Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Int J Mol Sci ; 23(23)2022 Nov 25.
Article in English | MEDLINE | ID: mdl-36499060

ABSTRACT

Eryptosis is a physiological mechanism for the clearance of senescent or damaged erythrocytes by phagocytes. Excessive eryptosis is stimulated under several pathologies and associated with endothelial injury and thrombosis. Cigarette smoke (CS) is an established risk factor for vascular diseases and cigarette smokers have high-levels of eryptotic erythrocytes. This study, for the first time, investigates the mechanism by which CS damages red blood cells (RBCs). CS extract (CSE) from commercial cigarettes was prepared and standardized for nicotine content. Cytofluorimetric analysis demonstrated that treatment of human RBCs with CSE caused dose-dependent, phosphatidylserine externalization and cell shrinkage, hallmarks of apoptotic death. CSE did not affect cellular levels of Ca2+, reactive oxygen species (ROS) or glutathione (GSH). Immununoprecipitation and immunoblotting revealed the assembly of the death-inducing signaling complex (DISC) and oligomerization of Fas receptor as well as cleaved caspase-8 and caspase-3 within 6 h from the treatment. At the same time-interval, CSE elicited neutral sphyngomielinase (nSMase) activity-dependent ceramide formation and phosphorylation of p38 MAPK. Through specific inhibitors' nSMase, caspase-8 or p38 MAPK activities, we demonstrated that p38 MAPK activation is required for caspase-8-mediated eryptosis and that ceramide generation is initiator caspase-dependent. Finally, ex vivo analysis detected phosphorylated p38 MAPK (p-p38) and Fas-associated signaling complex in erythrocytes from cigarette smokers. In conclusion, our study demonstrates that CSE exposure induces in erythrocytes an extrinsic apoptotic pathway involving p38 MAPK-initiated DISC formation followed by activation of caspase-8/caspase-3 via ceramide formation.


Subject(s)
Eryptosis , Smoke , p38 Mitogen-Activated Protein Kinases , Humans , Caspase 3/metabolism , Caspase 8/metabolism , Ceramides/metabolism , Erythrocytes/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Nicotiana/adverse effects , Smoke/adverse effects
2.
Clin Immunol ; 191: 94-99, 2018 06.
Article in English | MEDLINE | ID: mdl-29108854

ABSTRACT

Smoke inhalation leads to acute lung injury (ALI), a devastating clinical problem associated with high mortality. Suppressor of cytokine signaling-1 (SOCS-1) is a negative regulator of apoptosis and pro-inflammatory cytokine signaling, two major contributors to the pathogenesis of ALI. We have found that SOCS-1 protects lung epithelial cells from smoke-induced apoptosis through two mechanisms. One is that SOCS-1 enhances degradation of ASK-1 and diminishes cleavage of pro-caspase-3 to repress smoke-triggered apoptosis in lung epithelial cells. The other is that SOCS-1 represses smoke-triggered DISC formation through altering TRADD-caspase-8 interaction rather than TNFR-1-TRADD interaction or TNFR-1-TRAF-2 interaction. In conclusion, SOCS-1 relieves smoke inhalation-induced lung injury by repressing ASK-1 and DISC-mediated epithelium apoptosis.


Subject(s)
Acute Lung Injury/prevention & control , Death Domain Receptor Signaling Adaptor Proteins/antagonists & inhibitors , MAP Kinase Kinase Kinase 5/antagonists & inhibitors , Smoke Inhalation Injury/prevention & control , Suppressor of Cytokine Signaling 1 Protein/physiology , Apoptosis , Caspase 8/physiology , Cells, Cultured , Humans , Lung/pathology , TNF Receptor-Associated Death Domain Protein/physiology , TNF Receptor-Associated Factor 2/physiology
3.
Molecules ; 23(12)2018 Dec 13.
Article in English | MEDLINE | ID: mdl-30551620

