Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Biochem Biophys Res Commun ; 503(1): 249-256, 2018 09 03.
Article in English | MEDLINE | ID: mdl-29885833

ABSTRACT

Malignant melanoma has shown increased incidence and high mortality rate in the last three decades. In this study, we investigated whether combination therapy with ch282-5 (a novel BH3 mimetic) and microwave hyperthermia could display synergistic antitumor effects against melanoma. Our results indicated that combination therapy reduced the viability and proliferation of melanoma cells. Through inhibiting the expression of anti-apoptotic proteins of Bcl-2 and IAP family and activating MAPK proteins, combined hyperthermia enhanced ch282-5-induced apoptosis. Combination therapy also synergistically disturbed the mTOR/p70S6k signaling pathway, which is important for cell survival and migration. Moreover, our results showed that combination therapy remarkably suppressed melanoma cell migration in vitro and significantly reduced experimental pulmonary metastasis in vivo. In conclusion, our results indicate that combination therapy with ch282-5 and hyperthermia has synergistic antitumor effects and provides a possible therapeutic strategy for advanced melanoma.


Subject(s)
BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , Hyperthermia, Induced , Melanoma/therapy , Skin Neoplasms/therapy , Animals , Apoptosis/drug effects , Biomimetics , Cell Line, Tumor , Cell Movement/drug effects , Combined Modality Therapy , Gossypol/analogs & derivatives , Gossypol/pharmacology , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Male , Melanoma/metabolism , Melanoma/pathology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Microwaves/therapeutic use , Signal Transduction/drug effects , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
2.
J Bioenerg Biomembr ; 49(1): 65-74, 2017 Feb.
Article in English | MEDLINE | ID: mdl-26698318

ABSTRACT

Mitochondrial Apoptotic Channel inhibitors or iMACs are di-bromocarbazole derivatives with anti-apoptotic function which have been tested and validated in several mouse models of brain injury and neurodegeneration. Owing to the increased therapeutic potential of these compounds, we sought to expand our knowledge of their mechanism of action. We investigated the kinetics of MAC inhibition in mitochondria from wild type, Bak, and Bax knockout cell lines using patch clamp electrophysiology, fluorescence microscopy, ELISA, and semiquantitative western blot analyses. Our results show that iMACs work through at least two mechanisms: 1) by blocking relocation of the cytoplasmic Bax protein to mitochondria and 2) by disassembling Bax and Bak oligomers in the mitochondrial outer membrane. iMACs exert comparable effects on channel conductance of Bax or Bak and similarly affect cytochrome c release from Bax or Bak-containing mitochondria. Interestingly, wild type mitochondria were more susceptible to inhibition than the Bak or Bax knockouts. Western blot analysis showed that wild type mitochondria had lower steady state levels of Bak in the absence of apoptotic stimulation.


Subject(s)
Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , Carbazoles/pharmacology , Mitochondria/metabolism , Protein Multimerization/drug effects , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Cell Line , Cytochromes c/metabolism , Fibroblasts/cytology , Mice
3.
Proc Natl Acad Sci U S A ; 111(46): 16520-5, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25368155

ABSTRACT

Colorectal tumorigenesis is driven by genetic alterations in the adenomatous polyposis coli (APC) tumor suppressor pathway and effectively inhibited by nonsteroidal antiinflammatory drugs (NSAIDs). However, how NSAIDs prevent colorectal tumorigenesis has remained obscure. We found that the extrinsic apoptotic pathway and the BH3 interacting-domain death agonist (BID) are activated in adenomas from NSAID-treated patients. Loss of BID abolishes NSAID-mediated tumor suppression, survival benefit, and apoptosis in tumor-initiating stem cells in APC(Min/+) mice. BID-mediated cross-talk between the extrinsic and intrinsic apoptotic pathways is responsible for selective killing of neoplastic cells by NSAIDs. We further demonstrate that NSAIDs induce death receptor signaling in both cancer and normal cells, but only activate BID in cells with APC deficiency and ensuing c-Myc activation. Our results suggest that NSAIDs suppress intestinal tumorigenesis through BID-mediated synthetic lethality triggered by death receptor signaling and gatekeeper mutations, and provide a rationale for developing more effective cancer prevention strategies and agents.


Subject(s)
Adenomatous Polyposis Coli/prevention & control , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis , BH3 Interacting Domain Death Agonist Protein/physiology , Genes, APC , Adenomatous Polyposis Coli/pathology , Animals , Apoptosis Regulatory Proteins/physiology , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , BH3 Interacting Domain Death Agonist Protein/deficiency , BH3 Interacting Domain Death Agonist Protein/genetics , Caspases/physiology , Cell Line, Tumor , Colon/pathology , Gene Expression Regulation, Neoplastic , Genes, myc , Humans , Indomethacin/pharmacology , Intestine, Small/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mitochondria/metabolism , Organ Specificity , Pyrazoles/pharmacology , RNA, Small Interfering/pharmacology , Receptors, Death Domain/physiology , Stem Cells/metabolism , Stem Cells/pathology , Sulfonamides/pharmacology , Sulindac/pharmacology
4.
J Hepatol ; 64(3): 699-707, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26555271

