1
|
Seyrek K, Espe J, Reiss E, Lavrik IN. The Crosstalk of Apoptotic and Non-Apoptotic Signaling in CD95 System. Cells 2024; 13:1814. [PMID: 39513921 PMCID: PMC11545656 DOI: 10.3390/cells13211814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/24/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
The mechanisms of CD95 (Fas/APO-1)-mediated extrinsic apoptotic pathway in cancer cells have been extensively studied. The majority of human cells express CD95, but not all these cells can induce extrinsic apoptosis. Accumulating evidence has shown that CD95 is a multifunctional protein, and its stimulation can also elicit non-apoptotic or even survival signals. It has become clear that under certain cellular contexts, due to the various checkpoints, CD95 activation can trigger both apoptotic and non-apoptotic signals. The crosstalk of death and survival signals may occur at different levels of signal transduction. The strength of the CD95 stimulation, initial levels of anti-apoptotic proteins, and posttranslational modifications of the core DISC components have been proposed to be the most important factors in the life/death decisions at CD95. Successful therapeutic targeting of CD95 signaling pathways will require a better understanding of the crosstalk between CD95-induced apoptotic and cell survival pathways. In this review, in order to gain a systematic understanding of the crosstalk between CD95-mediated apoptosis and non-apoptotic signaling, we will discuss these issues in a step-by-step way.
Collapse
Affiliation(s)
| | | | | | - Inna N. Lavrik
- Translational Inflammation Research, Medical Faculty, Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany; (K.S.); (J.E.); (E.R.)
| |
Collapse
|
2
|
Guerrache A, Micheau O. TNF-Related Apoptosis-Inducing Ligand: Non-Apoptotic Signalling. Cells 2024; 13:521. [PMID: 38534365 PMCID: PMC10968836 DOI: 10.3390/cells13060521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/01/2024] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL or Apo2 or TNFSF10) belongs to the TNF superfamily. When bound to its agonistic receptors, TRAIL can induce apoptosis in tumour cells, while sparing healthy cells. Over the last three decades, this tumour selectivity has prompted many studies aiming at evaluating the anti-tumoral potential of TRAIL or its derivatives. Although most of these attempts have failed, so far, novel formulations are still being evaluated. However, emerging evidence indicates that TRAIL can also trigger a non-canonical signal transduction pathway that is likely to be detrimental for its use in oncology. Likewise, an increasing number of studies suggest that in some circumstances TRAIL can induce, via Death receptor 5 (DR5), tumour cell motility, potentially leading to and contributing to tumour metastasis. While the pro-apoptotic signal transduction machinery of TRAIL is well known from a mechanistic point of view, that of the non-canonical pathway is less understood. In this study, we the current state of knowledge of TRAIL non-canonical signalling.
Collapse
Affiliation(s)
- Abderrahmane Guerrache
- Université de Bourgogne, 21000 Dijon, France
- INSERM Research Center U1231, «Equipe DesCarTes», 21000 Dijon, France
| | - Olivier Micheau
- Université de Bourgogne, 21000 Dijon, France
- INSERM Research Center U1231, «Equipe DesCarTes», 21000 Dijon, France
- Laboratoire d’Excellence LipSTIC, 21000 Dijon, France
| |
Collapse
|
3
|
Santacroce L, Magrone T. Molluscum Contagiosum Virus: Biology and Immune Response. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1451:151-170. [PMID: 38801577 DOI: 10.1007/978-3-031-57165-7_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Molluscum contagiosum virus is a poxvirus belonging to the Poxviridae family, which includes Orthopoxvirus, Parapoxvirus, Yantapoxvirus, Molluscipoxvirus, Smallpox virus, Cowpox virus and Monkeypox virus. MCV belongs to the genus Molluscipoxvirus and has a tropism for skin tissue. MCV infects keratinocytes and, after an incubation period of 2 weeks to 6 weeks, causes a breakdown of the skin barrier with the development of papules of variable size depending on the proper functioning of the immune response (both adaptive and acquired). MCV only infects humans and does not cause viraemia. MCV encodes for several inhibitory proteins responsible to circumvent the immune response through different signalling pathways. Individuals who can be infected with MCV are children, immunocompromised individuals such as organ transplant recipients and Human Immunodeficiency Virus (HIV)-infected individuals. Current treatments to manage MCV-induced lesions are different and include the use of immunomodulators, which, however, do not provide an effective response.
Collapse
Affiliation(s)
- Luigi Santacroce
- Section of Microbiology and Virology, Interdisciplinary Department of Medicine, School of Medicine, University of Bari, Bari, Italy.
| | - Thea Magrone
- Section of Microbiology and Virology, Interdisciplinary Department of Medicine, School of Medicine, University of Bari, Bari, Italy
| |
Collapse
|
4
|
Sadakane H, Matsumura M, Murakami M, Itoyama E, Shimokawa F, Sakota S, Yoshioka H, Kawabata H, Matsui T, Funaba M. Weak response of bovine hepcidin induction to iron through decreased expression of Smad4. FASEB J 2023; 37:e23243. [PMID: 37800888 DOI: 10.1096/fj.202301186rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/31/2023] [Accepted: 09/25/2023] [Indexed: 10/07/2023]
Abstract
Hepcidin negatively regulates systemic iron levels by inhibiting iron entry into the circulation. Hepcidin production is increased in response to an increase in systemic iron via the activation of the bone morphogenetic protein (BMP) pathway. Regulation of hepcidin expression by iron status has been proposed on the basis of evidence mainly from rodents and humans. We evaluated the effect of iron administration on plasma hepcidin concentrations in calves and the expression of bovine hepcidin by the BMP pathway in a cell culture study. Hematocrit as well as levels of blood hemoglobin and plasma iron were lower than the reference level in calves aged 1-4 weeks. Although intramuscular administration of iron increased iron-related parameters, plasma hepcidin concentrations were unaffected. Treatment with BMP6 increased hepcidin expression in human liver-derived cells but not in bovine liver-derived cells. A luciferase-based reporter assay revealed that Smad4 was required for hepcidin reporter transcription induced by Smad1. The reporter activity of hepcidin was lower in the cells transfected with bovine Smad4 than in those transfected with murine Smad4. The lower expression levels of bovine Smad4 were responsible for the lower activity of the hepcidin reporter, which might be due to the instability of bovine Smad4 mRNA. In fact, the endogenous Smad4 protein levels were lower in bovine cells than in human and murine cells. Smad4 also confers TGF-β/activin-mediated signaling. Induction of TGF-β-responsive genes was also lower after treatment with TGF-β1 in bovine hepatocytes than in human hepatoma cells. We revealed the unique regulation of bovine hepcidin expression and the characteristic TGF-β family signaling mediated by bovine Smad4. The present study suggests that knowledge of the regulatory expression of hepcidin as well as TGF-β family signaling obtained in murine and human cells is not always applicable to bovine cells.
Collapse
Affiliation(s)
- Hiroyuki Sadakane
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Manami Matsumura
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Masaru Murakami
- Laboratory of Molecular Biology, Azabu University School of Veterinary Medicine, Sagamihara, Japan
| | | | - Fumie Shimokawa
- Laboratory of Molecular Biology, Azabu University School of Veterinary Medicine, Sagamihara, Japan
| | - Shotaro Sakota
- Laboratory of Molecular Biology, Azabu University School of Veterinary Medicine, Sagamihara, Japan
| | | | - Hiroshi Kawabata
- National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Tohru Matsui
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Masayuki Funaba
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| |
Collapse
|
5
|
Reactive oxygen species-dependent upregulation of death receptor, tumor necrosis factor receptor 1, is responsible for theophylline-mediated cytotoxicity in MDA-MB-231 breast cancer cells. Anticancer Drugs 2022; 33:731-740. [PMID: 35946512 DOI: 10.1097/cad.0000000000001322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Theophylline, a methylxanthine drug, has been used as a therapy for respiratory diseases. Recently, it has also been shown to have a potential in treating different cancers. Also, it has shown promising results in clinical trials for AML in combination therapy. Subsequently, studies have shown theophylline to kill breast cancer cells but not normal breast cells. Therefore, in this study, we have explored the molecular mechanism underlying the cytotoxic effect of theophylline on breast cancer cells. Theophylline-treated cancer cells were analyzed for the transcript and protein expression of candidate apoptotic genes such as TNFR1, caspase-8, -9, -3 using qPCR and immunoblotting, respectively. Cell viability and apoptosis was measured in the presence or absence of TNFR1 inhibitor, R7050, using AO/EtBr staining and MTT assay, respectively. Similarly, oxidative stress was studied by analyzing ROS in the presence or absence of ROS inhibitor, NAC, using DCFDA assay. Theophylline caused reduced cell viability in cancer but not normal cells. Theophylline-treated breast cancer cells showed increased expression of death receptor, TNFR1, along with elevated levels of active caspase-8, -9 and -3. Inhibition of TNFR1 reduced caspase-dependent apoptosis even in the presence of theophylline. Theophylline further caused increased ROS generation, inhibition of which resulted in reduced TNFR1-mediated apoptosis. Theophylline also increased cathepsin activity, which was reduced on exposure of cells to TNFR1 inhibitor, R7050. We conclude that ROS-mediated activation of TNFR1 is responsible for caspase-3 and cathepsin-dependent cell death in breast cancer cells on exposure to theophylline.
Collapse
|
6
|
Risso V, Lafont E, Le Gallo M. Therapeutic approaches targeting CD95L/CD95 signaling in cancer and autoimmune diseases. Cell Death Dis 2022; 13:248. [PMID: 35301281 PMCID: PMC8931059 DOI: 10.1038/s41419-022-04688-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 02/09/2022] [Accepted: 02/24/2022] [Indexed: 12/14/2022]
Abstract
Cell death plays a pivotal role in the maintenance of tissue homeostasis. Key players in the controlled induction of cell death are the Death Receptors (DR). CD95 is a prototypic DR activated by its cognate ligand CD95L triggering programmed cell death. As a consequence, alterations in the CD95/CD95L pathway have been involved in several disease conditions ranging from autoimmune diseases to inflammation and cancer. CD95L-induced cell death has multiple roles in the immune response since it constitutes one of the mechanisms by which cytotoxic lymphocytes kill their targets, but it is also involved in the process of turning off the immune response. Furthermore, beyond the canonical pro-death signals, CD95L, which can be membrane-bound or soluble, also induces non-apoptotic signaling that contributes to its tumor-promoting and pro-inflammatory roles. The intent of this review is to describe the role of CD95/CD95L in the pathophysiology of cancers, autoimmune diseases and chronic inflammation and to discuss recently patented and emerging therapeutic strategies that exploit/block the CD95/CD95L system in these diseases.
Collapse
Affiliation(s)
- Vesna Risso
- INSERM U1242, Oncogenesis Stress Signaling, University of Rennes, Rennes, France
- Centre de lutte contre le cancer Eugène Marquis, Rennes, France
| | - Elodie Lafont
- INSERM U1242, Oncogenesis Stress Signaling, University of Rennes, Rennes, France
- Centre de lutte contre le cancer Eugène Marquis, Rennes, France
| | - Matthieu Le Gallo
- INSERM U1242, Oncogenesis Stress Signaling, University of Rennes, Rennes, France.
