Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 128
Filter
1.
J Cell Biol ; 223(3)2024 03 04.
Article in English | MEDLINE | ID: mdl-38329462

ABSTRACT

TNF signaling does not result in cell death unless multiple inhibitory signals are overcome, which can be accomplished by simultaneous signaling through IFNγ. In this issue, Deng and colleagues (http://doi.org/10.1083/jcb.202305026) dissect the mechanisms by which IFNγ signaling combines with TNF to mediate cell death through caspase-8, discussed by James E. Vince.


Subject(s)
Cell Death , Interferon-gamma , Signal Transduction , Interferon-gamma/physiology , Caspase 8/physiology , Tumor Necrosis Factors/physiology
2.
PLoS Biol ; 19(8): e3001304, 2021 08.
Article in English | MEDLINE | ID: mdl-34437534

ABSTRACT

Tumor necrosis factor receptor-1 (TNFR1) signaling, apart from its pleiotropic functions in inflammation, plays a role in embryogenesis as deficiency of varieties of its downstream molecules leads to embryonic lethality in mice. Caspase-8 noncleavable receptor interacting serine/threonine kinase 1 (RIPK1) mutations occur naturally in humans, and the corresponding D325A mutation in murine RIPK1 leads to death at early midgestation. It is known that both the demise of Ripk1D325A/D325A embryos and the death of Casp8-/- mice are initiated by TNFR1, but they are mediated by apoptosis and necroptosis, respectively. Here, we show that the defects in Ripk1D325A/D325A embryos occur at embryonic day 10.5 (E10.5), earlier than that caused by Casp8 knockout. By analyzing a series of genetically mutated mice, we elucidated a mechanism that leads to the lethality of Ripk1D325A/D325A embryos and compared it with that underlies Casp8 deletion-mediated lethality. We revealed that the apoptosis in Ripk1D325A/D325A embryos requires a scaffold function of RIPK3 and enzymatically active caspase-8. Unexpectedly, caspase-1 and caspase-11 are downstream of activated caspase-8, and concurrent depletion of Casp1 and Casp11 postpones the E10.5 lethality to embryonic day 13.5 (E13.5). Moreover, caspase-3 is an executioner of apoptosis at E10.5 in Ripk1D325A/D325A mice as its deletion extends life of Ripk1D325A/D325A mice to embryonic day 11.5 (E11.5). Hence, an unexpected death pathway of TNFR1 controls RIPK1 D325A mutation-induced lethality at E10.5.


Subject(s)
Caspase 8/physiology , Embryonic Development , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Receptors, Tumor Necrosis Factor, Type I/metabolism , Animals , Caspases/metabolism , Cell Death , Mice , Primary Cell Culture , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
3.
Cancer Sci ; 112(7): 2664-2678, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33934451

ABSTRACT

Immunotherapy targeting the PD-L1/PD-1 pathway is a novel type of clinical cancer treatment, but only small subsets of patients can benefit from it because of multiple factors. PD-L1/PD-1 expression is a biomarker for predicting the efficacy of anti-PD-L1/PD-1 therapy, which highlights the importance of understanding the regulatory mechanisms of PD-L1 expression in cancer cells. Casp8 is an apical caspase protease involved in mediating cell apoptosis, but it also has multiple nonapoptotic functions. Casp8 mutations are associated with increased risks of cancer, and low expression of Casp8 is closely connected with poor prognosis in patients with cancer. In addition, mutations of Casp8 in lymphocytes also lead to human immunodeficiency, thereby causing dysfunction of the innate immune system, but the roles of Casp8 in antitumor immunity remain unclear. Here, we found that knocking down Casp8 in mouse melanoma cells promoted tumor progression in an immune system-dependent manner. Mechanistically, Casp8 induced PD-L1 degradation by upregulating TNFAIP3 (A20) expression, a ubiquitin-editing enzyme that results in PD-L1 ubiquitination. In addition, compared with Casp8fl/fl mice, mice with conditional deletion of Casp8 in natural killer (NK) cells (Ncr1iCre/+ Casp8fl/fl mice) showed a decreased frequency of IFN-γ+ and CD107a+ NK cells but an increased frequency of PD-1+ and CTLA-4+ NK cells. Melanoma cells with Casp8 knocked down exhibited sensitivity to anti-PD-1 or anti-CTLA-4 antibody treatments, particularly in Ncr1iCre/+Casp8fl/fl mice. Together, the results indicate that Casp8 induces PD-L1 degradation by upregulating A20 expression and that decreased Casp8 expression is a potential biomarker for predicting the sensitivity to anti-PD-L1/PD-1 immunotherapy.


