Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cancer Res ; 82(16): 2848-2859, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35731919

ABSTRACT

African-American (AA) men are more likely to be diagnosed with and die from prostate cancer than European American (EA) men. Despite the central role of the androgen receptor (AR) transcription factor in prostate cancer, little is known about the contribution of epigenetics to observed racial disparities. We performed AR chromatin immunoprecipitation sequencing on primary prostate tumors from AA and EA men, finding that sites with greater AR binding intensity in AA relative to EA prostate cancer are enriched for lipid metabolism and immune response genes. Integration with transcriptomic and metabolomic data demonstrated coinciding upregulation of lipid metabolism gene expression and increased lipid levels in AA prostate cancer. In a metastatic prostate cancer cohort, upregulated lipid metabolism associated with poor prognosis. These findings offer the first insights into ancestry-specific differences in the prostate cancer AR cistrome. The data suggest a model whereby increased androgen signaling may contribute to higher levels of lipid metabolism, immune response, and cytokine signaling in AA prostate tumors. Given the association of upregulated lipogenesis with prostate cancer progression, our study provides a plausible biological explanation for the higher incidence and aggressiveness of prostate cancer observed in AA men. SIGNIFICANCE: With immunotherapies and inhibitors of metabolic enzymes in clinical development, the altered lipid metabolism and immune response in African-American men provides potential therapeutic opportunities to attenuate racial disparities in prostate cancer.


Subject(s)
Prostatic Neoplasms , Receptors, Androgen , Black or African American/genetics , Humans , Immunity , Lipid Metabolism/genetics , Male , Prostatic Neoplasms/pathology , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Up-Regulation
2.
Endocr Relat Cancer ; 29(1): 15-31, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34636746

ABSTRACT

Castration-resistant prostate cancer (CRPC) remains highly lethal and in need of novel, actionable therapeutic targets. The pioneer factor GATA2 is a significant prostate cancer (PC) driver and is linked to poor prognosis. GATA2 directly promotes androgen receptor (AR) gene expression (both full-length and splice-variant) and facilitates AR binding to chromatin, recruitment of coregulators, and target gene transcription. Unfortunately, there is no clinically applicable GATA2 inhibitor available at the moment. Using a bioinformatics algorithm, we screened in silico 2650 clinically relevant drugs for a potential GATA2 inhibitor. Validation studies used cytotoxicity and proliferation assays, global gene expression analysis, RT-qPCR, reporter assay, reverse phase protein array analysis (RPPA), and immunoblotting. We examined target engagement via cellular thermal shift assay (CETSA), ChIP-qPCR, and GATA2 DNA-binding assay. We identified the vasodilator dilazep as a potential GATA2 inhibitor and confirmed on-target activity via CETSA. Dilazep exerted anticancer activity across a broad panel of GATA2-dependent PC cell lines in vitro and in a PDX model in vivo. Dilazep inhibited GATA2 recruitment to chromatin and suppressed the cell-cycle program, transcriptional programs driven by GATA2, AR, and c-MYC, and the expression of several oncogenic drivers, including AR, c-MYC, FOXM1, CENPF, EZH2, UBE2C, and RRM2, as well as of several mediators of metastasis, DNA damage repair, and stemness. In conclusion, we provide, via an extensive compendium of methodologies, proof-of-principle that a small molecule can inhibit GATA2 function and suppress its downstream AR, c-MYC, and other PC-driving effectors. We propose GATA2 as a therapeutic target in CRPC.