- Centre de lutte contre le cancer Eugène Marquis, Rennes, France.
| |
Collapse
|
7
|
Shanmugam MK, Sethi G. Molecular mechanisms of cell death. MECHANISMS OF CELL DEATH AND OPPORTUNITIES FOR THERAPEUTIC DEVELOPMENT 2022:65-92. [DOI: 10.1016/b978-0-12-814208-0.00002-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
|
8
|
Zhu H, Sun B, Shen Q. TNF-α induces apoptosis of human nucleus pulposus cells via activating the TRIM14/NF-κB signalling pathway. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:3004-3012. [PMID: 31322007 DOI: 10.1080/21691401.2019.1643733] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Hao Zhu
- Department of Orthopaedics, The Affiliated Shanghai General Hospital of Nanjing Medical University, Shanghai, China
- Department of Orthopaedics, The Fourth Affiliated Hospital of Nantong University, Yancheng, China
| | - Bao Sun
- Department of Orthopaedics, The Affiliated Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| | - Qiang Shen
- Department of Orthopaedics, The Affiliated Shanghai General Hospital of Nanjing Medical University, Shanghai, China
| |
Collapse
|
9
|
Sharma S, Carmona A, Skowronek A, Yu F, Collins MO, Naik S, Murzeau CM, Tseng PL, Erdmann KS. Apoptotic signalling targets the post-endocytic sorting machinery of the death receptor Fas/CD95. Nat Commun 2019; 10:3105. [PMID: 31308371 PMCID: PMC6629679 DOI: 10.1038/s41467-019-11025-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 06/12/2019] [Indexed: 02/07/2023] Open
Abstract
Fas plays a major role in regulating ligand-induced apoptosis in many cell types. It is well known that several cancers demonstrate reduced cell surface levels of Fas and thus escape a potential control system via ligand-induced apoptosis, although underlying mechanisms are unclear. Here we report that the endosome associated trafficking regulator 1 (ENTR1), controls cell surface levels of Fas and Fas-mediated apoptotic signalling. ENTR1 regulates, via binding to the coiled coil domain protein Dysbindin, the delivery of Fas from endosomes to lysosomes thereby controlling termination of Fas signal transduction. We demonstrate that ENTR1 is cleaved during Fas-induced apoptosis in a caspase-dependent manner revealing an unexpected interplay of apoptotic signalling and regulation of endolysosomal trafficking resulting in a positive feedback signalling-loop. Our data provide insights into the molecular mechanism of Fas post-endocytic trafficking and signalling, opening possible explanations on how cancer cells regulate cell surface levels of death receptors. Fas is a death receptor that regulates apoptosis in many cell types and is downregulated on the cell surface in many cancers. Here, Sharma et al. show that endosome associated trafficking regulator ENTR1 regulates delivery of Fas to lysosomes, thereby controlling its degradation and signalling.
Collapse
Affiliation(s)
- Shruti Sharma
- Department of Biomedical Science & Centre of Membrane Interactions and Dynamics, University of Sheffield, Sheffield, S10 2TN, UK
| | - Antonio Carmona
- Department of Biomedical Science & Centre of Membrane Interactions and Dynamics, University of Sheffield, Sheffield, S10 2TN, UK
| | - Agnieszka Skowronek
- Department of Biomedical Science & Centre of Membrane Interactions and Dynamics, University of Sheffield, Sheffield, S10 2TN, UK
| | - Fangyan Yu
- Department of Biomedical Science & Centre of Membrane Interactions and Dynamics, University of Sheffield, Sheffield, S10 2TN, UK.,Department of Radiation Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Mark O Collins
- Department of Biomedical Science & Centre of Membrane Interactions and Dynamics, University of Sheffield, Sheffield, S10 2TN, UK
| | - Sindhu Naik
- Department of Biomedical Science & Centre of Membrane Interactions and Dynamics, University of Sheffield, Sheffield, S10 2TN, UK
| | - Claire M Murzeau
- Department of Biomedical Science & Centre of Membrane Interactions and Dynamics, University of Sheffield, Sheffield, S10 2TN, UK
| | - Pei-Li Tseng
- Department of Biomedical Science & Centre of Membrane Interactions and Dynamics, University of Sheffield, Sheffield, S10 2TN, UK
| | - Kai S Erdmann
- Department of Biomedical Science & Centre of Membrane Interactions and Dynamics, University of Sheffield, Sheffield, S10 2TN, UK.
| |
Collapse
|
10
|
Wallach D, Kang TB. Programmed Cell Death in Immune Defense: Knowledge and Presumptions. Immunity 2019; 49:19-32. [PMID: 30021143 DOI: 10.1016/j.immuni.2018.06.019] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 06/14/2018] [Accepted: 06/29/2018] [Indexed: 01/06/2023]
Abstract
Cell-culture studies are our main source of knowledge of the various forms of programmed cell death. Yet genetic perturbations of death-protein function in animal models are almost the only source of our knowledge of the physiological roles of these programs. Shortcomings in the state of knowledge acquired by these two experimental approaches are exemplified in this Perspective by reference to research on the contribution of apoptosis to lymphocyte development, a subject on which there is already much knowledge, and on the role of necroptosis in inflammation, about which information is just beginning to emerge. To address these shortcomings, there is need to find ways to verify the notions obtained through the current experimental approaches by directly monitoring death programs within specific cells in vivo.
Collapse
Affiliation(s)
- David Wallach
- Department of Biomolecular Sciences, The Weizmann Institute of Science, 76100 Rehovot, Israel.
| | - Tae-Bong Kang
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chung-Ju 27478, Republic of Korea
| |
Collapse
|
11
|
Kang TB, Jeong JS, Yang SH, Kovalenko A, Wallach D. Caspase-8 deficiency in mouse embryos triggers chronic RIPK1-dependent activation of inflammatory genes, independently of RIPK3. Cell Death Differ 2018; 25:1107-1117. [PMID: 29666472 PMCID: PMC5988659 DOI: 10.1038/s41418-018-0104-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 02/26/2018] [Accepted: 03/12/2018] [Indexed: 12/22/2022] Open
Abstract
Deletion of the Casp8 gene in epithelial tissues of mice results in severe inflammatory pathologies. Its ubiquitous deletion, or its specific deletion in endothelial cells, results in intrauterine death associated with capillary damage. These pathologies are all preventable by co-deletion of Casp8 and the genes encoding either the RIPK1 or the RIPK3 protein kinase. Since activation of RIPK3 in Caspase-8-deficient cells can trigger necroptotic cell death, and since RIPK1 can activate RIPK3, it is widely assumed that the inflammatory states resulting from Caspase-8 deficiency occur as a consequence of RIPK3-induced necroptosis. Here, we report that although on a Ripk3-null background Casp8 deletion in mice does not result in outright pathological changes, it triggers enhanced expression of a variety of inflammatory genes in utero, which gradually subsides after birth. Deletion of Ripk1, or even of only one of its two alleles, obliterates this activation. Resembling the embryonic pathology observed in RIPK3-expressing cells, the activation of inflammatory genes observed on a Ripk3-null background seems to be initiated in endothelial cells. Analysis of endothelial cells isolated from livers of Caspase-8-deficient embryos revealed neither an increase in the amount of RIPK1 in these cells after Casp8 deletion, nor triggering of RIPK1 phosphorylation. These findings indicate that the triggering of inflammation by Casp8 deletion in mice occurs, in part, independently of necroptosis or other functions of RIPK3, and rather reflects enhanced RIPK1-dependent signaling for activation of inflammatory genes.
Collapse
Affiliation(s)
- Tae-Bong Kang
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, Chung-Ju, 27478, Korea
| | - Ju-Seong Jeong
- Department of Biomolecular Sciences, The Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Seung-Hoon Yang
- Department of Biomolecular Sciences, The Weizmann Institute of Science, 76100, Rehovot, Israel.,Systems Biotechnology Research Center, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Korea
| | - Andrew Kovalenko
- Department of Biomolecular Sciences, The Weizmann Institute of Science, 76100, Rehovot, Israel
| | - David Wallach
- Department of Biomolecular Sciences, The Weizmann Institute of Science, 76100, Rehovot, Israel.
| |
Collapse
|
12
|
Bachmann M, Pfeilschifter J, Mühl H. A Prominent Role of Interleukin-18 in Acetaminophen-Induced Liver Injury Advocates Its Blockage for Therapy of Hepatic Necroinflammation. Front Immunol 2018; 9:161. [PMID: 29472923 PMCID: PMC5809456 DOI: 10.3389/fimmu.2018.00161] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 01/18/2018] [Indexed: 12/19/2022] Open
Abstract
Acetaminophen [paracetamol, N-acetyl-p-aminophenol (APAP)]-induced acute liver injury (ALI) not only remains a persistent clinical challenge but likewise stands out as well-characterized paradigmatic model of drug-induced liver damage. APAP intoxication associates with robust hepatic necroinflammation the role of which remains elusive with pathogenic but also pro-regenerative/-resolving functions being ascribed to leukocyte activation. Here, we shine a light on and put forward a unique role of the interleukin (IL)-1 family member IL-18 in experimental APAP-induced ALI. Indeed, amelioration of disease as previously observed in IL-18-deficient mice was further substantiated herein by application of the IL-18 opponent IL-18-binding protein (IL-18BPd:Fc) to wild-type mice. Data altogether emphasize crucial pathological action of this cytokine in APAP toxicity. Adding recombinant IL-22 to IL-18BPd:Fc further enhanced protection from liver injury. In contrast to IL-18, the role of prototypic pro-inflammatory IL-1 and tumor necrosis factor-α is controversially discussed with lack of effects or even protective action being repeatedly reported. A prominent detrimental function for IL-18 in APAP-induced ALI as proposed herein should relate to its pivotal role for hepatic expression of interferon-γ and Fas ligand, both of which aggravate APAP toxicity. As IL-18 serum levels increase in patients after APAP overdosing, targeting IL-18 may evolve as novel therapeutic option in those hard-to-treat patients where standard therapy with N-acetylcysteine is unsuccessful. Being a paradigmatic experimental model of ALI, current knowledge on ill-fated properties of IL-18 in APAP intoxication likewise emphasizes the potential of this cytokine to serve as therapeutic target in other entities of inflammatory liver diseases.
Collapse
Affiliation(s)
- Malte Bachmann
- Pharmazentrum Frankfurt/ZAFES, University Hospital Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt/ZAFES, University Hospital Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| | - Heiko Mühl
- Pharmazentrum Frankfurt/ZAFES, University Hospital Goethe-University Frankfurt am Main, Frankfurt am Main, Germany
| |
Collapse
|
13
|
Leonard BC, Johnson DE. Signaling by cell surface death receptors: Alterations in head and neck cancer. Adv Biol Regul 2018; 67:170-178. [PMID: 29066276 PMCID: PMC5854325 DOI: 10.1016/j.jbior.2017.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/13/2017] [Accepted: 10/15/2017] [Indexed: 12/01/2022]
Abstract
Cell surface death receptors are members of the tumor necrosis factor receptor (TNFR) superfamily and mediate signals leading to the induction of apoptosis or necroptosis, as well as NF-κB-mediated cell survival. These biochemical processes play key roles in cell growth, development, tissue homeostasis, and immune responses. The downstream signaling complexes activated by different death receptors can differ significantly and are subject to multiple, distinct regulatory mechanisms. Dysregulation of signaling by the TNFR superfamily contributes to a variety of pathologic conditions, including defective immune responses and cancer. Caspase-8 signaling is important for mediating death receptor signals leading to either apoptosis or NF-κB activation. By contrast, inactivation of caspase-8 or loss of caspase-8 expression shifts death receptor signaling to the necroptosis pathway. Notably, the gene encoding caspase-8 is mutated in roughly ten percent of head and neck cancers. These findings support the hypothesis that alterations in the biochemical pathways mediated by death receptors have important consequences for the development of head and neck, and possibly other, cancers.
Collapse
Affiliation(s)
- Brandon C Leonard
- Department of Otolaryngology - Head and Neck Surgery, University of California at San Francisco, San Francisco, CA, USA
| | - Daniel E Johnson
- Department of Otolaryngology - Head and Neck Surgery, University of California at San Francisco, San Francisco, CA, USA.
| |
Collapse
|
14
|
Lafont E, Hartwig T, Walczak H. Paving TRAIL's Path with Ubiquitin. Trends Biochem Sci 2017; 43:44-60. [PMID: 29195774 DOI: 10.1016/j.tibs.2017.11.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/31/2017] [Accepted: 11/01/2017] [Indexed: 12/22/2022]
Abstract
Despite its name, signalling induced by the tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is versatile. Besides eliciting cell death by both apoptosis and necroptosis, TRAIL can also induce migration, proliferation, and cytokine production in cancerous and non-cancerous cells. Unravelling the mechanisms regulating the intricate balance between these different outputs could therefore facilitate our understanding of the role of TRAIL in tissue homeostasis, immunity, and cancer. Ubiquitination and its reversal, deubiquitination, are crucial modulators of immune receptor signalling. This review discusses recent progress on the orchestration of TRAIL signalling outcomes by ubiquitination of various components of the signalling complexes, our understanding of the molecular switches that decide between cell death and gene activation, and what remains to be discovered.
