Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
ACS Nano ; 16(3): 4084-4101, 2022 03 22.
Article in English | MEDLINE | ID: mdl-35230826

ABSTRACT

The complex involvement of neutrophils in inflammatory diseases makes them intriguing but challenging targets for therapeutic intervention. Here, we tested the hypothesis that varying endocytosis capacities would delineate functionally distinct neutrophil subpopulations that could be specifically targeted for therapeutic purposes. By using uniformly sized (∼120 nm in diameter) albumin nanoparticles (ANP) to characterize mouse neutrophils in vivo, we found two subsets of neutrophils, one that readily endocytosed ANP (ANPhigh neutrophils) and another that failed to endocytose ANP (ANPlow population). These ANPhigh and ANPlow subsets existed side by side simultaneously in bone marrow, peripheral blood, spleen, and lungs, both under basal conditions and after inflammatory challenge. Human peripheral blood neutrophils showed a similar duality. ANPhigh and ANPlow neutrophils had distinct cell surface marker expression and transcriptomic profiles, both in naive mice and in mice after endotoxemic challenge. ANPhigh and ANPlow neutrophils were functionally distinct in their capacities to kill bacteria and to produce inflammatory mediators. ANPhigh neutrophils produced inordinate amounts of reactive oxygen species and inflammatory chemokines and cytokines. Targeting this subset with ANP loaded with the drug piceatannol, a spleen tyrosine kinase (Syk) inhibitor, mitigated the effects of polymicrobial sepsis by reducing tissue inflammation while fully preserving neutrophilic host-defense function.


Subject(s)
Nanoparticles , Neutrophils , Albumins/metabolism , Animals , Endocytosis , Inflammation/drug therapy , Inflammation/metabolism , Mice , Neutrophils/metabolism
2.
BMC Genomics ; 18(1): 664, 2017 Aug 29.
Article in English | MEDLINE | ID: mdl-28851267

ABSTRACT

BACKGROUND: Sphingosine- 1-Phosphate (S1P) is a bioactive lipid and an intracellular as well as an extracellular signaling molecule. S1P ligand specifically binds to five related cell surface G-protein-coupled receptors (S1P1-5). S1P levels are tightly regulated by its synthesis catalyzed by sphingosine kinases (SphKs) 1 & 2 and catabolism by S1P phosphatases, lipid phosphate phosphatases and S1P lyase. We previously reported that knock down of SphK1 (Sphk1 -/- ) in a neonatal mouse BPD model conferred significant protection against hyperoxia induced lung injury. To better understand the underlying molecular mechanisms, genome-wide gene expression profiling was performed on mouse lung tissue using Affymetrix MoGene 2.0 array. RESULTS: Two-way ANOVA analysis was performed and differentially expressed genes under hyperoxia were identified using Sphk1 -/- mice and their wild type (WT) equivalents. Pathway (PW) enrichment analyses identified several signaling pathways that are likely to play a key role in hyperoxia induced lung injury in the neonates. These included signaling pathways that were anticipated such as those involved in lipid signaling, cell cycle regulation, DNA damage/apoptosis, inflammation/immune response, and cell adhesion/extracellular matrix (ECM) remodeling. We noted hyperoxia induced downregulation of the expression of genes related to mitotic spindle formation in the WT which was not observed in Sphk1 -/- neonates. Our data clearly suggests a role for SphK1 in neonatal hyperoxic lung injury through elevated inflammation and apoptosis in lung tissue. Further, validation by RT-PCR on 24 differentially expressed genes showed 83% concordance both in terms of fold change and vectorial changes. Our findings are in agreement with previously reported human BPD microarray data and completely support our published in vivo findings. In addition, the data also revealed a significant role for additional unanticipitated signaling pathways involving Wnt and GADD45. CONCLUSION: Using SphK1 knockout mice and differential gene expression analysis, we have shown here that S1P/SphK1 signaling plays a key role in promoting hyperoxia induced DNA damage, inflammation, apoptosis and ECM remodeling in neonatal lungs. It also appears to suppress pro-survival cellular responses involved in normal lung development. We therefore propose SphK1 as a therapeutic target for the development drugs to combat BPD.