Subject(s)
B7-H1 Antigen/metabolism , Caspase 8/physiology , Immunotherapy, Adoptive/methods , Melanoma/therapy , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Animals , B7-H1 Antigen/genetics , CTLA-4 Antigen/metabolism , Caspase 8/genetics , Cell Line, Tumor , Disease Progression , Down-Regulation , GTPase-Activating Proteins/metabolism , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lysosomal Membrane Proteins/metabolism , Melanoma/immunology , Melanoma/pathology , Mice , Mice, Inbred C57BL , Mice, Nude , NF-kappa B/metabolism , Ubiquitination , Up-Regulation
4.
Diabetes Res Clin Pract ; 176: 108837, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33901624

ABSTRACT

BACKGROUND: CircRNAs are reported to be aberrantly expressed and perform biological functions in diverse processes. This study aimed to investigate the potential involvement of hsa_circ_0054633 in high glucose (HG)­induced diabetic model and its potential mechanism. METHODS: The expression of hsa_circ_0054633, miR-409-3p and caspase-8 was detected by real-time PCR and western blotting. Cell viability, apoptosis and the protein levels of apoptosis-related factors were revealed by CCK-8 colorimetry, flow cytometry and western blotting, respectively. Insulin secretion was determined by enzyme-linked immunosorbent assay (ELISA) and the measurement of insulin-related transcription factors. The target association between miR-409-3p and hsa_circ_0054633 or caspase-8 was confirmed by dual-luciferase reporter assays and biotin-based pulldown assay. RESULTS: Hsa_circ_0054633 was highly expressed and the expression of miR-409-3p was downregulated in serum of DM patients and HG-treated human pancreatic ß cell line NES2Y. Further investigation indicated that hsa_circ_0054633 suppression promoted cell proliferation, inhibited apoptosis and facilitated insulin secretion in HG-treated NES2Y cells. Mechanical analysis revealed that hsa_circ_0054633 regulated caspase-8 expression via sponging miR-409-3p. Rescue experiments demonstrated that miR-409-3p knockdown or caspase-8 overexpression reversed the effects of hsa_circ_0054633 in HG-stimulated NES2Y cells. CONCLUSION: Inhibition of hsa_circ_0054633 protected against HG-induced NES2Y cell apoptosis and impairment of insulin secretion by regulating miR-409-3p/caspase-8 axis.


Subject(s)
Apoptosis/genetics , Insulin Secretion/genetics , Insulin-Secreting Cells/physiology , RNA, Circular/physiology , Adult , Aged , Aged, 80 and over , Case-Control Studies , Caspase 8/genetics , Caspase 8/physiology , Cell Proliferation/genetics , Cells, Cultured , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Female , Humans , Insulin-Secreting Cells/metabolism , Male , MicroRNAs/genetics , MicroRNAs/physiology , Middle Aged , Signal Transduction/genetics , Young Adult
5.
Cancer Metastasis Rev ; 40(1): 303-318, 2021 03.
Article in English | MEDLINE | ID: mdl-33026575

ABSTRACT

Caspase-8 is an aspartate-specific cysteine protease, which is best known for its apoptotic functions. Caspase-8 is placed at central nodes of multiple signal pathways, regulating not only the cell cycle but also the invasive and metastatic cell behavior, the immune cell homeostasis and cytokine production, which are the two major components of the tumor microenvironment (TME). Ovarian cancer often has dysregulated caspase-8 expression, leading to imbalance between its apoptotic and non-apoptotic functions within the tumor and the surrounding milieu. The downregulation of caspase-8 in ovarian cancer seems to be linked to high aggressiveness with chronic inflammation, immunoediting, and immune resistance. Caspase-8 plays therefore an essential role not only in the primary tumor cells but also in the TME by regulating the immune response, B and T lymphocyte activation, and macrophage differentiation and polarization. The switch between M1 and M2 macrophages is possibly associated with changes in the caspase-8 expression. In this review, we are discussing the non-apoptotic functions of caspase-8, highlighting this protein as a modulator of the immune response and the cytokine composition in the TME. Considering the low survival rate among ovarian cancer patients, it is urgently necessary to develop new therapeutic strategies to optimize the response to the standard treatment. The TME is highly heterogenous and provides a variety of opportunities for new drug targets. Given the variety of roles of caspase-8 in the TME, we should focus on this protein in the development of new therapeutic strategies against the TME of ovarian cancer.