Subject(s)
Prostatic Neoplasms, Castration-Resistant , Cell Line, Tumor , Chromatin , Dilazep/therapeutic use , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Gene Expression Regulation, Neoplastic , Humans , Male , Oncogenes , Prostatic Neoplasms, Castration-Resistant/genetics , Receptors, Androgen/metabolism
3.
Food Funct ; 12(22): 11399-11407, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34673869

ABSTRACT

To maximize the biological activity of branched-chain amino acids (BCAAs), it is necessary to find a new excipient agent to increase the bioavailability of BCAAs in protein mixtures. The aim of the current study was to investigate the effects of soy lecithin (SLC), zinc oxide (ZnO), and methylsulfonylmethane (MSM) on the bioaccessibility and intestinal transport of BCAAs from animal and plant protein mixtures (PMs) via an in vitro digestion model with human intestinal epithelial (Caco-2) cells. The bioaccessibility of total BCAAs in PMs considerably increased by 107.51 ± 1.50% with the addition of SLC, and the combined effects of SLC, ZnO, and MSM on enhancing the bioaccessibility of total BCAAs was observed (107.14 ± 0.18%). Interestingly, SLC showed a major role in binding bile acid, showing 65.78 ± 1.66% of binding capacity. Intestinal transport of BCAAs was measured to be at 100.48, 110.86, and 130.29 µg mL-1 for leucine, isoleucine, and valine, respectively, in PMs with SLC + ZnO + MSM, and it eventually amplified the amount of the total transported BCAAs (341.63 ± 6.34 µg mL-1), which was about 8.72 times higher than that of PM only. The cellular integrity of digesta-treated Caco-2 cells tended to decrease according to the incubation time, but it was recovered in the treatment of PM + SLC + ZnO + MSM, and nearly reached the control levels with 92.82 ± 0.53%. Results from the current study suggest that the co-consumption of proteins equally consisting of plant and animal sources with SLC, ZnO, and MSM could improve the bioavailability of total BCAAs, resulting in the improvement of health benefits.


Subject(s)
Amino Acids, Branched-Chain , Dimethyl Sulfoxide/chemistry , Excipients/chemistry , Plant Proteins , Sulfones/chemistry , Zinc Oxide/chemistry , Amino Acids, Branched-Chain/chemistry , Amino Acids, Branched-Chain/pharmacokinetics , Animals , Biological Availability , Caco-2 Cells , Humans , Lecithins/chemistry , Plant Proteins/chemistry , Plant Proteins/pharmacokinetics
4.
J Immunol ; 207(2): 661-670, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34193605

ABSTRACT

Inflammation contributes to the pathogenesis and morbidity of wide spectrum of human diseases. The inflammatory response must be actively controlled to prevent bystander damage to tissues. Yet, the mechanisms controlling excessive inflammatory responses are poorly understood. NLRP3 inflammasome plays an important role in innate immune response to cellular infection or stress. Its activation must be tightly regulated because uncontrolled inflammasome activation is associated with a number of human diseases. p38 MAPK signaling plays an essential role in the regulation of inflammation. The role of p38 MAPK in inflammatory response associated with the expression of proinflammatory molecules is known. However, the anti-inflammatory functions of p38 MAPK are largely unknown. In this study, we show that pharmacologic inhibition or genetic deficiency of p38 MAPK leads to hyperactivation of NLRP3 inflammasome, resulting in enhanced Caspase 1 activation and IL-1ß and IL-18 production. The deficiency of p38 MAPK activity induced an increase of cytosolic Ca2+ and excessive mitochondrial Ca2+ uptake, leading to exacerbation of mitochondrial damage, which was associated with hyperactivation of NLRP3 inflammasome. In addition, mice with deficiency of p38 MAPK in granulocytes had evidence of in vivo hyperactivation of NLRP3 inflammasome and were more susceptible to LPS-induced sepsis compared with wild-type mice. Our results suggest that p38 MAPK negatively regulates NLRP3 inflammasome through control of Ca2+ mobilization. Hyperactivity of inflammasome in p38-deficient mice causes lung inflammation and increased susceptibility to septic shock.


