Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
2.
Breast Cancer Res ; 25(1): 10, 2023 01 26.
Article in English | MEDLINE | ID: mdl-36703228

ABSTRACT

Necroptosis is a form of regulated necrosis and is executed by MLKL when MLKL is engaged in triggering the rupture of cell plasma membrane. MLKL activation also leads to the protease, ADAMs-mediated ectodomain shedding of cell surface proteins of necroptotic cells. Tumor necroptosis often happens in advanced solid tumors, and blocking necroptosis by MLKL deletion in breast cancer dramatically reduces tumor metastasis. It has been suggested that tumor necroptosis affects tumor progression through modulating the tumor microenvironment. However, the exact mechanism by which tumor necroptosis promotes tumor metastasis remains elusive. Here, we report that the ectodomain shedding of cell surface proteins of necroptotic cells is critical for the promoting effect of tumor necroptosis in tumor metastasis through inhibiting the anti-tumor activity of T cells. We found that blocking tumor necroptosis by MLKL deletion led to the dramatic reduction of tumor metastasis and significantly elevated anti-tumor activity of tumor-infiltrating and peripheral blood T cells. Importantly, the increased anti-tumor activity of T cells is a key cause for the reduced metastasis as the depletion of CD8+ T cells completely restored the level of metastasis in the Mlkl KO mice. Interestingly, the levels of some soluble cell surface proteins including sE-cadherin that are known to promote metastasis are also dramatically reduced in MLKL null tumors/mice. Administration of ADAMs pan inhibitor reduces the levels of soluble cell surface proteins in WT tumors/mice and leads to the dramatic decrease in metastasis. Finally, we showed the sE-cadherin/KLRG1 inhibitory receptor is the major pathway for necroptosis-mediated suppression of the anti-tumor activity of T cells and the promotion of metastasis. Hence, our study reveals a novel mechanism of tumor necroptosis-mediated promotion of metastasis and suggests that tumor necroptosis and necroptosis-activated ADAMs are potential targets for controlling metastasis.


Subject(s)
Breast Neoplasms , Membrane Proteins , Necroptosis , Neoplasm Metastasis , Animals , Mice , Cadherins , Membrane Proteins/metabolism , Mice, Knockout , Protein Kinases , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/pharmacology , Tumor Microenvironment , Breast Neoplasms/pathology
3.
Trends Cancer ; 8(1): 21-27, 2022 01.
Article in English | MEDLINE | ID: mdl-34627742

ABSTRACT

Necroptosis, a form of programmed necrotic cell death, is a gatekeeper of host defense against certain pathogen invasions. The deregulation of necroptosis is also a key factor of many inflammatory diseases. Recent studies have revealed an important role of necroptosis in tumorigenesis and metastasis and imply the potential of targeting necroptosis as a novel cancer therapy. While its molecular mechanism has been well studied, details of the regulation and function of necroptosis of tumor cells in tumorigenesis and metastasis only began to emerge recently, and we discuss these herein.


Subject(s)
Necroptosis , Receptor-Interacting Protein Serine-Threonine Kinases , Apoptosis/genetics , Humans , Necroptosis/genetics , Necrosis , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
4.
Nat Commun ; 12(1): 2666, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33976222

ABSTRACT

Tumor necrosis happens commonly in advanced solid tumors. We reported that necroptosis plays a major role in tumor necrosis. Although several key necroptosis regulators including receptor interacting protein kinase 1 (RIPK1) have been identified, the regulation of tumor necroptosis during tumor development remains elusive. Here, we report that Z-DNA-binding protein 1 (ZBP1), not RIPK1, mediates tumor necroptosis during tumor development in preclinical cancer models. We found that ZBP1 expression is dramatically elevated in necrotic tumors. Importantly, ZBP1, not RIPK1, deletion blocks tumor necroptosis during tumor development and inhibits metastasis. We showed that glucose deprivation triggers ZBP1-depedent necroptosis in tumor cells. Glucose deprivation causes mitochondrial DNA (mtDNA) release to the cytoplasm and the binding of mtDNA to ZBP1 to activate MLKL in a BCL-2 family protein, NOXA-dependent manner. Therefore, our study reveals ZBP1 as the key regulator of tumor necroptosis and provides a potential drug target for controlling tumor metastasis.