ABSTRACT

BACKGROUND & AIMS: Liver fibrosis is the most worrisome feature of non-alcoholic steatohepatitis (NASH). Growing evidence supports a link between hepatocyte apoptosis and liver fibrogenesis. Our aim was to determine the therapeutic efficacy and safety of liver Bid, a key pro-apoptotic molecule, suppression using RNA interference (RNAi) for the treatment of fibrosis. METHODS: First, we optimized the delivery system for Bid siRNA in mice using ten different stealth RNAi siRNAs and two lipid formulations -Invivofectamine2.0 and a newly developed Invivofectamine3.0 - that have been designed for high efficacy accumulation in the liver, assessed via real-time PCR of Bid mRNA. Next, C57BL/6 mice were placed on a choline-deficient L-amino acid defined (CDAA) diet. After 19weeks of the CDAA diet, a time point that results in severe fibrotic NASH, mice were injected with the selected Bid siRNA-Invivofectamine3.0 biweekly for three weeks. Additionally hepatocyte-specific Bid deficient (Bid(Δhep)) mice were placed on CDAA diet for 20weeks. RESULTS: A maximum Bid knockdown was achieved at 1.5mg/kg siRNA with Invivofectamine3.0, whereas it was at 7mg/kg with Invivofectamine2.0. In NASH mice, after 3weeks of treatment, BID protein was reduced to 10% and this was associated with an improvement in liver fibrosis and inflammation associated with a marked reduction in TUNEL positive cells, caspase 3 activation, and a reduction in mitochondrial BAX and BAK. Bid(Δhep) mice showed similar protection from fibrotic changes. CONCLUSION: Our data demonstrate that liver Bid suppression by RNAi technology, as well as hepatocyte-specific Bid deficiency, improves liver fibrosis coupled with a reduction of inflammation in experimental NASH. These findings are consistent with existing evidence that hepatocyte apoptosis triggers hepatic stellate cell activation and liver fibrosis and suggest that Bid inhibition may be useful as an antifibrotic NASH therapy.


Subject(s)
BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , Liver Cirrhosis, Experimental/therapy , Non-alcoholic Fatty Liver Disease/complications , RNA Interference , Animals , Apoptosis , BH3 Interacting Domain Death Agonist Protein/genetics , Extracellular Vesicles/physiology , Hep G2 Cells , Humans , Liver/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitochondria/physiology
5.
Anal Chem ; 88(16): 8050-7, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27425825

ABSTRACT

A simple and effective method for identifying inhibitors of protein-protein interactions (PPIs) was developed by using capillary electrophoresis frontal analysis (CE-FA). Antiapoptotic B-cell-2 (Bcl-2) family member Bcl-XL protein, a 5-carboxyfluorescein labeled peptide truncated from the BH3 domain of Bid (F-Bid) as the ligand, and a known Bcl-XL-Bid interaction inhibitor ABT-263 were employed as an experimental model for the proof of concept. In CE-FA, the free ligand is separated from the protein and protein-ligand complex to permit the measurement of the equilibrium concentration of the ligand, hence the dissociation constant of the protein-ligand complex. In the presence of inhibitors, formation of the protein-ligand complex is hindered, thereby the inhibition can be easily identified by the raised plateau height of the ligand and the decayed plateau of the complex. Further, we proposed an equation used to convert the IC50 value into the inhibition constant Ki value, which is more useful than the former for comparison. In addition, the sample pooling strategy was employed to improve the screening throughput more than 10 times. A small chemical library composed of synthetic compounds and natural extracts were screened with the method, two natural products, namely, demethylzeylasteral and celastrol, were identified as new inhibitors to block the Bcl-XL-Bid interaction. Cell-based assay was performed to validate the activity of the identified compounds. The result demonstrated that CE-FA represents a straightforward and robust technique for screening of PPI inhibitors.


Subject(s)
BH3 Interacting Domain Death Agonist Protein/metabolism , Electrophoresis, Capillary/methods , Small Molecule Libraries/metabolism , bcl-X Protein/metabolism , Aniline Compounds/chemistry , Aniline Compounds/metabolism , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , Binding, Competitive , Fluoresceins/chemistry , HT29 Cells , Humans , Kinetics , Ligands , Pentacyclic Triterpenes , Protein Interaction Domains and Motifs/drug effects , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Sulfonamides/chemistry , Sulfonamides/metabolism , Triterpenes/chemistry , Triterpenes/metabolism , Triterpenes/pharmacology , bcl-X Protein/antagonists & inhibitors
6.
Cell Physiol Biochem ; 36(3): 1037-48, 2015.
Article in English | MEDLINE | ID: mdl-26112407