Collapse
Affiliation(s)
- Elodie Lafont
- Centre for Cell Death, Cancer, and Inflammation, UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK; These authors contributed equally to this work
| | - Torsten Hartwig
- Centre for Cell Death, Cancer, and Inflammation, UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK; These authors contributed equally to this work
| | - Henning Walczak
- Centre for Cell Death, Cancer, and Inflammation, UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK.
| |
Collapse
|
15
|
Siegmund D, Lang I, Wajant H. Cell death-independent activities of the death receptors CD95, TRAILR1, and TRAILR2. FEBS J 2016; 284:1131-1159. [PMID: 27865080 DOI: 10.1111/febs.13968] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 11/10/2016] [Accepted: 11/17/2016] [Indexed: 12/25/2022]
Abstract
Since their identification more than 20 years ago, the death receptors CD95, TRAILR1, and TRAILR2 have been intensively studied with respect to their cell death-inducing activities. These receptors, however, can also trigger a variety of cell death-independent cellular responses reaching from the activation of proinflammatory gene transcription programs over the stimulation of proliferation and differentiation to induction of cell migration. The cell death-inducing signaling mechanisms of CD95 and the TRAIL death receptors are well understood. In contrast, despite the increasing recognition of the biological and pathophysiological relevance of the cell death-independent activities of CD95, TRAILR1, and TRAILR2, the corresponding signaling mechanisms are less understood and give no fully coherent picture. This review is focused on the cell death-independent activities of CD95 and the TRAIL death receptors and addresses mainly three questions: (a) how are these receptors linked to noncell death pathways at the molecular level, (b) which factors determine the balance of cell death and cell death-independent activities of CD95 and the TRAIL death receptors at the cellular level, and (c) what are the consequences of the cell death-independent functions of these receptors for their role in cancer and inflammatory diseases.
Collapse
Affiliation(s)
- Daniela Siegmund
- Division of Molecular Internal Medicine, Medical Clinic and Polyclinic II, University Hospital Würzburg, Germany
| | - Isabell Lang
- Division of Molecular Internal Medicine, Medical Clinic and Polyclinic II, University Hospital Würzburg, Germany
| | - Harald Wajant
- Division of Molecular Internal Medicine, Medical Clinic and Polyclinic II, University Hospital Würzburg, Germany
| |
Collapse
|
16
|
CD95 Signaling Inhibits B Cell Receptor-Mediated Gammaherpesvirus Replication in Apoptosis-Resistant B Lymphoma Cells. J Virol 2016; 90:9782-9796. [PMID: 27558422 DOI: 10.1128/jvi.00668-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 08/13/2016] [Indexed: 11/20/2022] Open
Abstract
While CD95 is an apoptosis-inducing receptor and has emerged as a potential anticancer therapy target, mounting evidence shows that CD95 is also emerging as a tumor promoter by activating nonapoptotic signaling pathways. Gammaherpesviral infection is closely associated with lymphoproliferative diseases, including B cell lymphomas. The nonapoptotic function of CD95 in gammaherpesvirus-associated lymphomas is largely unknown. Here, we show that stimulation of CD95 agonist antibody drives the majority of sensitive gammaherpesvirus-transformed B cells to undergo caspase-dependent apoptosis and promotes the survival and proliferation of a subpopulation of apoptosis-resistant B cells. Surprisingly, CD95-mediated nonapoptotic signaling induced beta interferon (IFN-β) expression and correlatively inhibited B cell receptor (BCR)-mediated gammaherpesviral replication in the apoptosis-resistant lymphoma cells without influencing BCR signaling. Further analysis showed that IFN-β alone or synergizing with CD95 blocked the activation of lytic switch proteins and the gene expression of gammaherpesviruses. Our findings indicate that, independent of its apoptotic activity, CD95 signaling activity plays an important role in blocking viral replication in apoptosis-resistant, gammaherpesvirus-associated B lymphoma cells, suggesting a novel mechanism that indicates how host CD95 prototype death receptor controls the life cycle of gammaherpesviruses independent of its apoptotic activity. IMPORTANCE Gammaherpesviruses are closely associated with lymphoid malignancies and other cancers. Viral replication and persistence strategies leading to cancer involve the activation of antiapoptotic and proliferation programs, as well as evasion of the host immune response. Here, we provide evidence that the stimulation of CD95 agonist antibody, mimicking one of the major mechanisms of cytotoxic T cell killing, inhibits B cell receptor-mediated gammaherpesviral replication in CD95 apoptosis-resistant lymphoma cells. CD95-induced type I interferon (IFN-β) contributes to the inhibition of gammaherpesviral replication. This finding sheds new light on the CD95 nonapoptotic function and provides a novel mechanism for gammaherpesviruses that helps them to escape host immune surveillance.
Collapse
|
17
|
Li W, Chen Z, Yan M, He P, Chen Z, Dai H. The protective role of isorhamnetin on human brain microvascular endothelial cells from cytotoxicity induced by methylglyoxal and oxygen-glucose deprivation. J Neurochem 2016; 136:651-659. [PMID: 26578299 DOI: 10.1111/jnc.13436] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 10/25/2015] [Accepted: 11/05/2015] [Indexed: 01/04/2023]
Abstract
As the first target of stroke, cerebral endothelial cells play a key role in brain vascular repair and maintenance, and their function is impeded in diabetes. Methylglyoxal (MGO), a reactive dicarbonyl produced during glucose metabolism, accumulates in diabetic patients. MGO and MGO-induced advanced glycation end-products (AGEs) could ameliorate stroke-induced brain vascular damage, closely related with ECs dysfunction. Using MGO plus oxygen-glucose deprivation (OGD) to mimic diabetic stroke, we reported the protective effect of isorhamnetin on OGD-induced cytotoxicity after MGO treatment on primary human brain microvascular endothelial cells (HBMEC) and explored the underlying mechanisms. Treatment of MGO for 24 h significantly enhanced 3-h OGD-induced HBMEC toxic effect, which was inhibited by pretreatment of isorhamnetin (100 μmol/L). Moreover, the protective effect of isorhamnetin is multiple function dependent, which includes anti-inflammation, anti-oxidative stress and anti-apoptosis effects. Besides its well-known inhibition on the mitochondria-dependent or intrinsic apoptotic pathway, isorhamnetin also reduced activation of the extrinsic apoptotic pathway, as characterized by the decreased expression and activity of caspase 3 and caspase 8. Furthermore, pretreatment with isorhamnetin specifically inhibited FAS/FASL expression and suppressed nuclear factor-kappa B nuclear translocation. Taken together, our results indicated that isorhamnetin protected against OGD-induced cytotoxicity after MGO treatment in cultured HBMEC due to its multiple protective effects and could inhibit Fas-mediated extrinsic apoptosis. Therefore, isorhamnetin is a promising reagent for the treatment of hyperglycemia and ischemia-induced cerebral vascular degeneration. A proposed model of the potential protective mechanism of isorhamnetin, a metabolite of quercetin, on methylglyoxal (MGO) treatment plus oxygen-glucose deprivation (OGD) exposure-induced cytotoxicity in cultured human brain microvascular endothelial cells. Isorhamnetin inhibits FasL-mediated extrinsic apoptosis and neurotrophic factor κB (NF-κB) nuclear translocation, which can induce the cell DNA damage. Therefore, the protective effect of isorhamnetin occurs through multiple functions, including anti-inflammation, anti-oxidative stress and anti-apoptosis. Therefore, isorhamnetin is a promising reagent for the treatment of hyperglycemia and ischemia-induced cerebral vascular degeneration.
Collapse
Affiliation(s)
- Wenlu Li
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhigang Chen
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Institute, Zhejiang University, Hangzhou, China
| | - Min Yan
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ping He
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhong Chen
- Department of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haibin Dai
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Institute, Zhejiang University, Hangzhou, China
| |
Collapse
|
18
|
Hultgårdh-Nilsson A, Borén J, Chakravarti S. The small leucine-rich repeat proteoglycans in tissue repair and atherosclerosis. J Intern Med 2015; 278:447-61. [PMID: 26477596 PMCID: PMC4616156 DOI: 10.1111/joim.12400] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Proteoglycans consist of a protein core with one or more covalently attached glycosaminoglycan (GAG) side chains and have multiple roles in the initiation and progression of atherosclerosis. Here we discuss the potential and known functions of a group of small leucine-rich repeat proteoglycans (SLRPs) in atherosclerosis. We focus on five SLRPs, decorin, biglycan, lumican, fibromodulin and PRELP, because these have been detected in atherosclerotic plaques or demonstrated to have a role in animal models of atherosclerosis. Decorin and biglycan are modified post-translationally by substitution with chondroitin/dermatan sulphate GAGs, whereas lumican, fibromodulin and PRELP have keratan sulphate side chains, and the core proteins have leucine-rich repeat (LRR) motifs that are characteristic of the LRR superfamily. The chondroitin/dermatan sulphate GAG side chains have been implicated in lipid retention in atherosclerosis. The core proteins are discussed here in the context of (i) interactions with collagens and their implications in tissue integrity, fibrosis and wound repair and (ii) interactions with growth factors, cytokines, pathogen-associated molecular patterns and cell surface receptors that impact normal physiology and disease processes such as inflammation, innate immune responses and wound healing (i.e. processes that are all important in plaque development and progression). Thus, studies of these SLRPs in the context of wound healing are providing clues about their functions in early stages of atherosclerosis to plaque vulnerability and cardiovascular disease at later stages. Understanding of signal transduction pathways regulated by the core protein interactions is leading to novel roles and therapeutic potential for these proteins in wound repair and atherosclerosis.
Collapse
Affiliation(s)
| | - J Borén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - S Chakravarti
- Departments of Medicine, Ophthalmology and Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
19
|
NLRP3 mediates NF-κB activation and cytokine induction in microbially induced and sterile inflammation. PLoS One 2015; 10:e0119179. [PMID: 25761061 PMCID: PMC4356585 DOI: 10.1371/journal.pone.0119179] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 01/10/2015] [Indexed: 12/21/2022] Open
Abstract
Nucleotide-binding domain and leucine-rich repeat-containing family, pyrin domain containing 3 (NLRP3) has recently emerged as a central regulator of innate immunity and inflammation in response to both sterile inflammatory and microbial invasion signals. Although its ability to drive proteolytic procaspase-1 processing has drawn more attention, NLPR3 can also activate NF-κB. To clarify the physiological relevance of this latter function, we examined the effect of NLRP3 on NF-κB activation and cytokine induction in RNA-interference-based NLRP3-knockdown cell lines generated from the human monocytic cell line THP-1. Knocking down NLRP3 reduced NF-κB activation and cytokine induction in the early stages of Staphylococcus aureus infection. Expression of cytokine genes induced by Staphylococcus aureus was not inhibited by a caspase-1 inhibitor, and did not occur through an autocrine mechanism in response to newly synthesized cytokines. We also demonstrated that NLRP3 could activate NF-κB and induce cytokines in response to sterile signals, monosodium urate crystals and aluminum adjuvant. Thus, NLRP3 mediates NF-κB activation in both sterile and microbially induced inflammation. Our findings show that not only does NLRP3 activate caspase-1 post-translationally, but it also induces multiple cytokine genes in the innate immune system.