Subject(s)
Caspase 8/physiology , Ovarian Neoplasms , Tumor Microenvironment , Female , Humans , Macrophages , Signal Transduction
6.
JCI Insight ; 5(23)2020 12 03.
Article in English | MEDLINE | ID: mdl-33108350

ABSTRACT

Caspase-8 (CASP8) is one of the most frequently mutated genes in head and neck squamous carcinomas (HNSCCs), and CASP8 mutations are associated with poor survival. The distribution of these mutations in HNSCCs suggests that they are likely to be inactivating. Inhibition of CASP8 has been reported to sensitize cancer cells to necroptosis, a regulated cell death mechanism. Here, we show that knockdown of CASP8 renders HNSCCs susceptible to necroptosis by a second mitochondria-derived activator of caspase (SMAC) mimetic, birinapant, in combination with pan-caspase inhibitors Z-VAD-FMK or emricasan and radiation. In a syngeneic mouse model of oral cancer, birinapant, particularly when combined with radiation, delayed tumor growth and enhanced survival under CASP8 loss. Exploration of molecular underpinnings of necroptosis sensitivity confirmed that the level of functional receptor-interacting serine/threonine protein kinase 3 (RIP3) determines susceptibility to this mode of death. Although an in vitro screen revealed that low RIP3 levels rendered many HNSCC cell lines resistant to necroptosis, patient tumors maintained RIP3 expression and should therefore remain sensitive. Collectively, these results suggest that targeting the necroptosis pathway with SMAC mimetics, especially in combination with radiation, may be relevant therapeutically in HNSCC with compromised CASP8 status, provided that RIP3 function is maintained.


Subject(s)
Caspase 8/metabolism , Necroptosis/physiology , Squamous Cell Carcinoma of Head and Neck/metabolism , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Biomimetics , Caspase 8/genetics , Caspase 8/physiology , Caspase Inhibitors/metabolism , Caspase Inhibitors/pharmacology , Caspases/metabolism , Cell Line, Tumor , Databases, Genetic , Dipeptides/metabolism , Dipeptides/pharmacology , Humans , Indoles/metabolism , Indoles/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Necroptosis/genetics , Squamous Cell Carcinoma of Head and Neck/genetics
7.
Cell Rep ; 32(4): 107959, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32726624

ABSTRACT

Pyroptosis has emerged as a key mechanism by which inflammasomes promote host defense against microbial pathogens and sterile inflammation. Gasdermin D (GSDMD)-mediated cell lysis is a hallmark of pyroptosis, but our understanding of cell death signaling during pyroptosis is fragmented. Here, we show that independently of GSDMD-mediated plasma membrane permeabilization, inflammasome receptors engage caspase-1 and caspase-8, both of which redundantly promote activation of apoptotic executioner caspase-3 and caspase-7 in pyroptotic macrophages. Impaired GSDMD pore formation downstream of caspase-1 and caspase-8 activation suffices to unmask the apoptotic phenotype of pyroptotic macrophages. Combined inactivation of initiator caspase-1 and caspase-8, or executioner caspase-3 and caspase-7, is required to abolish inflammasome-induced DEVDase activity during pyroptosis and in apoptotic Gsdmd-/- cells. Collectively, these results unveil a robust apoptotic caspase network that is activated in parallel to GSDMD-mediated plasma membrane permeabilization and safeguards cell death induction in pyroptotic macrophages.