ABSTRACT

BACKGROUND/AIMS: Phosphatidylinositol (PI) regulates a variety of cell processes. The present study investigated the antitumor action of 1,2-dioleoyl-sn-glycero-3-phospho-(1'-myo-inositol)(DOPI) and 1,2-dipalmitoyl-sn-glycero-3-phospho-(1'-myo-inositol)(DPPI) on human malignant pleural mesothelioma (MPM) cell lines such as NCI-H28, NCI-H2052, NCI-H2452, and MSTO-211H cells. METHODS: MTT assay, TUNEL staining, flow cytometry using propidium iodide (PI) and annexin V (AV), enzymatic caspase assay, and nuclear staining using DAPI were carried out, and mitochondrial membrane potentials and intracellular distribution of apoptosis-inducing factor (AIF) were monitored in cells with and without the siRNA silencing the Bid-targeted gene. RESULTS: Both DOPI and DPPI reduced cell viability for all the investigated MPM cell lines in a concentration (0.01-100 µM)-dependent manner. DOPI and DPPI significantly increased TUNEL-positive cells and the population of PI-negative/AV-positive and PI-positive/AV-positive cells, corresponding to early apoptosis and late apoptosis/secondary necrosis, respectively. DOPI and DPPI perturbed mitochondrial membrane potentials in MSTO-211H cells, but no significant activation of caspase-3, -4, -8, and -9 was obtained. DOPI and DPPI upregulated expression of Bid in MSTO-211H cells. DOPI and DPPI significantly increased nuclear localization of AIF without affecting expression of the mRNAs and proteins in MSTO-211H cells, which was inhibited by knocking-down Bid. In the DAPI staining, nuclear fragmentation and condensation were found. CONCLUSION: The results of the present study indicate that DOPI and DPPI facilitate Bid-mediated AIF release from the mitochondria, to accumulate AIF in the nucleus and induce caspase-independent apoptosis of MPM cells.


Subject(s)
Apoptosis Inducing Factor/genetics , Apoptosis/drug effects , Cell Nucleus/drug effects , Epithelial Cells/drug effects , Gene Expression Regulation, Neoplastic , Apoptosis Inducing Factor/metabolism , BH3 Interacting Domain Death Agonist Protein/agonists , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , BH3 Interacting Domain Death Agonist Protein/genetics , BH3 Interacting Domain Death Agonist Protein/metabolism , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Survival/drug effects , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , In Situ Nick-End Labeling , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Phosphatidylinositols , Pleura/drug effects , Pleura/metabolism , Pleura/pathology , Protein Transport/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction
7.
Org Biomol Chem ; 13(19): 5520-31, 2015 May 21.
Article in English | MEDLINE | ID: mdl-25877523

ABSTRACT

The synthesis, on a large scale, with very good yield and er via an efficient strategy, of a chiral 4-substituted 2-cyclohexenone intermediate, was a milestone in the synthesis of seven analogues of meiogynin A, a natural sesquiterpenoid dimer. These compounds were elaborated in ten linear steps. Their binding affinities for Bcl-xL and Mcl-1, two proteins of the Bcl-2 family, overexpressed in various types of cancers, were evaluated. This enabled to further SAR studies en route to the elaboration of potent dual inhibitors of anti-apoptotic proteins of the Bcl-2 family.


Subject(s)
Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Sesquiterpenes/pharmacology , bcl-X Protein/antagonists & inhibitors , Aldehydes/chemistry , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , BH3 Interacting Domain Death Agonist Protein/metabolism , Chromatography, High Pressure Liquid , Cyclohexanones/chemical synthesis , Cyclohexanones/chemistry , Ligands , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Naphthalenes/chemistry , Sesquiterpenes/chemical synthesis , Sesquiterpenes/chemistry , bcl-2 Homologous Antagonist-Killer Protein/antagonists & inhibitors , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-X Protein/metabolism
8.
Glia ; 62(6): 855-80, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24616078

ABSTRACT

The roles of cathepsins in the ischemic astrocytic injury remain unclear. Here, we test the hypothesis that activation of cathepsin B and L contributes to the ischemic astrocyte injury via the tBid-mitochondrial apoptotic signaling pathways. In the rat models of pMCAO, CA-074Me or Clik148, a selective inhibitor of cathepsin B or cathepsin L, reduced the infarct volume, improved the neurological deficits and increased the MAP2 and GFAP levels. In OGD-induced astrocyte injury, CA-074Me or Clik148 decreased the LDH leakage and increased the GFAP levels. In the ischemic cortex or OGD-induced astrocytes injury, Clik148 or CA-074Me reversed pMCAO or OGD-induced increase in active cathepsin L or cathepsin B at 3 h or 6 h, increase in tBid, reduction in mitochondrial cytochrome-c (Cyt-c) and increase in cytoplastic Cyt-c and active caspase-3 at 12-24 h of the late stage of pMCAO or OGD. CA-074Me or Clik148 also reduced cytosolic and mitochondrial tBid, increased mitochondrial Cyt-c and decreased cytoplastic Cyt-c and active caspase-3 at 6 h of the early stage of Bid activation. CA-074Me or Clik148 blocked the pMCAO-induced release of cathepsin B or L from the lysosomes into the cytoplasm and activation of caspase-3 in ischemic astrocytes at 12 h after ischemia. Concurrent inhibition of cathepsin B and cathepsin L provided better protection on the OGD-induced astrocytic apoptosis than obtained with separate use of each inhibitor. These results suggest that inhibition of the cysteine cathepsin B and cathepsin L activation in ischemic astrocytes contributes to neuroprotection via blocking the tBid-mitochondrial apoptotic signaling pathway.