Subject(s)
Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , HEK293 Cells , Humans , Immunity, Innate/physiology , Inflammation/metabolism , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Sepsis/metabolism , Shock, Septic/metabolism , Signal Transduction/physiology
5.
Int J Mol Sci ; 21(12)2020 Jun 17.
Article in English | MEDLINE | ID: mdl-32560429

ABSTRACT

Alpha one antitrypsin (α1AT), a serine proteinase inhibitor primarily produced by the liver, protects pulmonary tissue from neutrophil elastase digestion. Mutations of the SERPINA1 gene results in a misfolded α1AT protein which aggregates inside hepatocytes causing cellular damage. Therefore, inhibition of mutant α1AT production is one practical strategy to alleviate liver damage. Here we show that proteasome inhibitors can selectively downregulate α1AT expression in human hepatocytes by suppressing the translation of α1AT. Translational suppression of α1AT is mediated by phosphorylation of eukaryotic translation initiation factor 2α and increased association of RNA binding proteins, especially stress granule protein Ras GAP SH3 binding protein (G3BP1), with α1AT mRNA. Treatment of human-induced pluripotent stem cell-derived hepatocytes with a proteasome inhibitor also results in translational inhibition of mutant α1AT in a similar manner. Together we revealed a previously undocumented role of proteasome inhibitors in the regulation of α1AT translation.


Subject(s)
Gene Expression Regulation/drug effects , Proteasome Inhibitors/pharmacology , RNA Processing, Post-Transcriptional/drug effects , alpha 1-Antitrypsin/genetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Protein Biosynthesis/drug effects , Stress, Physiological , alpha 1-Antitrypsin/biosynthesis
6.
Cell Rep ; 12(11): 1731-9, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26344765

ABSTRACT

Autophagy, an intracellular degradation and energy recycling mechanism, is emerging as an important regulator of immune responses. However, the role of autophagy in regulating neutrophil functions is not known. We investigated neutrophil biology using myeloid-specific autophagy-deficient mice and found that autophagy deficiency reduced neutrophil degranulation in vitro and in vivo. Mice with autophagy deficiency showed reduced severity of several neutrophil-mediated inflammatory and autoimmune disease models, including PMA-induced ear inflammation, LPS-induced breakdown of blood-brain barrier, and experimental autoimmune encephalomyelitis. NADPH oxidase-mediated reactive oxygen species generation was also reduced in autophagy-deficient neutrophils, and inhibition of NADPH oxidase reduced neutrophil degranulation, suggesting NADPH oxidase to be a player at the intersection of autophagy and degranulation. Overall, this study establishes autophagy as an important regulator of neutrophil functions and neutrophil-mediated inflammation in vivo.


Subject(s)
Autophagy/physiology , Inflammation/metabolism , Inflammation/pathology , Neutrophils/metabolism , Neutrophils/pathology , Animals , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism
7.
J Biol Chem ; 288(50): 35886-95, 2013 Dec 13.
Article in English | MEDLINE | ID: mdl-24178293

ABSTRACT

The association between altered proteostasis and inflammatory disorders has been increasingly recognized, but the underlying mechanisms are not well understood. In this study, we show that deficiency of either autophagy or sequestosome 1 (p62 or SQSTM) led to inflammasome hyperactivation in response to LPS and ATP in primary macrophages and in mice in vivo. Importantly, induction of protein misfolding by puromycin, thapsigargin, or geldanamycin resulted in inflammasome activation that was more pronounced in autophagy- or p62-deficient macrophages. Accumulation of misfolded proteins caused inflammasome activation by inducing generation of nonmitochondrial reactive oxygen species and lysosomal damage, leading to release of cathepsin B. Our results suggest that altered proteostasis results in inflammasome activation and thus provide mechanisms for the association of altered proteostasis with inflammatory disorders.


Subject(s)
Homeostasis , Inflammasomes/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Animals , Autophagy , Carrier Proteins/metabolism , Cathepsin B/metabolism , Cell Line , Heat-Shock Proteins/deficiency , Humans , Lysosomes/metabolism , Macrophages/cytology , Macrophages/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Folding , Reactive Oxygen Species/metabolism , Sequestosome-1 Protein
8.
Biochem Biophys Res Commun ; 435(2): 234-8, 2013 May 31.
Article in English | MEDLINE | ID: mdl-23665015