Subject(s)
Bronchopulmonary Dysplasia/complications , Gene Expression Profiling , Hyperoxia/etiology , Hyperoxia/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Animals , Animals, Newborn , Apoptosis/genetics , Bronchopulmonary Dysplasia/drug therapy , Cell Cycle/genetics , Disease Models, Animal , Gene Deletion , Humans , Hyperoxia/pathology , Lysophospholipids/metabolism , Mice , Molecular Targeted Therapy , Oligonucleotide Array Sequence Analysis , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Transcription, Genetic
3.
PLoS One ; 11(7): e0158865, 2016.
Article in English | MEDLINE | ID: mdl-27442243

ABSTRACT

Protein Kinase C (PKC) plays a significant role in thrombin-induced loss of endothelial cell (EC) barrier integrity; however, the existence of more than 10 isozymes of PKC and tissue-specific isoform expression has limited our understanding of this important second messenger in vascular homeostasis. In this study, we show that PKCδ isoform promotes thrombin-induced loss of human pulmonary artery EC barrier integrity, findings substantiated by PKCδ inhibitory studies (rottlerin), dominant negative PKCδ construct and PKCδ silencing (siRNA). In addition, we identified PKCδ as a signaling mediator upstream of both thrombin-induced MLC phosphorylation and Rho GTPase activation affecting stress fiber formation, cell contraction and loss of EC barrier integrity. Our inhibitor-based studies indicate that thrombin-induced PKCδ activation exerts a positive feedback on Rho GTPase activation and contributes to Rac1 GTPase inhibition. Moreover, PKD (or PKCµ) and CPI-17, two known PKCδ targets, were found to be activated by PKCδ in EC and served as modulators of cytoskeleton rearrangement. These studies clarify the role of PKCδ in EC cytoskeleton regulation, and highlight PKCδ as a therapeutic target in inflammatory lung disorders, characterized by the loss of barrier integrity, such as acute lung injury and sepsis.


Subject(s)
Endothelial Cells/pathology , Lung/enzymology , Lung/pathology , Protein Kinase C-delta/metabolism , Thrombin/pharmacology , Acetophenones/pharmacology , Benzopyrans/pharmacology , Enzyme Activation/drug effects , Gene Silencing/drug effects , Humans , Intracellular Signaling Peptides and Proteins , Models, Biological , Muscle Proteins , Myosin Light Chains/metabolism , Phosphoprotein Phosphatases , Phosphorylation/drug effects , Protein Kinase C , Protein Kinase C-delta/antagonists & inhibitors , Signal Transduction/drug effects , Stress Fibers/drug effects , Stress Fibers/metabolism , rho GTP-Binding Proteins/metabolism
4.
Am J Physiol Lung Cell Mol Physiol ; 311(2): L337-51, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27343196

ABSTRACT

Hyperoxia-induced lung injury adversely affects ICU patients and neonates on ventilator assisted breathing. The underlying culprit appears to be reactive oxygen species (ROS)-induced lung damage. The major contributor of hyperoxia-induced ROS is activation of the multiprotein enzyme complex NADPH oxidase. Sphingosine-1-phosphate (S1P) signaling is known to be involved in hyperoxia-mediated ROS generation; however, the mechanism(s) of S1P-induced NADPH oxidase activation is unclear. Here, we investigated various steps in the S1P signaling pathway mediating ROS production in response to hyperoxia in lung endothelium. Of the two closely related sphingosine kinases (SphKs)1 and 2, which synthesize S1P from sphingosine, only Sphk1(-/-) mice conferred protection against hyperoxia-induced lung injury. S1P is metabolized predominantly by S1P lyase and partial deletion of Sgpl1 (Sgpl1(+/-)) in mice accentuated lung injury. Hyperoxia stimulated S1P accumulation in human lung microvascular endothelial cells (HLMVECs), and downregulation of S1P transporter spinster homolog 2 (Spns2) or S1P receptors S1P1&2, but not S1P3, using specific siRNA attenuated hyperoxia-induced p47(phox) translocation to cell periphery and ROS generation in HLMVECs. These results suggest a role for Spns2 and S1P1&2 in hyperoxia-mediated ROS generation. In addition, p47(phox) (phox:phagocyte oxidase) activation and ROS generation was also reduced by PF543, a specific SphK1 inhibitor in HLMVECs. Our data indicate a novel role for Spns2 and S1P1&2 in the activation of p47(phox) and production of ROS involved in hyperoxia-mediated lung injury in neonatal and adult mice.


Subject(s)
Endothelial Cells/enzymology , Hyperoxia/enzymology , NADPH Oxidases/metabolism , Aldehyde-Lyases/metabolism , Animals , Anion Transport Proteins/metabolism , Cells, Cultured , Endothelium, Vascular/pathology , Enzyme Activation , Female , Humans , Lung/blood supply , Lysophospholipids/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microvessels/pathology , Protein Transport , Reactive Oxygen Species/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism
5.
J Cell Biochem ; 115(2): 261-70, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24038283

ABSTRACT

Mitogen-activated kinase activating death domain containing protein (MADD) is abundantly expressed in cancer cells and necessary for maintaining cancer cell survival. However, this survival function of MADD is dependent upon its phosphorylation by protein kinase B (Akt). The tumour suppressor PTEN (phosphatase and tensin homolog deleted on chromosome 10) is a lipid phosphatase that negatively regulates the phosphatidylinositol 3-kinase (PI3K)-Akt signaling pathway. The downstream targets of PTEN in triggering apoptosis have not yet been completely identified. Here, we report that MADD can act as a pro-apoptotic factor to initiate TRAIL-induced apoptosis when its phosphorylation is attenuated by PTEN. Our data show that tumor necrosis factor α-related apoptosis-inducing ligand (TRAIL) induced a reduction in MADD phosphorylation with a concomitant up-regulation of PTEN. Knock down of PTEN using a specific siRNA prevented TRAIL-induced reduction in pMADD levels. Surprisingly, Akt non-phosphorylated MADD translocated from the plasma membrane to cytoplasm where it bound to 14-3-3 and displaced 14-3-3 associated Bax, which translocated to mitochondria resulting in cytochrome c release. Taken together, our data reveal that PTEN can convey the death signal by preventing MADD phosphorylation by Akt.


Subject(s)
Apoptosis/genetics , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Guanine Nucleotide Exchange Factors/metabolism , PTEN Phosphohydrolase/metabolism , Tumor Necrosis Factor-alpha/metabolism , 14-3-3 Proteins/metabolism , Cell Line, Tumor , Death Domain Receptor Signaling Adaptor Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , Humans , Mitochondria/metabolism , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering , Tumor Necrosis Factor-alpha/genetics
6.
Am J Pathol ; 183(4): 1169-1182, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23933064

ABSTRACT

Bronchopulmonary dysplasia of the premature newborn is characterized by lung injury, resulting in alveolar simplification and reduced pulmonary function. Exposure of neonatal mice to hyperoxia enhanced sphingosine-1-phosphate (S1P) levels in lung tissues; however, the role of increased S1P in the pathobiological characteristics of bronchopulmonary dysplasia has not been investigated. We hypothesized that an altered S1P signaling axis, in part, is responsible for neonatal lung injury leading to bronchopulmonary dysplasia. To validate this hypothesis, newborn wild-type, sphingosine kinase1(-/-) (Sphk1(-/-)), sphingosine kinase 2(-/-) (Sphk2(-/-)), and S1P lyase(+/-) (Sgpl1(+/-)) mice were exposed to hyperoxia (75%) from postnatal day 1 to 7. Sphk1(-/-), but not Sphk2(-/-) or Sgpl1(+/-), mice offered protection against hyperoxia-induced lung injury, with improved alveolarization and alveolar integrity compared with wild type. Furthermore, SphK1 deficiency attenuated hyperoxia-induced accumulation of IL-6 in bronchoalveolar lavage fluids and NADPH oxidase (NOX) 2 and NOX4 protein expression in lung tissue. In vitro experiments using human lung microvascular endothelial cells showed that exogenous S1P stimulated intracellular reactive oxygen species (ROS) generation, whereas SphK1 siRNA, or inhibitor against SphK1, attenuated hyperoxia-induced S1P generation. Knockdown of NOX2 and NOX4, using specific siRNA, reduced both basal and S1P-induced ROS formation. These results suggest an important role for SphK1-mediated S1P signaling-regulated ROS in the development of hyperoxia-induced lung injury in a murine neonatal model of bronchopulmonary dysplasia.


