukemic infiltrates, prolonging survival of mice that underwent xenotransplantation without inducing hematologic toxicity. CA4P decreases the interaction of leukemic cells with neovessels by down-regulating the expression of the adhesion molecule VCAM-1 thereby augmenting leukemic cell death. These data suggest that CA4P targets both circulating and vascular-adherent leukemic cells through mitochondrial damage and down-regulation of VCAM-1 without incurring hematologic toxicities. As such, CA4P provides for an effective means to treat refractory organ-infiltrating leukemias.


Antineoplastic Agents, Phytogenic/pharmacology , Cell Death/drug effects , Leukemia, Myeloid, Acute/drug therapy , Mitochondria/physiology , Stilbenes/pharmacology , Annexin A5/drug effects , Annexin A5/metabolism , Apoptosis/drug effects , Cell Division/drug effects , DNA Damage/drug effects , Humans , Leukemia, Myeloid, Acute/pathology , Lymphoma/drug therapy , Lymphoma/pathology , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured
15.
Toxicol Appl Pharmacol ; 225(3): 318-28, 2007 Dec 15.
Article En | MEDLINE | ID: mdl-17881028

Undecylprodigiosin (UP) is a bacterial bioactive metabolite produced by Streptomyces and Serratia. In this study, we explored the anticancer effect of UP. Human breast carcinoma cell lines BT-20, MCF-7, MDA-MB-231 and T47D and one nonmalignant human breast epithelial cell line, MCF-10A, were tested in this study. We found that UP exerted a potent cytotoxicity against all breast carcinoma cell lines in a dose- and time-dependent manner. In contrast, UP showed limited toxicity to MCF-10A cells, indicating UP's cytotoxic effect is selective for malignant cells. UP's cytotoxic effect was due to apoptosis, as confirmed by positive TUNEL signals, annexin V-binding, caspase 9 activation and PARP cleavage. Notably, UP-induced apoptosis was blocked by the pan-caspase inhibitor z-VAD.fmk, further indicating the involvement of caspase activity. Moreover, UP caused a marked decrease of the levels of antiapoptotic BCL-X(L), Survivin and XIAP while enhancing the levels of proapoptotic BIK, BIM, MCL-1S and NOXA, consequently favoring induction of apoptosis. Additionally, we found that cells with functional p53 (MCF-7, T47D) or mutant p53 (BT-20, MDA-MB-231) were both susceptible to UP's cytotoxicity. Importantly, UP was able to induce apoptosis in MCF-7 cells with p53 knockdown by RNA interference, confirming the dispensability of p53 in UP-induced apoptosis. Overall, our results establish that UP induces p53-independent apoptosis in breast carcinoma cells with no marked toxicity to nonmalignant cells, raising the possibility of its use as a new chemotherapeutic drug for breast cancer irrespective of p53 status.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Tumor Suppressor Protein p53/metabolism , Annexin A5/drug effects , Annexin A5/metabolism , Antineoplastic Agents/administration & dosage , Apoptosis Regulatory Proteins/metabolism , Bcl-2-Like Protein 11 , Caspase 9/drug effects , Caspase 9/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Humans , In Situ Nick-End Labeling , Inhibitor of Apoptosis Proteins , Membrane Proteins/metabolism , Microtubule-Associated Proteins/drug effects , Microtubule-Associated Proteins/metabolism , Mitochondrial Proteins , Myeloid Cell Leukemia Sequence 1 Protein , Neoplasm Proteins/drug effects , Neoplasm Proteins/metabolism , Poly(ADP-ribose) Polymerases/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Prodigiosin/administration & dosage , Prodigiosin/analogs & derivatives , Prodigiosin/pharmacology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Serratia marcescens/chemistry , Survivin , Time Factors , Tumor Suppressor Protein p53/drug effects , X-Linked Inhibitor of Apoptosis Protein/drug effects , X-Linked Inhibitor of Apoptosis Protein/metabolism , bcl-X Protein/drug effects , bcl-X Protein/metabolism
16.
BMC Cancer ; 7: 237, 2007 Dec 31.
Article En | MEDLINE | ID: mdl-18166137