Subject(s)
Apoptosis Inducing Factor/antagonists & inhibitors , Astrocytes/drug effects , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , Brain Ischemia/prevention & control , Cathepsin B/antagonists & inhibitors , Cathepsin L/antagonists & inhibitors , Animals , Apoptosis Inducing Factor/metabolism , Astrocytes/metabolism , BH3 Interacting Domain Death Agonist Protein/metabolism , Brain Ischemia/metabolism , Cathepsin B/metabolism , Cathepsin L/metabolism , Cells, Cultured , Cysteine/antagonists & inhibitors , Cysteine/metabolism , Epoxy Compounds/pharmacology , Epoxy Compounds/therapeutic use , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Rats , Rats, Sprague-Dawley
9.
Neurobiol Dis ; 70: 99-107, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24956542

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the loss of motoneurons in the spinal cord, brainstem and motor cortex. Mutations in the superoxide dismutase 1 (SOD1) gene represent a frequent genetic determinant and recapitulate a disease phenotype similar to ALS when expressed in mice. Previous studies using SOD1(G93A) transgenic mice have suggested a paracrine mechanism of neuronal loss, in which cytokines and other toxic factors released from astroglia or microglia trigger motoneuron degeneration. Several pro-inflammatory cytokines activate death receptors and may downstream from this activate the Bcl-2 family protein, Bid. We here sought to investigate the role of Bid in astrocyte activation and non-cell autonomous motoneuron degeneration. We found that spinal cord Bid protein levels increased significantly during disease progression in SOD1(G93A) mice. Subsequent experiments in vitro indicated that Bid was expressed at relatively low levels in motoneurons, but was enriched in astrocytes and microglia. Bid was strongly induced in astrocytes in response to pro-inflammatory cytokines or exposure to lipopolysaccharide. Experiments in bid-deficient astrocytes or astrocytes treated with a small molecule Bid inhibitor demonstrated that Bid was required for the efficient activation of transcription factor nuclear factor-κB in response to these pro-inflammatory stimuli. Finally, we found that conditioned medium from wild-type astrocytes, but not from bid-deficient astrocytes, was toxic when applied to primary motoneuron cultures. Collectively, our data demonstrate a new role for the Bcl-2 family protein Bid as a mediator of astrocyte activation during neuroinflammation, and suggest that Bid activation may contribute to non-cell autonomous motoneuron degeneration in ALS.


Subject(s)
Astrocytes/immunology , BH3 Interacting Domain Death Agonist Protein/metabolism , Superoxide Dismutase/metabolism , Amyotrophic Lateral Sclerosis , Animals , Anterior Horn Cells/physiology , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , BH3 Interacting Domain Death Agonist Protein/genetics , Cell Death/physiology , Cells, Cultured , Humans , Lipopolysaccharides , Mice, Knockout , Mice, Transgenic , Microglia/immunology , Motor Neurons/physiology , NF-kappa B/metabolism , Neurodegenerative Diseases/physiopathology , Neuroimmunomodulation/physiology , Superoxide Dismutase/genetics , Superoxide Dismutase-1
10.
Bioorg Med Chem Lett ; 24(21): 5086-8, 2014 Nov 01.
Article in English | MEDLINE | ID: mdl-25266781

ABSTRACT

The biological evaluation of a natural sesquiterpene dimer meiogynin A 1, is described as well as that of five non-natural analogues. Although active on a micromolar range on the inhibition of Bcl-xL/Bak and Mcl-1/Bid interaction, meiogynin A 1 is not cytotoxic on three cell lines that overexpress Bcl-xL and Mcl-1. Contrarily, one of its analogues 6 with an inverted configuration on the side chain and an aromatic moiety replacing the cyclohexane ring was active on both target proteins, cytotoxic on a micromolar range and was found to induce apoptosis through a classical pathway.


Subject(s)
Benzoates/chemistry , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Naphthalenes/chemistry , Sesquiterpenes/chemistry , bcl-X Protein/metabolism , Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , BH3 Interacting Domain Death Agonist Protein/metabolism , Benzoates/chemical synthesis , Benzoates/pharmacology , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Humans , Molecular Docking Simulation , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Naphthalenes/chemical synthesis , Naphthalenes/pharmacology , Poly(ADP-ribose) Polymerases/metabolism , Protein Structure, Tertiary , Sesquiterpenes/chemical synthesis , Sesquiterpenes/pharmacology , bcl-2 Homologous Antagonist-Killer Protein/antagonists & inhibitors , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-X Protein/antagonists & inhibitors
11.
J Biol Chem ; 287(18): 15016-23, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22416135

ABSTRACT

The molecular basis of the interaction between mitochondrial carrier homologue 2 (MTCH2) and truncated BID (tBID) was characterized. These proteins participate in the apoptotic pathway, and the interaction between them may serve as a target for anticancer lead compounds. In response to apoptotic signals, MTCH2 recruits tBID to the mitochondria, where it activates apoptosis. A combination of peptide arrays screening with biochemical and biophysical techniques was used to characterize the mechanism of the interaction between tBID and MTCH2 at the structural and molecular levels. The regions that mediate the interaction between the proteins were identified. The two specific binding sites between the proteins were determined to be tBID residues 59-73 that bind MTCH2 residues 140-161, and tBID residues 111-125 that bind MTCH2 residues 240-290. Peptides derived from tBID residues 111-125 and 59-73 induced cell death in osteosarcoma cells. These peptides may serve as lead compounds for anticancer drugs that act by targeting the tBID-MTCH2 interaction.