ABSTRACT

The natural product 23-hydroxyursolic acid (23-HUA) is a derivative of ursolic acid, which is known to induce cancer cell apoptosis. However, apoptotic effects and mechanisms of 23-HUA have not been well characterized yet. Herein, we investigated the molecular mechanisms of 23-HUA-induced apoptosis in HL-60 human promyelocytic leukemia cells. 23-HUA-treated HL-60 cells showed apoptotic features including internucleosomal DNA condensation and fragmentation as well as externalization of phosphatidylserine residues. 23-HUA induced a series of mitochondrial events including disruption of mitochondrial membrane potential (ΔΨm), cytochrome c and Smac/DIABLO release and loss of balance between pro-apoptotic and anti-apoptotic Bcl-2 proteins in HL-60 cells. In addition, 23-HUA activated caspase-8, caspase-9 and caspase-3. Pretreatment with a broad caspase inhibitor (z-VAD-fmk), a caspase-3 inhibitor (z-DEVD-fmk), and a caspase-8 inhibitor (z-IETD-fmk) significantly attenuated 23-HUA-induced DNA fragmentation. After 23-HUA-induced apoptosis, proteins expression levels of FasL, Fas and FADD constituting the death-inducing signaling complex (DISC) were upregulated in HL-60 cells. Moreover, transfection with Fas or FADD siRNA significantly blocked 23-HUA-induced DNA fragmentation and caspases activation. Taken together, these findings indicate that 23-HUA induces apoptosis in HL-60 human promyelocytic leukemia cells through formation of DISC and caspase-8 activation leading to loss of ΔΨm and caspase-3 activation.


Subject(s)
Apoptosis/drug effects , Araliaceae/chemistry , Caspase 8/metabolism , Leukemia, Promyelocytic, Acute/pathology , Plant Bark/chemistry , Plant Stems/chemistry , Triterpenes/pharmacology , fas Receptor/metabolism , Cell Proliferation/drug effects , Death Domain Receptor Signaling Adaptor Proteins/metabolism , HL-60 Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Models, Biological , Triterpenes/chemistry , Triterpenes/isolation & purification
4.
Structure ; 30(2): 229-239.e5, 2022 02 03.
Article in English | MEDLINE | ID: mdl-34800372

ABSTRACT

Cellular FLICE-like inhibitory protein (cFLIP) is a member of the Death Domain superfamily with pivotal roles in many cellular processes and disease states, including cancer and autoimmune disorders. In the context of the death-inducing signaling complex (DISC), cFLIP isoforms regulate extrinsic apoptosis by controlling procaspase-8 activation. The function of cFLIP is mediated through a series of protein-protein interactions, engaging the two N-terminal death effector domains (DEDs). Here, we solve the structure of an engineered DED1 domain of cFLIP using solution nuclear magnetic resonance (NMR) and we define the interaction with FADD and calmodulin, protein-protein interactions that regulate the function of cFLIP in the DISC. cFLIP DED1 assumes a canonical DED fold characterized by six α helices and is able to bind calmodulin and FADD through two separate interfaces. Our results clearly demonstrate the role of DED1 in the cFLIP/FADD association and contribute to the understanding of the assembly of DISC filaments.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/chemistry , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Calmodulin/metabolism , Fas-Associated Death Domain Protein/metabolism , Protein Engineering/methods , Binding Sites , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Circular Dichroism , Magnetic Resonance Spectroscopy , Models, Molecular , Protein Binding , Protein Domains , Protein Interaction Maps , Protein Structure, Secondary
5.
Methods Mol Biol ; 1857: 101-107, 2018.
Article in English | MEDLINE | ID: mdl-30136234

ABSTRACT

FAS-associated protein with death domain (FADD) is a signaling molecule required by members of the TNF receptor superfamily (TNFRSF) such as FAS and TNFR1 to induce apoptosis. FADD is a small adapter molecule that functions as a scaffold to recruit procaspase-8 and other regulators. The FADD-containing signaling complex that initiates the apoptotic cascade has been termed the death inducing signaling complex (DISC). In the absence of FADD, death receptors cannot induce apoptosis and in appropriate cell types, these death receptors then induce necroptosis. Necroptosis can also be induced by death receptors in FADD-sufficient cells when caspase-8 is inhibited, usually accomplished by the addition of caspase inhibitors. Under such necroptotic conditions, the immunoprecipitation of FADD to isolate the DISC can be utilized to examine components of this complex. Here, we describe the immunoprecipitation of FADD and subsequent western-blotting to identify RIPK1 in this complex during necroptosis.