ABSTRACT

MUDENG, also known as AP5M1, was originally identified as an adaptin domain-containing gene that induced cell death in lymphoma cell lines. However, little is known of the mechanism responsible for MUDENG-mediated cell death. In this study, we investigated MUDENG changes during TRAIL-induced cell death. We found that MUDENG is rapidly processed in response to TRAIL in Jurkat and BJAB cells with time line similar to that of caspase activation. Caspase-3-mediated MUDENG cleavage was confirmed by an in vitro cleavage assay using recombinant active caspase proteins. Caspase cleavage sites (D276 and D290) were located in the adaptin domain of MUDENG, and cleaved MUDENG showed the reduced killing activity. These results suggest that the adaptin domain plays a key role in MUDENG-mediated cell death.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis/drug effects , Caspase 3/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Enzyme Activation , HeLa Cells , Humans , Jurkat Cells , Neoplasms, Experimental , Protein Binding
10.
BMB Rep ; 42(8): 511-5, 2009 Aug 31.
Article in English | MEDLINE | ID: mdl-19712588

ABSTRACT

Downstream of Bid (DOBI) known as Pus10, has been identified as a modulator of TRAIL-induced cell death using RNAi library screening. The crystal structure of DOBI has revealed that it is a crescent-shaped protein containing the pseudouridine synthase catalytic domain and a THUMP-containing domain. Here, we demonstrated that DOBI is expressed in various tissues such as heart and lung, and is also expressed in various tumor cells such as HeLa and A549. Although ectopic expression of DOBI does not promote TRAIL death signaling in HeLa cells, knock-down of DOBI expression using shRNA inhibited TRAIL death signaling. DOBI is cleaved into a 54 kD cleaved DOBI during cell death, and the recombinant DOBI protein can be directly cleaved by caspases-3, or -8 in vitro. Together, these data suggest that the cleaved DOBI may acquire a new function, possibly by cooperating with tBid in the mitochondrial event of cell death caused by TRAIL.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Hydro-Lyases/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Animals , HeLa Cells , Humans , Mice , Mitochondria, Liver/drug effects , Mitochondria, Liver/metabolism , Models, Biological , Recombinant Proteins/pharmacology , Transfection
11.
J Gynecol Oncol ; 20(2): 113-6, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19590723

ABSTRACT

OBJECTIVE: Platinum (Pt) based drugs including cisplatin and carboplatin are widely used as anticancer drugs in various human cancers. Many studies have shown that chemotherapeutic agents synergistically enhance cell death induced by death ligands. However it has been recently reported that cisplatin may inhibit tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced cell death through inactivation of caspases. Thus, we investigated whether carboplatin also inhibits TRAIL-induced cell death. METHODS: HeLa cells were treated with TRAIL in the presence of cisplatin or carboplatin, and cell death was analyzed using the crystal violet staining method. Caspase activation was checked through detection of Bid cleavage by Western blotting using anti-Bid antibody. RESULTS: Cisplatin inhibits TRAIL-induced cell death in HeLa cells; however, carboplatin enhanced TRAIL-induced cell death. Whereas cisplatin inhibited caspase-8-mediated Bid cleavage, carboplatin had no effect on caspase-8 activity. CONCLUSION: Although cisplatin and carboplatin are platinum-containing cancer therapeutic agents, they have the opposite effects on TRAIL-induced cell death.

12.
Exp Cell Res ; 315(13): 2249-55, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19376111

ABSTRACT

TRAIL is an apoptotic cell death-inducing ligand that belongs to a TNF superfamily. To identify the regulators that govern the susceptibility to TRAIL, TRAIL-resistant HeLa (TR) cells were established by repeatedly treating HeLa cells with TRAIL. Here we showed that scaffolding protein Homer1 plays a decisive role in regulating the apoptotic susceptibility to TRAIL. TR cells showing the normal susceptibility to FasL and chemotherapeutic agent etoposide expressed the lower protein levels of Homer1 than parental HeLa cells. They showed the delayed activation of caspases-8, Bid cleavage and Bax translocation to mitochondria in response to TRAIL. Reconstitution of Homer1 expression in TR cells significantly restored the susceptibility to TRAIL. In addition, knock-down of Homer1 using interfering shRNA in parental HeLa cells lost the susceptibility to TRAIL. Together, our data indicate that Homer1 plays a critical role in determining the apoptotic susceptibility to TRAIL.