Subject(s)
Breast Neoplasms/genetics , Necroptosis/genetics , RNA-Binding Proteins/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , HEK293 Cells , Humans , MCF-7 Cells , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , RNA-Binding Proteins/metabolism , RNAi Therapeutics/methods , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Xenograft Model Antitumor Assays/methods
5.
Methods Mol Biol ; 2248: 73-80, 2021.
Article in English | MEDLINE | ID: mdl-33185868

ABSTRACT

Tumor necrosis factor (TNF) plays a key role in inflammatory responses and in various cellular events such as apoptosis and necroptosis. The interaction of TNF with its receptor, TNFR1, drives the initiation of complex molecular pathways leading to inflammation and cell death. RARγ is released from the nucleus to orchestrate the formation of the cytosolic death complexes, and it is cytosolic RARγ that plays a pivotal role in switching TNF-induced inflammatory responses to RIPK1-initiated cell death. Thus, RARγ provides a checkpoint for the transition from inflammatory signaling to death machinery of RIPK1-initiated cell death in response to TNF. Here, we use techniques to identify RARγ as a downstream mediator of TNFR1 signaling complex. We use confocal imaging to show the localization of RARγ upon activation of cell death. Immunoprecipitation of RARγ identified the interacting proteins.


Subject(s)
Apoptosis , Inflammation/etiology , Inflammation/metabolism , Signal Transduction , Tumor Necrosis Factors/metabolism , Animals , Apoptosis/genetics , Biomarkers , Blotting, Western , Cell Line , Disease Susceptibility , Humans , Immunoprecipitation , Inflammation/pathology , Tumor Necrosis Factors/genetics
6.
Cell Commun Signal ; 18(1): 161, 2020 10 09.
Article in English | MEDLINE | ID: mdl-33036630

ABSTRACT

BACKGROUND: Toll-like receptor 3 (TLR3) ligand which activates TLR3 signaling induces both cancer cell death and activates anti-tumor immunity. However, TLR3 signaling can also harbor pro-tumorigenic consequences. Therefore, we examined the status of TLR3 in cholangiocarcinoma (CCA) cases to better understand TLR3 signaling and explore the potential therapeutic target in CCA. METHODS: The expression of TLR3 and receptor-interacting protein kinase 1 (RIPK1) in primary CCA tissues was assayed by Immunohistochemical staining and their associations with clinicopathological characteristics and survival data were evaluated. The effects of TLR3 ligand, Poly(I:C) and Smac mimetic, an IAP antagonist on CCA cell death and invasion were determined by cell death detection methods and Transwell invasion assay, respectively. Both genetic and pharmacological inhibition of RIPK1, RIPK3 and MLKL and inhibitors targeting NF-κB and MAPK signaling were used to investigate the underlying mechanisms. RESULTS: TLR3 was significantly higher expressed in tumor than adjacent normal tissues. We demonstrated in a panel of CCA cell lines that TLR3 was frequently expressed in CCA cell lines, but was not detected in a nontumor cholangiocyte. Subsequent in vitro study demonstrated that Poly(I:C) specifically induced CCA cell death, but only when cIAPs were removed by Smac mimetic. Cell death was also switched from apoptosis to necroptosis when caspases were inhibited in CCA cells-expressing RIPK3. In addition, RIPK1 was required for Poly(I:C) and Smac mimetic-induced apoptosis and necroptosis. Of particular interest, high TLR3 or low RIPK1 status in CCA patients was associated with more invasiveness. In vitro invasion demonstrated that Poly(I:C)-induced invasion through NF-κB and MAPK signaling. Furthermore, the loss of RIPK1 enhanced Poly(I:C)-induced invasion and ERK activation in vitro. Smac mimetic also reversed Poly(I:C)-induced invasion, partly mediated by RIPK1. Finally, a subgroup of patients with high TLR3 and high RIPK1 had a trend toward longer disease-free survival (p = 0.078, 28.0 months and 10.9 months). CONCLUSION: RIPK1 plays a pivotal role in TLR3 ligand, Poly(I:C)-induced cell death when cIAPs activity was inhibited and loss of RIPK1 enhanced Poly(I:C)-induced invasion which was partially reversed by Smac mimetic. Our results suggested that TLR3 ligand in combination with Smac mimetic could provide therapeutic benefits to the patients with CCA. Video abstract.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Mitochondrial Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Toll-Like Receptor 3/metabolism , Aged , Caspase 8/metabolism , Cell Death/drug effects , Cell Line, Tumor , Enzyme Activation/drug effects , Female , Humans , Ligands , Male , Models, Biological , Necroptosis/drug effects , Neoplasm Invasiveness , Poly I-C/pharmacology , Survival Analysis
7.
PLoS One ; 15(1): e0227454, 2020.
Article in English | MEDLINE | ID: mdl-31914150