Subject(s)
Apoptosis , BH3 Interacting Domain Death Agonist Protein/metabolism , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Antineoplastic Agents/pharmacology , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , BH3 Interacting Domain Death Agonist Protein/genetics , Cell Line, Tumor , Humans , Mitochondria/genetics , Mitochondrial Membrane Transport Proteins/genetics , Osteosarcoma/drug therapy , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology , Peptides/metabolism , Peptides/pharmacology , Protein Array Analysis
12.
Apoptosis ; 18(11): 1437-1446, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23851982

ABSTRACT

PP2A activator FTY720 has been shown to possess the anti-leukemic activity for chronic myelogenous leukemia (CML), however, the cell killing mechanism underlying its anti-leukemic activity has remained to be verified. We investigated the precise mechanisms underlying the apoptosis induction by FTY720, especially focusing on the roles of BH3-only proteins, and the therapeutic potency of FTY720 for CML. Enforced expression of either BCL2 or the dominant-negative protein of FADD (FADD.DN) partly protected CML cells from apoptosis by FTY720, indicating the involvement of both cell extrinsic and intrinsic apoptosis pathways. FTY720 activates pro-apoptotic BH3-only proteins: BIM, which is essential for apoptosis by BCR-ABL1 tyrosine kinase inhibitors (TKIs), and BID, which accelerates the extrinsic apoptosis pathway. Gene knockdown of either BIM or BID partly protected K562 cells from apoptosis by FTY720, but the extent of cell protection was not as much as that by overexpression of either BCL2 or FADD.DN. Moreover, knockdown of both BIM and BID did not provide additional protection compared with knockdown of only BIM or BID, indicating that BIM and BID complement each other in apoptosis by FTY720, especially when either is functionally impaired. FTY720 can overcome TKI resistance caused by ABL kinase domain mutations, dysfunction of BIM resulting from gene deletion polymorphism, and galectin-3 overexpression. In addition, ABT-263, a BH3-mimetic, significantly augmented cell death induction by FTY720 both in TKI-sensitive and -resistant leukemic cells. These results provide the rationale that FTY720, with its unique effects on BIM and BID, could lead to new therapeutic strategies for CML.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/genetics , BH3 Interacting Domain Death Agonist Protein/genetics , Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Membrane Proteins/genetics , Propylene Glycols/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/genetics , Sphingosine/analogs & derivatives , Apoptosis/drug effects , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/metabolism , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , BH3 Interacting Domain Death Agonist Protein/metabolism , Bcl-2-Like Protein 11 , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Fingolimod Hydrochloride , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mutation , Primary Cell Culture , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Sphingosine/pharmacology , bcl-Associated Death Protein/genetics , bcl-Associated Death Protein/metabolism
13.
Mol Psychiatry ; 17(8): 770-80, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21727899

ABSTRACT

The BH3-interacting domain death agonist (Bid) is a pro-apoptotic member of the B-cell lymphoma-2 (Bcl-2) protein family. Previous studies have shown that stress reduces levels of Bcl-2 in brain regions implicated in the pathophysiology of mood disorders, whereas antidepressants and mood stabilizers increase Bcl-2 levels. The Bcl-2 protein family has an essential role in cellular resilience as well as synaptic and neuronal plasticity and may influence mood and affective behaviors. This study inhibited Bid in mice using two pharmacological antagonists (BI-11A7 and BI-2A7); the selective serotonin reuptake inhibitor citalopram was used as a positive control. These agents were studied in several well-known rodent models of depression-the forced swim test (FST), the tail suspension test (TST), and the learned helplessness (LH) paradigm-as well as in the female urine sniffing test (FUST), a measure of sex-related reward-seeking behavior. Citalopram and BI-11A7 both significantly reduced immobility time in the FST and TST and attenuated escape latencies in mice that underwent the LH paradigm. In the FUST, both agents significantly improved duration of female urine sniffing in mice that had developed helplessness. LH induction increased the activation of apoptosis-inducing factor (AIF), a caspase-independent cell death constituent activated by Bid, and mitochondrial AIF expression was attenuated by chronic BI-11A7 infusion. Taken together, the results suggest that functional perturbation of apoptotic proteins such as Bid and, alternatively, enhancement of Bcl-2 function, is a putative strategy for developing novel therapeutics for mood disorders.


Subject(s)
Aniline Compounds/therapeutic use , Antidepressive Agents/therapeutic use , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , Citalopram/therapeutic use , Depression/drug therapy , Drug Delivery Systems/psychology , Sulfonamides/therapeutic use , Aniline Compounds/administration & dosage , Aniline Compounds/pharmacology , Animals , Antidepressive Agents/administration & dosage , Antidepressive Agents/pharmacology , Apoptosis Inducing Factor/metabolism , Apoptosis Regulatory Proteins , Behavior, Animal/drug effects , Carrier Proteins/metabolism , Citalopram/administration & dosage , Cytochromes c/metabolism , Disease Models, Animal , Drug Delivery Systems/methods , Infusions, Intraventricular , Male , Mice , Mice, Inbred Strains , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Sulfides/administration & dosage , Sulfides/pharmacology , Sulfides/therapeutic use , Sulfonamides/administration & dosage , Sulfonamides/pharmacology
14.
Exp Cell Res ; 318(13): 1564-76, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22513214