Subject(s)
Apoptosis/physiology , Carrier Proteins/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Carrier Proteins/genetics , Gene Expression Profiling , HeLa Cells , Homer Scaffolding Proteins , Humans , Mitochondria/metabolism , Oligonucleotide Array Sequence Analysis , RNA Interference , TNF-Related Apoptosis-Inducing Ligand/genetics , bcl-2-Associated X Protein/metabolism
13.
Biochem Biophys Res Commun ; 370(3): 504-8, 2008 Jun 06.
Article in English | MEDLINE | ID: mdl-18395520

ABSTRACT

A screening system comprised of a randomized hybrid-ribozyme library has previously been used to identify pro-death genes in Fas-mediated apoptosis, and short sequence information of candidate genes from this system was previously reported by Kawasaki and Taira [H. Kawasaki, K. Taira, A functional gene discovery in the Fas-mediated pathway to apoptosis by analysis of transiently expressed randomized hybrid-ribozyme libraries, Nucleic Acids Res. 30 (2002) 3609-3614]. In this study, we have cloned the full-length of the candidate's open reading frames and found that one of the candidates, referred to as MUDENG (Mu-2 related death-inducing gene), which is composed of 490 amino acids that contain the adaptin domain found in the mu2 subunit of APs related to clathrin-mediated endocytosis, is able to induce cell death by itself. Ectopic expression of MUDENG induced cell death in Jurkat T cells and HeLa cells. In addition, when MUDENG expression was evaluated by immnuohistochemical staining, it was found in most tissues, including the intestine and testis. Furthermore, MUDENG appears to be evolutionary conserved from mammals to amphibians, suggesting that it may have a common role in cell death. Taken together, these results suggest that MUDENG is likely to play an important role in cell death in various tissues.


Subject(s)
Apoptosis , T-Lymphocytes, Cytotoxic/immunology , Amino Acid Sequence , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/analysis , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cattle , Cloning, Molecular , Dogs , Evolution, Molecular , HeLa Cells , Humans , Jurkat Cells , Mice , Molecular Sequence Data , Rats
14.
Exp Cell Res ; 312(19): 3892-8, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-16996498

ABSTRACT

TRAIL has been suggested to induce the cell death in various tumor cells but not in normal cells; however, several studies have provided the evidence that TRAIL can induce the cell death in some normal cells including human normal hepatocytes, suggesting that TRAIL may show hepatic toxicity in human. In this study, we designed a pro-form of TRAIL (sTRAIL:IL-18) in that soluble TRAIL (sTRAIL) is fused to IL-18, and a matrix metalloproteinases (MMPs) cleavage site is introduced at the connecting site. We showed that sTRAIL:IL-18 has significantly diminished the killing activity in HeLa cells but regains the activity by releasing the free sTRAIL through MMP-2-mediated cleavage. In addition, the killing activity of sTRAIL:IL-18 was significantly increased in HeLa cells when active MMP-2 was produced by TNF-alpha. Taken together, the data suggested that the sTRAIL:IL-18 can be reactivated at the specialized areas where MMPs are pathologically produced.


Subject(s)
Matrix Metalloproteinases/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Apoptosis , Base Sequence , DNA, Complementary/genetics , HeLa Cells , Humans , Interleukin-18/genetics , Interleukin-18/metabolism , Matrix Metalloproteinase 2/metabolism , Protein Precursors/genetics , Protein Precursors/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , TNF-Related Apoptosis-Inducing Ligand/genetics , Tumor Necrosis Factor-alpha/pharmacology
15.
J Biol Chem ; 280(11): 10509-15, 2005 Mar 18.
Article in English | MEDLINE | ID: mdl-15634686