BACKGROUND: Medicinal plant is a main source of cancer drug development. Some of the cycloartane triterpenoids isolated from the aerial part of Cimicifuga dahurica showed cytotoxicity in several cancer cell lines. It is of great interest to examine the antiproliferative activity and mechanisms of total triterpenoid glycosides of C. dahurica and therefore might eventually be useful in the prevention or treatment of Hepatoma. METHODS: The total glycosides from the aerial part (TGA) was extracted and its cytotoxicity was evaluated in HepG2 cells and primary cultured normal mouse hepatocytes by an MTT assay. Morphology observation, Annexin V-FITC/PI staining, cell cycle analysis and western blot were used to further elucidate the cytotoxic mechanism of TGA. Implanted mouse H22 hepatoma model was used to demonstrate the tumor growth inhibitory activity of TGA in vivo. RESULTS: The IC50 values of TGA in HepG2 and primary cultured normal mouse hepatocytes were 21 and 105 mug/ml, respectively. TGA induced G0/G1 cell cycle arrest at lower concentration (25 mug/ml), and triggered G2/M arrest and apoptosis at higher concentrations (50 and 100 mug/ml respectively). An increase in the ratio of Bax/Bcl-2 was implicated in TGA-induced apoptosis. In addition, TGA inhibited the growth of the implanted mouse H22 tumor in a dose-dependent manner. CONCLUSION: TGA may potentially find use as a new therapy for the treatment of hepatoma.


Cimicifuga/chemistry , Glycosides/pharmacology , Hepatoblastoma/drug therapy , Hepatoblastoma/pathology , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Plant Extracts/pharmacology , Animals , Annexin A5/drug effects , Annexin A5/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Glycosides/chemistry , Glycosides/isolation & purification , Hepatoblastoma/metabolism , Humans , Inhibitory Concentration 50 , Liver Neoplasms/metabolism , Mice , Mice, Inbred ICR , Neoplasm Transplantation , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Cells, Cultured , bcl-2-Associated X Protein/drug effects , bcl-2-Associated X Protein/metabolism
17.
Cell Physiol Biochem ; 17(5-6): 201-10, 2006.
Article En | MEDLINE | ID: mdl-16790996

Prostaglandin-E2 (PGE2) is known to trigger suicidal death of nucleated cells (apoptosis) and enucleated erythrocytes (eryptosis). In erythrocytes PGE2 induced suicidal cell death involves activation of nonselective cation channels leading to Ca2+ entry followed by cell shrinkage and triggering of Ca2+ sensitive cell membrane scrambling with phosphatidylserine (PS) exposure at the cell surface. The present study was performed to explore whether PGE2 induces apoptosis of nucleated cells similarly through cation channel activation and to possibly disclose the molecular identity of the cation channels involved. To this end, Ca2+ activity was estimated from Fluo3 fluorescence, mitochondrial potential from DePsipher fluorescence, phosphatidylserine exposure from annexin binding, caspase activation from caspAce fluorescence, cell volume from FACS forward scatter, and DNA fragmentation utilizing a photometric enzyme immunoassay. Stimulation of K562 human leukaemia cells with PGE2 (50 microM) increased cytosolic Ca2+ activity, decreased forward scatter, depolarized the mitochondrial potential, increased annexin binding, led to caspase activation and resulted in DNA fragmentation. Gene silencing of the Ca2+-permeable transient receptor potential cation channel TRPC7 significantly blunted PGE2-induced triggering of PS exposure and DNA fragmentation. In conclusion, K562 cells express Ca2+-permeable TRPC7 channels, which are activated by PGE2 and participate in the triggering of apoptosis.


Apoptosis/drug effects , Apoptosis/physiology , Dinoprostone/pharmacology , Acid Sensing Ion Channels , Amiloride/analogs & derivatives , Amiloride/pharmacology , Annexin A5/drug effects , Annexin A5/metabolism , Calcium/metabolism , Caspases/drug effects , Caspases/metabolism , Cell Death/drug effects , Cell Size/drug effects , Cytosol/drug effects , Cytosol/metabolism , DNA Fragmentation , Dose-Response Relationship, Drug , Enzyme Activation , Gene Silencing , Humans , Ion Channels/drug effects , Ion Channels/metabolism , K562 Cells/drug effects , Membrane Potentials/drug effects , Membrane Proteins/drug effects , Membrane Proteins/genetics , Mitochondria/drug effects , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/genetics , Phosphatidylserines/metabolism , Sodium Channels/drug effects , Sodium Channels/genetics , TRPC Cation Channels/drug effects , TRPC Cation Channels/genetics
19.
Mol Imaging ; 4(1): 1-6, 2005.
Article En | MEDLINE | ID: mdl-15967121

We examined the accumulation of Cy5.5-labeled annexin V in the paws of mice with and without collagen-induced arthritis, with and without methotrexate (MTX) treatment, by near-infrared fluorescence imaging. Fluorescence reflectance imaging (FRI) of paws was performed 48 hr after MTX injection and at 10 min and 3 hr after the injection of Cy5.5-annexin V (1 nmol dye per mouse). With arthritic paws, MTX treatment caused a 7-fold increase in fluorescence intensity compared with the paws of untreated mice and a 4-fold increase compared to nonarthritic paws of MTX-treated mice (p < .001 each). Tissue samples of paws were examined histologically for Cy5.5 fluorescence and by TUNEL staining for apoptosis. Cy5.5-annexin V was seen in the hyperplastic synovia of MTX-treated mice, and TUNEL staining for apoptosis showed apoptotic cells in the hyperplastic synovia. Monitoring the uptake of Cy5.5-annexin V in arthritic paws by FRI provided a method of assessing a response to MTX, a response that was readily quantitated with simple instrumentation and that occurred before conventional measurements of treatment response.