Collapse
|
20
|
Lipid rafts and raft-mediated supramolecular entities in the regulation of CD95 death receptor apoptotic signaling. Apoptosis 2015; 20:584-606. [DOI: 10.1007/s10495-015-1104-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
21
|
Abstract
Molluscum contagiosum virus (MCV) is the causative agent of molluscum contagiosum (MC), the third most common viral skin infection in children, and one of the five most prevalent skin diseases worldwide. No FDA-approved treatments, vaccines, or commercially available rapid diagnostics for MCV are available. This review discusses several aspects of this medically important virus including: physical properties of MCV, MCV pathogenesis, MCV replication, and immune responses to MCV infection. Sequencing of the MCV genome revealed novel immune evasion molecules which are highlighted here. Special attention is given to the MCV MC159 and MC160 proteins. These proteins are FLIPs with homologs in gamma herpesviruses and in the cell. They are of great interest because each protein regulates apoptosis, NF-κB, and IRF3. However, the mechanism that each protein uses to impart its effects is different. It is important to elucidate how MCV inhibits immune responses; this knowledge contributes to our understanding of viral pathogenesis and also provides new insights into how the immune system neutralizes virus infections.
Collapse
|
22
|
Abstract
Cellular FLICE-inhibitory protein (cFLIP) is structurally related to caspase-8, but lacks its protease activity. Cflip gene encodes several splicing variants including short form (cFLIPs) and long form (cFLIPL). cFLIPL is composed of two death effector domains at the N terminus and a C-terminal caspase-like domain, and cFLIPs lacks the caspase-like domain. Our studies reveal that cFLIP plays a central role in NF-κB-dependent survival signals that control apoptosis and programmed necrosis. Germline deletion of Cflip results in embryonic lethality due to enhanced apoptosis and programmed necrosis; however, the combined deletion of the death-signaling regulators, Fadd and Ripk3, prevents embryonic lethality in Cflip-deficient mice. Moreover, tissue-specific deletion of Cflip reveals cFLIP as a crucial regulator that maintains tissue homeostasis of immune cells, hepatocytes, intestinal epithelial cells, and epidermal cells by preventing apoptosis and programmed necrosis.
Collapse
|
23
|
Abstract
Cell turnover is a fundamental feature in metazoans. Cells can die passively, as a consequence of severe damage to their structural integrity, or actively, owing to a more confined biological disruption such as DNA damage. Passive cell death is uncontrolled and often harmful to the organism. In contrast, active cell death is tightly regulated and serves to support the organism's life. Apoptosis-the primary form of regulated cell death-is relatively well defined. Necroptosis-an alternative, distinct kind of regulated cell death discovered more recently-is less well understood. Apoptosis and necroptosis can be triggered either from within the cell or by extracellular stimuli. Certain signaling components, including several death ligands and receptors, can regulate both processes. Whereas apoptosis is triggered and executed via intracellular proteases called caspases, necroptosis is suppressed by caspase activity. Here we highlight current understanding of the key signaling mechanisms that control regulated cell death.
Collapse
Affiliation(s)
- Avi Ashkenazi
- Genentech Inc., South San Francisco, California 94080;
| | | |
Collapse
|
24
|
Welsby I, Hutin D, Gueydan C, Kruys V, Rongvaux A, Leo O. PARP12, an interferon-stimulated gene involved in the control of protein translation and inflammation. J Biol Chem 2014; 289:26642-26657. [PMID: 25086041 DOI: 10.1074/jbc.m114.589515] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Transcriptome analyses have recently identified PARP12, a member of a large family of ADP-ribosyl transferases, as an interferon-induced gene (ISG), whose function remains incompletely characterized. We demonstrate herein that PARP12 is a genuine ISG, whose expressed protein displays at least two distinct subcellular locations and related functions. Upon ectopic expression or exposure to oxidative stress, PARP12 is recruited to stress-granules (SGs), known sites of mRNA translational arrest. Accordingly, PARP12 was found to block mRNA translation, possibly upon association to the translational machinery. Both the N-terminal domain (containing an RNA-binding domain characterized by the presence of five CCCH-type Zn-fingers) and integrity of the catalytic domain are required for this suppressive function. In contrast, stimulation with LPS leads to the localization of PARP12 to p62/SQSTM1 (an adaptor protein involved in innate signaling and autophagy) containing structures, unrelated to SGs. Deletion of the N-terminal domain promotes the association of the protein to p62/SQSTM1, suggesting that the RNA-binding domain is responsible for the subcellular localization of PARP12. Association to p62/SQSTM1 was found to correlate with increased NF-κB signaling, suggesting a role for PARP12 in inflammation. Collectively, these observations suggest that PARP12 can alternate between two distinct subcellular compartments associated to two distinct cellular functions. The present work therefore identifies PARP12 as an ISG with a potential role in cellular defenses against viral infections.
Collapse
Affiliation(s)
- Iain Welsby
- Laboratoire d'Immunobiologie, Université Libre de Bruxelles, Gosselies, Belgium; Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
| | - David Hutin
- Laboratoire d'Immunobiologie, Université Libre de Bruxelles, Gosselies, Belgium
| | - Cyril Gueydan
- Laboratoire de Biologie Moléculaire du Gène, Université Libre de Bruxelles, Gosselies, Belgium
| | - Veronique Kruys
- Laboratoire de Biologie Moléculaire du Gène, Université Libre de Bruxelles, Gosselies, Belgium; Center for Microscopy and Molecular Imaging (CMMI), Gosselies 6041, Belgium, and
| | - Anthony Rongvaux
- Department of Immunobiology, Yale University, New Haven, Connecticut 06520
| | - Oberdan Leo
- Laboratoire d'Immunobiologie, Université Libre de Bruxelles, Gosselies, Belgium; Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium,.
| |
Collapse
|
25
|
Wueest S, Mueller R, Blüher M, Item F, Chin ASH, Wiedemann MSF, Takizawa H, Kovtonyuk L, Chervonsky AV, Schoenle EJ, Manz MG, Konrad D. Fas (CD95) expression in myeloid cells promotes obesity‐induced muscle insulin resistance. EMBO Mol Med 2014; 6:43-56. [PMID: 24203314 PMCID: PMC3936487 DOI: 10.1002/emmm.201302962] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 09/30/2013] [Accepted: 10/01/2013] [Indexed: 12/12/2022] Open
Abstract
Low-grade inflammation in adipose tissue and liver has been implicated in obesity-associated insulin resistance and type 2 diabetes. Yet, the contribution of inflammatory cells to the pathogenesis of skeletal muscle insulin resistance remains elusive. In a large cohort of obese human individuals, blood monocyte Fas (CD95) expression correlated with systemic and skeletal muscle insulin resistance. To test a causal role for myeloid cell Fas expression in the development of skeletal muscle insulin resistance, we generated myeloid/haematopoietic cell-specific Fas-depleted mice. Myeloid/haematopoietic Fas deficiency prevented the development of glucose intolerance in high fat-fed mice, in ob/ob mice, and in mice acutely challenged by LPS. In vivo, ex vivo and in vitro studies demonstrated preservation of muscle insulin responsiveness with no effect on adipose tissue or liver. Studies using neutralizing antibodies demonstrated a role for TNFα as mediator between myeloid Fas and skeletal muscle insulin resistance, supported by significant correlations between monocyte Fas expression and circulating TNFα in humans. In conclusion, our results demonstrate an unanticipated crosstalk between myeloid cells and skeletal muscle in the development of obesity-associated insulin resistance.
Collapse
Affiliation(s)
- Stephan Wueest
- Division of Pediatric Endocrinology and Diabetology, University Children's HospitalZurich, Switzerland
- Children's Research Centre, University Children's HospitalZurich, Switzerland
| | - Rouven Mueller
- Division of Hematology, University Hospital ZurichZurich, Switzerland
| | - Matthias Blüher
- University of Leipzig, Department of MedicineLeipzig, Germany
| | - Flurin Item
- Division of Pediatric Endocrinology and Diabetology, University Children's HospitalZurich, Switzerland
- Children's Research Centre, University Children's HospitalZurich, Switzerland
| | - Annie S H Chin
- Division of Pediatric Endocrinology and Diabetology, University Children's HospitalZurich, Switzerland
- Children's Research Centre, University Children's HospitalZurich, Switzerland
- Zurich Centre for Integrative Human Physiology, University of ZurichZurich, Switzerland
| | - Michael S F Wiedemann
- Division of Pediatric Endocrinology and Diabetology, University Children's HospitalZurich, Switzerland
- Children's Research Centre, University Children's HospitalZurich, Switzerland
| | - Hitoshi Takizawa
- Division of Hematology, University Hospital ZurichZurich, Switzerland
| | - Larisa Kovtonyuk
- Division of Hematology, University Hospital ZurichZurich, Switzerland
| | | | - Eugen J Schoenle
- Division of Pediatric Endocrinology and Diabetology, University Children's HospitalZurich, Switzerland
- Children's Research Centre, University Children's HospitalZurich, Switzerland
| | - Markus G Manz
- Division of Hematology, University Hospital ZurichZurich, Switzerland
| | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology, University Children's HospitalZurich, Switzerland
- Children's Research Centre, University Children's HospitalZurich, Switzerland
- Zurich Centre for Integrative Human Physiology, University of ZurichZurich, Switzerland
- *Corresponding author: Tel: +41 44 266 7966; Fax: +41 44 266 7983; E-mail:
| |
Collapse
|
26
|
Trairatphisan P, Mizera A, Pang J, Tantar AA, Sauter T. optPBN: an optimisation toolbox for probabilistic Boolean networks. PLoS One 2014; 9:e98001. [PMID: 24983623 PMCID: PMC4077690 DOI: 10.1371/journal.pone.0098001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 04/27/2014] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND There exist several computational tools which allow for the optimisation and inference of biological networks using a Boolean formalism. Nevertheless, the results from such tools yield only limited quantitative insights into the complexity of biological systems because of the inherited qualitative nature of Boolean networks. RESULTS We introduce optPBN, a Matlab-based toolbox for the optimisation of probabilistic Boolean networks (PBN) which operates under the framework of the BN/PBN toolbox. optPBN offers an easy generation of probabilistic Boolean networks from rule-based Boolean model specification and it allows for flexible measurement data integration from multiple experiments. Subsequently, optPBN generates integrated optimisation problems which can be solved by various optimisers. In term of functionalities, optPBN allows for the construction of a probabilistic Boolean network from a given set of potential constitutive Boolean networks by optimising the selection probabilities for these networks so that the resulting PBN fits experimental data. Furthermore, the optPBN pipeline can also be operated on large-scale computational platforms to solve complex optimisation problems. Apart from exemplary case studies which we correctly inferred the original network, we also successfully applied optPBN to study a large-scale Boolean model of apoptosis where it allows identifying the inverse correlation between UVB irradiation, NFκB and Caspase 3 activations, and apoptosis in primary hepatocytes quantitatively. Also, the results from optPBN help elucidating the relevancy of crosstalk interactions in the apoptotic network. SUMMARY The optPBN toolbox provides a simple yet comprehensive pipeline for integrated optimisation problem generation in the PBN formalism that can readily be solved by various optimisers on local or grid-based computational platforms. optPBN can be further applied to various biological studies such as the inference of gene regulatory networks or the identification of the interaction's relevancy in signal transduction networks.