ABSTRACT

Cisplatin is a platinum-containing chemotherapeutic drug that has been widely used to treat various human cancers. It acts by forming inter- and intracross-links of DNA, which is believed to be a major cause for its therapeutic efficacy. However, little attention has been paid to the effect of cisplatin on death ligand-induced cell death. Here we demonstrate that cisplatin inhibits death ligand-induced cell death in cell lines in a p53-independent manner. This inhibitory effect of cisplatin on cell death is direct, whereby cisplatin forms a complex with caspases leading to their inactivation. The cisplatin-caspase complex is reversed by the addition of reducing agent dithiothreitol, and caspase activity is regained. In addition, cisplatin shows a death-inhibition effect in in vivo animal models of fulminant liver damage induced by Fas activation and lipopolysaccharide-induced liver shock mediated by tumor necrosis factor-alpha. Together, we demonstrate that cisplatin inhibits cell death induced by death ligands in cell lines and in mice through caspase inactivation.


Subject(s)
Antineoplastic Agents/pharmacology , Caspase Inhibitors , Cisplatin/pharmacology , Enzyme Inhibitors/pharmacology , Liver/injuries , Animals , Apoptosis Regulatory Proteins , BH3 Interacting Domain Death Agonist Protein , Blotting, Western , Carrier Proteins/metabolism , Caspase 3 , Caspase 8 , Caspases/chemistry , Caspases/metabolism , Cell Death , Cell Line, Tumor , Cisplatin/metabolism , Dithiothreitol/pharmacology , Enzyme Activation , HeLa Cells , Humans , In Vitro Techniques , Ligands , Lipopolysaccharides/metabolism , Liver/pathology , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred BALB C , Recombinant Proteins/chemistry , TNF-Related Apoptosis-Inducing Ligand , Time Factors , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/metabolism
16.
Mol Biol Cell ; 15(7): 3061-72, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15107464

ABSTRACT

Release of cytochrome c from the mitochondrial intermembrane space is critical to apoptosis induced by a variety of death stimuli. Bid is a BH3-only prodeath Bcl-2 family protein that can potently activate this efflux. In the current study, we investigated the mitochondrial localization of Bid and its interactions with mitochondrial phospholipids, focusing on their relationships with Bid-induced cytochrome c release. We found that Bid binding to the mitochondria required only three of its eight helical structures (alpha4-alpha6), but not the BH3 domain, and the binding could not be inhibited by the antideath molecule Bcl-x(L). Membrane fractionations indicated that tBid bound to mitochondrial outer membranes at both contact and noncontact sites. Bid could interact with specific cardiolipin species on intact mitochondria as identified by mass spectrometry. Like the binding to the mitochondria, this interaction could not be blocked by the mutation in the BH3 domain or by Bcl-x(L.) However, a cardiolipin-specific dye, 10-N-nonyl acridine orange, could preferentially suppress Bid binding to the mitochondrial contact site and inhibit Bid-induced mitochondrial cristae reorganization and cytochrome c release. These findings thus suggest that interactions of Bid with mitochondrial cardiolipin at the contact site can contribute significantly to its functions.


Subject(s)
Cardiolipins/metabolism , Carrier Proteins/metabolism , Cytochromes c/metabolism , Mitochondria/metabolism , Mitochondria/ultrastructure , Aminoacridines/pharmacology , Animals , BH3 Interacting Domain Death Agonist Protein , Cardiolipins/analysis , Carrier Proteins/analysis , Carrier Proteins/genetics , Membrane Proteins/analysis , Membrane Proteins/metabolism , Mice , Mitochondria/immunology , Protein Structure, Tertiary/genetics , Protein Transport/drug effects , Proto-Oncogene Proteins c-bcl-2/analysis , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2 Homologous Antagonist-Killer Protein , bcl-X Protein
17.
J Biol Chem ; 278(48): 48292-9, 2003 Nov 28.
Article in English | MEDLINE | ID: mdl-14500711