Subject(s)
Caspases/metabolism , Macrophages/metabolism , Pyroptosis/physiology , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins/metabolism , Caspase 1/metabolism , Caspase 1/physiology , Caspase 3/metabolism , Caspase 7/metabolism , Caspase 8/metabolism , Caspase 8/physiology , Cell Death , Cell Membrane/metabolism , Female , Inflammasomes/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Phosphate-Binding Proteins/metabolism
8.
Curr Opin Cell Biol ; 63: 186-193, 2020 04.
Article in English | MEDLINE | ID: mdl-32163825

ABSTRACT

Necroptosis and pyroptosis are inflammatory forms of regulated necrotic cell death as opposed to apoptosis that is generally considered immunologically silent. Recent studies revealed unexpected links in the pathways regulating and executing cell death in response to activation of signaling cascades inducing apoptosis, necroptosis, and pyroptosis. Emerging evidence suggests that receptor interacting protein kinase 1 and caspase-8 control the cross-talk between apoptosis, necroptosis, and pyroptosis and determine the type of cell death induced in response to activation of cell death signaling.


Subject(s)
Apoptosis/genetics , Caspase 8/physiology , Necroptosis/genetics , Pyroptosis/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Caspase 8/genetics , Caspase 8/metabolism , Humans , Necrosis/genetics , Necrosis/pathology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics
10.
Am J Physiol Gastrointest Liver Physiol ; 317(4): G493-G507, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31411503

ABSTRACT

Genetically modified mice have been successfully used as models for inflammatory bowel diseases; however, dietary effects were poorly examined. Here, we studied the impact of particular nutrients and supplements on gut functions related to the knockout of the epithelial caspase-8 gene. Caspase-8 knockout (Casp8∆IEC) and control (Casp8fl) mice were fed for 4 wk a control diet (CD) enriched with 10% inulin (CD-Inu) or 5% sodium butyrate (CD-But) while having free access to plain water or water supplemented with 30% fructose (+F). Body weight changes, intestinal inflammation, and selected markers for barrier function and of liver steatosis were assessed. Casp8∆IEC mice developed ileocolitis accompanied by changes in intestinal barrier morphology and reduced expression of barrier-related genes such as mucin-2 (Muc2) and defensins in the ileum and Muc2 in the colon. Casp8∆IEC mice fed a CD also showed impaired body weight gain compared with Casp8fl mice, which was even more pronounced in mice receiving water supplemented with fructose. Furthermore, we observed a marked liver steatosis and inflammation in some but not all Casp8∆IEC mice under a CD, which was on average similar to that observed in control mice under a fructose-rich diet. Hepatic lipid accumulation, as well as markers of ileal barrier function, but not intestinal pathohistology or body weight loss, were attenuated by diets enriched with inulin or butyrate, especially in the absence of fructose supplementation. Our data show that ileocolitis, barrier dysfunction, and malassimilation in Caspase-8 knockout mice can be partially attenuated by oral inulin or butyrate supplementation.NEW & NOTEWORTHY Genetic mouse models for ileocolitis are important to understand inflammatory bowel disease in humans. We examined dietetic factors that might aggravate or attenuate ileocolitis and related pathologies in such a model. Deletion of the caspase-8 gene results not only in ileocolitis but also in gut barrier dysfunction, liver steatosis, and malassimilation, which can be partially attenuated by oral inulin or sodium butyrate. Our data indicate that diet modifications can contribute to disease variability and therapy.


Subject(s)
Butyric Acid/pharmacology , Caspase 8/genetics , Caspase 8/physiology , Crohn Disease/genetics , Crohn Disease/pathology , Intestinal Mucosa/pathology , Inulin/pharmacology , Animals , Body Weight/genetics , Crohn Disease/drug therapy , Diet, Western , Dietary Supplements , Female , Gastrointestinal Microbiome , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucin-2/genetics , Mucin-2/metabolism , Nutrients
11.
Nat Commun ; 10(1): 1729, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30988283