Collapse
Affiliation(s)
- Panuwat Trairatphisan
- Systems Biology Group, Life Sciences Research Unit, University of Luxembourg, Luxembourg, Luxembourg
| | - Andrzej Mizera
- Computer Science and Communications Research Unit, University of Luxembourg, Luxembourg, Luxembourg
| | - Jun Pang
- Computer Science and Communications Research Unit, University of Luxembourg, Luxembourg, Luxembourg
- Interdisciplinary Centre for Security, Reliability and Trust, University of Luxembourg, Luxembourg, Luxembourg
| | - Alexandru Adrian Tantar
- Interdisciplinary Centre for Security, Reliability and Trust, University of Luxembourg, Luxembourg, Luxembourg
| | - Thomas Sauter
- Systems Biology Group, Life Sciences Research Unit, University of Luxembourg, Luxembourg, Luxembourg
| |
Collapse
|
27
|
Konrath F, Witt J, Sauter T, Kulms D. Identification of new IκBα complexes by an iterative experimental and mathematical modeling approach. PLoS Comput Biol 2014; 10:e1003528. [PMID: 24675998 PMCID: PMC3967930 DOI: 10.1371/journal.pcbi.1003528] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 02/03/2014] [Indexed: 11/21/2022] Open
Abstract
The transcription factor nuclear factor kappa-B (NFκB) is a key regulator of pro-inflammatory and pro-proliferative processes. Accordingly, uncontrolled NFκB activity may contribute to the development of severe diseases when the regulatory system is impaired. Since NFκB can be triggered by a huge variety of inflammatory, pro-and anti-apoptotic stimuli, its activation underlies a complex and tightly regulated signaling network that also includes multi-layered negative feedback mechanisms. Detailed understanding of this complex signaling network is mandatory to identify sensitive parameters that may serve as targets for therapeutic interventions. While many details about canonical and non-canonical NFκB activation have been investigated, less is known about cellular IκBα pools that may tune the cellular NFκB levels. IκBα has so far exclusively been described to exist in two different forms within the cell: stably bound to NFκB or, very transiently, as unbound protein. We created a detailed mathematical model to quantitatively capture and analyze the time-resolved network behavior. By iterative refinement with numerous biological experiments, we yielded a highly identifiable model with superior predictive power which led to the hypothesis of an NFκB-lacking IκBα complex that contains stabilizing IKK subunits. We provide evidence that other but canonical pathways exist that may affect the cellular IκBα status. This additional IκBα:IKKγ complex revealed may serve as storage for the inhibitor to antagonize undesired NFκB activation under physiological and pathophysiological conditions. In unstimulated cells, the transcription factor NFκB resides in the cytosol bound to its inhibitor IκBα. Canonical activation of NFκB by numerous stimuli leads to proteasomal depletion of IκBα, thereby liberating NFκB to translocate into the nucleus to induce transcription of genes leading to proliferation, angiogenesis, metastasis, or chronic inflammation. Consequently, only transient activity needs to be warranted by immediate NFκB-dependent induction of negative regulatory mechanisms, including up-regulation of its inhibitor IκBα. Resynthesized IκBα consequently terminates NFκB activity by binding to its nuclear localization sequence. However, under physiological or pathophysiological conditions, random NFκB activation may occur, which needs to be avoided in order to guarantee proper cellular function. Using detailed dynamical modeling, we have now identified an additional IκBα containing complex to exist in un-stimulated cells which lacks NFκB but includes IKKγ (IκBα:IKKγ complex). This additional IκBα is not depleted from cells in the canonical fashion and may therefore serve as a cellular backup to avoid random NFκB activation.
Collapse
Affiliation(s)
- Fabian Konrath
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Institute for System Dynamics, University of Stuttgart, Stuttgart, Germany
| | - Johannes Witt
- Institute for System Dynamics, University of Stuttgart, Stuttgart, Germany
| | - Thomas Sauter
- Life Sciences Research Unit, University of Luxembourg, Luxembourg, Luxembourg
| | - Dagmar Kulms
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
- Experimental Dermatology, Department of Dermatology, TU-Dresden, Dresden, Germany
- * E-mail:
| |
Collapse
|
28
|
Liu Q, Tan Q, Zheng Y, Chen K, Qian C, Li N, Wang Q, Cao X. Blockade of Fas signaling in breast cancer cells suppresses tumor growth and metastasis via disruption of Fas signaling-initiated cancer-related inflammation. J Biol Chem 2014; 289:11522-11535. [PMID: 24627480 DOI: 10.1074/jbc.m113.525014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Mechanisms for cancer-related inflammation remain to be fully elucidated. Non-apoptotic functions of Fas signaling have been proposed to play an important role in promoting tumor progression. It has yet to be determined if targeting Fas signaling can control tumor progression through suppression of cancer-related inflammation. In the current study we found that breast cancer cells with constitutive Fas expression were resistant to apoptosis induction by agonistic anti-Fas antibody (Jo2) ligation or Fas ligand cross-linking. Higher expression of Fas in human breast cancer tissue has been significantly correlated with poorer prognosis in breast cancer patients. To determine whether blockade of Fas signaling in breast cancer could suppress tumor progression, we prepared an orthotopic xenograft mouse model with mammary cancer cells 4T1 and found that blockade of Fas signaling in 4T1 cancer cells markedly reduced tumor growth, inhibited tumor metastasis in vivo, and prolonged survival of tumor-bearing mice. Mechanistically, blockade of Fas signaling in cancer cells significantly decreased systemic or local recruitment of myeloid derived suppressor cells (MDSCs) in vivo. Furthermore, blockade of Fas signaling markedly reduced IL-6, prostaglandin E2 production from breast cancer cells by impairing p-p38, and activity of the NFκB pathway. In addition, administration of a COX-2 inhibitor and anti-IL-6 antibody significantly reduced MDSC accumulation in vivo. Therefore, blockade of Fas signaling can suppress breast cancer progression by inhibiting proinflammatory cytokine production and MDSC accumulation, indicating that Fas signaling-initiated cancer-related inflammation in breast cancer cells may be a potential target for treatment of breast cancer.
Collapse
Affiliation(s)
- Qiuyan Liu
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai 200433, China,.
| | - Qinchun Tan
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China, and
| | - Yuanyuan Zheng
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | - Kun Chen
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China, and
| | - Cheng Qian
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | - Nan Li
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai 200433, China
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China, and
| | - Xuetao Cao
- National Key Laboratory of Medical Immunology and Institute of Immunology, Second Military Medical University, Shanghai 200433, China,; Institute of Immunology, Zhejiang University School of Medicine, Hangzhou 310058, China, and; National Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing 100005, China.
| |
Collapse
|
29
|
Matsuda I, Matsuo K, Matsushita Y, Haruna Y, Niwa M, Kataoka T. The C-terminal domain of the long form of cellular FLICE-inhibitory protein (c-FLIPL) inhibits the interaction of the caspase 8 prodomain with the receptor-interacting protein 1 (RIP1) death domain and regulates caspase 8-dependent nuclear factor κB (NF-κB) activation. J Biol Chem 2014; 289:3876-87. [PMID: 24398693 DOI: 10.1074/jbc.m113.506485] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Caspase 8 plays an essential role in the regulation of apoptotic and non-apoptotic signaling pathways. The long form of cellular FLICE-inhibitory protein (c-FLIPL) has been shown previously to regulate caspase 8-dependent nuclear factor κB (NF-κB) activation by receptor-interacting protein 1 (RIP1) and TNF receptor-associated factor 2 (TRAF2). In this study, the molecular mechanism by which c-FLIPL regulates caspase 8-dependent NF-κB activation was further explored in the human embryonic kidney cell line HEK 293 and variant cells barely expressing caspase 8. The caspase inhibitor benzyloxycarbonyl-Val-Ala-Asp(OMe)-fluoromethylketone greatly diminished caspase 8-dependent NF-κB activation induced by Fas ligand (FasL) when c-FLIPL, but not its N-terminal fragment c-FLIP(p43), was expressed. The prodomain of caspase 8 was found to interact with the RIP1 death domain and to be sufficient to mediate NF-κB activation induced by FasL or c-FLIP(p43). The interaction of the RIP1 death domain with caspase 8 was inhibited by c-FLIPL but not c-FLIP(p43). Thus, these results reveal that the C-terminal domain of c-FLIPL specifically inhibits the interaction of the caspase 8 prodomain with the RIP1 death domain and, thereby, regulates caspase 8-dependent NF-κB activation.
Collapse
Affiliation(s)
- Iyo Matsuda
- From the Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan and
| | | | | | | | | | | |
Collapse
|
30
|
Yunusova T, Akhtar M, Poltoratsky V. Analysis of LPS-induced, NFκB-dependent interleukin-8 transcription in kidney embryonic cell line expressing TLR4 using luciferase assay. Methods Mol Biol 2014; 1172:305-14. [PMID: 24908317 DOI: 10.1007/978-1-4939-0928-5_28] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Gene expression is orchestrated by a complex network of signal transduction pathways that typically originate on cell surface receptors and culminate in DNA-binding transcription factors, which translocate to the nucleus and bind cis-regulatory elements in promoter regions of genes, thereby inducing de novo synthesis of the nascent RNA transcripts and their splicing. Gene expression arrays monitor abundance of the matured, spliced cDNA, which undergoes additional posttranscriptional modifications that greatly affect the half-life of the cDNA. Thus, the relative abundance of cDNA is not necessarily commensurable with the activity of promoters of the corresponding genes. In contrast, reporter gene assays provide valuable insight into the regulation of gene expression at the level of transcription and allow for discerning the contribution of individual transcription factors into changes in gene expression. Here, we describe a robust reporter gene assay method that is useful for exploration of transcription regulatory network, which regulates gene expression in response to inflammation. The method is exemplified by using the promoter region of the prototypic pro-inflammatory chemokine interleukin-8 (IL-8, CXCL8), which plays an important role in immune response as well as carcinogenesis. Using the luciferase reporter gene assay, we analyze the activation status of the IL-8 promoter in lipopolysaccharide (LPS)-stimulated human embryonic kidney cells.
Collapse
Affiliation(s)
- Tamara Yunusova
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439, USA
| | | | | |
Collapse
|
31
|
Kragh CL, Gysbers AM, Rockenstein E, Murphy K, Halliday GM, Masliah E, Jensen PH. Prodegenerative IκBα expression in oligodendroglial α-synuclein models of multiple system atrophy. Neurobiol Dis 2013; 63:171-83. [PMID: 24361600 DOI: 10.1016/j.nbd.2013.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 12/04/2013] [Indexed: 12/11/2022] Open
Abstract
Multiple system atrophy is a progressive, neurodegenerative disease characterized by parkinsonism, ataxia, autonomic dysfunction, and accumulation of α-synuclein in oligodendrocytes. To understand how α-synuclein aggregates impact oligodendroglial homeostasis, we investigated an oligodendroglial cell model of α-synuclein dependent degeneration and identified responses linked to the NF-κB transcription factor stress system. Coexpression of human α-synuclein and the oligodendroglial protein p25α increased the expression of IκBα mRNA and protein early during the degenerative process and this was dependent on both aggregation and Ser129 phosphorylation of α-synuclein. This response was prodegenerative because blocking IκBα expression by siRNA rescued the cells. IκBα is an inhibitor of NF-κB and acts by binding and retaining NF-κB p65 in the cytoplasm. The protection obtained by silencing IκBα was accompanied by a strong increase in nuclear p65 translocation indicating that NF-κB activation protects against α-synuclein aggregate stress. In the cellular model, two different phenotypes were observed; degenerating cells retracting their microtubules and resilient cells tolerating the coexpression of α-synuclein and p25α. The resilient cells displayed a significant higher nuclear translocation of p65 and activation of the NF-κB system relied on stress elicited by aggregated and Ser129 phosphorylated α-synuclein. To validate the relationship between oligodendroglial α-synuclein expression and IκBα, we analyzed two different lines of transgenic mice expressing human α-synuclein under the control of the oligodendrocytic MBP promotor (intermediate-expresser line 1 and high-expresser line 29). IκBα mRNA expression was increased in both lines and immunofluorescence microscopy and in situ hybridization revealed that IκBα mRNA and protein is expressed in oligodendrocytes. IκBα mRNA expression was demonstrated prior to activation of microglia and astrocytes in line 1. Human brain tissue affected by MSA displayed increased expression of IκBα and NF-κB p65 in some oligodendrocytes containing glial cytoplasmic inclusions. Our data suggest that oligodendroglial IκBα expression and NF-κB are activated early in the course of MSA and their balance contributes to the decision of cellular demise. Favoring oligodendroglial NF-κB activation may represent a therapeutic strategy for this devastating disease.