Subject(s)
Apoptosis , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Embryo, Mammalian/pathology , Fas-Associated Death Domain Protein/metabolism , Fibroblasts/pathology , Necrosis , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Cells, Cultured , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Mice , Signal Transduction
6.
Methods Mol Biol ; 1557: 41-48, 2017.
Article in English | MEDLINE | ID: mdl-28078581

ABSTRACT

Distribution of the death receptor CD95 into lipid rafts (aggregation) and/or its internalization may contribute to the implementation of the apoptotic signal at the detriment of the non-apoptotic signaling pathway [1-6]. Also CD95 can form different protein complexes via dynamic protein-protein interactions (PPIs) according to its interaction with soluble or transmembrane CD95L. Therefore, spatiotemporal identification of these PPIs is pivotal to anticipate the signaling pathway implemented in cells stimulated with different forms of CD95L. Also, many disorders result from dysfunctions in terms of PPI subcellular distribution and/or their intensity, rendering evaluation of these features crucial to better understand pathogenesis.In situ proximity ligation assay (PLA) is a methodology that offers the possibility to identify PPIs and to determine where these PPIs occur in subcellular location (Fig. 1). Moreover, based on imaging, this method allows a quantification of PPIs at the cellular level and with a higher specificity than classical immunofluorescence assays. We here describe PLA used to confirm CD95/FADD interaction, a protocol that may serve to highlight other CD95 partners.


Subject(s)
Biological Assay/methods , Fas-Associated Death Domain Protein/metabolism , Multiprotein Complexes/metabolism , Signal Transduction , Apoptosis , Fas Ligand Protein/metabolism , Humans , Jurkat Cells , Ligands , Protein Binding , fas Receptor/metabolism
7.
Neurol Res ; 39(1): 54-64, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27832728

ABSTRACT

OBJECTIVES: Apoptosis plays an important role in the progression of the ischemic penumbra after reperfusion. Estrogen and progesterone have neuroprotective effects against ischemic brain damage, however the exact mechanisms of neuroprotection and signaling pathways is not completely understood. In this study, we investigated the possible regulatory effects of a combined steroid treatment on extrinsic and intrinsic apoptotic signaling pathways after cerebral ischemia. METHODS: Adult male Wistar rats were subjected to transient middle cerebral artery occlusion (tMCAO) using an intraluminal filament technique for 1 h followed by 23 h reperfusion. Estrogen and progesterone were immediately injected after tMCAO subcutaneously. Sensorimotor functional tests and the infarct volume were evaluated 24 h after ischemia. Protein expression of calpain-1 and Fas receptor (FasR), key members of intrinsic and extrinsic apoptosis, were determined in the penumbra region of the ischemic brain using western blot analysis, immunohistochemistry, and TUNEL staining. RESULTS: Neurological deficits and infarct volume were significantly reduced following hormone therapy. Calpain-1 up-regulation and caspase-3 activation were apparent 24 h after ischemia in the peri-infarct area of the cerebral cortex. Steroid hormone treatment reduced infarct pathology and attenuated the induction of both proteases. FasR protein levels were not affected by ischemia and hormone application. CONCLUSION: We conclude that a combined steroid treatment inhibits ischemia-induced neuronal apoptosis through the regulation of intrinsic pathways.


Subject(s)
Apoptosis/drug effects , Calpain/metabolism , Infarction, Middle Cerebral Artery , Signal Transduction/physiology , Steroids/therapeutic use , Animals , Brain Infarction/drug therapy , Brain Infarction/etiology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cerebrovascular Circulation/drug effects , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , In Situ Nick-End Labeling , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/drug therapy , Infarction, Middle Cerebral Artery/pathology , Laser-Doppler Flowmetry , Male , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Wistar , Signal Transduction/drug effects , Statistics, Nonparametric
8.
Mutat Res ; 777: 11-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25912078