ABSTRACT

RIPK1 regulates cell death and inflammation through kinase-dependent and -independent mechanisms. As a scaffold, RIPK1 inhibits caspase-8-dependent apoptosis and RIPK3/MLKL-dependent necroptosis. As a kinase, RIPK1 paradoxically induces these cell death modalities. The molecular switch between RIPK1 pro-survival and pro-death functions remains poorly understood. We identify phosphorylation of RIPK1 on Ser25 by IKKs as a key mechanism directly inhibiting RIPK1 kinase activity and preventing TNF-mediated RIPK1-dependent cell death. Mimicking Ser25 phosphorylation (S > D mutation) protects cells and mice from the cytotoxic effect of TNF in conditions of IKK inhibition. In line with their roles in IKK activation, TNF-induced Ser25 phosphorylation of RIPK1 is defective in TAK1- or SHARPIN-deficient cells and restoring phosphorylation protects these cells from TNF-induced death. Importantly, mimicking Ser25 phosphorylation compromises the in vivo cell death-dependent immune control of Yersinia infection, a physiological model of TAK1/IKK inhibition, and rescues the cell death-induced multi-organ inflammatory phenotype of the SHARPIN-deficient mice.


Subject(s)
Apoptosis , Models, Immunological , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Caspase 8/genetics , Caspase 8/metabolism , Caspase 8/physiology , Cell Line , I-kappa B Kinase/metabolism , I-kappa B Kinase/physiology , Immunity/physiology , Mice , Phosphorylation , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Serine/chemistry , Serine/metabolism , Yersinia , Yersinia Infections/immunology
12.
Mol Nutr Food Res ; 63(12): e1801206, 2019 06.
Article in English | MEDLINE | ID: mdl-30900802

ABSTRACT

SCOPE: Pyrrolizidine alkaloids (PAs) are common phytotoxins. Intoxication can lead to liver damage. Previous studies showed PA-induced apoptosis in liver cells. However, the exact role of the extrinsic apoptotic pathway has not been investigated yet. This study aims to analyze whether the PA representative lasiocarpine sensitizes human liver cells toward extrinsic Fas-mediated apoptosis. METHODS AND RESULTS: HepG2 cells with limited xenobiotic metabolic activity are used to analyze metabolism-dependent effects. External in vitro metabolism is simulated using rat or human liver enzymes. Additionally, metabolically competent HepaRG cells are used to confirm the observed effects in a human liver cell system with internal xenobiotic metabolism. Metabolized lasiocarpine decreases cell viability and induces Fas receptor gene expression in both cell lines. Increased Fas receptor protein expression on the cell surface is demonstrated by flow cytometry. The addition of a Fas ligand-simulating antibody induces apoptosis. Induction of extrinsic Fas-mediated apoptosis is verified by Western blotting for cleaved caspase 8, the initiator caspase of extrinsic apoptosis. All effects are dependent on lasiocarpine metabolism. CONCLUSION: The results demonstrate that metabolically metabolized lasiocarpine sensitizes human liver cells toward Fas-mediated apoptosis. They broaden our knowledge on the hepatotoxic molecular mechanisms of PA as widely distributed food contaminants.


Subject(s)
Apoptosis/drug effects , Hepatocytes/drug effects , Pyrrolizidine Alkaloids/pharmacology , fas Receptor/physiology , Activation, Metabolic , Animals , Caspase 8/physiology , Fas Ligand Protein/pharmacology , Hep G2 Cells , Hepatocytes/physiology , Humans , Male , Pyrrolizidine Alkaloids/pharmacokinetics , Rats , Rats, Wistar
13.
BMC Vet Res ; 15(1): 20, 2019 Jan 08.
Article in English | MEDLINE | ID: mdl-30621683

ABSTRACT

BACKGROUND: Caspases are a family of conserved intracellular cysteine-dependent aspartate-specific cysteine proteases that play important roles in regulating cell death and inflammation. Our previous study revealed the importance of the inflammatory caspase 1 gene in extracellular ATP-mediated immune signaling in Japanese flounder, Paralichthys olivaceus. To explore the potential roles of other caspases in P. olivaceus innate immunity, we extended our study by characterizing of the responses of four additional P. olivaceus caspase genes, termed JfCaspase 2, 3, 6 and 8, to inflammatory challenge and extracellular ATP stimulation. RESULTS: Sequence analysis revealed that the domain structures of all the Japanese flounder caspase proteins are evolutionarily conserved. Quantitative real-time PCR analysis showed that the JfCaspase 2, 3, 6 and 8 genes were expressed ubiquitously but at unequal levels in all examined Japanese flounder normal tissues. In addition, the basal gene expression levels of JfCaspase 2, 3, 6 and 8 were higher than those of JfCaspase 1 in both Japanese flounder head kidney macrophages (HKMs) and peripheral blood leukocytes (PBLs). Furthermore, immune challenge experiments showed that the inflammatory stimuli LPS and poly(I:C) significantly modulated the expression of the JfCaspase 2, 3, 6 and 8 genes in Japanese flounder immune cells. Finally, DNA fragmentation, associated with increased extracellular ATP-induced JfCaspase 2, 3, 6 and 8 gene expression and enzymatic activity, was inhibited by the caspase inhibitor Z-VAD-FMK in the HKMs. CONCLUSION: Our findings demonstrate broad participation of multiple caspase genes in response to inflammatory stimulation in Japanese flounder immune cells and provide new evidence for the involvement of caspase(s) in extracellular ATP-induced apoptosis in fish.