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is often resistant to conventional chemotherapy and thus requires novel treatment regimens. Here, we investigated the effects of the proteasome inhibitor MG132 in combination with tumor necrosis factor-related apoptosis inducing ligand (TRAIL) or agonistic TRAIL receptor 1 (DR4)-specific monoclonal antibody, AY4, on sensitization of TRAIL- and AY4-resistant human HNSCC cell lines. Combination treatment of HNSCC cells synergistically induced apoptotic cell death accompanied by caspase-8, caspase-9, and caspase-3 activation and Bid cleavage into truncated Bid (tBid). Generation and accumulation of tBid through the cooperative action of MG132 with TRAIL or AY4 and Bik accumulation through MG132-mediated proteasome inhibition are critical to the synergistic apoptosis. In HNSCC cells, Bak was constrained by Mcl-1 and Bcl-X(L), but not by Bcl-2. Conversely, Bax did not interact with Mcl-1, Bcl-X(L), or Bcl-2. Importantly, tBid plays a major role in Bax activation, and Bik indirectly activates Bak by displacing it from Mcl-1 and Bcl-X(L), pointing to the synergistic mechanism of the combination treatment. In addition, knockdown of both Mcl-1 and Bcl-X(L) significantly sensitized HNSCC cells to TRAIL and AY4 as a single agent, suggesting that Bak constraint by Mcl-1 and Bcl-X(L) is an important resistance mechanism of TRAIL receptor-mediated apoptotic cell death. Our results provide a novel molecular mechanism for the potent synergy between MG132 proteasome inhibitor and TRAIL receptor agonists in HNSCC cells, suggesting that the combination of these agents may offer a new therapeutic strategy for HNSCC treatment.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Head and Neck Neoplasms/drug therapy , Leupeptins/administration & dosage , Proteasome Inhibitors , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Antibodies, Monoclonal/administration & dosage , Apoptosis/drug effects , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , BH3 Interacting Domain Death Agonist Protein/genetics , BH3 Interacting Domain Death Agonist Protein/metabolism , Base Sequence , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Drug Synergism , Gene Knockdown Techniques , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondrial Proteins , Myeloid Cell Leukemia Sequence 1 Protein , Protein Stability/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering/genetics , TNF-Related Apoptosis-Inducing Ligand/administration & dosage , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/genetics , bcl-X Protein/metabolism
15.
Proc Natl Acad Sci U S A ; 107(24): 10967-71, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20534453

ABSTRACT

Death by apoptosis shapes tissue homeostasis. Apoptotic mechanisms are so universal that harnessing them for tailored immune intervention would seem challenging; however, the range and different expression levels of pro- and anti-apoptotic molecules among tissues offer hope that targeting only a subset of such molecules may be therapeutically useful. We examined the effects of the drug ABT-737, a mimetic of the killer BH3 domain of the Bcl-2 family of proteins that induces apoptosis by antagonizing Bcl-2, Bcl-X(L), and Bcl-W (but not Mcl-1 and A1), on the mouse immune system. Treatment with ABT-737 reduced the numbers of selected lymphocyte and dendritic cell subpopulations, most markedly in lymph nodes. It inhibited the persistence of memory B cells, the establishment of newly arising bone marrow plasma cells, and the induction of a cytotoxic T cell response. Preexisting plasma cells and germinal centers were unaffected. Notably, ABT-737 was sufficiently immunomodulatory to allow long-term survival of pancreatic allografts, reversing established diabetes in this model. These results provide an insight into the selective mechanisms of immune cell survival and how this selectivity avails a different strategy for immune modulation.


Subject(s)
BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , Peptide Fragments/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Animals , Apoptosis/drug effects , Apoptosis/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Biphenyl Compounds/pharmacology , Graft Rejection/prevention & control , Immunity, Humoral/drug effects , Immunologic Factors/pharmacology , Islets of Langerhans Transplantation , Leukocytes/classification , Leukocytes/cytology , Leukocytes/drug effects , Leukocytes/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Inbred NOD , Mice, Knockout , Nitrophenols/pharmacology , Piperazines/pharmacology , Sulfonamides/pharmacology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Transplantation, Homologous
16.
J Lipid Res ; 53(10): 2102-2114, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22761256

ABSTRACT

Upon apoptotic stimuli, lysosomal proteases, including cathepsins and chymotrypsin, are released into cytosol due to lysosomal membrane permeabilization (LMP), where they trigger apoptosis via the lysosomal-mitochondrial pathway of apoptosis. Herein, the mechanism of LMP was investigated. We found that caspase 8-cleaved Bid (tBid) could result in LMP directly. Although Bax or Bak might modestly enhance tBid-triggered LMP, they are not necessary for LMP. To study this further, large unilamellar vesicles (LUVs), model membranes mimicking the lipid constitution of lysosomes, were used to reconstitute the membrane permeabilization process in vitro. We found that phosphatidic acid (PA), one of the major acidic phospholipids found in lysosome membrane, is essential for tBid-induced LMP. PA facilitates the insertion of tBid deeply into lipid bilayers, where it undergoes homo-oligomerization and triggers the formation of highly curved nonbilayer lipid phases. These events induce LMP via pore formation mechanisms because encapsulated fluorescein-conjugated dextran (FD)-20 was released more significantly than FD-70 or FD-250 from LUVs due to its smaller molecular size. On the basis of these data, we proposed tBid-PA interactions in the lysosomal membranes form lipidic pores and result in LMP. We further noted that chymotrypsin-cleaved Bid is more potent than tBid at binding to PA, inserting into the lipid bilayer, and promoting LMP. This amplification mechanism likely contributes to the culmination of apoptotic signaling.