ABSTRACT

Cholangiocarcinoma (CCA), a malignant tumor originating in the biliary tract, is well known to be associated with adverse clinical outcomes and high mortality rates due to the lack of effective therapy. Evasion of apoptosis is considered a key contributor to therapeutic success and chemotherapy resistance in CCA, highlighting the need for novel therapeutic strategies. In this study, we demonstrated that the induction of necroptosis, a novel regulated form of necrosis, could potentially serve as a novel therapeutic approach for CCA patients. The RNA sequencing data in The Cancer Genome Atlas (TCGA) database were analyzed and revealed that both receptor-interacting protein kinase 3 (RIPK3) and mixed lineage kinase domain-like (MLKL), two essential mediators of necroptosis, were upregulated in CCA tissues when compared with the levels in normal bile ducts. We demonstrated in a panel of CCA cell lines that RIPK3 was differentially expressed in CCA cell lines, while MLKL was more highly expressed in CCA cell lines than in nontumor cholangiocytes. We therefore showed that treatment with both tumor necrosis factor-α (TNF-α) and Smac mimetic, an inhibitor of apoptosis protein (IAP) antagonist, induced RIPK1/RIPK3/MLKL-dependent necroptosis in CCA cells when caspases were blocked. The necroptotic induction in a panel of CCA cells was correlated with RIPK3 expression. Intriguingly, we demonstrated that Smac mimetic sensitized CCA cells to a low dose of standard chemotherapy, gemcitabine, and induced necroptosis in an RIPK1/RIPK3/MLKL-dependent manner upon caspase inhibition but not in nontumor cholangiocytes. We further demonstrated that Smac mimetic and gemcitabine synergistically induced an increase in TNF-α mRNA levels and that Smac mimetic reversed gemcitabine-induced cell cycle arrest, leading to cell killing. Collectively, our present study demonstrated that TNF-α and gemcitabine induced RIPK1/RIPK3/MLKL-dependent necroptosis upon IAP depletion and caspase inhibition; therefore, our findings have pivotal implications for designing a novel necroptosis-based therapeutic strategy for CCA patients.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Deoxycytidine/analogs & derivatives , Necroptosis/drug effects , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Triazoles/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Biliary Tract Neoplasms/metabolism , Biliary Tract Neoplasms/pathology , Caspase Inhibitors/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Deoxycytidine/pharmacology , Drug Synergism , Humans , Protein Kinases/chemistry , Protein Kinases/genetics , Protein Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Gemcitabine
8.
Transl Oncol ; 13(1): 32-41, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31760267

ABSTRACT

BACKGROUND: Escaping cell death pathways is an important event during carcinogenesis. We previously identified anti-TNFα-induced apoptosis (ATIA, also known as vasorin) as an antiapoptotic factor that suppresses reactive oxygen species (ROS) production. However, the role of vasorin in lung carcinogenesis has not been investigated. METHODS: Vasorin expression was examined in human lung cancer tissues with immunohistochemistry and database analysis. Genetic and pharmacological approaches were used to manipulate protein expression and autophagy activity in human bronchial epithelial cells (HBECs). ROS generation was measured with fluorescent indicator, apoptosis with release of lactate dehydrogenase, and cell transformation was assessed with colony formation in soft agar. RESULTS: Vasorin expression was increased in human lung cancer tissues and cell lines, which was inversely associated with lung cancer patient survival. Cigarette smoke extract (CSE) and benzo[a]pyrene diol epoxide (BPDE)-induced vasorin expression in HBECs. Vasorin knockdown in HBECs significantly suppressed CSE-induced transformation in association with enhanced ROS accumulation and autophagy. Scavenging ROS attenuated autophagy and cytotoxicity in vasorin knockdown cells, suggesting that vasorin potentiates transformation by impeding ROS-mediated CSE cytotoxicity and improving survival of the premalignant cells. Suppression of autophagy effectively inhibited CSE-induced apoptosis, suggesting that autophagy was pro-apoptotic in CSE-treated cells. Importantly, blocking autophagy strongly potentiated CSE-induced transformation. CONCLUSION: These results suggest that vasorin is a potential lung cancer-promoting factor that facilitates cigarette smoke-induced bronchial epithelial cell transformation by suppressing autophagy-mediated apoptosis, which could be exploited for lung cancer prevention.

9.
iScience ; 17: 74-86, 2019 Jul 26.
Article in English | MEDLINE | ID: mdl-31255985

ABSTRACT

DNA-damaging compounds, commonly used as chemotherapeutic drugs, are known to trigger cells to undergo programmed cell death such as apoptosis and necroptosis. However, the molecular mechanism of DNA damage-induced cell death is not fully understood. Here, we report that RARγ has a critical role in DNA damage-induced programmed cell death, specifically in necroptosis. The loss of RARγ abolishes the necroptosis induced by DNA damage. In addition, cells that lack RARγ are less susceptible to extrinsic apoptotic pathway activated by DNA-damaging agents whereas the intrinsic apoptotic pathway is not affected. We demonstrate that RARγ is essential for the formation of RIPK1/RIPK3 death complex, known as Ripoptosome, in response to DNA damage. Furthermore, we show that RARγ plays a role in skin cancer development by using RARγ1 knockout mice and human squamous cell carcinoma biopsies. Hence, our study reveals that RARγ is a critical component of DNA damage-induced cell death.

10.
Sci Rep ; 9(1): 1759, 2019 02 11.
Article in English | MEDLINE | ID: mdl-30741975

ABSTRACT

The inflammatory response, modulated both by tissue resident macrophages and recruited monocytes from peripheral blood, plays a critical role in human diseases such as cancer and neurodegenerative disorders. Here, we sought a model to interrogate human immune behavior in vivo. We determined that primary human monocytes and macrophages survive in zebrafish for up to two weeks. Flow cytometry revealed that human monocytes cultured at the physiological temperature of the zebrafish survive and differentiate comparable to cohorts cultured at human physiological temperature. Moreover, key genes that encode for proteins that play a role in tissue remodeling were also expressed. Human cells migrated within multiple tissues at speeds comparable to zebrafish macrophages. Analysis of gene expression of in vivo educated human macrophages confirmed expression of activated macrophage phenotypes. Here, human cells adopted phenotypes relevant to cancer progression, suggesting that we can define the real time immune modulation of human tumor cells during the establishment of a metastatic lesion in zebrafish.


Subject(s)
Genotype , Macrophage Activation/genetics , Macrophage Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Animals , Biomarkers , Cell Survival/genetics , Cell Survival/immunology , Humans , Phenotype , Zebrafish
12.
Mol Cell Oncol ; 5(1): e1392402, 2018.
Article in English | MEDLINE | ID: mdl-29404390

ABSTRACT

TNFR1-mediated cell signaling involves complex molecular pathways leading to inflammation and death. Cytosolic RARγ plays a pivotal role in converting TNF-induced inflammatory responses to RIP1 initiated cell death and this finely regulated function of RARγ serves as a checkpoint to engage death pathways in response to TNF.