Subject(s)
Adenosine Triphosphate/pharmacology , Caspase 2/genetics , Caspase 3/genetics , Caspase 6/genetics , Caspase 8/genetics , Fish Proteins/genetics , Flounder/immunology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Caspase 2/physiology , Caspase 3/physiology , Caspase 6/physiology , Caspase 8/physiology , Fish Proteins/physiology , Flounder/genetics , Gene Expression Regulation/drug effects , Genes/drug effects , Immunity, Innate/drug effects , Immunity, Innate/immunology , Lipopolysaccharides/pharmacology , Phylogeny , Real-Time Polymerase Chain Reaction/veterinary , Sequence Alignment/veterinary , Sequence Analysis, DNA/veterinary
14.
Mol Biol Rep ; 45(6): 1715-1722, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30141104

ABSTRACT

The RIPoptosome, composed of RIP1 and caspase-8, plays an important role in the regulation of apoptosis and necroptosis; however, the mechanism of complex formation by oligomerization and how the caspase-activating process and necroptosis are mediated by the formation of the RIPoptosome is not well-understood. This study revealed that the assembly mechanism of the RIPoptosome core is dependent on salt concentration and not on pH and time. In addition, we demonstrated that three RIP1 mutations, E626K, M637K, and S657K, have dominant negative effects. These dominant negative mutations in RIP1 may have potential applications in therapeutic intervention.


Subject(s)
Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Apoptosis/genetics , Apoptosis/physiology , Caspase 8/genetics , Caspase 8/physiology , Cloning, Molecular/methods , Death Domain/genetics , Death Domain/physiology , Fas-Associated Death Domain Protein/metabolism , Genes, Dominant/genetics , Humans , Hydrogen-Ion Concentration , Inhibitor of Apoptosis Proteins/metabolism , Necrosis/genetics , Necrosis/physiopathology , RNA-Binding Proteins/genetics , Salts , Signal Transduction
15.
Med Sci Monit ; 24: 5247-5257, 2018 Jul 29.
Article in English | MEDLINE | ID: mdl-30057402

ABSTRACT

BACKGROUND Periodontal ligament stem cells (PDLSCs) possess characteristics of multi-potential differentiation and immuno-modulation, and PDLSCs-mediated periodontal tissue regeneration is regarded as a hopeful method for periodontitis treatment. Recent studies demonstrated that RIP3 and caspase8 regulate bacteria-induced innate immune response and programmed necrosis, which is also called necroptosis. This study aimed to determine the role of the RIP3/Caspase8 signal pathway on necroptosis of PDLSCs under the inflammatory microenvironment, both [i]in vitro[/i] and [i]in vivo[/i]. MATERIAL AND METHODS PDLSCs were cultured, and transmission electron microscopy and flow cytometry were used to detect necroptosis. PCR, ALP, and Alizarin Red S staining were used to assess the effect of necroptosis on osteogenesis differentiation of PDLSCs [i]in vitro[/i], while HE and Masson staining were taken after the nude mouse subcutaneous transplant experiment. RESULTS Our research indicates that RIP3/caspase8 can regulate the immune response of PDLSCs, and blockade of RIP3/caspase8 can protect the biological characteristics of the PDLSCs, effectively promoting periodontal tissue regeneration in the inflammatory microenvironment. CONCLUSIONS Inhibiting RIP3/caspase8 can effectively promote periodontal tissue regeneration in the inflammatory microenvironment.