ABSTRACT

UNLABELLED: The tumor necrosis factor (TNF) signaling pathway is a classical immune system pathway that plays a key role in regulating cell survival and apoptosis. The TNF receptor-associated death domain (TRADD) protein is recruited to the death domain of TNF receptor 1 (TNFR1), where it interacts with TNF receptor-associated factor 2 (TRAF2) and receptor-interacting protein (RIP) for the induction of apoptosis, necrosis, nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB), and mitogen-activated protein (MAP) kinase activation. In this study, we found that the human MutY homolog (hMYH) interacted with human TRADD (hTRADD) via the C-terminal domain of hMYH. Moreover, under conditions promoting TNF-α-induced cell death or survival in HeLa cells, this interaction was weakened or enhanced, respectively. The interaction between hMYH and hTRADD was important for signaling pathways mediated by TNF-α. Our results also suggested that the hTRADD-hMYH association was involved in the nuclear translocation of NFκB and formation of the TNFR1-TRADD complex. Thus, this study identified a novel mechanism through which the hMYH-hTRADD interaction may affect the TNF-α signaling pathway. IMPLICATIONS: In HeLa cells, the hTRADD-hMYH interaction functioned in both cell survival and apoptosis pathways following TNF-α stimulation.


Subject(s)
DNA Glycosylases/metabolism , TNF Receptor-Associated Death Domain Protein/metabolism , Tumor Necrosis Factor-alpha/metabolism , Apoptosis , DNA Glycosylases/genetics , HeLa Cells , Humans , NF-kappa B/genetics , NF-kappa B/metabolism , Necrosis , Protein Interaction Domains and Motifs , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Signal Transduction , TNF Receptor-Associated Death Domain Protein/genetics , Tumor Necrosis Factor-alpha/genetics
9.
Autophagy ; 11(2): 214-24, 2015.
Article in English | MEDLINE | ID: mdl-25607248

ABSTRACT

Both apoptosis ("self-killing") and autophagy ("self-eating") are evolutionarily conserved processes, and their crosstalk influences anticancer drug sensitivity and cell death. However, the underlying mechanism remains unclear. Here, we demonstrated that HMGB1 (high mobility group box 1), normally a nuclear protein, is a crucial regulator of TNFSF10/TRAIL (tumor necrosis factor [ligand] superfamily, member 10)-induced cancer cell death. Activation of PARP1 (poly [ADP-ribose] polymerase 1) was required for TNFSF10-induced ADP-ribosylation of HMGB1 in cancer cells. Moreover, pharmacological inhibition of PARP1 activity or knockdown of PARP1 gene expression significantly inhibited TNFSF10-induced HMGB1 cytoplasmic translocation and subsequent HMGB1-BECN1 complex formation. Furthermore, suppression of the PARP1-HMGB1 pathway diminished autophagy, increased apoptosis, and enhanced the anticancer activity of TNFSF10 in vitro and in a subcutaneous tumor model. These results indicate that PARP1 acts as a prominent upstream regulator of HMGB1-mediated autophagy and maintains a homeostatic balance between apoptosis and autophagy, which provides new insight into the mechanism of TNFSF10 resistance.


Subject(s)
Autophagy/physiology , HMGB1 Protein/metabolism , Poly(ADP-ribose) Polymerases/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Humans , Mice , Poly (ADP-Ribose) Polymerase-1 , Signal Transduction/physiology
10.
Cancer Biol Ther ; 16(6): 821-33, 2015.
Article in English | MEDLINE | ID: mdl-25897893

ABSTRACT

Protein phosphatase 2A (PP2A) is a serine/threonine phosphatase that plays a significant role in mitotic progression and cellular responses to DNA damage. While traditionally viewed as a tumor suppressor, inhibition of PP2A has recently come to attention as a novel therapeutic means of driving senescent cancer cells into mitosis and promoting cell death via mitotic catastrophe. These findings have been corroborated in numerous studies utilizing naturally produced compounds that selectively inhibit PP2A. To overcome the known human toxicities associated with these compounds, a water-soluble small molecule inhibitor, LB100, was recently developed to competitively inhibit the PP2A protein. This review summarizes the pre-clinical studies to date that have demonstrated the anti-cancer activity of LB100 via its chemo- and radio-sensitizing properties. These studies demonstrate the tremendous therapeutic potential of LB100 in a variety of cancer types. The results of an ongoing phase 1 trial are eagerly anticipated.