13.
Nat Commun ; 8(1): 425, 2017 09 04.
Article in English | MEDLINE | ID: mdl-28871172

ABSTRACT

Tumor necrosis factor (TNF) has a critical role in diverse cellular events including inflammation, apoptosis and necroptosis through different signaling complexes. However, little is known about how the transition from inflammatory signaling to the engagement of death pathways is modulated. Here we report that the cytoplasmic retinoic acid receptor gamma (RARγ) controls receptor-interacting protein kinase 1 (RIP1)-initiated cell death when cellular inhibitor of apoptosis (cIAP) activity is blocked. Through screening a short hairpin RNA library, we found that RARγ was essential for TNF-induced RIP1-initiated apoptosis and necroptosis. Our data suggests that RARγ initiates the formation of death signaling complexes by mediating RIP1 dissociation from TNF receptor 1. We demonstrate that RARγ is released from the nucleus to orchestrate the formation of the cytosolic death complexes. In addition, we demonstrate that RARγ has a similar role in TNF-induced necroptosis in vivo. Thus, our study suggests that nuclear receptor RARγ provides a key checkpoint for the transition from life to death.The molecular switch between how tumour necrosis factor (TNF) controls inflammation versus cell death is less well defined. Here, the authors show that the nuclear receptor retinoic acid receptor gamma is released from the nucleus to disrupt TNF initiated cell death complexes in the cytoplasm.


Subject(s)
Cell Nucleus/metabolism , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/metabolism , Receptors, Retinoic Acid/metabolism , Animals , Apoptosis/drug effects , Cell Line , Cytoprotection/drug effects , Cytosol/drug effects , Cytosol/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Mice, Knockout , Models, Biological , Receptors, Tumor Necrosis Factor/metabolism , TNF Receptor-Associated Death Domain Protein/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Retinoic Acid Receptor gamma
14.
PLoS Genet ; 12(8): e1006244, 2016 08.
Article in English | MEDLINE | ID: mdl-27541266

ABSTRACT

Cancer-associated fibroblasts (CAFs), the most common constituent of the tumor stoma, are known to promote tumor initiation, progression and metastasis. However, the mechanism of how cancer cells transform normal fibroblasts (NFs) into CAFs is largely unknown. In this study, we determined the contribution of miRNAs in the transformation of NFs into CAFs. We found that miR-1 and miR-206 were down-regulated, whereas miR-31 was up-regulated in lung CAFs when compared with matched NFs. Importantly, modifying the expression of these three deregulated miRNAs induced a functional conversion of NFs into CAFs and vice versa. When the miRNA-reprogrammed NFs and CAFs were co-cultured with lung cancer cells (LCCs), a similar pattern of cytokine expression profiling were observed between two groups. Using a combination of cytokine expression profiling and miRNAs algorithms, we identified VEGFA/CCL2 and FOXO3a as direct targets of miR-1, miR-206 and miR-31, respectively. Importantly, systemic delivery of anti-VEGFA/CCL2 or pre-miR-1, pre-miR-206 and anti-miR-31 significantly inhibited tumor angiogenesis, TAMs accumulation, tumor growth and lung metastasis. Our results show that miRNAs-mediated FOXO3a/VEGF/CCL2 signaling plays a prominent role in LCCs-mediated NFs into CAFs, which may have clinical implications for providing novel biomarker(s) and potential therapeutic target(s) of lung cancer in the future.


Subject(s)
Chemokine CCL2/biosynthesis , Lung Neoplasms/genetics , MicroRNAs/biosynthesis , Vascular Endothelial Growth Factor A/biosynthesis , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Cellular Reprogramming/genetics , Chemokine CCL2/genetics , Forkhead Box Protein O3/biosynthesis , Forkhead Box Protein O3/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , MicroRNAs/genetics , Neoplasm Metastasis , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Signal Transduction/genetics , Vascular Endothelial Growth Factor A/genetics
15.
J Biol Chem ; 291(38): 20030-41, 2016 09 16.
Article in English | MEDLINE | ID: mdl-27489105

ABSTRACT

NADPH oxidases (NOXs) are involved in inflammation, angiogenesis, tumor growth, and osteoclast differentiation. However, the role of NOX1 and NOX2 in macrophage differentiation and tumor progression is still elusive. Here we report that NOX1 and NOX2 are critical for the differentiation of monocytes to macrophages, the polarization of M2-type but not M1-type macrophages, and the occurrence of tumor-associated macrophages (TAMs). We found that deletion of both NOX1 and NOX2 led to a dramatic decrease in ROS production in macrophages and resulted in impaired efficiency in monocyte-to-macrophage differentiation and M2-type macrophage polarization. We further showed that NOX1 and NOX2 were critical for the activation of the MAPKs JNK and ERK during macrophage differentiation and that the deficiency of JNK and ERK activation was responsible for the failure of monocyte-to-macrophage differentiation, in turn affecting M2 macrophage polarization. Furthermore, we demonstrated that the decrease in M2 macrophages and TAMs, concomitant with the reduction of cytokine and chemokine secretion, contributed to the delay in wound healing and the inhibition of tumor growth and metastasis in NOX1/2 double knockout mice compared with WT mice. Collectively, these data provide direct evidence that NOX1 and NOX2 deficiency impairs macrophage differentiation and the occurrence of M2-type TAMs during tumor development.