ABSTRACT

Genotoxic stresses stabilize the p53 tumor suppressor protein which, in turn, transactivates target genes to cause apoptosis. Although Noxa, a "BH3-only" member of the Bcl-2 family, was shown to be a target of p53-mediated transactivation and to function as a mediator of p53-dependent apoptosis through mitochondrial dysfunction, the molecular mechanism by which Noxa causes mitochondrial dysfunction is largely unknown. Here we show that two domains (BH3 domain and mitochondrial targeting domain) in Noxa are essential for the release of cytochrome c from mitochondria. Noxa-induced cytochrome c release is inhibited by permeability transition pore inhibitors such as CsA or MgCl2, and Noxa induces an ultra-structural change of mitochondria yielding "swollen" mitochondria that are unlike changes induced by tBid. This indicates that Noxa may activate the permeability transition-related pore to release cytochrome c from mitochondria into cytosol. Moreover, Bak-oligomerization, which is an essential event for tBid-induced cytochrome c release in the extrinsic death signaling pathway, is not associated with Noxa-induced cytochrome c release. This finding suggests that the pathway of Noxa-induced mitochondrial dysfunction is distinct from the one of tBid-induced mitochondrial dysfunction. Thus, we propose that there are at least two different pathways of mitochondrial dysfunction; one mediated through Noxa in response to genotoxic stresses and the other through tBid in response to death ligands.


Subject(s)
Apoptosis , Mitochondria/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , BH3 Interacting Domain Death Agonist Protein , Blotting, Western , Carrier Proteins/metabolism , Cytochromes c/metabolism , Cytosol/metabolism , Endodeoxyribonucleases/metabolism , Green Fluorescent Proteins , HeLa Cells , Humans , Ligands , Luminescent Proteins/metabolism , Magnesium Chloride/chemistry , Membrane Proteins/metabolism , Microscopy, Fluorescence , Mitochondria/ultrastructure , Molecular Sequence Data , Plasmids/metabolism , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Signal Transduction , Transcriptional Activation , Tumor Necrosis Factor-alpha/metabolism , bcl-2 Homologous Antagonist-Killer Protein
18.
J Biol Chem ; 277(10): 8346-53, 2002 Mar 08.
Article in English | MEDLINE | ID: mdl-11751921

ABSTRACT

A variety of surface receptors eliciting diverse cellular responses have been shown to recruit tumor necrosis factor receptor-associated factor (TRAF) adaptor molecules. However, a few TRAF-interacting intracellular proteins that serve as downstream targets or regulators of TRAF function have been identified. In search of new intracellular molecules that bind TRAF6, we carried out a yeast two-hybrid cDNA library screening with an N-terminal segment of TRAF6 as the bait. A novel human C(2)H(2)-type zinc finger family protein was identified, which when coexpressed with TRAF6 led to a suppression of TRAF6-induced activation of NF-kappa B and c-Jun N-terminal kinase. This novel protein was designated TIZ (for TRAF6-inhibitory zinc finger protein). TIZ expression also inhibited the signaling of RANK (receptor activator of NF-kappa B), which together with TRAF6 has been shown to be essential for osteoclastogenesis. Furthermore, the expression level of TIZ appeared to be regulated during the differentiation of human peripheral blood monocytes into osteoclasts. More significantly, transfection of TIZ into the monocyte/macrophage cell line Raw264.7 reduced the RANK ligand-induced osteoclastogenesis of this cell line. Our findings suggest that the novel zinc finger protein TIZ may play a role during osteoclast differentiation by modulating TRAF6 signaling activity.


Subject(s)
Carrier Proteins/biosynthesis , Carrier Proteins/chemistry , Osteoclasts/cytology , Osteoclasts/metabolism , Proteins/physiology , Transcription Factors , Zinc Fingers , Amino Acid Sequence , Animals , Carrier Proteins/metabolism , Cell Differentiation , Cell Line , Cloning, Molecular , DNA, Complementary/metabolism , Enzyme Activation , Gene Library , Glutathione Transferase/metabolism , Humans , JNK Mitogen-Activated Protein Kinases , Luciferases/metabolism , Macrophages/metabolism , Membrane Glycoproteins/metabolism , Mice , Microscopy, Confocal , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , NF-kappa B/metabolism , Protein Binding , Protein Structure, Tertiary , Proteins/metabolism , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Recombinant Fusion Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , TNF Receptor-Associated Factor 6 , Time Factors , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...