Collapse
Affiliation(s)
- Christine L Kragh
- Department of Biomedicine & Danish Research Institute of Translational Neuroscience-DANDRITE, University of Aarhus, Aarhus, Denmark
| | - Amanda M Gysbers
- Neuroscience Research Australia, Sydney, NSW, Australia; University of New South Wales, Sydney, NSW, Australia
| | - Edward Rockenstein
- Department of Neurosciences and Pathology, University of California, San Diego, School of Medicine, La Jolla, CA 92093-0624, USA
| | - Karen Murphy
- Neuroscience Research Australia, Sydney, NSW, Australia; University of New South Wales, Sydney, NSW, Australia
| | - Glenda M Halliday
- Neuroscience Research Australia, Sydney, NSW, Australia; University of New South Wales, Sydney, NSW, Australia
| | - Eliezer Masliah
- Department of Neurosciences and Pathology, University of California, San Diego, School of Medicine, La Jolla, CA 92093-0624, USA
| | - Poul Henning Jensen
- Department of Biomedicine & Danish Research Institute of Translational Neuroscience-DANDRITE, University of Aarhus, Aarhus, Denmark.
| |
Collapse
|
32
|
Rationale and Means to Target Pro-Inflammatory Interleukin-8 (CXCL8) Signaling in Cancer. Pharmaceuticals (Basel) 2013; 6:929-59. [PMID: 24276377 PMCID: PMC3817732 DOI: 10.3390/ph6080929] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 07/03/2013] [Accepted: 07/29/2013] [Indexed: 12/13/2022] Open
Abstract
It is well established that chronic inflammation underpins the development of a number of human cancers, with pro-inflammatory signaling within the tumor microenvironment contributing to tumor progression and metastasis. CXCL8 is an ELR+ pro-inflammatory CXC-chemokine which mediates its effects via signaling through two G protein-coupled receptors, CXCR1 and CXCR2. Elevated CXCL8-CXCR1/2 signaling within the tumor microenvironment of numerous cancers is known to enhance tumor progression via activation of signaling pathways promoting proliferation, angiogenesis, migration, invasion and cell survival. This review provides an overview of established roles of CXCL8-CXCR1/2 signaling in cancer and subsequently, discusses the possible strategies of targeting CXCL8-CXCR1/2 signaling in cancer, covering indirect strategies (e.g., anti-inflammatories, NFκB inhibitors) and direct CXCL8 or CXCR1/2 inhibition (e.g., neutralizing antibodies, small molecule receptor antagonists, pepducin inhibitors and siRNA strategies). Reports of pre-clinical cancer studies and clinical trials using CXCL8-CXCR1/2-targeting strategies for the treatment of inflammatory diseases will be discussed. The future translational opportunities for use of such agents in oncology will be discussed, with emphasis on exploitation in stratified populations.
Collapse
|
33
|
Herrero R, Tanino M, Smith LS, Kajikawa O, Wong VA, Mongovin S, Matute-Bello G, Martin TR. The Fas/FasL pathway impairs the alveolar fluid clearance in mouse lungs. Am J Physiol Lung Cell Mol Physiol 2013; 305:L377-88. [PMID: 23812636 DOI: 10.1152/ajplung.00271.2012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Alveolar epithelial damage is a critical event that leads to protein-rich edema in acute lung injury (ALI), but the mechanisms leading to epithelial damage are not completely understood. Cell death by necrosis and apoptosis occurs in alveolar epithelial cells in the lungs of patients with ALI. Fas activation induces apoptosis of alveolar epithelial cells, but its role in the formation of lung edema is unclear. The main goal of this study was to determine whether activation of the Fas/Fas ligand pathway in the lungs could alter the function of the lung epithelium, and the mechanisms involved. The results show that Fas activation alters the alveolar barrier integrity and impairs the ability of the lung alveolar epithelium to reabsorb fluid from the air spaces. This result was dependent on the presence of a normal Fas receptor and was not affected by inflammation induced by Fas activation. Alteration of the fluid transport properties of the alveolar epithelium was partially restored by β-adrenergic stimulation. Fas activation also caused apoptosis of alveolar endothelial cells, but this effect was less pronounced than the effect on the alveolar epithelium. Thus, activation of the Fas pathway impairs alveolar epithelial function in mouse lungs by mechanisms involving caspase-dependent apoptosis, suggesting that targeting apoptotic pathways could reduce the formation of lung edema in ALI.
Collapse
Affiliation(s)
- Raquel Herrero
- Medical Research Service of the Veterans Affairs Puget Sound Health Care Center, Seattle, WA, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Messer MP, Kellermann P, Weber SJ, Hohmann C, Denk S, Klohs B, Schultze A, Braumüller S, Huber-Lang MS, Perl M. Silencing of fas, fas-associated via death domain, or caspase 3 differentially affects lung inflammation, apoptosis, and development of trauma-induced septic acute lung injury. Shock 2013; 39:19-27. [PMID: 23247118 DOI: 10.1097/shk.0b013e318277d856] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Activation of Fas signaling is a potentially important pathophysiological mechanism in the development of septic acute lung injury (ALI). However, so far the optimal targets within this signaling cascade remain elusive. Thus, we tested the hypothesis that in vivo gene silencing of Fas, Fas-associated via death domain (FADD), or caspase 3 by intratracheal administration of small interfering RNA would ameliorate ALI in a clinically relevant double-hit mouse model of trauma induced septic lung injury. Male C57Bl/6 mice received small interfering (Fas, FADD, caspase 3) or control RNA 24 h before and 12 h after blunt chest trauma or sham procedures. Polymicrobial sepsis was induced by cecal ligation and puncture 24 h after chest trauma. Twelve or 24 h later, lung tissue, plasma, and bronchoalveolar lavage fluid were harvested. During ALI, lung apoptosis (active caspase 3 Western blotting, TUNEL staining) was substantially increased when compared with sham. Silencing of caspase 3 or FADD both markedly reduced pulmonary apoptosis. Fas- and FADD-small interfering RNA administration substantially decreased lung cytokine concentration, whereas caspase 3 silencing did not reduce lung inflammation. In addition, Fas silencing markedly decreased lung neutrophil infiltration. Interestingly, only in response to caspase 3 silencing, ALI-induced lung epithelial barrier dysfunction was substantially improved, and histological appearance was beneficially affected. Taken together, downstream inhibition of lung apoptosis via caspase 3 silencing proved to be superior in mitigating ALI when compared with upstream inhibition of apoptosis via Fas or FADD silencing, even in the presence of additional anti-inflammatory effects. This indicates a major pathophysiological role of lung apoptosis and suggests the importance of other than Fas-driven apoptotic pathways in trauma-induced septic ALI.
Collapse
Affiliation(s)
- Mirko Philipp Messer
- Department of Trauma, Hand, Plastic, and Reconstructive Surgery, University of Ulm, Ulm, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Gerez J, Fuertes M, Tedesco L, Silberstein S, Sevlever G, Paez-Pereda M, Holsboer F, Turjanski AG, Arzt E. In silico structural and functional characterization of the RSUME splice variants. PLoS One 2013; 8:e57795. [PMID: 23469069 PMCID: PMC3585135 DOI: 10.1371/journal.pone.0057795] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Accepted: 01/29/2013] [Indexed: 01/09/2023] Open
Abstract
RSUME (RWD-containing SUMO Enhancer) is a small protein that increases SUMO conjugation to proteins. To date, four splice variants that codify three RSUME isoforms have been described, which differ in their C-terminal end. Comparing the structure of the RSUME isoforms we found that, in addition to the previously described RWD domain in the N-terminal, all these RSUME variants also contain an intermediate domain. Only the longest RSUME isoform presents a C-terminal domain that is absent in the others. Given these differences, we used the shortest and longest RSUME variants for comparative studies. We found that the C-terminal domain is dispensable for the SUMO-conjugation enhancer properties of RSUME. We also demonstrate that these two RSUME variants are equally induced by hypoxia. The NF-κB signaling pathway is inhibited and the HIF-1 pathway is increased more efficiently by the longest RSUME, by means of a greater physical interaction of RSUME267 with the target proteins. In addition, the mRNA and protein levels of these isoforms differ in human glioma samples; while the shortest RSUME isoform is expressed in all the tumors analyzed, the longest variant is expressed in most but not all of them. The results presented here show a degree of redundancy of the RSUME variants on the SUMO pathway. However, the increased inhibition conferred by RSUME267 over the NF-κB signaling pathway, the increased activation over the HIF-1 pathway and the different expression of the RSUME isoforms suggest specific roles for each RSUME isoform which may be relevant in certain types of brain tumors that express RSUME, like human pituitary adenomas and gliomas.
Collapse
Affiliation(s)
- Juan Gerez
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET- Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Mariana Fuertes
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET- Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Lucas Tedesco
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET- Partner Institute of the Max Planck Society, Buenos Aires, Argentina
| | - Susana Silberstein
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET- Partner Institute of the Max Planck Society, Buenos Aires, Argentina
- Departamento de Fisiología y Biología Molecular, FCEN, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gustavo Sevlever
- Departamento de Neuropatología y Biología Molecular, FLENI, Buenos Aires, Argentina
| | | | | | - Adrián G. Turjanski
- Laboratorio de Bioinformática Estructural, Departamento de Química Biológica, INQUIMAE-CONICET, FCEN, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Eduardo Arzt
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA)-CONICET- Partner Institute of the Max Planck Society, Buenos Aires, Argentina
- Departamento de Fisiología y Biología Molecular, FCEN, Universidad de Buenos Aires, Buenos Aires, Argentina
- Max Planck Institute of Psychiatry, Munich, Germany
- * E-mail:
| |
Collapse
|
36
|
Gaud G, Guillemot D, Jacob Y, Favre M, Vuillier F. EVER2 protein binds TRADD to promote TNF-α-induced apoptosis. Cell Death Dis 2013; 4:e499. [PMID: 23429285 PMCID: PMC3734840 DOI: 10.1038/cddis.2013.27] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
EVER1 and 2 confer resistance to cutaneous oncogenic human papillomavirus infections by downregulating the activating protein 1 (AP-1) signaling pathway. Defects in their expression are associated with susceptibility to epidermodysplasia verruciformis, which is characterized by persistent β-HPV infection, tumor necrosis factor alpha (TNF-α) overproduction in keratinocytes and the development of skin cancers. TNF-α-induced apoptosis is a key defense strategy, preventing the persistence of the virus within cells, but the role of EVER proteins in this cell death mechanism triggered by extrinsic stimuli is unknown. We show here that EVER2 induces TNF-α- and TRAIL-dependant apoptosis. It interacts with the N-terminal domain of TRADD, impairs the recruitment of TRAF2 and RIPK1 and promotes apoptosis. The skin cancer-associated EVER2 I306 allele results in an impaired TRADD-EVER2 interaction, with lower levels of cell death following treatment with TNF-α. These data highlight a new, critical function of EVER2 in controlling cell survival in response to death stimuli.
Collapse
Affiliation(s)
- G Gaud
- Unité de Génétique, Papillomavirus et Cancer Humain, Institut Pasteur, Paris, France
| | | | | | | | | |
Collapse
|
37
|
Kragh CL, Fillon G, Gysbers A, Hansen HD, Neumann M, Richter-Landsberg C, Haass C, Zalc B, Lubetzki C, Gai WP, Halliday GM, Kahle PJ, Jensen PH. FAS-dependent cell death in α-synuclein transgenic oligodendrocyte models of multiple system atrophy. PLoS One 2013; 8:e55243. [PMID: 23372841 PMCID: PMC3555893 DOI: 10.1371/journal.pone.0055243] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 12/27/2012] [Indexed: 12/26/2022] Open
Abstract
Multiple system atrophy is a parkinsonian neurodegenerative disorder. It is cytopathologically characterized by accumulation of the protein p25α in cell bodies of oligodendrocytes followed by accumulation of aggregated α-synuclein in so-called glial cytoplasmic inclusions. p25α is a stimulator of α-synuclein aggregation, and coexpression of α-synuclein and p25α in the oligodendroglial OLN-t40-AS cell line causes α-synuclein aggregate-dependent toxicity. In this study, we investigated whether the FAS system is involved in α-synuclein aggregate dependent degeneration in oligodendrocytes and may play a role in multiple system atrophy. Using rat oligodendroglial OLN-t40-AS cells we demonstrate that the cytotoxicity caused by coexpressing α-synuclein and p25α relies on stimulation of the death domain receptor FAS and caspase-8 activation. Using primary oligodendrocytes derived from PLP-α-synuclein transgenic mice we demonstrate that they exist in a sensitized state expressing pro-apoptotic FAS receptor, which makes them sensitive to FAS ligand-mediated apoptosis. Immunoblot analysis shows an increase in FAS in brain extracts from multiple system atrophy cases. Immunohistochemical analysis demonstrated enhanced FAS expression in multiple system atrophy brains notably in oligodendrocytes harboring the earliest stages of glial cytoplasmic inclusion formation. Oligodendroglial FAS expression is an early hallmark of oligodendroglial pathology in multiple system atrophy that mechanistically may be coupled to α-synuclein dependent degeneration and thus represent a potential target for protective intervention.