Subject(s)
Cell Differentiation/immunology , Macrophages/immunology , Membrane Glycoproteins/immunology , Monocytes/immunology , NADH, NADPH Oxidoreductases/immunology , NADPH Oxidases/immunology , Reactive Oxygen Species/immunology , Animals , Cell Differentiation/genetics , Chemokines/genetics , Chemokines/immunology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/immunology , Macrophages/enzymology , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Monocytes/enzymology , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , NADPH Oxidase 1 , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism
16.
Cell Res ; 26(8): 886-900, 2016 08.
Article in English | MEDLINE | ID: mdl-27444869

ABSTRACT

Necroptosis is a programmed, caspase-independent cell death that is morphologically similar to necrosis. TNF-induced necroptosis is mediated by receptor-interacting protein kinases, RIP1 and RIP3, and the mixed lineage kinase domain-like (MLKL). After being phosphorylated by RIP3, MLKL is translocated to the plasma membrane and mediates necroptosis. However, the execution of necroptosis and its role in inflammation and other cellular responses remain largely elusive. In this study, we report that MLKL-mediated activation of cell-surface proteases of the a disintegrin and metalloprotease (ADAM) family promotes necroptosis, inflammation and cell migration. ADAMs are specifically activated at the early stage of necroptosis when MLKL is phosphorylated and translocated to the cell plasma membrane. Activation of ADAMs induces ectodomain shedding of diverse cell-surface proteins including adhesion molecules, receptors, growth factors and cytokines. Importantly, the shedding of cell-surface proteins disrupts cell adhesion and accelerates necroptosis, while the soluble fragments of the cleaved proteins trigger the inflammatory responses. We also demonstrate that the shedding of E-cadherin ectodomain from necroptotic cells promotes cell migration. Thus, our study provides a novel mechanism of necroptosis-induced inflammation and new insights into the physiological and pathological functions of this unique form of cell death.


Subject(s)
Apoptosis , Cell Membrane/enzymology , Cell Movement , Inflammation/pathology , Peptide Hydrolases/metabolism , ADAM Proteins/metabolism , Animals , Cadherins/metabolism , Enzyme Activation , HEK293 Cells , HT29 Cells , Humans , Mice, Inbred C57BL , Necrosis , Neoplasm Invasiveness , Protein Kinases/metabolism
17.
Methods Mol Biol ; 1155: 15-22, 2014.
Article in English | MEDLINE | ID: mdl-24788169

ABSTRACT

Receptor signaling can be evaluated in multiple ways, including analysis of phosphorylation of downstream molecules and analysis of proteins that are recruited to the receptor upon ligand binding. Majority of studies on the mechanism of DR3 signaling were performed using overexpression systems that can often lead to artifacts. In this chapter we describe how to analyze DR3 downstream events with most attention being paid to endogenous immunoprecipitation.


Subject(s)
Blotting, Western/methods , Immunoprecipitation/methods , Receptors, Tumor Necrosis Factor, Member 25/metabolism , Fluoresceins , Fluorescent Dyes , Humans , Lymphocyte Activation/drug effects , NF-kappa B/genetics , NF-kappa B/metabolism , Phosphorylation , Receptors, Tumor Necrosis Factor, Member 25/genetics , Signal Transduction , Succinimides
18.
Nat Cell Biol ; 16(1): 55-65, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24316671

ABSTRACT

The mixed lineage kinase domain-like protein (MLKL) has recently been identified as a key RIP3 (receptor interacting protein 3) downstream component of tumour necrosis factor (TNF)-induced necroptosis. MLKL is phosphorylated by RIP3 and is recruited to the necrosome through its interaction with RIP3. However, it is still unknown how MLKL mediates TNF-induced necroptosis. Here, we report that MLKL forms a homotrimer through its amino-terminal coiled-coil domain and locates to the cell plasma membrane during TNF-induced necroptosis. By generating different MLKL mutants, we demonstrated that the plasma membrane localization of trimerized MLKL is critical for mediating necroptosis. Importantly, we found that the membrane localization of MLKL is essential for Ca(2+) influx, which is an early event of TNF-induced necroptosis. Furthermore, we identified that TRPM7 (transient receptor potential melastatin related 7) is a MLKL downstream target for the mediation of Ca(2+) influx and TNF-induced necroptosis. Hence, our study reveals a crucial mechanism of MLKL-mediated TNF-induced necroptosis.


Subject(s)
Apoptosis/drug effects , Cell Membrane/metabolism , Necrosis/pathology , Protein Kinases/metabolism , Protein Multimerization , Tumor Necrosis Factor-alpha/pharmacology , Animals , Calcium/metabolism , Cell Membrane/drug effects , HEK293 Cells , HT29 Cells , Humans , Imidazoles/pharmacology , Indoles/pharmacology , Jurkat Cells , Mice , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Kinases/chemistry , Protein Multimerization/drug effects , Protein Serine-Threonine Kinases , Protein Structure, Tertiary , Protein Transport/drug effects , RNA, Small Interfering/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , TRPM Cation Channels/metabolism
19.
Cancers (Basel) ; 5(4): 1643-54, 2013 Dec 04.
Article in English | MEDLINE | ID: mdl-24305654

ABSTRACT

Tumor-associated macrophages (TAMs) promote tumorigenesis because of their proangiogenic and immune-suppressive functions. Here, we report that butylated hydroxyanisole (BHA) blocks occurrence of tumor associated macrophages (TAMs) in tobacco smoke carcinogen-induced lung tumorigenesis. Continuous administration of butylated hydroxyanisole (BHA), a ROS inhibitor, before or after NNK treatment significantly blocked tumor development, although less effectively when BHA is administered after NNK treatment. Strikingly, BHA abolished the occurrence of F4/80+ macrophages with similar efficiency no matter whether it was administered before or after NNK treatment. Detection of cells from bronchioalveolar lavage fluid (BALF) confirmed that BHA markedly inhibited the accumulation of macrophages while slightly reducing the number of lymphocytes that were induced by NNK. Immunohistological staining showed that BHA specifically abolished the occurrence of CD206+ TAMs when it was administered before or after NNK treatment. Western blot analysis of TAMs markers, arginase I and Ym-1, showed that BHA blocked NNK-induced TAMs accumulation. Our study clearly demonstrated that inhibiting the occurrence of TAMs by BHA contributes to the inhibition of tobacco smoke carcinogen-induced tumorigenesis, suggesting ROS inhibitors may serve as a therapeutic target for treating smoke-induced lung cancer.

20.
Cell Res ; 23(7): 898-914, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23752925

ABSTRACT

Differentiation to different types of macrophages determines their distinct functions. Tumor-associated macrophages (TAMs) promote tumorigenesis owing to their proangiogenic and immune-suppressive functions similar to those of alternatively activated (M2) macrophages. We report that reactive oxygen species (ROS) production is critical for macrophage differentiation and that inhibition of superoxide (O(2-)) production specifically blocks the differentiation of M2 macrophages. We found that when monocytes are triggered to differentiate, O(2-) is generated and is needed for the biphasic ERK activation, which is critical for macrophage differentiation. We demonstrated that ROS elimination by butylated hydroxyanisole (BHA) and other ROS inhibitors blocks macrophage differentiation. However, the inhibitory effect of ROS elimination on macrophage differentiation is overcome when cells are polarized to classically activated (M1), but not M2, macrophages. More importantly, the continuous administration of the ROS inhibitor BHA efficiently blocked the occurrence of TAMs and markedly suppressed tumorigenesis in mouse cancer models. Targeting TAMs by blocking ROS can be a potentially effective method for cancer treatment.


Subject(s)
Macrophages/metabolism , Reactive Oxygen Species/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cell Movement/genetics , Cell Movement/physiology , Cell Proliferation , Cells, Cultured , Female , HeLa Cells , Humans , Inflammation/genetics , Inflammation/metabolism , Mice , Mice, Nude , Monocytes/metabolism , Superoxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...