Subject(s)
Antineoplastic Agents/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Enzyme Inhibitors/pharmacology , Piperazines/pharmacology , Protein Phosphatase 2/antagonists & inhibitors , Radiation-Sensitizing Agents/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm , Humans , Mitosis/drug effects , Mitosis/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Tumor Protein, Translationally-Controlled 1 , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Wnt Signaling Pathway/drug effects
11.
Acta Pharm Sin B ; 5(6): 544-53, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26713269

ABSTRACT

Fucoidan is a traditional Chinese medicine suggested to possess anti-tumor effects. In this study the anti-metastatic effects of fucoidan were investigated in vitro in human hepatocellular carcinoma (HCC) cells (Huh-7 and SNU-761) under normoxic and hypoxic conditions and in vivo using a distant liver metastasis model involving injection of MH134 cells into spleen via the portal vein. Its ability to protect hepatocytes against bile acid (BA)-induced apoptosis was investigated in primary hepatocytes. Fucoidan was found to suppress the invasion of HCC cells through up-regulation of p42/44 MAPK-dependent NDRG-1/CAP43 and partly, under normoxic conditions, through up-regulation of p42/44 MAPK-dependent VMP-1 expression. It also significantly decreased liver metastasis in vivo. As regards its hepatoprotective effect, fucoidan decreased BA-induced hepatocyte apoptosis as shown by the attenuation of caspase-8, and -7 cleavages and suppression of the mobilization of caspase-8 and Fas associated death domain (FADD) into the death-inducing signaling complex. In summary, fucoidan displays inhibitory effects on proliferation of HCC cells and protective effects on hepatocytes. The results suggest fucoidan is a potent suppressor of tumor invasion with hepatoprotective effects.

12.
Acta Pharm Sin B ; 5(5): 402-18, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26579471

ABSTRACT

Cancer metastasis is the major cause of cancer morbidity and mortality, and accounts for about 90% of cancer deaths. Although cancer survival rate has been significantly improved over the years, the improvement is primarily due to early diagnosis and cancer growth inhibition. Limited progress has been made in the treatment of cancer metastasis due to various factors. Current treatments for cancer metastasis are mainly chemotherapy and radiotherapy, though the new generation anti-cancer drugs (predominantly neutralizing antibodies for growth factors and small molecule kinase inhibitors) do have the effects on cancer metastasis in addition to their effects on cancer growth. Cancer metastasis begins with detachment of metastatic cells from the primary tumor, travel of the cells to different sites through blood/lymphatic vessels, settlement and growth of the cells at a distal site. During the process, metastatic cells go through detachment, migration, invasion and adhesion. These four essential, metastatic steps are inter-related and affected by multi-biochemical events and parameters. Additionally, it is known that tumor microenvironment (such as extracellular matrix structure, growth factors, chemokines, matrix metalloproteinases) plays a significant role in cancer metastasis. The biochemical events and parameters involved in the metastatic process and tumor microenvironment have been targeted or can be potential targets for metastasis prevention and inhibition. This review provides an overview of these metastasis essential steps, related biochemical factors, and targets for intervention.

13.
Cell Cycle ; 14(11): 1631-42, 2015.
Article in English | MEDLINE | ID: mdl-25927598

ABSTRACT

Apoptosis is a primary characteristic in the pathogenesis of liver disease. Hepatic apoptosis is regulated by autophagic activity. However, mechanisms mediating their interaction remain to be determined. Basal level of autophagy ensures the physiological turnover of old and damaged organelles. Autophagy also is an adaptive response under stressful conditions. Autophagy can control cell fate through different cross-talk signals. A complex interplay between hepatic autophagy and apoptosis determines the degree of hepatic apoptosis and the progression of liver disease as demonstrated by pre-clinical models and clinical trials. This review summarizes recent advances on roles of autophagy that plays in pathophysiology of liver. The autophagic pathway can be a novel therapeutic target for liver disease.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Liver Diseases/physiopathology , Models, Biological , Receptor Cross-Talk/physiology , Signal Transduction/physiology , Disease Progression , Humans
14.
Cancer Biol Ther ; 16(7): 1029-41, 2015.
Article in English | MEDLINE | ID: mdl-26011589