Collapse
Affiliation(s)
| | - Gwenaëlle Fillon
- Laboratory for Alzheimer's and Parkinson's Disease Research, Department of Biochemistry, Ludwig Maximilians University, Munich, Germany
| | - Amanda Gysbers
- Neuroscience Research Australia and University of New South Wales, Sydney, New South Wales, Australia
| | - Hanne D. Hansen
- Laboratory of Functional Neurogenetics, Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | - Manuela Neumann
- Institute for Neuropathology, University of Zürich, Zürich, Switzerland
| | | | - Christian Haass
- Laboratory for Alzheimer's and Parkinson's Disease Research, Department of Biochemistry, Ludwig Maximilians University, Munich, Germany
| | - Bernard Zalc
- Centre de Recherche de l'Institut du Cerveau et de la Moelle épinière, Université Pierre et Marie Curie, Paris, France
| | - Catherine Lubetzki
- Centre de Recherche de l'Institut du Cerveau et de la Moelle épinière, Université Pierre et Marie Curie, Paris, France
| | - Wei-Ping Gai
- Department of Human Physiology and Centre for Neuroscience, Flinders University School of Medicine, Bedford Park, South Australia, Australia
| | - Glenda M. Halliday
- Neuroscience Research Australia and University of New South Wales, Sydney, New South Wales, Australia
| | - Philipp J. Kahle
- Laboratory of Functional Neurogenetics, Department of Neurodegeneration, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Center for Neurodegenerative Diseases, Tübingen, Germany
- * E-mail: (PHJ); (PJK)
| | - Poul H. Jensen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
- * E-mail: (PHJ); (PJK)
| |
Collapse
|
38
|
Oh J, Kim SH, Ahn S, Lee CE. Suppressors of cytokine signaling promote Fas-induced apoptosis through downregulation of NF-κB and mitochondrial Bfl-1 in leukemic T cells. THE JOURNAL OF IMMUNOLOGY 2012; 189:5561-71. [PMID: 23152563 DOI: 10.4049/jimmunol.1103415] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Suppressors of cytokine signaling (SOCS) are known as negative regulators of cytokine- and growth factor-induced signal transduction. Recently they have emerged as multifunctional proteins with regulatory roles in inflammation, autoimmunity, and cancer. We have recently reported that SOCS1 has antiapoptotic functions against the TNF-α- and the hydrogen peroxide-induced T cell apoptosis through the induction of thioredoxin, which protects protein tyrosine phosphatases and attenuates Jaks. In this study, we report that SOCS, on the contrary, promote death receptor Fas-mediated T cell apoptosis. The proapoptotic effect of SOCS1 was manifested with increases in Fas-induced caspase-8 activation, truncated Bid production, and mitochondrial dysfunctions. Both caspase-8 inhibitor c-Flip and mitochondrial antiapoptotic factor Bfl-1 were significantly reduced by SOCS1. These proapoptotic responses were not associated with changes in Jak or p38/Jnk activities but were accompanied with downregulation of NF-κB and NF-κB-dependent reporter gene expression. Indeed, p65 degradation via ubiquitination was accelerated in SOCS1 overexpressing cells, whereas it was attenuated in SOCS1 knockdown cells. With high NF-κB levels, the SOCS1-ablated cells displayed resistance against Fas-induced apoptosis, which was abrogated upon siBfl-1 transfection. The results indicate that the suppression of NF-κB-dependent induction of prosurvival factors, such as Bfl-1 and c-Flip, may serve as a mechanism for SOCS action to promote Fas-mediated T cell apoptosis. SOCS3 exhibited a similar proapoptotic function. Because both SOCS1 and SOCS3 are induced upon TCR stimulation, SOCS would play a role in activation-induced cell death by sensitizing activated T cells toward Fas-mediated apoptosis to maintain T cell homeostasis.
Collapse
Affiliation(s)
- Jiyoung Oh
- Laboratory of Immunology, Department of Biological Science, Sungkyunkwan University, Suwon 440-746, Korea
| | | | | | | |
Collapse
|
39
|
He X, Wang Y, Hu H, Zhang Z. In vitro and in vivo antimammary tumor activities and mechanisms of the apple total triterpenoids. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2012; 60:9430-6. [PMID: 22924395 DOI: 10.1021/jf3026925] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Consumption of apples has been linked to the prevention of various chronic diseases, including tumors and cardiovascular diseases. The apple total triterpenoid content (ATT) was extracted and concentrated from apple peels. The in vitro and in vivo antitumor activities, related antitumor mechanisms, were investigated. In vitro, ATT showed potent antiproliferative activities against human breast cancer (MCF-7, MDA-MB-231), human colon cancer (Caco-2), and human liver cancer (HepG2) cell lines. In vivo antitumor experiments showed that ATT could substantially reduce the occurrence and growth of mammary tumor with a good dose-dependent manner in a rat model. During the apoptosis in MDA-MB-231 cells induced by ATT, the caspase-independent pathway was involved in the process of apoptosis, and the mitochondrial transmembrane potential was markedly reduced. Also the PI3K/Akt/NF-κB pathway was activated. These results indicated that ATT-induced apoptosis of MDA-MB-231 cells may involve a mitochondrial-related pathway.
Collapse
Affiliation(s)
- Xiangjiu He
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | | | | | | |
Collapse
|
40
|
Furuichi K, Kokubo S, Hara A, Imamura R, Wang Q, Kitajima S, Toyama T, Okumura T, Matsushima K, Suda T, Mukaida N, Kaneko S, Wada T. Fas Ligand Has a Greater Impact than TNF-α on Apoptosis and Inflammation in Ischemic Acute Kidney Injury. NEPHRON EXTRA 2012; 2:27-38. [PMID: 22479266 PMCID: PMC3318938 DOI: 10.1159/000335533] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Background/Aim Fas ligand (FasL) and tumor necrosis factor (TNF)-α are major pro-apoptotic molecules and also induce inflammation through cytokine and chemokine production. Although precise intracellular mechanisms of action have been reported for each molecule, the differential impact of these molecules on kidney injury in vivo still requires clarification. Methods We explored the differential impact of FasL and TNF-α upon apoptosis and inflammation in ischemic acute kidney injury using neutralizing anti-FasL antibodies and TNF-α receptor 1 (TNFR1)-deficient mice. Results TNFR1 deficiency was associated with a lesser anti-inflammatory effect upon leukocyte infiltration and tubular necrosis than treatment with anti-FasL antibody. Furthermore, the number of TUNEL-positive cells was significantly reduced in anti-FasL antibody-treated mice, whereas it was only partially diminished in TNFR1-deficient mice. In vitro studies confirmed these findings. FasL administration induced both apoptosis and cytokine/chemokine production from cultured tubular epithelial cells. However, TNF-α had a limited effect upon tubular epithelial cells. Conclusion In ischemic acute kidney injury, FasL has a greater impact than TNF-α on the apoptosis and inflammatory reaction through cytokine/chemokine production from tubular epithelial cells.
Collapse
Affiliation(s)
- Kengo Furuichi
- Division of Blood Purification, Kanazawa University Hospital, Kanazawa University, Kanazawa, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ishizawa YH, Tamura K, Yamaguchi T, Matsumoto K, Komiyama M, Takamatsu N, Shiba T, Ito M. Xenopus death-domain-containing proteins FADD and RIP1 synergistically activate JNK and NF-κB. Biol Cell 2012; 98:465-78. [PMID: 16597320 DOI: 10.1042/bc20050091] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND INFORMATION Death receptors (DRs) induce intracellular signalling upon engagement of their cognate ligands, leading to apoptosis, cell survival or pro-inflammatory responses. In mammals, DR signalling is mediated by the recruitment of several DD (death domain)-containing molecules, such as FADD (Fas-associated DD) and RIP1 (receptor-interacting protein 1). RESULTS To elucidate the molecular mechanisms of intracellular DR signalling in Xenopus, we have isolated cDNAs encoding xFADD (Xenopus FADD), and xRIP1 and its short isoform xRIP1beta, which is produced by alternative splicing of the xRIP1 gene. These DD-containing proteins interacted with Xenopus DR members xDR-M1 and xDR-M2 through their DDs in co-transfected HEK-293T cells. Overexpression of xFADD activated not only xCaspase 8, but also AP-1 (activator protein 1), which reflects activation of JNK (c-Jun N-terminal kinase) and NF-kappaB (nuclear factor kappaB). A comparative analysis of xRIP1, a kinase-dead mutant of xRIP1 and xRIP1beta indicated that the kinase activity of xRIP1 was required for the activation of AP-1 and NF-kappaB. Interestingly, xFADD and xRIP1 interacted with each other via their DDs, and the expression of a mutant xRIP1 containing only the DD (xRIP1-DD) repressed the xFADD-induced activation of NF-kappaB and AP-1. xFADD and xRIP1 synergistically induced the activation of AP-1 and NF-kappaB, both of which were partially mediated by TRAF2 (tumour-necrosis-factor-receptor-associated factor 2) and TAK1 (transforming-growth-factor-beta-activated kinase 1). We also found that the activation pathways of NF-kappaB induced by xDR-M2 were inhibited by xRIP1-DD. CONCLUSIONS Xenopus FADD, RIP1 and its splice variant RIP1beta have been characterized. Interaction of xFADD and xRIP1 induced synergistic activation of JNK and NF-kappaB. In addition, the NF-kappaB activation induced by xDR-M2 was partially mediated by xRIP1.
Collapse
Affiliation(s)
- Yo-hei Ishizawa
- Department of Bioscience, School of Science, Kitasato University, 1-15-1 Kitasato, Sagamihara 228-8555, Japan
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Chen L. Okadaic acid induces apoptosis through the PKR, NF-κB and caspase pathway in human osteoblastic osteosarcoma MG63 cells. Toxicol In Vitro 2011; 25:1796-802. [PMID: 21964477 DOI: 10.1016/j.tiv.2011.09.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 09/05/2011] [Accepted: 09/18/2011] [Indexed: 11/24/2022]
Abstract
Okadaic acid (OA) is the major component of diarrheic shellfish poisoning toxins and a potent inhibitor of protein phosphatase 1 and 2A. However, the underlying regulatory mechanisms involved in OA-induced cell death are not well understood. In the present study, we examined the effects of OA on apoptosis of MG63 cells by characterizing apoptotic morphological changes of the cells and DNA fragmentation. The roles of double-stranded RNA-dependent protein kinase (PKR), nuclear factor-κB (NF-κB) and caspase in OA-mediated apoptosis in MG63 cells were also examined. Results showed that OA induced cytotoxicity and apoptosis in MG63 cells at IC50 of 75 nM. A functional PKR pathway is required to induce apoptosis in response to OA treatment. Blockade of NF-κB by ammonium pyrrolidinedithiocarbamate (PDTC) resulted in down-regulation of apoptosis. The caspase-3 and caspase-8 inhibitors blocked apoptosis in MG63 cells. In conclusion, our results imply that OA can induce MG63 cell apoptosis through the PKR, NF-κB and caspase pathway.