Subject(s)
Apoptosis , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , Lysosomes/metabolism , Phosphatidic Acids/metabolism , Animals , BH3 Interacting Domain Death Agonist Protein/metabolism , Humans , Intracellular Membranes/metabolism , Mice , Mitochondria/metabolism , Permeability , Rats , bcl-2-Associated X Protein/metabolism
17.
Glia ; 59(3): 397-412, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21264947

ABSTRACT

Apoptosis is a controlled cell-death process mediated inter alia by proteins of the Bcl-2 family. Some proteins previously shown to promote the apoptotic process were found to have nonapoptotic functions as well. Microglia, the resident immune cells of the central nervous system, respond to brain derangements by becoming activated to contend with the brain damage. Activated microglia can also undergo activation-induced cell death. Previous studies have addressed the role of core apoptotic proteins in the death process, but whether these proteins also play a role or not in the activation process is not been reported. Here we explore the effect of the BH3-only protein Bid on the immunological features of microglia and macrophages. Our results showed that Bid regulates both the phagocytotic activities and the inflammatory profiles of these cells. Deficiency of Bid attenuated the phagocytotic activity of primary microglia and peritoneal macrophages. It also changed the expression profile of distinct inflammation-related genes in lipopolysaccharide-activated microglia and peritoneal macrophages in vitro and in an in vivo sepsis-like paradigm. Notably, similar changes followed downregulation of Bid in the N9 microglial cell line. Cell death could not be detected in any of the systems examined. Our findings demonstrate that Bid can regulate the immunological profiles of activated microglial and macrophages, via a novel nonapoptotic activity. In view of the critical role of these cells in various pathologies, including acute and chronic brain insults, our findings suggest that impairments in Bid expression may contribute to these pathologies also via a nonapoptotic activity.


Subject(s)
BH3 Interacting Domain Death Agonist Protein/physiology , Macrophages/immunology , Microglia/immunology , Nerve Degeneration/immunology , Animals , Apoptosis/immunology , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , BH3 Interacting Domain Death Agonist Protein/genetics , Cell Line , Cells, Cultured , Female , HEK293 Cells , Humans , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Lipopolysaccharides/toxicity , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/pathology , Nerve Degeneration/genetics , Nerve Degeneration/pathology , Phagocytosis/immunology , Signal Transduction/immunology
18.
Int J Cancer ; 127(2): 462-73, 2010 Jul 15.
Article in English | MEDLINE | ID: mdl-19908232

ABSTRACT

NF-kappaB activation is known to reduce the efficiency of chemotherapy in cancer treatment. Ursolic acid, a minimally toxic compound, has shown the capability to inhibit NF-kappaB activation in living cells. Here, for the first time, we investigated the effects and mechanisms of NF-kappaB inhibition by ursolic acid on chemotherapy treatment (Taxol or cisplatin) of cancer. ASTC-a-1 (human lung adenocarcinoma), Hela (human cervical cancer) cells, primary normal mouse cells of lung and liver and mouse in vivo model were used. Activity of signal factors (NF-kappaB, Akt, Fas/FasL, BID, Bcl-2, cytochrome c and caspase-8, 3) was used to analyze the mechanisms of ursolic acid-chemo treatment. Ursolic acid-mediated suppression of NF-kappaB drastically reduced the required dosage of the chemotherapeutic agents to achieve identical biological endpoints and enhanced the chemotherapeutic agent-induced cancer cells apoptosis. Chemosensitization by ursolic acid in cancer cells was dependent on the amplified activation of intrinsic pathway (caspase-8-BID-mitochondria-cytochrome c-caspase-3) by augmentation of BID cleavage and activation of Fas/FasL-caspase-8 pathway. Prolonged treatment with relatively low doses of ursolic acid also sensitized cancer cells to the chemotherapeutic agents through suppression of NF-kappaB. Chemosensitization by ursolic acid was observed only in cancer cells, but not in primary normal cells. The inhibitive effect of ursolic acid on NF-kappaB was reversible, and the reversal was not accompanied by a loss in cells viability. By supplementing chemotherapy with minimally toxic ursolic acid, it is possible to improve the efficacy of cancer treatment by significantly reducing the necessary drug dose without sacrificing the treatment results.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Lung Neoplasms/pathology , NF-kappa B/antagonists & inhibitors , Signal Transduction/drug effects , Uterine Cervical Neoplasms/pathology , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , BH3 Interacting Domain Death Agonist Protein/genetics , BH3 Interacting Domain Death Agonist Protein/metabolism , Blotting, Western , Caspase 8/metabolism , Cells, Cultured , Cisplatin/administration & dosage , Cytochromes c/metabolism , Drug Synergism , Fas Ligand Protein/metabolism , Female , Flow Cytometry , Humans , Liver/cytology , Liver/drug effects , Liver/metabolism , Luciferases/metabolism , Lung/cytology , Lung/drug effects , Lung/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Protein Transport , RNA, Small Interfering/pharmacology , Triterpenes/administration & dosage , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism , fas Receptor/metabolism , Ursolic Acid
19.
J Pharmacol Exp Ther ; 332(1): 316-25, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19812371