ABSTRACT

Interferon Regulatory Factor (IRF)-1, originally identified as a transcription factor of the human interferon (IFN)-ß gene, mediates tumor suppression and may inhibit oncogenesis. We have shown that IRF-1 in human breast cancer cells results in the down-regulation of survivin, tumor cell death, and the inhibition of tumor growth in vivo in xenogeneic mouse models. In this current report, we initiate studies comparing the effect of IRF-1 in human nonmalignant breast cell and breast cancer cell lines. While IRF-1 in breast cancer cells results in growth inhibition and cell death, profound growth inhibition and cell death are not observed in nonmalignant human breast cells. We show that TNF-α or IFN-γ induces IRF-1 in breast cancer cells and results in enhanced cell death. Abrogation of IRF-1 diminishes TNF-α and IFN-γ-induced apoptosis. We test the hypothesis that IRF-1 augments TNF-α-induced apoptosis in breast cancer cells. Potential signaling networks elicited by IRF-1 are investigated by evaluating the NF-κB pathway. TNF-α and/or IFN-γ results in decreased presence of NF-κB p65 in the nucleus of breast cancer cells. While TNF-α and/or IFN-γ can induce IRF-1 in nonmalignant breast cells, a marked change in NF-κB p65 is not observed. Moreover, the ectopic expression of IRF-1 in breast cancer cells results in caspase-3, -7, -8 cleavage, inhibits NF-κB activity, and suppresses the expression of molecules involved in the NF-κB pathway. These data show that IRF-1 in human breast cancer cells elicits multiple signaling networks including intrinsic and extrinsic cell death and down-regulates molecules involved in the NF-κB pathway.


Subject(s)
Inhibitor of Apoptosis Proteins/metabolism , Interferon Regulatory Factor-1/metabolism , NF-kappa B/metabolism , TNF Receptor-Associated Factor 2/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Caspases/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Humans , Immunoblotting , Interferon Regulatory Factor-1/genetics , Interferon-gamma/pharmacology , RNA Interference , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription Factor RelA/metabolism , Tumor Necrosis Factor-alpha/pharmacology
15.
Methods Enzymol ; 545: 201-42, 2014.
Article in English | MEDLINE | ID: mdl-25065892

ABSTRACT

This chapter describes reports of the structural characterization of death ligands and death receptors (DRs) from the tumor necrosis factor (TNF) and TNF receptor families. The review discusses the interactions of these proteins with agonist ligands, inhibitors, and downstream signaling molecules. Though historically labeled as being implicated in programmed cell death, the function of these proteins extends to nonapoptotic pathways. The review highlights, from a structural biology perspective, the complexity of DR signaling and the ongoing challenge to discern the precise mechanisms that occur at the point of DR activation, including how the degree to which the receptors are induced to cluster may be related to the nature of the impact upon the cell. The potential for posttranslational modification and receptor internalization to play roles in DR signaling is briefly discussed.


Subject(s)
Apoptosis/genetics , Receptors, Death Domain/chemistry , Signal Transduction , Tumor Necrosis Factor-alpha/chemistry , Crystallography, X-Ray , Humans , Ligands , Magnetic Resonance Spectroscopy , Protein Conformation , Protein Processing, Post-Translational/genetics , Receptors, Death Domain/metabolism
16.
J Ethnopharmacol ; 151(1): 292-8, 2014.
Article in English | MEDLINE | ID: mdl-24212079

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Tripterygium hypoglaucum (levl.) Hutch (Celastraceae) (THH) root is a traditional Chinese medicinal herb commonly used for treating autoimmune diseases and cancer. Alkaloid is one of the most bioactive components of THH extract. To evaluate the in vitro and in vivo antitumor properties of the total alkaloids of THH (THHta). MATERIALS AND METHODS: THHta was extracted in pilot-scale. HCT116 cells were chose to establish human colon cancer xenograft model. The in vitro anti-tumor activity of THHta was tested by Cell malignant transformation test, Soft agar colony formation assay and MTT assay. The in vivo anti-tumor effect of THHta was confirmed by xenograft mouse model. THHta-induced apoptosis was examined by flow cytometry. The levels of apoptosis-related proteins were investigated by Western blot. RESULTS: TPA-induced cell transformation was significantly inhibited by THHta in JB6 Cl41 cells. THHta inhibits the growth of colon cancer cells in vitro in a significant dose-dependent manner. Compared to the control set, i.p. administration of THHta to xenograft mice significantly reduced both tumor weight and volume. Apoptosis induction of THHta was mediated by activation of caspase-3, PARP and inhibiting of Bcl-2, Bcl-xL and XIAP. CONCLUSION: THHta was effective in inhibiting tumor growth both in vitro and in vivo at less toxic concentrations by inducing apoptosis which suggested it could be developed as a potential anticancer agent.