Collapse
Affiliation(s)
- Ling Chen
- Department of Histology and Oral Histology, Institute of Health Biosciences, University of Tokushima Graduate School, Kuramoto, Tokushima 770-8504, Japan.
| |
Collapse
|
43
|
Herrero R, Kajikawa O, Matute-Bello G, Wang Y, Hagimoto N, Mongovin S, Wong V, Park DR, Brot N, Heinecke JW, Rosen H, Goodman RB, Fu X, Martin TR. The biological activity of FasL in human and mouse lungs is determined by the structure of its stalk region. J Clin Invest 2011; 121:1174-90. [PMID: 21285513 DOI: 10.1172/jci43004] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2010] [Accepted: 12/08/2010] [Indexed: 01/20/2023] Open
Abstract
Acute lung injury (ALI) is a life-threatening condition in critically ill patients. Injury to the alveolar epithelium is a critical event in ALI, and accumulating evidence suggests that it is linked to proapoptotic Fas/FasL signals. Active soluble FasL (sFasL) is detectable in the bronchoalveolar lavage (BAL) fluid of patients with ALI, but the mechanisms controlling its bioactivity are unclear. We therefore investigated how the structure of sFasL influences cellular activation in human and mouse lungs and the role of oxidants and proteases in modifying sFasL activity. The sFasL in BAL fluid from patients with ALI was bioactive and present in high molecular weight multimers and aggregates. Oxidants generated from neutrophil myeloperoxidase in BAL fluid promoted aggregation of sFasL in vitro and in vivo. Oxidation increased the biological activity of sFasL at low concentrations but degraded sFasL at high concentrations. The amino-terminal extracellular stalk region of human sFasL was required to induce lung injury in mice, and proteolytic cleavage of the stalk region by MMP-7 reduced the bioactivity of sFasL in human cells in vitro. The sFasL recovered from the lungs of patients with ALI contained both oxidized methionine residues and the stalk region. These data provide what we believe to be new insights into the structural determinants of sFasL bioactivity in the lungs of patients with ALI.
Collapse
Affiliation(s)
- Raquel Herrero
- Medical Research Service of the VA Puget Sound Health Center, Seattle, Washington, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Pro- and anti-apoptotic CD95 signaling in T cells. Cell Commun Signal 2011; 9:7. [PMID: 21477291 PMCID: PMC3090738 DOI: 10.1186/1478-811x-9-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Accepted: 04/08/2011] [Indexed: 12/20/2022] Open
Abstract
The TNF receptor superfamily member CD95 (Fas, APO-1, TNFRSF6) is known as the prototypic death receptor in and outside the immune system. In fact, many mechanisms involved in apoptotic signaling cascades were solved by addressing consequences and pathways initiated by CD95 ligation in activated T cells or other "CD95-sensitive" cell populations. As an example, the binding of the inducible CD95 ligand (CD95L) to CD95 on activated T lymphocytes results in apoptotic cell death. This activation-induced cell death was implicated in the control of immune cell homeostasis and immune response termination. Over the past years, however, it became evident that CD95 acts as a dual function receptor that also exerts anti-apoptotic effects depending on the cellular context. Early observations of a potential non-apoptotic role of CD95 in the growth control of resting T cells were recently reconsidered and revealed quite unexpected findings regarding the costimulatory capacity of CD95 for primary T cell activation. It turned out that CD95 engagement modulates TCR/CD3-driven signal initiation in a dose-dependent manner. High doses of immobilized CD95 agonists or cellular CD95L almost completely silence T cells by blocking early TCR-induced signaling events. In contrast, under otherwise unchanged conditions, lower amounts of the same agonists dramatically augment TCR/CD3-driven activation and proliferation. In the present overview, we summarize these recent findings with a focus on the costimulatory capacity of CD95 in primary T cells and discuss potential implications for the T cell compartment and the interplay between T cells and CD95L-expressing cells including antigen-presenting cells.
Collapse
|
45
|
Lim DS, Bae YS. Metastatic lymph node 51 and fibroblast-like synoviocyte hyperproliferation in rheumatoid arthritis pathogenesis. Rheumatol Int 2011; 31:843-7. [PMID: 21327427 DOI: 10.1007/s00296-011-1818-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Accepted: 01/30/2011] [Indexed: 12/26/2022]
Abstract
One of the varied characteristic features of the pathogenesis of rheumatoid arthritis (RA) is synovial hyperplasia. Fibroblast-like synoviocytes (FLSs) play a key role in the development of sustained inflammation in arthritic joints. We have reported previously that metastatic lymph node 51 (MLN51) is involved in the proliferation of FLSs in the pathogenesis of RA. Interestingly, the overexpression of MLN51 was observed only in RA FLSs, but not in osteoarthritis FLSs, possibly expecting that MLN51 may be a RA-specific marker. Additionally, we found that granulocyte-macrophage colony-stimulating factor signaling activates mitogen-activated protein kinase, followed by the upregulation of MLN51 and FLICE-inhibitory protein, resulting in FLS hyperplasia in RA. Based on these studies, we could be firm that MLN51 is a key factor in FLS hyperplasia of RA patients.
Collapse
Affiliation(s)
- Dae-Seog Lim
- Department of Applied Bioscience, Laboratory for Immune Cell-based Therapy, CHA University, 222 Yatap-dong, Bundang-gu, Sungnam, Gyeonggi-do, 463-836, Korea.
| | | |
Collapse
|
46
|
Meley D, Spiller DG, White MRH, McDowell H, Pizer B, Sée V. p53-mediated delayed NF-κB activity enhances etoposide-induced cell death in medulloblastoma. Cell Death Dis 2010; 1:e41. [PMID: 21364648 PMCID: PMC3032310 DOI: 10.1038/cddis.2010.16] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Medulloblastoma (MB) is an embryonic brain tumour that arises in the cerebellum. Using several MB cell lines, we have demonstrated that the chemotherapeutic drug etoposide induces a p53- and caspase-dependent cell death. We have observed an additional caspase-independent cell death mechanism involving delayed nuclear factor κB (NF-κB) activity. The delayed induction was controlled by a p53-dependent transcription step and the production of death receptors (especially CD95/Fas). We further demonstrated that in both MB and glioblastoma (GM) cell lines, in which the p53 pathway was not functional, no p65 activation could be detected upon etoposide treatment. MB cell lines that have mutations in p53 or NF-κB are either less sensitive (NF-κB mutant) or even completely resistant (p53 mutant) to chemotherapeutic intervention. The optimal cell death was only achieved when both p53 and NF-κB were switched on. Taken together, our results shed light on the mechanism of NF-κB activation by etoposide in brain tumours and show that the genetic background of MB and GM cells determines their sensitivity to chemotherapy and has to be taken into account for efficient therapeutic intervention.
Collapse
Affiliation(s)
- D Meley
- Centre for Cell Imaging, School of Biological Sciences, University of Liverpool, Liverpool, UK
| | | | | | | | | | | |
Collapse
|
47
|
Imamura R, Wang Y, Kinoshita T, Suzuki M, Noda T, Sagara J, Taniguchi S, Okamoto H, Suda T. Anti-inflammatory activity of PYNOD and its mechanism in humans and mice. THE JOURNAL OF IMMUNOLOGY 2010; 184:5874-84. [PMID: 20393137 DOI: 10.4049/jimmunol.0900779] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Many members of the nucleotide-binding and oligomerization domain (NOD)- and leucine-rich-repeat-containing protein (NLR) family play important roles in pathogen recognition and inflammation. However, we previously reported that human PYNOD/NLRP10, an NLR-like protein consisting of a pyrin domain and a NOD, inhibits inflammatory signal mediated by caspase-1 and apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) in reconstitution experiments using HEK293 cells. In this study, we investigated the molecular mechanism of PYNOD's anti-inflammatory activity in vitro and its expression and function in mice. Human PYNOD inhibited the autoprocessing of caspase-1 and caspase-1-mediated IL-1beta processing and suppressed the aggregation of ASC, a hallmark of ASC activation. Interestingly, the NOD of human PYNOD was sufficient to inhibit caspase-1-mediated IL-1beta secretion, whereas its pyrin domain was sufficient to inhibit ASC-mediated NF-kappaB activation and apoptosis and to reduce ASC's ability to promote caspase-1-mediated IL-1beta production. Mouse PYNOD protein was detected in the skin, tongue, heart, colon, peritoneal macrophages, and several cell lines of hematopoietic and myocytic lineages. Mouse PYNOD colocalized with ASC aggregates in LPS + R837-stimulated macrophages; however, unlike human PYNOD, mouse PYNOD failed to inhibit ASC aggregation. Macrophages and neutrophils from PYNOD-transgenic mice exhibited reduced IL-1beta processing and secretion upon microbial infection, although mouse PYNOD failed to inhibit caspase-1 processing, which was inhibited by caspase-4 inhibitor z-LEED-fluoromethylketone. These results suggest that mouse PYNOD colocalizes with ASC and inhibits caspase-1-mediated IL-1beta processing without inhibiting caspase-4 (mouse caspase-11)-mediated caspase-1 processing. Furthermore, PYNOD-transgenic mice were resistant to lethal endotoxic shock. Thus, PYNOD is the first example of an NLR that possesses an anti-inflammatory function in vivo.
Collapse
Affiliation(s)
- Ryu Imamura
- Division of Immunology and Molecular Biology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Ehrenschwender M, Siegmund D, Wicovsky A, Kracht M, Dittrich-Breiholz O, Spindler V, Waschke J, Kalthoff H, Trauzold A, Wajant H. Mutant PIK3CA licenses TRAIL and CD95L to induce non-apoptotic caspase-8-mediated ROCK activation. Cell Death Differ 2010; 17:1435-47. [PMID: 20379197 DOI: 10.1038/cdd.2010.36] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Constitutively active PI3K catalytic subunit alpha (PIK3CA) interfered with apoptosis induction downstream of death receptor-signaling complex formation allowing robust caspase-8 activation without triggering the execution steps of apoptosis. In mutant PIK3CA-expressing cells, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and CD95L stimulated nuclear factor kappaB (NFkappaB) activation, invasion, and transition to an amoeboid-like morphology. NFkappaB activation and adoption of amoeboid shape were inhibited by caspase-8 knockdown or FLIP-S expression, but only the cell morphology alterations required caspase-8 activity. Furthermore, we identified caspase-8-mediated, caspase-3-independent cleavage of the protein kinase rho-associated, coiled-coil containing protein kinase 1 as a novel mechanism for acquiring amoeboid shape and enhanced invasiveness in response to TRAIL and CD95L. Taken together, we provide evidence that mutated PIK3CA converts the 'tumor surveillance' activity of cancer cell-expressed death receptors and caspase-8 toward tumor promotion.
Collapse
Affiliation(s)
- M Ehrenschwender
- Department of Internal Medicine II, University Hospital Würzburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Affiliation(s)
- Lionel Hebbard
- Storr Liver Unit, Westmead Millennium InstituteThe University of Sydney and Westmead Hospital Westmead, NSW, Australia
| | | |
Collapse
|
50
|
Evodiamine-induced human melanoma A375-S2 cell death was mediated by PI3K/Akt/caspase and Fas-L/NF-kappaB signaling pathways and augmented by ubiquitin-proteasome inhibition. Toxicol In Vitro 2009; 24:898-904. [PMID: 20005289 DOI: 10.1016/j.tiv.2009.11.019] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 10/29/2009] [Accepted: 11/26/2009] [Indexed: 11/22/2022]
Abstract
Evodiamine, a major alkaloidal component of Evodiae fructus exhibits anti-tumor activities. We have previously reported that evodiamine has a marked inhibitory effect on IL-1 sensitive human melanoma A375-S2 cells proliferation, and this action might be through inactivation of PI3K signaling. However, the detailed molecular mechanisms of evodiamine-induced cell death remains poorly understood. In present study, we further confirmed that Akt is the main effector molecule involved in this pathway. Evodiamine also led to IkappaBalpha phosphorylation and degradation that reflect translocation of NF-kappaB. Pretreatment of A375-S2 cells with ubiquitin-proteasome inhibitor MG132 was shown to aggregate the evodiamine caused cell death at 24h. In addition, MG132 reduced ERK phosphorylation, increased caspase-3 activation, Fas-L expression and Bcl-2 cleavage in evodiamine-treated A375-S2 cells. These results suggested the PI3K/Akt/caspase and Fas-L/NF-kappaB signaling pathways might account for the responses of A375-S2 cell death induced by evodiamine, and these signals could be augmented by ubiquitin-proteasome pathway.
Collapse
|