ABSTRACT

The topoisomerase I inhibitor topotecan (TPT) is used in the therapy of different tumors including high-grade gliomas. We previously showed that TPT-induced apoptosis depends on p53 with p53 wild-type (wt) cells being more resistant because of p53-controlled degradation of topoisomerase I. Here, we show that p53-deficient (p53(-/-)) fibroblasts undergo excessive mitochondrial apoptosis featuring H2AX phosphorylation, Bcl-x(L) decline, cytochrome c release, caspase-9/-3/-2 activation, and cleavage of Bid. In wt and apaf-1(-/-) cells, caspase-2 did not become activated and Bid was not cleaved. In addition, p53(-/-) cells cotreated with TPT and caspase-3 inhibitor showed neither caspase-2 activation nor Bid cleavage, implying that caspase-2 is processed downstream of the apoptosome by caspase-3. Although processing of caspase-9/-3 was similar in wt and p53(-/-) cells, only p53(-/-) cells displayed active caspase-3. This was due to the proteasomal degradation of X-chromosome-linked inhibitor of apoptosis (XIAP) and survivin that inhibits caspase-3 activity. Accordingly, TPT-induced apoptosis in wt cells was increased after XIAP/survivin knockdown. Silencing of Bid led to reduction of TPT-triggered apoptosis. Data obtained with mouse fibroblasts could be extended to human glioma cells. In U87MG (p53wt) cells cotreated with TPT and pifithrin-alpha, or transfected with p53-siRNA, caspase-2 and Bid were significantly cleaved and XIAP/survivin was degraded. Furthermore, the knockdown of XIAP and survivin led to increased TPT-triggered apoptosis. Overall, the data show that p53-deficient/depleted cells are hypersensitive to TPT because they down-regulate XIAP and survivin, and thus amplify the intrinsic apoptotic pathway via caspase-3-mediated Bid cleavage. Therefore, in gliomas harboring wild-type p53, TPT-based therapy might be improved by targeted down-regulation of XIAP and survivin.


Subject(s)
Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , Caspase 3/metabolism , Microtubule-Associated Proteins/antagonists & inhibitors , Topotecan/pharmacology , Tumor Suppressor Protein p53/deficiency , X-Linked Inhibitor of Apoptosis Protein/antagonists & inhibitors , Animals , Apoptotic Protease-Activating Factor 1/genetics , Blotting, Western , Caspase 2/metabolism , Cell Line, Tumor , Down-Regulation , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Flow Cytometry , Histones/metabolism , Humans , Inhibitor of Apoptosis Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , RNA, Small Interfering/genetics , Repressor Proteins , Survivin , Topoisomerase I Inhibitors , Transfection , Tumor Suppressor Protein p53/genetics
20.
Mol Pharmacol ; 76(5): 998-1010, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19713356

ABSTRACT

The Bcl-2 homology 3-only protein Bid is an important mediator of death receptor-induced apoptosis. Recent reports and this study suggest that Bid may also mediate genotoxic drug-induced apoptosis of various human cancer cells. Here, we characterized the role of Bid and the mechanism of Bid activation during oxaliplatin-induced apoptosis of HeLa cervical cancer cells. Small hairpin RNA-mediated silencing of Bid protected HeLa cells against both death receptor- and oxaliplatin-induced apoptosis. Expression of a Bid mutant in which caspase-8 cleavage site was mutated (D59A) reactivated oxaliplatin-induced apoptosis in Bid-deficient cells but failed to reactivate death receptor-induced apoptosis, suggesting that caspase-8-mediated Bid cleavage did not contribute to oxaliplatin-induced apoptosis. Overexpression of bcl-2 or treatment with the pan-caspase inhibitor N-benzyloxycarbonyl-Val-Ala-dl-Asp-fluoromethylketone abolished caspase-2, -8, -9, and -3 activation as well as Bid cleavage in response to oxaliplatin, suggesting that Bid cleavage occurred downstream of mitochondrial permeabilization and was predominantly mediated by caspases. We also detected an early activation of calpains in response to oxaliplatin. Calpain inhibition reduced Bid cleavage, mitochondrial depolarization, and activation of caspase-9, -3, -2, and -8 in response to oxaliplatin. Further experiments, however, suggested that Bid cleavage by calpains was not a prerequisite for oxaliplatin-induced apoptosis: single-cell imaging experiments using a yellow fluorescent protein-Bid-cyan fluorescent protein probe demonstrated translocation of full-length Bid to mitochondria that was insensitive to calpain or caspase inhibition. Moreover, calpain inhibition showed a potent protective effect in Bid-silenced cells. In conclusion, our data suggest that calpains and Bid act in a cooperative, but mutually independent, manner to mediate oxaliplatin-induced apoptosis of HeLa cells.


Subject(s)
Apoptosis/drug effects , BH3 Interacting Domain Death Agonist Protein/physiology , Calpain/physiology , Organoplatinum Compounds/pharmacology , Uterine Cervical Neoplasms/metabolism , Apoptosis/physiology , BH3 Interacting Domain Death Agonist Protein/antagonists & inhibitors , Calpain/antagonists & inhibitors , Female , Gene Knockdown Techniques/methods , HCT116 Cells , HeLa Cells , Humans , Organoplatinum Compounds/therapeutic use , Oxaliplatin , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL