Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 978, 2023 02 22.
Article in English | MEDLINE | ID: mdl-36813768

ABSTRACT

Lymphatic malformation (LM) is a vascular anomaly originating from lymphatic endothelial cells (ECs). While it mostly remains a benign disease, a fraction of LM patients progresses to malignant lymphangiosarcoma (LAS). However, very little is known about underlying mechanisms regulating LM malignant transformation to LAS. Here, we investigate the role of autophagy in LAS development by generating EC-specific conditional knockout of an essential autophagy gene Rb1cc1/FIP200 in Tsc1iΔEC mouse model for human LAS. We find that Fip200 deletion blocked LM progression to LAS without affecting LM development. We further show that inhibiting autophagy by genetical ablation of FIP200, Atg5 or Atg7, significantly inhibited LAS tumor cell proliferation in vitro and tumorigenicity in vivo. Transcriptional profiling of autophagy-deficient tumor cells and additional mechanistic analysis determine that autophagy plays a role in regulating Osteopontin expression and its down-stream Jak/Stat3 signaling in tumor cell proliferation and tumorigenicity. Lastly, we show that specifically disrupting FIP200 canonical autophagy function by knocking-in FIP200-4A mutant allele in Tsc1iΔEC mice blocked LM progression to LAS. These results demonstrate a role for autophagy in LAS development, suggesting new strategies for preventing and treating LAS.


Subject(s)
Lymphangiosarcoma , Humans , Mice , Animals , Autophagy-Related Proteins , Endothelial Cells , Osteopontin , Autophagy/genetics , STAT3 Transcription Factor
2.
Cell Rep ; 35(11): 109235, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34133924

ABSTRACT

T regulatory (Treg) cells are crucial to maintain immune tolerance and repress antitumor immunity, but the mechanisms governing their cellular redox homeostasis remain elusive. We report that glutathione peroxidase 4 (Gpx4) prevents Treg cells from lipid peroxidation and ferroptosis in regulating immune homeostasis and antitumor immunity. Treg-specific deletion of Gpx4 impairs immune homeostasis without substantially affecting survival of Treg cells at steady state. Loss of Gpx4 results in excessive accumulation of lipid peroxides and ferroptosis of Treg cells upon T cell receptor (TCR)/CD28 co-stimulation. Neutralization of lipid peroxides and blockade of iron availability rescue ferroptosis of Gpx4-deficient Treg cells. Moreover, Gpx4-deficient Treg cells elevate generation of mitochondrial superoxide and production of interleukin-1ß (IL-1ß) that facilitates T helper 17 (TH17) responses. Furthermore, Treg-specific ablation of Gpx4 represses tumor growth and concomitantly potentiates antitumor immunity. Our studies establish a crucial role for Gpx4 in protecting activated Treg cells from lipid peroxidation and ferroptosis and offer a potential therapeutic strategy to improve cancer treatment.


Subject(s)
Ferroptosis , Immunity , Lipid Peroxidation , Lymphocyte Activation/immunology , Neoplasms/immunology , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Cell Line, Tumor , Forkhead Transcription Factors/metabolism , Gene Deletion , Homeostasis , Interleukin-1beta/metabolism , Iron/metabolism , Lipid Peroxides/metabolism , Lymphoid Tissue/immunology , Mice, Inbred C57BL , Mitochondria/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/deficiency , Superoxides/metabolism , Th17 Cells/immunology
3.
Br J Cancer ; 122(12): 1791-1802, 2020 06.
Article in English | MEDLINE | ID: mdl-32336756

ABSTRACT

BACKGOUND: The mechanistic target of rapamycin complex 1 (mTORC1) is important in the development and progression of many cancers. Targeted cancer therapy using mTORC1 inhibitors is used for treatment of cancers; however, their clinical efficacies are still limited. METHODS: We recently created a new mouse model for human lymphangiosarcoma by deleting Tsc1 in endothelial cells and consequent hyper-activation of mTORC1. Using Tsc1iΔEC tumour cells from this mouse model, we assessed the efficacies of histone deacetylase (HDAC) inhibitors as anti-tumour agents for mTORC1-driven tumours. RESULTS: Unlike the cytostatic effect of mTORC1 inhibitors, HDAC inhibitors induced Tsc1iΔEC tumour cell death in vitro and their growth in vivo. Analysis of several HDAC inhibitors suggested stronger anti-tumour activity of class I HDAC inhibitor than class IIa or class IIb inhibitors, but these or pan HDAC inhibitor SAHA did not affect mTORC1 activation in these cells. Moreover, HDAC inhibitor-induced cell death required elevated autophagy, but was not affected by disrupting caspase-dependent apoptosis pathways. We also observed increased reactive oxygen species and endoplasmic reticulum stress in SAHA-treated tumour cells, suggesting their contribution to autophagic cell death, which were dependent on mTORC1 hyper-activation. CONCLUSION: These studies suggest a potential new treatment strategy for mTORC1-driven cancers like lymphangiosarcoma through an alternative mechanism.


Subject(s)
Antineoplastic Agents/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Lymphangiosarcoma/pathology , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Molecular Targeted Therapy/methods , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Mice , Neoplasms, Experimental/pathology
4.
Sci Rep ; 8(1): 2550, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29416084

ABSTRACT

Focal adhesion kinase (FAK) is essential in embryonic angiogenesis by regulating endothelial cell (EC) survival and barrier functions through its kinase-independent and -dependent activities. Here, we generated EC-specific tamoxifen-inducible FAK knockout and FAK kinase-defective (KD) mutant knockin mice to investigate the role of FAK and its kinase activity in angiogenesis of adult animals. Unlike previous observations of their differential defects in embryonic vascular development, both FAK ablation and inactivation of its kinase activity resulted in deficient angiogenesis in wound-healing as well as retinal angiogenesis models. Consistent with these phenotypes, loss of FAK or its kinase activity decreased EC proliferation and migration to similar extents, suggesting FAK primarily acts as a kinase for the regulation of adult EC-mediated angiogenesis. Further mechanistic analyses were carried out using an established mouse EC line MS1 cells. Interestingly, we found that FAK regulated the expression of VEGFR2, a central mediator of various EC functions and angiogenesis, which requires both FAK kinase activity and its translocation into the nucleus. Moreover, nuclear FAK was detected in the RNA polymerase II complex associated with VEGFR2 promoter, suggesting its direct participation in the transcriptional regulation of VEGFR2. Together, our results provide significant insights into the signaling mechanisms of FAK in angiogenesis that may contribute to future design of more effective angiogenesis related therapy.


Subject(s)
Endothelial Cells/metabolism , Focal Adhesion Kinase 1/metabolism , Retinal Neovascularization/genetics , Transcription, Genetic , Vascular Endothelial Growth Factor Receptor-2/genetics , Animals , Cell Movement , Cell Nucleus/metabolism , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Female , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/physiology , Gene Expression Regulation , Male , Mice , Mice, Inbred C57BL , Signal Transduction
5.
Cell Cycle ; 15(14): 1801-2, 2016 07 17.
Article in English | MEDLINE | ID: mdl-27105012
6.
Genes Dev ; 30(7): 856-69, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27013233

ABSTRACT

Autophagy is an evolutionarily conserved cellular process controlled through a set of essential autophagy genes (Atgs). However, there is increasing evidence that most, if not all, Atgs also possess functions independent of their requirement in canonical autophagy, making it difficult to distinguish the contributions of autophagy-dependent or -independent functions of a particular Atg to various biological processes. To distinguish these functions for FIP200 (FAK family-interacting protein of 200 kDa), an Atg in autophagy induction, we examined FIP200 interaction with its autophagy partner, Atg13. We found that residues 582-585 (LQFL) in FIP200 are required for interaction with Atg13, and mutation of these residues to AAAA (designated the FIP200-4A mutant) abolished its canonical autophagy function in vitro. Furthermore, we created a FIP200-4A mutant knock-in mouse model and found that specifically blocking FIP200 interaction with Atg13 abolishes autophagy in vivo, providing direct support for the essential role of the ULK1/Atg13/FIP200/Atg101 complex in the process beyond previous studies relying on the complete knockout of individual components. Analysis of the new mouse model showed that nonautophagic functions of FIP200 are sufficient to fully support embryogenesis by maintaining a protective role in TNFα-induced apoptosis. However, FIP200-mediated canonical autophagy is required to support neonatal survival and tumor cell growth. These studies provide the first genetic evidence linking an Atg's autophagy and nonautophagic functions to different biological processes in vivo.


Subject(s)
Autophagy/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Proteins , Cell Proliferation/genetics , Disease Models, Animal , Embryonic Development/genetics , Female , Gene Knock-In Techniques , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Nude , Mutation , Survival Analysis , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism
7.
Breast Cancer Res Treat ; 149(3): 619-29, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25638397

ABSTRACT

Chemotherapy is the mainstay of systemic treatment for triple negative breast cancer (TNBC); however, the development of drug resistance limits its effectiveness. Therefore, we investigated the underlying mechanism for drug resistance and potential approaches to overcome it for a more effective treatment for TNBCs. Using a pulse-stimulated selection strategy to mimic chemotherapy administration in the clinic, we developed a new paclitaxel-resistant MDA-MB-231 cell line and analyzed these cells for changes in autophagy activity, and the role and mechanisms of the increased autophagy in promoting drug resistance were determined. We found that the pulse-stimulated selection strategy with paclitaxel resulted in MDA-MB-231 variant cells with enhanced resistance to paclitaxel. These resistant cells were found to have enhanced basal autophagy activity, which confers a cytoprotective function under paclitaxel treatment stress. Inhibition of autophagy enhanced paclitaxel-induced cell death in these paclitaxel-resistant cells. We further revealed that up-regulated autophagy in resistant cells enhanced the clearance of damaged mitochondria. Last, we showed that the paclitaxel-resistant cancer cells acquired cross resistance to epirubicin and cisplatin. Together, these results suggest that combining autophagy inhibition with chemotherapy may be an effective strategy to improve treatment outcome in paclitaxel-resistant TNBC patients.


Subject(s)
Autophagy/drug effects , Drug Resistance, Neoplasm/genetics , Paclitaxel/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols , Apoptosis/drug effects , Cell Line, Tumor , Cisplatin/administration & dosage , Female , Humans , Induction Chemotherapy , Mitochondria/drug effects , Mitochondria/pathology , Treatment Outcome , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
8.
J Bone Miner Res ; 30(7): 1195-205, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25639352

ABSTRACT

Tuberous sclerosis complex (TSC) is an autosomal dominant disorder caused by mutations in either TSC1 or TSC2. TSC has high frequency of osseous manifestations such as sclerotic lesions in the craniofacial region. However, an animal model that replicates TSC craniofacial bone lesions has not yet been described. The roles of Tsc1 and the sequelae of Tsc1 dysfunction in bone are unknown. In this study, we generated a mouse model of TSC with a deletion of Tsc1 in neural crest-derived (NCD) cells that recapitulated the sclerotic craniofacial bone lesions in TSC. Analysis of this mouse model demonstrated that TSC1 deletion led to enhanced mTORC1 signaling in NCD bones and the increase in bone formation is responsible for the aberrantly increased bone mass. Lineage mapping revealed that TSC1 deficient NCD cells overpopulated the NCD bones. Mechanistically, hyperproliferation of osteoprogenitors at an early postnatal stage accounts for the increased osteoblast pool. Intriguingly, early postnatal treatment with rapamycin, an mTORC1 inhibitor, can completely rescue the aberrant bone mass, but late treatment cannot. Our data suggest that enhanced mTOR signaling in NCD cells can increase bone mass through enlargement of the osteoprogenitor pool, which likely explains the sclerotic bone lesion observed in TSC patients.


Subject(s)
Bone and Bones/metabolism , Bone and Bones/pathology , Craniofacial Abnormalities/pathology , Gene Deletion , Neural Crest/metabolism , Tumor Suppressor Proteins/deficiency , Animals , Animals, Newborn , Bone and Bones/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Integrases/metabolism , Mice, Knockout , Neural Crest/drug effects , Organ Size , Organ Specificity/drug effects , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteogenesis/drug effects , Phenotype , Sclerosis , Sirolimus/pharmacology , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/metabolism
9.
Cancer Cell ; 28(6): 758-772, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26777415

ABSTRACT

Angiosarcoma/lymphangiosarcoma is a rare malignancy with poor prognosis. We generated a mouse model with inducible endothelial-cell-specific deletion of Tsc1 to examine mTORC1 signaling in lymphangiosarcoma. Tsc1 loss increased retinal angiogenesis in neonates and led to endothelial proliferative lesions from vascular malformations to vascular tumors in adult mice. Sustained mTORC1 signaling was required for lymphangiosarcoma development and maintenance. Increased VEGF expression in tumor cells was seen, and blocking autocrine VEGF signaling abolished vascular tumor development and growth. We also found significant correlations between mTORC1 activation and VEGF, HIF1α, and c-Myc expression in human angiosarcoma samples. These studies demonstrated critical mechanisms of aberrant mTORC1 activation in lymphangiosarcoma and validate the mice as a valuable model for further study.


Subject(s)
Autocrine Communication , Cell Transformation, Neoplastic/metabolism , Endothelial Cells/enzymology , Lymphangiosarcoma/metabolism , Multiprotein Complexes/metabolism , Neovascularization, Pathologic , TOR Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Autocrine Communication/drug effects , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/pathology , Enzyme Activation , Genotype , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lymphangiosarcoma/drug therapy , Lymphangiosarcoma/genetics , Lymphangiosarcoma/pathology , Mechanistic Target of Rapamycin Complex 1 , Mice, Knockout , Mice, Nude , Multiprotein Complexes/antagonists & inhibitors , Neoplasm Invasiveness , Phenotype , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/analysis , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Interference , Receptors, Vascular Endothelial Growth Factor/metabolism , Retinal Neovascularization/genetics , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , Transfection , Tuberous Sclerosis Complex 1 Protein , Tumor Burden , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Vascular Endothelial Growth Factor A/analysis , Vascular Endothelial Growth Factor A/antagonists & inhibitors
11.
J Biol Chem ; 288(5): 3322-33, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23255596

ABSTRACT

Tyrosine kinases have been shown to play critical roles in cancer development and progression, and their inhibitors hold the potential as effective targeted therapies for breast and other cancers. However, some of these kinases like focal adhesion kinase (FAK) also possess scaffolding functions in intracellular signaling, but such kinase-independent functions of FAK or other kinases have not been examined in cancer directly in vivo. Here, we report that disruption of the function of FAK scaffolding through its Pro-878/881 motif suppressed mammary tumor growth and metastasis in a well characterized murine model of human breast cancer. P878A/P881A mutation in the endogenous FAK gene decreased the expression of markers for epithelial-mesenchymal transition (EMT) and mammary cancer stem cell (MaCSC) activities in tumors derived from mutant mice. This mutation disrupted the function of FAK scaffolding to mediate endophilin A2 phosphorylation at Tyr-315 by Src, leading to the decreased surface expression of MT1-MMP, as observed previously in transformed fibroblasts in vitro. Inhibition of the downstream components of this FAK scaffolding function by Y315F endophilin A2 mutant or MT1-MMP knockdown reduced markers for EMT and MaCSC activities. Conversely, bypass of the scaffolding function using the phosphorylation mimic mutant Y315E endophilin A2 or endophilin A2 knockdown rescued the decreased markers for EMT and MaCSCs as well as surface expression of MT1-MMP in tumor cells harboring the P878A/P881A mutation. Together, these results identify a novel role of FAK scaffolding function in breast cancer, which could serve as a new target in combination with kinase inhibition for more effective treatment strategies.


Subject(s)
Acyltransferases/metabolism , Epithelial-Mesenchymal Transition , Focal Adhesion Kinase 1/metabolism , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Neoplastic Stem Cells/enzymology , Neoplastic Stem Cells/pathology , Animals , Biomarkers, Tumor/metabolism , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Female , Gene Knock-In Techniques , Gene Knockdown Techniques , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Matrix Metalloproteinase 14/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation/genetics , Phosphorylation , Protein Binding , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Suppression, Genetic , Tumor Cells, Cultured
12.
J Immunol ; 186(1): 539-48, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-21098220

ABSTRACT

NF-κB is a family of important transcription factors that modulate immunity, development, inflammation, and cancer. The biological activity of NF-κB is subjected to various spatial and temporal regulations. Bioinformatics analysis predicts that CITED2 is topologically close to NF-κB in the protein interaction networks. In this study, we show that ectopic expression or knockdown of CITED2 attenuates or potentiates, respectively, the expression of NF-κB-responsive genes. Mechanistically, CITED2 constitutively localizes inside the nucleus and interacts specifically with the coactivator p300. This prevents p65 from binding to p300, impairs p65 acetylation, and attenuates p65 binding to its cognate promoters. Furthermore, LPS induces CITED2 expression via NF-κB in macrophages. CITED2 sensitizes cells to TNF-α-induced apoptosis. Collectively, this study identifies CITED2 as a novel regulator of NF-κB in the nucleus, which reveals a negative feedback mechanism for NF-κB signaling.


Subject(s)
Cell Nucleus/immunology , Cell Nucleus/metabolism , Down-Regulation/immunology , Feedback, Physiological/physiology , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Repressor Proteins/physiology , Trans-Activators/physiology , Animals , Cell Line , HEK293 Cells , Humans , Jurkat Cells , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Protein Interaction Mapping , Repressor Proteins/biosynthesis , Repressor Proteins/deficiency , Signal Transduction/genetics , Signal Transduction/immunology , Trans-Activators/biosynthesis , Trans-Activators/deficiency
13.
J Cell Biol ; 189(6): 955-65, 2010 Jun 14.
Article in English | MEDLINE | ID: mdl-20530207

ABSTRACT

Focal adhesion kinase (FAK) is essential for vascular development as endothelial cell (EC)-specific knockout of FAK (conditional FAK knockout [CFKO] mice) leads to embryonic lethality. In this study, we report the differential kinase-independent and -dependent functions of FAK in vascular development by creating and analyzing an EC-specific FAK kinase-defective (KD) mutant knockin (conditional FAK knockin [CFKI]) mouse model. CFKI embryos showed apparently normal development through embryonic day (E) 13.5, whereas the majority of CFKO embryos died at the same stage. Expression of KD FAK reversed increased EC apoptosis observed with FAK deletion in embryos and in vitro through suppression of up-regulated p21. However, vessel dilation and defective angiogenesis of CFKO embryos were not rescued in CFKI embryos. ECs without FAK or expressing KD FAK showed increased permeability, abnormal distribution of vascular endothelial cadherin (VE-cadherin), and reduced VE-cadherin Y658 phosphorylation. Together, our data suggest that kinase-independent functions of FAK can support EC survival in vascular development through E13.5 but are insufficient for maintaining EC function to allow for completion of embryogenesis.


Subject(s)
Cell Survival , Embryo, Mammalian/physiology , Embryonic Development/physiology , Endothelial Cells/physiology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Cells, Cultured , Embryo, Mammalian/anatomy & histology , Endothelial Cells/cytology , Female , Focal Adhesion Protein-Tyrosine Kinases/genetics , Gene Knock-In Techniques , Male , Mice , Mice, Knockout , Neovascularization, Physiologic/physiology , Pregnancy
14.
Cell Res ; 19(4): 412-28, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19153595

ABSTRACT

Interferon regulatory factor (IRF)3 is critical for the transcriptional induction of chemokines and cytokines during viral or bacterial invasion. The kinases Tank binding kinase (TBK)1 and Ikappa B kinase (IKK)epsilon can phosphorylate the C-terminal part of IRF3 and play important roles in IRF3 activation. In this study, we show that another kinase, c-Jun-NH(2)-terminal kinase (JNK), phosphorylates IRF3 on its N-terminal serine 173 residue, and TAK1 can stimulate IRF3 phosphorylation via JNK. JNK specific inhibitor SP600125 inhibits the N-terminal phosphorylation without affecting the C-terminal phosphorylation. In addition, IRF3-mediated gene expressions on lipopolysaccharide (LPS) or polyinosinic-cytidylic acid (polyI:C) treatment are severely impaired by SP600125, as well as for reporter gene assay of IRF3 activation. Knockdown of TAK1 further confirmed these observations. Interestingly, constitutive active IRF3(5D) can be inhibited by SP600125; JNK1 can synergize the action of IRF3(5D), but not the S173A-IRF3(5D) mutant. More importantly, polyI:C failed to induce the phosphorylation of mutant S173A and SP600125 dramatically abrogated IRF3 phosphorylation and dimerization that was stimulated by polyI:C. Thus, this study demonstrates that the TAK1-JNK cascade is required for IRF3 function, in addition to TBK1/IKKvarepsilon, uncovering a new mechanism for mitogen-activated protein (MAP) kinase to regulate the innate immunity.


Subject(s)
Immunity, Innate , Interferon Regulatory Factor-3/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Amino Acid Substitution , Animals , Anthracenes/chemistry , Anthracenes/pharmacology , Cell Line , Gene Expression Regulation , Gene Knockdown Techniques , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Lipopolysaccharides/pharmacology , MAP Kinase Signaling System , Mice , Mutant Proteins/metabolism , Poly I-C/pharmacology , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/metabolism
15.
J Cell Biol ; 178(2): 231-44, 2007 Jul 16.
Article in English | MEDLINE | ID: mdl-17620405