Annexin A5/metabolism , Antirheumatic Agents/pharmacology , Arthritis, Experimental/metabolism , Methotrexate/pharmacology , Animals , Annexin A5/drug effects , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Carbocyanines/metabolism , Collagen/toxicity , Fluorescent Dyes/metabolism , Joints/drug effects , Joints/metabolism , Joints/pathology , Male , Mice , Mice, Inbred DBA , Molecular Biology/methods , Synovial Fluid/metabolism
20.
Transfusion ; 45(6): 911-9, 2005 Jun.
Article En | MEDLINE | ID: mdl-15934989

BACKGROUND: Several strategies are being developed to reduce the risk of pathogen transmission associated with platelet (PLT) transfusion. STUDY DESIGN AND METHODS: The impact of a new technology for pathogen reduction based on riboflavin plus illumination (Mirasol PRT, Navigant Biotechnologies, Inc.) at 6.2 and 12.3 J per mL on functional and biochemical characteristics of PLTs was evaluated. PLT concentrates (PCs) obtained by apheresis were treated with Mirasol PRT and stored at 22 degrees C. Modifications in major PLT glycoproteins (GPIbalpha, GPIV, and GPIIb-IIIa), adhesive ligands (von Willebrand factor [VWF], fibrinogen [Fg], and fibronectin), activation antigens (P-selectin and LIMP), and apoptotic markers (annexin V binding and factor [F]Va) were analyzed by flow cytometry. Adhesive and cohesive PLT functions were evaluated with well-established perfusion models. Studies were performed on the preparation day (Day 0) and during PCs storage (Days 3 and 5). RESULTS: Levels of glycoproteins remained stable during storage in PCs treated with 6.2 J per mL pathogen reduction technology (PRT) and similar to those observed in nontreated PCs. When 12.3 J per mL PRT was applied, however, levels of GPIbalpha moderately decreased on Days 3 and 5. VWF, Fg, and FVa were not modified in their expression levels, either by treatment or by storage period. Fibronectin appeared more elevated in all PRT samples. A progressive increase in P-selectin and LIMP expression and in annexin V binding was observed during storage of PRT-treated PCs. Functional studies indicated that 6.2 J per mL Mirasol PRT-treated PLTs preserved adhesive and cohesive functions to levels compatible with those observed in the respective control PCs. CONCLUSION: PLT function was well preserved in PCs treated with 6.2 J per mL Mirasol PRT and stored for 5 days.


Blood Platelets , Blood Platelets/drug effects , Blood Platelets/radiation effects , Blood Preservation , Riboflavin/pharmacology , Ultraviolet Rays , Annexin A5/analysis , Annexin A5/drug effects , Annexin A5/radiation effects , Antigens, CD/analysis , Antigens, CD/drug effects , Antigens, CD/radiation effects , Blood Platelets/chemistry , Blood Platelets/cytology , Blood Platelets/physiology , Fibrinogen/analysis , Fibrinogen/drug effects , Fibrinogen/radiation effects , Fibronectins/analysis , Fibronectins/drug effects , Fibronectins/radiation effects , Flow Cytometry , Humans , Lysosomal Membrane Proteins , P-Selectin/analysis , P-Selectin/drug effects , P-Selectin/radiation effects , Platelet Activation/drug effects , Platelet Activation/radiation effects , Platelet Adhesiveness/drug effects , Platelet Adhesiveness/radiation effects , Platelet Count , Platelet Glycoprotein GPIIb-IIIa Complex/analysis , Platelet Glycoprotein GPIIb-IIIa Complex/drug effects , Platelet Glycoprotein GPIIb-IIIa Complex/radiation effects , Platelet Glycoprotein GPIb-IX Complex/analysis , Platelet Glycoprotein GPIb-IX Complex/drug effects , Platelet Glycoprotein GPIb-IX Complex/radiation effects , Platelet Membrane Glycoprotein IIb/analysis , Platelet Membrane Glycoprotein IIb/drug effects , Platelet Membrane Glycoprotein IIb/radiation effects , Platelet Transfusion , Plateletpheresis , Temperature , Time Factors , von Willebrand Factor/analysis , von Willebrand Factor/drug effects , von Willebrand Factor/radiation effects
...