Subject(s)
Bronchopulmonary Dysplasia/enzymology , Bronchopulmonary Dysplasia/prevention & control , Hyperoxia/complications , Lysophospholipids/metabolism , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Sphingosine/analogs & derivatives , Aldehyde-Lyases/deficiency , Aldehyde-Lyases/metabolism , Animals , Animals, Newborn , Bronchopulmonary Dysplasia/etiology , Bronchopulmonary Dysplasia/pathology , Disease Models, Animal , Down-Regulation/drug effects , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Cells/pathology , Humans , Hyperoxia/enzymology , Hyperoxia/pathology , Mice , Mice, Inbred C57BL , NADPH Oxidase 2 , NADPH Oxidase 4 , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Pneumonia/complications , Pneumonia/pathology , Pulmonary Alveoli/enzymology , Pulmonary Alveoli/pathology , Reactive Oxygen Species/metabolism , Signal Transduction , Sphingosine/metabolism , rac1 GTP-Binding Protein/metabolism
7.
PLoS One ; 8(2): e56817, 2013.
Article in English | MEDLINE | ID: mdl-23457619

ABSTRACT

The Map kinase Activating Death Domain containing protein (MADD) isoform of the IG20 gene is over-expressed in different types of cancer tissues and cell lines and it functions as a negative regulator of apoptosis. Therefore, we speculated that MADD might be over-expressed in human breast cancer tissues and that MADD knock-down might synergize with chemotherapeutic or TRAIL-induced apoptosis of breast cancer cells. Analyses of breast tissue microarrays revealed over-expression of MADD in ductal and invasive carcinomas relative to benign tissues. MADD knockdown resulted in enhanced spontaneous apoptosis in human breast cancer cell lines. Moreover, MADD knockdown followed by treatment with TRAIL or doxorubicin resulted in increased cell death compared to either treatment alone. Enhanced cell death was found to be secondary to increased caspase-8 activation. These data indicate that strategies to decrease MADD expression or function in breast cancer may be utilized to increase tumor cell sensitivity to TRAIL and doxorubicin induced apoptosis.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/pathology , Death Domain Receptor Signaling Adaptor Proteins/deficiency , Death Domain Receptor Signaling Adaptor Proteins/genetics , Doxorubicin/pharmacology , Gene Knockdown Techniques , Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/genetics , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Amino Acid Sequence , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Death Domain Receptor Signaling Adaptor Proteins/chemistry , Gene Expression Regulation, Neoplastic/drug effects , Guanine Nucleotide Exchange Factors/chemistry , Humans , Molecular Sequence Data , RNA, Small Interfering/genetics , Receptors, Death Domain/metabolism
8.
J Biol Chem ; 288(4): 2191-200, 2013 Jan 25.
Article in English | MEDLINE | ID: mdl-23212923

ABSTRACT

Vascular endothelial cell (EC) barrier integrity is critical to vessel homeostasis whereas barrier dysfunction is a key feature of inflammatory disorders and tumor angiogenesis. We previously reported that hepatocyte growth factor (HGF)-mediated increases in EC barrier integrity are signaled through a dynamic complex present in lipid rafts involving its receptor, c-Met. We extended these observations to confirm that S1PR1 (sphingosine 1-phosphate receptor 1) and integrin ß4 (ITGB4) are essential participants in HGF-induced EC barrier enhancement. Immunoprecipitation experiments demonstrated HGF-mediated recruitment of c-Met, ITGB4 and S1PR1 to caveolin-enriched lipid rafts in human lung EC with direct interactions of c-Met with both S1PR1 and ITGB4 accompanied by c-Met-dependent S1PR1 and ITGB4 transactivation. Reduced S1PR1 expression (siRNA) attenuated both ITGB4 and Rac1 activation as well as c-Met/ITGB4 interaction and resulted in decreased transendothelial electrical resistance. Furthermore, reduced ITGB4 expression attenuated HGF-induced c-Met activation, c-Met/S1PR1 interaction, and effected decreases in S1P- and HGF-induced EC barrier enhancement. Finally, the c-Met inhibitor, XL880, suppressed HGF-induced c-Met activation as well as S1PR1 and ITGB4 transactivation. These results support a critical role for S1PR1 and ITGB4 transactivation as rate-limiting events in the transduction of HGF signals via a dynamic c-Met complex resulting in enhanced EC barrier integrity.


Subject(s)
Endothelial Cells/cytology , Hepatocyte Growth Factor/metabolism , Integrin beta4/metabolism , Proto-Oncogene Proteins c-met/metabolism , Receptors, Lysosphingolipid/metabolism , Cell Membrane/metabolism , Electrophysiology , Humans , Lung/metabolism , Membrane Microdomains/chemistry , Membrane Microdomains/metabolism , Microcirculation , Models, Biological , RNA, Small Interfering/metabolism , Sphingosine-1-Phosphate Receptors , Threonine/chemistry , Transcriptional Activation , Tyrosine/chemistry , rac1 GTP-Binding Protein/metabolism
9.
PLoS One ; 7(9): e45330, 2012.
Article in English | MEDLINE | ID: mdl-23028939

ABSTRACT

BACKGROUND: Malignant pleural mesothelioma (MPM) is a devastating disease with an overall poor prognosis. Despite the recent advances in targeted molecular therapies, there is a clear and urgent need for the identification of novel mesothelioma targets for the development of highly efficacious therapeutics. METHODOLOGY/PRINCIPAL FINDINGS: In this study, we report that the expression of Sphingosine Kinase 1 (SphK1) protein was preferentially elevated in MPM tumor tissues (49 epithelioid and 13 sarcomatoid) compared to normal tissue (n = 13). In addition, we also observed significantly elevated levels of SphK1 and SphK2 mRNA and SphK1 protein expression in MPM cell lines such as H2691, H513 and H2461 compared to the non-malignant mesothelial Met5 cells. The underlying mechanism appears to be mediated by SphK1 induced upregulation of select gene transcription programs such as that of CBP/p300 and PCAF, two histone acetyl transferases (HAT), and the down regulation of cell cycle dependent kinase inhibitor genes such as p27Kip1 and p21Cip1. In addition, using immunoprecipitates of anti-acetylated histone antibody from SphK inhibitor, SphK-I2 treated Met5A and H2691 cell lysates, we also showed activation of other cell proliferation related genes, such as Top2A (DNA replication), AKB (chromosome remodeling and mitotic spindle formation), and suppression of p21 CIP1 and p27KIP1. The CDK2, HAT1 and MYST2 were, however, unaffected in the above study. Using SphK inhibitor and specific siRNA targeting either SphK1 or SphK2, we also unequivocally established that SphK1, but not SphK2, promotes H2691 mesothelioma cell proliferation. Using a multi-walled carbon nanotubes induced peritoneal mesothelioma mouse model, we showed that the SphK1-/- null mice exhibited significantly less inflammation and granulamatous nodules compared to their wild type counterparts. CONCLUSIONS/SIGNIFICANCE: The lipid kinase SphK1 plays a positive and essential role in the growth and development of malignant mesothelioma and is therefore a likely therapeutic target.


Subject(s)
Histones/metabolism , Mesothelioma/enzymology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Acetylation , Animals , Cell Line , Cell Line, Tumor , Cell Proliferation , Chromatin Immunoprecipitation , Humans , Immunoblotting , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Phosphotransferases (Alcohol Group Acceptor)/genetics , Tandem Mass Spectrometry
10.
J Biol Chem ; 285(29): 22713-22, 2010 Jul 16.
Article in English | MEDLINE | ID: mdl-20484047

ABSTRACT

MADD plays an essential role in cancer cell survival. Abrogation of endogenous MADD expression results in significant spontaneous apoptosis and enhanced susceptibility to tumor necrosis factor alpha-related apoptosis-inducing ligand (TRAIL)-induced apoptosis. However, the regulation of MADD function is largely unknown. Here, we demonstrate that endogenous MADD is phosphorylated at three highly conserved sites by Akt, and only the phosphorylated MADD can directly interact with the TRAIL receptor DR4 thereby preventing Fas-associated death domain recruitment. However, in cells susceptible to TRAIL treatment, TRAIL induces a reduction in MADD phosphorylation levels resulting in MADD dissociation from, and Fas-associated death domain association with DR4, which allows death-inducing signaling complex (DISC) formation leading to apoptosis. Thus, the pro-survival function of MADD is dependent upon its phosphorylation by Akt. Because Akt is active in most cancer cells and phosphorylated MADD confers resistance to TRAIL-induced apoptosis, co-targeting Akt-MADD axis is likely to increase efficacy of TRAIL-based therapies.


Subject(s)
Apoptosis/drug effects , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Fas-Associated Death Domain Protein/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Tumor Necrosis Factor/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Cell Line , Humans , Phosphorylation/drug effects , Protein Binding/drug effects , Receptors, TNF-Related Apoptosis-Inducing Ligand , Staining and Labeling
11.
J Environ Pathol Toxicol Oncol ; 28(1): 25-38, 2009.
Article in English | MEDLINE | ID: mdl-19392652

ABSTRACT

The proapoptotic protein Siva-1 plays an important role in some of the extrinsic and intrinsic apoptosis signaling pathways in cancer cells. Previously, we showed that Siva-1 inhibited the activity of the prosurvival transcription factor NF-kappaB. In the present study, upon TCR cross-linking of Jurkat T leukemia cells, we demonstrated that the inhibitory target of Siva-1 is upstream of the IKK complex in the NF-kappaB signaling pathway. Additionally, Siva-1 also suppressed the activity of another crucial transcription factor AP-1, and a common mediator of both these pathways is the adaptor protein TRAF2. Further, we observed that Siva-1 indeed interacted with TRAF2 and negatively regulated its activity by promoting K48-hnked polyubiquitination. Siva-1 specifically interacted with the ring finger domain of TRAF2, which is essential for its E3 hgase activity and its ability to subsequently activate NF-kappaB. TCR cross-linking of Jurkat T cells that lacked Siva-1 revealed significantly lowered K48- but elevated K63-ubiquitinated TRAF2 levels upon TCR cross-linking, suggesting that the differential pattern of ubiquitination in these cells essentially contributed to a robust and sustained activation of NF-kappaB. The above results demonstrated an important role for endogenous Siva-1 in negatively regulating NF-kappaB activation by targeting TRAF2.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , NF-kappa B/metabolism , Receptors, Antigen, T-Cell/physiology , TNF Receptor-Associated Factor 2/metabolism , Apoptosis Regulatory Proteins , Cell Line , Humans , I-kappa B Kinase/metabolism , Jurkat Cells , Lysine/metabolism , Protein Interaction Mapping , RING Finger Domains , TNF Receptor-Associated Factor 2/chemistry , TNF Receptor-Associated Factor 6/metabolism , Transcription Factor AP-1/metabolism , Ubiquitination
12.
J Clin Endocrinol Metab ; 94(4): 1467-71, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19190106

ABSTRACT

AIM: The aim of the study was to investigate the expression and function of the IG20 gene in thyroid cancer cell survival, proliferation, and apoptosis. METHODS: We determined the expression levels of the major isoforms of IG20 by quantitative RT-PCR in normal and thyroid tumor tissues/cell lines. We evaluated the functional consequence of IG20 knockdown in WRO (follicular carcinoma) and FRO (anaplastic carcinoma) thyroid cancer cell lines by measuring spontaneous, TNFalpha-related apoptosis-inducing ligand (TRAIL), and TNFalpha-induced apoptosis. RESULTS: The IG20 gene expression levels were higher in benign and malignant thyroid tumors and in WRO and FRO cells relative to normal tissues. Predominantly, MADD and DENN-SV isoforms of IG20 gene were expressed. IG20 knockdown resulted in increased spontaneous, TRAIL-, and TNFalpha-induced apoptosis in WRO, but not FRO, cells. FRO cell resistance to apoptosis is likely due to caspase-8 deficiency. CONCLUSION: IG20 knockdown renders WRO cells more susceptible to spontaneous, TRAIL-, and TNFalpha-induced apoptosis and thus demonstrates the prosurvival function of the IG20 gene in thyroid cancer. These observations, combined with overexpression of IG20 noted in thyroid tumor tissues, may suggest a potential role in thyroid cancer survival and growth and indicate that IG20 may be targeted either alone or in conjunction with TRAIL or TNFalpha treatment in certain thyroid cancers.


Subject(s)
Death Domain Receptor Signaling Adaptor Proteins/genetics , Guanine Nucleotide Exchange Factors/genetics , Thyroid Neoplasms/genetics , Adenocarcinoma, Follicular/genetics , Adenocarcinoma, Follicular/pathology , Alternative Splicing , Apoptosis , Cell Division , Cell Line, Tumor , DNA Primers , DNA, Neoplasm/genetics , Genetic Variation , Humans , Inverted Repeat Sequences/genetics , RNA, Neoplasm/genetics , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Neoplasms/pathology
13.
Cancer Res ; 65(12): 5301-9, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15958577

ABSTRACT

cis-Diaminedichloroplatinum (II) (cisplatin) is routinely used to treat various types of cancers; however, a significant number develop resistance. One of the underlying factors that contribute to cisplatin resistance is the elevated level of BCL-2 and/or BCL-XL, which promotes cell survival. A potential method of overcoming such resistance is to use a potentiator that is capable of neutralizing the antiapoptotic effects of BCL-2/BCL-XL, such as Siva-1. We previously cloned the proapoptotic protein Siva-1 and showed a possible role for it in both extrinsic and intrinsic apoptosis. Using an adenovirus-based expression system, we now show that Siva-1 can synergize with cisplatin in inducing apoptosis in MCF7 and MDA-MB-231 breast cancer cells. In an anchorage-independent clonogenicity assay, MCF7/caspase-3 cells stably expressing Siva-1, but not the control cells, showed a dramatic decrease in the number of colonies formed on one-time cisplatin treatment. Further, we show that the unique putative amphipathic helical region (SAH) in Siva-1 (amino acid residues 36-55) is necessary and sufficient for the observed enhancement in cisplatin-induced apoptosis by Siva-1. Although cisplatin treatment results in significant elevation in the expression of Fas ligand and intracellular p21 levels, expression of Siva-1 has no additional benefit. Instead, the enhancement in apoptosis seems to be due to activation of intrinsic pathway that involves caspase-9 activation. Moreover, Siva-1 augments cisplatin-mediated cell death in MCF7 cells stably expressing BCL-2. We therefore propose that Siva-1 or its SAH region can be used as a potentiator of cisplatin-based chemotherapy.


Subject(s)
Apoptosis/drug effects , Breast Neoplasms/drug therapy , Cisplatin/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Apoptosis Regulatory Proteins , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspase 3 , Caspases/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Inhibitory Concentration 50 , Intracellular Signaling Peptides and Proteins/genetics , Protein Structure, Secondary , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Transfection
14.
Oncogene ; 22(44): 6845-51, 2003 Oct 09.
Article in English | MEDLINE | ID: mdl-14534530

ABSTRACT

Inhibition of apoptosis or abnormal cell survival can result in tumorigenesis by facilitating the insurgence of various mutations. Immediate-early response gene X-1 (IEX-1), protects T cells from apoptosis induced by the ligation of Fas or the T-cell receptor (TCR)/CD3 complex in Emu-IEX-1 mice that direct the gene expression in both T and B cell lineages under the control of the Emu enhancer. Consistent with a biased effect of IEX-1 towards T cells, Emu-IEX-1 mice selectively developed T-cell lymphomas in the spleen, when they aged, which may be associated with increased levels of IEX-1 phosphorylation in T cells compared to B cells. The lymphomas were single positive (CD4+CD8-, CD4-CD8+), double positive (CD4+CD8+), or double negative (CD4-CD8-) T cells. They resulted from aberrantly clonal expansions of T cells expressing a specific TCR, as suggested by the TCR repertoire analysis using a panel of monoclonal antibodies recognizing TCR Vbeta chain, as well as by TCR beta gene rearrangements. The study provides, for the first time, unambiguous evidence of the oncogenic potential of IEX-1 in a cell-specific manner. The animal model may help our understanding of peripheral T-cell lymphoma development.


Subject(s)
Enhancer Elements, Genetic/physiology , Genes, Immediate-Early/physiology , Lymphoma, T-Cell/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocytes/metabolism , Aging , Animals , Antibodies, Monoclonal/metabolism , Apoptosis , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD3 Complex/metabolism , CD4 Antigens/genetics , CD8 Antigens/genetics , Gene Expression , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Lymphoma, T-Cell/immunology , Lymphoma, T-Cell/pathology , Mice , Mice, Transgenic , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Spleen/immunology , Spleen/pathology , T-Lymphocyte Subsets/classification , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...