Subject(s)
Caspase 8/physiology , Periodontitis/therapy , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Caspase 8/metabolism , Cell Differentiation/physiology , China , Female , Humans , Male , Mesenchymal Stem Cells/cytology , Mice , Necrosis/physiopathology , Osteogenesis/drug effects , Periodontal Ligament/cytology , Periodontal Ligament/physiology , Periodontitis/physiopathology , Primary Cell Culture , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Regeneration/physiology , Signal Transduction/physiology , Stem Cell Niche/physiology , Stem Cells/cytology , Stem Cells/physiology
16.
Sci Rep ; 8(1): 3788, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29491424

ABSTRACT

The NLRC4 inflammasome recognizes bacterial flagellin and components of the type III secretion apparatus. NLRC4 stimulation leads to caspase-1 activation followed by a rapid lytic cell death known as pyroptosis. NLRC4 is linked to pathogen-free auto-inflammatory diseases, suggesting a role for NLRC4 in sterile inflammation. Here, we show that NLRC4 activates an alternative cell death program morphologically similar to apoptosis in caspase-1-deficient BMDMs. By performing an unbiased genome-wide CRISPR/Cas9 screen with subsequent validation studies in gene-targeted mice, we highlight a critical role for caspase-8 and ASC adaptor in an alternative apoptotic pathway downstream of NLRC4. Furthermore, caspase-1 catalytically dead knock-in (Casp1 C284A KI) BMDMs genetically segregate pyroptosis and apoptosis, and confirm that caspase-1 does not functionally compete with ASC for NLRC4 interactions. We show that NLRC4/caspase-8-mediated apoptotic cells eventually undergo plasma cell membrane damage in vitro, suggesting that this pathway can lead to secondary necrosis. Unexpectedly, we found that DFNA5/GSDME, a member of the pore-forming gasdermin family, is dispensable for the secondary necrosis that follows NLRC4-mediated apoptosis in macrophages. Together, our data confirm the existence of an alternative caspase-8 activation pathway diverging from the NLRC4 inflammasome in primary macrophages.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , CARD Signaling Adaptor Proteins/physiology , Calcium-Binding Proteins/metabolism , Caspase 1/physiology , Caspase 8/physiology , Inflammasomes/metabolism , Macrophages/pathology , Animals , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , CRISPR-Cas Systems , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Genome , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout
17.
Am J Vet Res ; 79(4): 424-432, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29583049

ABSTRACT

OBJECTIVE To evaluate the effect of lipopolysaccharide (LPS) on apoptosis of equine neutrophils in vitro. SAMPLE Venous blood samples from 40 adult horses. PROCEDURES Neutrophils were isolated from blood samples and cultured with or without LPS from Escherichia coli O55:B5 for 12 or 24 hours. Neutrophil apoptosis was assessed by use of cytologic examination, annexin V and propidium iodide staining quantified with flow cytometry, coincubation with inducers of intrinsic and extrinsic apoptosis or a toll-like receptor (TLR) 4 inhibitor, and measurement of caspase-3, -8, and -9 activities. RESULTS Treatment with LPS resulted in a significant delay in apoptosis after incubation for 12 and 24 hours (neutrophils from blood samples of 40 horses). There was a significant correlation between increases in LPS dose and decreases in apoptosis after incubation for 24 hours (3 experiments, each of which involved neutrophils obtained from the same 3 horses at 3 separate times). Caspase-9 activity, but not caspase-3 or -8 activity, was significantly reduced in LPS-treated neutrophils after incubation for 12 hours (neutrophils from blood samples of 17 horses). Treatment with a TLR4 inhibitor or intrinsic and extrinsic inducers of apoptosis prevented LPS-delayed apoptosis. CONCLUSIONS AND CLINICAL RELEVANCE LPS treatment delayed apoptosis of equine neutrophils in vitro for up to 24 hours in a dose-dependent manner by alteration of the intrinsic pathway of apoptosis and was dependent on TLR4 signaling. Increased neutrophil life span may contribute to the development of a systemic inflammatory response syndrome in endotoxemic horses.