Subject(s)
Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Tripterygium/chemistry , Alkaloids/chemistry , Animals , Antineoplastic Agents/chemistry , Apoptosis , Cell Survival/drug effects , Dose-Response Relationship, Drug , HCT116 Cells , Humans , Male , Mice , Mice, Nude , Neoplasms, Experimental/drug therapy , Plant Roots/chemistry
17.
MAbs ; 6(6): 1560-70, 2014.
Article in English | MEDLINE | ID: mdl-25484045

ABSTRACT

Multiple therapeutic agonists of death receptor 5 (DR5) have been developed and are under clinical evaluation. Although these agonists demonstrate significant anti-tumor activity in preclinical models, the clinical efficacy in human cancer patients has been notably disappointing. One possible explanation might be that the current classes of therapeutic molecules are not sufficiently potent to elicit significant response in patients, particularly for dimeric antibody agonists that require secondary cross-linking via Fcγ receptors expressed on immune cells to achieve optimal clustering of DR5. To overcome this limitation, a novel multivalent Nanobody approach was taken with the goal of generating a significantly more potent DR5 agonist. In the present study, we show that trivalent DR5 targeting Nanobodies mimic the activity of natural ligand, and furthermore, increasing the valency of domains to tetramer and pentamer markedly increased potency of cell killing on tumor cells, with pentamers being more potent than tetramers in vitro. Increased potency was attributed to faster kinetics of death-inducing signaling complex assembly and caspase-8 and caspase-3 activation. In vivo, multivalent Nanobody molecules elicited superior anti-tumor activity compared to a conventional DR5 agonist antibody, including the ability to induce tumor regression in an insensitive patient-derived primary pancreatic tumor model. Furthermore, complete responses to Nanobody treatment were obtained in up to 50% of patient-derived primary pancreatic and colon tumor models, suggesting that multivalent DR5 Nanobodies may represent a significant new therapeutic modality for targeting death receptor signaling.


Subject(s)
Caspases/immunology , Neoplasms/immunology , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Single-Domain Antibodies/immunology , Animals , Antibody Affinity/immunology , Blotting, Western , Caspases/biosynthesis , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/immunology , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Induction/drug effects , HCT116 Cells , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Mice, Inbred NOD , Mice, Knockout , Mice, Nude , Mice, SCID , Neoplasms/drug therapy , Protein Multimerization , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/pharmacology , Surface Plasmon Resonance , Xenograft Model Antitumor Assays
18.
Autophagy ; 10(11): 1921-36, 2014.
Article in English | MEDLINE | ID: mdl-25470234

ABSTRACT

Endoplasmic reticulum (ER) stress-induced cell death is normally associated with activation of the mitochondrial apoptotic pathway, which is characterized by CYCS (cytochrome c, somatic) release, apoptosome formation, and caspase activation, resulting in cell death. In this study, we demonstrate that under conditions of ER stress cells devoid of CASP9/caspase-9 or BAX and BAK1, and therefore defective in the mitochondrial apoptotic pathway, still undergo a delayed form of cell death associated with the activation of caspases, therefore revealing the existence of an alternative stress-induced caspase activation pathway. We identified CASP8/caspase-8 as the apical protease in this caspase cascade, and found that knockdown of either of the key autophagic genes, ATG5 or ATG7, impacted on CASP8 activation and cell death induction, highlighting the crucial role of autophagy in the activation of this novel ER stress-induced death pathway. In line with this, we identified a protein complex composed of ATG5, FADD, and pro-CASP8 whose assembly coincides with caspase activation and cell death induction. Together, our results reveal the toxic potential of autophagy in cells undergoing ER stress that are defective in the mitochondrial apoptotic pathway, and suggest a model in which the autophagosome functions as a platform facilitating pro-CASP8 activation. Chemoresistance, a common problem in the treatment of cancer, is frequently caused by the downregulation of key mitochondrial death effector proteins. Alternate stress-induced apoptotic pathways, such as the one described here, may become of particular relevance for tackling the problem of chemoresistance in cancer cells.