ABSTRACT

As a latent transcription factor, nuclear factor kappaB (NF-kappaB) translocates from the cytoplasm into the nucleus upon stimulation and mediates the expression of genes that are important in immunity, inflammation, and development. However, little is known about how it is regulated inside the nucleus. By a two-hybrid approach, we identify a prefoldin-like protein, ubiquitously expressed transcript (UXT), that is expressed predominantly and interacts specifically with NF-kappaB inside the nucleus. RNA interference knockdown of UXT leads to impaired NF-kappaB activity and dramatically attenuates the expression of NF-kappaB-dependent genes. This interference also sensitizes cells to apoptosis by tumor necrosis factor-alpha. Furthermore, UXT forms a dynamic complex with NF-kappaB and is recruited to the NF-kappaB enhanceosome upon stimulation. Interestingly, the UXT protein level correlates with constitutive NF-kappaB activity in human prostate cancer cell lines. The presence of NF-kappaB within the nucleus of stimulated or constitutively active cells is considerably diminished with decreased endogenous UXT levels. Our results reveal that UXT is an integral component of the NF-kappaB enhanceosome and is essential for its nuclear function, which uncovers a new mechanism of NF-kappaB regulation.


Subject(s)
Enhancer Elements, Genetic/genetics , NF-kappa B/metabolism , Transcription, Genetic , Animals , Apoptosis/drug effects , Cell Cycle Proteins , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter , Humans , Luciferases/metabolism , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Molecular Chaperones , NF-kappa B/genetics , Neoplasm Proteins , Prostatic Neoplasms/pathology , RNA Interference , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Two-Hybrid System Techniques
16.
Mol Biol Cell ; 17(3): 1461-71, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16394098

ABSTRACT

Interferon regulatory factor 3 (IRF3) plays a crucial role in mediating cellular responses to virus intrusion. The protein kinase TBK1 is a key regulator inducing phosphorylation of IRF3. The regulatory mechanisms during IRF3 activation remain poorly characterized. In the present study, we have identified by yeast two-hybrid approach a specific interaction between IRF3 and chaperone heat-shock protein of 90 kDa (Hsp90). The C-terminal truncation mutant of Hsp90 is a strong dominant-negative inhibitor of IRF3 activation. Knockdown of endogenous Hsp90 by RNA interference attenuates IRF3 activation and its target gene expressions. Alternatively, Hsp90-specific inhibitor geldanamycin (GA) dramatically reduces expression of IRF3-regulated interferon-stimulated genes and abolishes the cytoplasm-to-nucleus translocation and DNA binding activity of IRF3 in Sendai virus-infected cells. Significantly, virus-induced IRF3 phosphorylation is blocked by GA, whereas GA does not affect the protein level of IRF3. In addition, TBK1 is found to be a client protein of Hsp90 in vivo. Treatment of 293 cells with GA interferes with the interaction of TBK1 and Hsp90, resulting in TBK1 destabilization and its subsequent proteasome-mediated degradation. Besides maintaining stability of TBK1, Hsp90 also forms a novel complex with TBK1 and IRF3, which brings TBK1 and IRF3 dynamically into proximity and facilitates signal transduction from TBK1 to IRF3. Our study uncovers an essential role of Hsp90 in the virus-induced activation of IRF3.


Subject(s)
Gene Expression Regulation , HSP90 Heat-Shock Proteins/metabolism , Interferon Regulatory Factor-3/metabolism , Protein Serine-Threonine Kinases/metabolism , Sendai virus/physiology , Animals , Benzoquinones , Binding Sites , Cells, Cultured , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/chemistry , HSP90 Heat-Shock Proteins/deficiency , Humans , Interferon Regulatory Factor-3/chemistry , Lactams, Macrocyclic , Mice , Multiprotein Complexes , Protein Binding , Protein Structure, Tertiary , Quinones/pharmacology , RNA Interference , Thermodynamics
17.
Exp Cell Res ; 308(1): 196-210, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-15896775

ABSTRACT

The immediate early protein ICP0 encoded by herpes simplex virus 1 (HSV-1) is believed to activate transcription and consequently productive infection. The precise mechanisms of ICP0-mediated transactivation are under intensive study. Here, we demonstrate that ICP0 can strongly activate AP-1 responsive genes specifically. This activation is inhibited by c-Jun (S73A), c-Jun (S63/73A), TAK1 (K63W), but not by p38 (AF), ERK1 (K71R), ERK2 (K52R) and TRAF6 (C85A/H87A). We further investigate the relevancy of ERK, JNK and p38 MAPK pathways using their respective inhibitors PD98059, SP600125 and SB202190. Only SP600125 significantly attenuates the AP-1 responsive gene activation by ICP0. Consistent with these, the JNK is remarkably activated in response to ICP0, and this JNK activation is shown to be significantly attenuated by TAK1 (K63W). It turns out that ICP0 interacts specifically with TAK1 and stimulates its kinase activity. These findings reveal a new molecular mechanism ICP0 explores to regulate gene expression.


Subject(s)
Immediate-Early Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Signal Transduction/physiology , Cell Line , Cell Line, Tumor , Humans , JNK Mitogen-Activated Protein Kinases/pharmacology , MAP Kinase Kinase Kinases/metabolism , Models, Biological , Transcription Factor AP-1/drug effects , Transcription Factor AP-1/metabolism , Transcription Factor AP-1/physiology , Ubiquitin-Protein Ligases , Up-Regulation/physiology
18.
Cell Signal ; 17(2): 217-29, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15494213

ABSTRACT

The immediate early proteins ICP0 and BICP0 from Herpes virus are promiscuous activators of both viral and cellular genes and play a critical role in virus life cycle. Here we report that ICP0 and BICP0 could induce NF-kappaB translocation from cytoplasm into nucleus and strongly activate NF-kappaB responsive genes specifically. This process was dependent on the RING domain of both proteins. In addition, ICP0 interacted specifically with IkappaBalpha and its activating effect was attenuated by Ubch5A(C85A) and MG132, but not by IkappaBalpha(S32A/S36A). Remarkably, IkappaBalpha was poly-ubiquitinated by both ICP0 and BICP0, in vitro and in vivo. These data indicate that ICP0 and BICP0, functioning as ubiquitin ligases, are bona fide activators of NF-kappaB signaling pathway. Our study identifies a new way ICP0 and BICP0 explore to regulate gene expression.


Subject(s)
I-kappa B Proteins/metabolism , Immediate-Early Proteins/physiology , NF-kappa B/metabolism , Trans-Activators/physiology , Ubiquitin-Protein Ligases/metabolism , Viral Proteins/physiology , Animals , Cattle , Cell Line , Cell Line, Tumor , Cysteine Proteinase Inhibitors/pharmacology , Electrophoretic Mobility Shift Assay , Gene Expression Regulation, Viral , Humans , I-kappa B Kinase , I-kappa B Proteins/genetics , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Iron-Binding Proteins/genetics , Iron-Binding Proteins/metabolism , Leupeptins/pharmacology , Mutation , NF-KappaB Inhibitor alpha , Protein Serine-Threonine Kinases/metabolism , Protein Transport/physiology , Simplexvirus , Terminal Repeat Sequences/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factor RelA , Transcription, Genetic/drug effects , Transcription, Genetic/genetics , Transfection , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
19.
Biochem Biophys Res Commun ; 324(1): 432-9, 2004 Nov 05.
Article in English | MEDLINE | ID: mdl-15465037

ABSTRACT

Tumor necrosis factor receptor-associated factor 6 (TRAF6) is a crucial signaling transducer that regulates a diverse array of physiological processes, including adaptive immunity, innate immunity, and bone metabolism. Importantly, it is essential for activating NF-kappaB signaling pathway in response to interleukin-1 and Toll-like receptor ligands. Previously, we characterized TRAF6 to be a ubiquitin ligase. In combination with the ubiquitin conjugating enzyme complex Ubc13/Uev1A, TRAF6 could catalyze the formation on itself of unique Lys-63 linked polyubiquitin chain that positively regulated NF-kappaB signaling pathway. However, it remains unknown how this auto-ubiquitination process is regulated. In this study, we found that the coiled-coil domain of TRAF6 was essential for its auto-ubiquitination and activating NF-kappaB signaling pathway. This domain served not as the specific target where the polyubiquitin chain was linked, but as a specific bridge to recruit Ubc13/Uev1A.


Subject(s)
NF-kappa B/metabolism , Protein Structure, Secondary , TNF Receptor-Associated Factor 6/chemistry , TNF Receptor-Associated Factor 6/metabolism , Ubiquitin/metabolism , Animals , Cell Line , Enzyme Activation , Genes, Reporter , Humans , I-kappa B Kinase , Lysine/metabolism , Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction/physiology , TNF Receptor-Associated Factor 6/genetics , Ubiquitin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...