Subject(s)
Apoptosis/physiology , Caspase 3/physiology , Caspase 8/physiology , Caspase 9/physiology , Lipopolysaccharides/pharmacology , Neutrophils/drug effects , Toll-Like Receptor 4/physiology , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cells, Cultured , Flow Cytometry , Horses , Neutrophils/physiology , Signal Transduction
18.
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
19.
Biochim Biophys Acta Gen Subj ; 1862(3): 557-566, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29203282

ABSTRACT

BACKGROUND: The development of approaches that increase therapeutic effects of anti-cancer drugs is one of the most important tasks of oncology. Caloric restriction in vivo or serum deprivation (SD) in vitro has been shown to be an effective tool for sensitizing cancer cells to chemotherapeutic drugs. However, the detailed mechanisms underlying the enhancement of apoptosis in cancer cells by SD remain to be elucidated. METHODS: Flow cytometry, caspase activity assay and western blotting were used for cell death rate evaluation. Western blotting, gel-filtration, siRNA approach and qRT-PCR were used to elucidate the mechanism underlying cell death potentiation upon SD. RESULTS: We demonstrated that SD sensitizes cancer cells to treatment with chemotherapeutic agent cisplatin. This effect is independent on activation of caspases-2 and -8, apical caspases triggering apoptosis in response to genotoxic stress. SD potentiates cell death via downregulation of the anti-apoptotic protein Mcl-1. In fact, SD reduces the Mcl-1 mRNA level, which consequently decreases the Mcl-1 protein level and renders cells more susceptible to apoptosis induction via the formation of apoptosome. CONCLUSIONS: Mcl-1 protein is an important regulator of sensitivity of cancer cells to apoptotic stimuli upon SD. GENERAL SIGNIFICANCE: This study identifies Mcl-1 as a new target for the sensitization of human cancer cells to cell death by SD, which is of great significance for the development of efficient anti-cancer therapies.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Cisplatin/pharmacology , Culture Media, Serum-Free/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Myeloid Cell Leukemia Sequence 1 Protein/biosynthesis , Neoplasm Proteins/biosynthesis , Apoptosis/physiology , Apoptosomes/physiology , Caspase 2/physiology , Caspase 8/physiology , Cell Line, Tumor , Cysteine Endopeptidases/physiology , Down-Regulation , Drug Resistance, Neoplasm/physiology , HeLa Cells , Humans , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/physiology , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , RNA Interference , RNA, Small Interfering/genetics
20.
Oncogene ; 36(31): 4434-4444, 2017 08.
Article in English | MEDLINE | ID: mdl-28368418

ABSTRACT

In the process of cancer spreading, different modes of invasion exist. One is expansive invasion, in which a group of cancer cells gradually expands along with cancer cell proliferation. Invasion of cancer cells is also modified by their interaction with stromal cells including cancer-associated fibroblasts (CAFs). Cancer cells co-invade with CAFs, and invasion by CAFs frequently precede invasion by cancer cells, which indicates CAF-led cancer cell invasion. Here, we show that CAFs induce apoptosis in gastric cancer cells, which prevented expansive invasion by cancer cells and instead facilitated CAF-led invasion. Death receptor 4 and activation of caspase-8 in cancer cells mediated cancer cell apoptosis induced by CAFs, which was dependent on contact between cancer cells and CAFs. Apoptotic cancer cells in turn released apoptotic vesicles and stimulated invasion of CAFs. Accordingly, cancer cells followed the migrating CAFs. Treatment with a caspase inhibitor, ZVAD, or forced expression of a death domain fragment in cancer cells prevented cancer cell apoptosis induced by CAFs and increased expansive invasion by cancer cells in extracellular gel invasion assays, while the rate of cancer cell invasion led by CAFs was decreased. Death domain-fragment expression also prevented intramural invasion by gastric cancer cells in the stomach. Because CAF-led invasion is characterized by the movement of individual cancer cells away from the tumour, adequate cancer cell apoptosis may promote cancer dissemination.


Subject(s)
Apoptosis , Cancer-Associated Fibroblasts/physiology , Neoplasm Invasiveness , Stomach Neoplasms/pathology , Animals , Caspase 8/physiology , Cell Communication , Cell Line, Tumor , Extracellular Vesicles/physiology , Female , Humans , Mice , Mice, Inbred BALB C , Receptors, TNF-Related Apoptosis-Inducing Ligand/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...