Subject(s)
Apoptosis , Autophagy , Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Mitochondria/metabolism , Animals , Autophagy-Related Protein 5 , Autophagy-Related Protein 7 , Caspase 3/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Endoplasmic Reticulum Chaperone BiP , Enzyme Activation , Fas-Associated Death Domain Protein/metabolism , Fibroblasts/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Mice , Microtubule-Associated Proteins/metabolism , Signal Transduction
19.
Free Radic Biol Med ; 63: 280-90, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23732517

ABSTRACT

One of the major problems associated with the chemotherapy of tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) that selectively kills tumor cells is decreased drug resistance. This warranted the development of safe novel pharmacological agents that could sensitize the tumor cells to TRAIL. Herein, we examined the role of aldose reductase (AR) in sensitizing cancer cells to TRAIL and potentiating TRAIL-induced apoptosis of human colon cancer cells. We demonstrate that AR inhibition potentiates TRAIL-induced cytotoxicity in cancer cells by upregulation of both death receptor (DR)-5 and DR4. Knockdown of DR5 and DR4 significantly (>85%) reduced the sensitizing effect of the AR inhibitor fidarestat on TRAIL-induced apoptosis. Further, AR inhibition also downregulates cell survival proteins (Bcl-xL, Bcl-2, survivin, XIAP, and FLIP) and upregulates the expression of proapoptotic proteins such as Bax and alters mitochondrial membrane potential, leading to cytochrome c release, caspases-3 activation, and PARP cleavage. We found that AR inhibition regulates AKT/PI3K-dependent activation of forkhead transcription factor FOXO3a. Knockdown of FOXO3a significantly (>80%) abolished AR inhibition-induced upregulation of DR5 and DR4 and apoptosis in colon cancer cells. Overall, our results show that fidarestat potentiates TRAIL-induced apoptosis through downregulation of cell survival proteins and upregulation of death receptors via activation of the AKT/FOXO3a pathway.


Subject(s)
Aldehyde Reductase/metabolism , Apoptosis/genetics , Colonic Neoplasms/metabolism , Forkhead Transcription Factors/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism , Aldehyde Reductase/antagonists & inhibitors , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Enzyme Inhibitors/administration & dosage , Forkhead Box Protein O3 , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HT29 Cells , Humans , Oncogene Protein v-akt/genetics , Oncogene Protein v-akt/metabolism , Receptors, Death Domain/genetics , Receptors, Death Domain/metabolism , Up-Regulation/drug effects
20.
Int Immunopharmacol ; 17(3): 638-50, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23994464

ABSTRACT

Neutrophils play a critical role in the host defense against bacterial and fungal infections, but their inappropriate activation also contributes to tissue damage during autoimmune and inflammatory diseases. Neutrophils express a large number of cell surface receptors for the recognition of pathogen invasion and the inflammatory environment. Those include G-protein-coupled chemokine and chemoattractant receptors, Fc-receptors, adhesion receptors such as selectins/selectin ligands and integrins, various cytokine receptors, as well as innate immune receptors such as Toll-like receptors and C-type lectins. The various cell surface receptors trigger very diverse signal transduction pathways including activation of heterotrimeric and monomeric G-proteins, receptor-induced and store-operated Ca(2+) signals, protein and lipid kinases, adapter proteins and cytoskeletal rearrangement. Here we provide an overview of the receptors involved in neutrophil activation and the intracellular signal transduction processes they trigger. This knowledge is crucial for understanding how neutrophils participate in antimicrobial host defense and inflammatory tissue damage and may also point to possible future targets of the pharmacological therapy of neutrophil-mediated autoimmune or inflammatory diseases.


Subject(s)
Neutrophils/immunology , Receptors, Cell Surface/immunology , Animals , Humans , Proto-Oncogene Mas , Selectins/immunology , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL