Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 121
Filter
1.
Cytokine Growth Factor Rev ; 77: 15-29, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38548490

ABSTRACT

Innate immunity is not only the first line of host defense against microbial infections but is also crucial for the host responses against a variety of noxious stimuli. Z-DNA binding protein 1 (ZBP1) is a cytosolic nucleic acid sensor that can induce inflammatory cell death in both immune and nonimmune cells upon sensing of incursive virus-derived Z-form nucleic acids and self-nucleic acids via its Zα domain. Mechanistically, aberrantly expressed or activated ZBP1 induced by pathogens or noxious stimuli enables recruitment of TANK binding kinase 1 (TBK1), interferon regulatory factor 3 (IRF3), receptor-interacting serine/threonine-protein kinase 1 (RIPK1) and RIPK3 to drive type I interferon (IFN-I) responses and activation of nuclear factor kappa B (NF-κB) signaling. Meanwhile, ZBP1 promotes the assembly of ZBP1- and absent in melanoma 2 (AIM2)-PANoptosome, which ultimately triggers PANoptosis through caspase 3-mediated apoptosis, mixed lineage kinase domain like pseudokinase (MLKL)-mediated necroptosis, and gasdermin D (GSDMD)-mediated pyroptosis. In response to damaged mitochondrial DNA, ZBP1 can interact with cyclic GMP-AMP synthase to augment IFN-I responses but inhibits toll like receptor 9-mediated inflammatory responses. This review summarizes the structure and expression pattern of ZBP1, discusses its roles in human diseases through immune-dependent (e.g., the production of IFN-I and pro-inflammatory cytokines) and -independent (e.g., the activation of cell death) functions, and highlights the attractive prospect of manipulating ZBP1 as a promising therapeutic target in diseases.


Subject(s)
Cell Death , Immunity, Innate , Inflammation , RNA-Binding Proteins , Humans , Animals , Inflammation/immunology , Cell Death/immunology , RNA-Binding Proteins/immunology , RNA-Binding Proteins/metabolism , Signal Transduction/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Necroptosis/immunology , Interferon Type I/immunology , Interferon Type I/metabolism , NF-kappa B/metabolism , NF-kappa B/immunology , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/immunology , Apoptosis
2.
J Virol ; 97(2): e0194522, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36651749

ABSTRACT

Receptor-interacting protein kinase 3 (RIPK3) and mixed lineage kinase domain-like pseudokinase (MLKL) are proteins that are critical for necroptosis, a mechanism of programmed cell death that is both activated when apoptosis is inhibited and thought to be antiviral. Here, we investigated the role of RIPK3 and MLKL in controlling the Orthopoxvirus ectromelia virus (ECTV), a natural pathogen of the mouse. We found that C57BL/6 (B6) mice deficient in RIPK3 (Ripk3-/-) or MLKL (Mlkl-/-) were as susceptible as wild-type (WT) B6 mice to ECTV lethality after low-dose intraperitoneal infection and were as resistant as WT B6 mice after ECTV infection through the natural footpad route. Additionally, after footpad infection, Mlkl-/- mice, but not Ripk3-/- mice, endured lower viral titers than WT mice in the draining lymph node (dLN) at three days postinfection and in the spleen or in the liver at seven days postinfection. Despite the improved viral control, Mlkl-/- mice did not differ from WT mice in the expression of interferons or interferon-stimulated genes or in the recruitment of natural killer (NK) cells and inflammatory monocytes (iMOs) to the dLN. Additionally, the CD8 T-cell responses in Mlkl-/- and WT mice were similar, even though in the dLNs of Mlkl-/- mice, professional antigen-presenting cells were more heavily infected. Finally, the histopathology in the livers of Mlkl-/- and WT mice at 7 dpi did not differ. Thus, the mechanism of the increased virus control by Mlkl-/- mice remains to be defined. IMPORTANCE The molecules RIPK3 and MLKL are required for necroptotic cell death, which is widely thought of as an antiviral mechanism. Here we show that C57BL/6 (B6) mice deficient in RIPK3 or MLKL are as susceptible as WT B6 mice to ECTV lethality after a low-dose intraperitoneal infection and are as resistant as WT B6 mice after ECTV infection through the natural footpad route. Mice deficient in MLKL are more efficient than WT mice at controlling virus loads in various organs. This improved viral control is not due to enhanced interferon, natural killer cell, or CD8 T-cell responses. Overall, the data indicate that deficiencies in the molecules that are critical to necroptosis do not necessarily result in worse outcomes following viral infection and may improve virus control.


Subject(s)
Ectromelia, Infectious , Animals , Mice , Ectromelia virus , Ectromelia, Infectious/immunology , Interferons/metabolism , Mice, Inbred C57BL , Necroptosis/immunology , Protein Kinases/genetics , Protein Kinases/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/immunology
3.
Int J Biol Sci ; 18(1): 199-213, 2022.
Article in English | MEDLINE | ID: mdl-34975327

ABSTRACT

Autoimmune hepatitis (AIH) is an immune-mediated chronic inflammatory liver disease, and its pathogenesis is not fully understood. Our previous study discovered that receptor interacting protein kinase 3 (RIP3) is correlated with serum transaminase levels in AIH patients. However, its role and underlying mechanism in AIH are poorly understood. Here, we detected the increased expression and activation of RIP3 in livers of patients and animal models with AIH. The inhibition of RIP3 kinase by GSK872 prevented concanavalin A (ConA)-induced immune-mediated hepatitis (IMH) by reduced hepatic proinflammatory cytokines and immune cells including Th17 cells and macrophages. Further experiments revealed that RIP3 inhibition resulted in an increase in CD11b+Gr1+ myeloid-derived suppressor cells (MDSCs) with immunoregulatory properties in the liver, spleen, and peripheral blood. Moreover, the depletion of Gr-1+ MDSCs abrogated the protective effect and immune suppression function of GSK872 in ConA-induced IMH. Altogether, our data demonstrate that RIP3 blockade prevents ConA-induced IMH through promoting MDSCs infiltration. Inhibition of RIP3 kinase may be a novel therapeutic avenue for AIH treatment.


Subject(s)
Hepatitis, Autoimmune/immunology , Hepatitis, Autoimmune/metabolism , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Concanavalin A , Disease Models, Animal , Female , Humans , Liver/immunology , Liver/metabolism , Mice , Mice, Inbred C57BL
4.
Front Immunol ; 12: 758407, 2021.
Article in English | MEDLINE | ID: mdl-34956189

ABSTRACT

Mutations in the scaffolding domain of Receptor Interacting Protein kinases (RIP) underlie the recently described human autoimmune syndrome, CRIA, characterized by lymphadenopathy, splenomegaly, and autoantibody production. While disease mechanisms for CRIA remain undescribed, RIP kinases work together with caspase-8 to regulate cell death, which is critical for normal differentiation of many cell types. Here, we describe a key role for RIP1 in facilitating innate B cell differentiation and subsequent activation. By comparing RIP1, RIP3, and caspase-8 triple deficient and RIP3, caspase-8 double deficient mice, we identified selective contributions of RIP1 to an accumulation of murine splenic Marginal Zone (MZ) B cells and B1-b cells. We used mixed bone-marrow chimeras to determine that innate B cell commitment required B cell-intrinsic RIP1, RIP3, and caspase-8 sufficiency. RIP1 regulated MZ B cell development rather than differentiation and RIP1 mediates its innate immune effects independent of the RIP1 kinase domain. NP-KLH/alum and NP-Ficoll vaccination of mice doubly deficient in both caspase-8 and RIP3 or deficient in all three proteins (RIP3, caspase-8, and RIP1) revealed uniquely delayed T-dependent and T-independent IgG responses, abnormal splenic germinal center architecture, and reduced extrafollicular plasmablast formation compared to WT mice. Thus, RIP kinases and caspase-8 jointly orchestrate B cell fate and delayed effector function through a B cell-intrinsic mechanism.


Subject(s)
B-Lymphocytes/immunology , Caspase 8/immunology , GTPase-Activating Proteins/immunology , Immunity, Innate/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Animals , Caspase 8/genetics , GTPase-Activating Proteins/deficiency , GTPase-Activating Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/genetics
5.
Front Immunol ; 12: 747737, 2021.
Article in English | MEDLINE | ID: mdl-34539677

ABSTRACT

Programmed cell death is a vital process in the life cycle of organisms. Necroptosis, an evolutionary form of programmed necrosis, contributes to the innate immune response by killing pathogen-infected cells. This virus-host interaction pathway is organized around two components: the receptor-interacting protein kinase 3 (RIPK3), which recruits and phosphorylates the mixed lineage kinase-like protein (MLKL), inducing cellular plasma membrane rupture and cell death. Critically, the presence of necroptotic inhibitors in viral genomes validates necroptosis as an important host defense mechanism. Here, we show, counterintuitively, that in different mammalian lineages, central components of necroptosis, such as RIPK3 and MLKL, are deleted or display inactivating mutations. Frameshifts or premature stop codons are observed in all the studied species of cetaceans and leporids. In carnivores' genomes, the MLKL gene is deleted, while in a small number of species from afrotheria and rodentia premature stop codons are observed in RIPK3 and/or MLKL. Interestingly, we also found a strong correlation between the disruption of necroptosis in leporids and cetaceans and the absence of the N-terminal domain of E3-like homologs (responsible for necroptosis inhibition) in their naturally infecting poxviruses. Overall, our study provides the first comprehensive picture of the molecular evolution of necroptosis in mammals. The loss of necroptosis multiple times during mammalian evolution highlights the importance of gene/pathway loss for species adaptation and suggests that necroptosis is not required for normal mammalian development. Moreover, this study highlights a co-evolutionary relationship between poxviruses and their hosts, emphasizing the role of host adaptation in shaping virus evolution.


Subject(s)
Necroptosis/immunology , Protein Kinases/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Virus Diseases/immunology , Animals , Humans , Mammals , Necroptosis/genetics , Protein Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics
6.
mBio ; 12(4): e0172321, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34372694

ABSTRACT

Receptor-interacting serine/threonine-protein kinase 1 (RIPK1) has emerged as a key upstream regulator of cell death and inflammation. RIPK1-mediated signaling governs the outcome of signaling pathways initiated by tumor necrosis factor receptor 1 (TNFR1), Toll-like receptor 3 (TLR3), TLR4, retinoic acid-inducible gene 1 (RIG-I)/melanoma differentiation-associated protein 5 (MDA-5), and Z-binding protein 1 (ZBP1) by signaling for NF-κB activation, mitogen-associated protein kinase (MAPK) and interferon regulatory factor 3/7 (IRF3/7) phosphorylation, and cell death via apoptosis and necroptosis. Both cell death and inflammatory responses play a major role in controlling virus infections. Therefore, viruses have evolved multifaceted mechanisms to exploit host immune responses by targeting RIPK1. This review focuses on the current understanding of RIPK1-mediated inflammatory and cell death pathways and multiple mechanisms by which viruses manipulate these pathways by targeting RIPK1. We also discuss gaps in our knowledge regarding RIPK1-mediated signaling pathways and highlight potential avenues for future research.


Subject(s)
Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Virus Diseases/immunology , Animals , Apoptosis , Cell Death , Humans , Inflammation/immunology , Inflammation/pathology , Mice , NF-kappa B/metabolism , Phosphorylation , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Receptors, Tumor Necrosis Factor, Type I/metabolism
7.
Fish Shellfish Immunol ; 115: 142-149, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34147612

ABSTRACT

Both the activation and attenuation of MAVS/IFN signaling are critical for host defensing against viral infection and thus lead to an elaborate regulation of MAVS-mediated signaling. However, the regulatory mechanisms concerning MAVS/IFN signaling in teleost fish are not well understood. RIPK3 has been identified as a key regulator of necroptosis, apoptosis, and inflammatory signaling in human and mammals. Here we report the identification of the RIPK3 homologue from black carp Mylopharyngodon piceus (bcRIPK3) and describe its role in regulating MAVS/IFN signaling. qPCR results demonstrated that bcRIPK3 was transcriptionally activated in response to poly (I:C) or LPS stimulation. Immunoblot assay and immunofluorescent staining assay showed that bcRIPK3 was a cytosolic protein with molecular weights of 47 kDa. Like its mammalian counterparts, bcRIPK3 exhibited a conserved function in inducing cell death. The reporter assay and plaque assay showed that overexpression of bcRIPK3 restricted bcMAVS-activated transcription of the interferon promoters of black carp and zebrafish, and suppressed bcMAVS-mediated antiviral activity. Notably, EPC cells co-expressing bcRIPK3, bcRIPK1 and bcMAVS presented much attenuated antiviral activity than the cells co-expressing bcRIPK3 and bcMAVS; and the subsequent co-IP assay identified the interaction between bcRIPK3 and bcRIPK1. Our findings collectively elucidate for the first time in teleost that black carp RIPK3 interacts with RIPK1 to inhibit MAVS-mediated antiviral signaling.


Subject(s)
Cyprinidae/genetics , Cyprinidae/immunology , Fish Diseases/immunology , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Regulation/immunology , Immunity, Innate/genetics , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Amino Acid Sequence , Animals , Gene Expression Profiling/veterinary , Phylogeny , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Rhabdoviridae/physiology , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/veterinary , Sequence Alignment/veterinary
9.
Am J Respir Cell Mol Biol ; 64(5): 579-591, 2021 05.
Article in English | MEDLINE | ID: mdl-33625952

ABSTRACT

Community-acquired pneumonia is the most common type of pneumonia and remains a leading cause of morbidity and mortality worldwide. Although many different pathogens can contribute to pneumonia, Streptococcus pneumoniae is one of the common bacterial pathogens that underlie community-acquired pneumonia. RIPK3 (receptor-interacting protein kinase 3) is widely recognized as a key modulator of inflammation and cell death. To elucidate a potential role of RIPK3 in pneumonia, we examined plasma from healthy control subjects and patients positive for streptococcal pneumonia. In human studies, RIPK3 protein concentrations were significantly elevated and were identified as a potential plasma marker of pneumococcal pneumonia. To expand these findings, we used an in vivo murine model of pneumococcal pneumonia to demonstrate that RIPK3 deficiency leads to reduced bacterial clearance, severe pathological damage, and high mortality. Our results illustrated that RIPK3 forms a complex with RIPK1, MLKL (mixed-lineage kinase domain-like protein), and MCU (mitochondrial calcium uniporter) to induce mitochondrial calcium uptake and mitochondrial reactive oxygen species(mROS) production during S. pneumoniae infection. In macrophages, RIPK3 initiated necroptosis via the mROS-mediated mitochondrial permeability transition pore opening and NLRP3 inflammasome activation via the mROS-AKT pathway to protect against S. pneumoniae. In conclusion, our study demonstrated a mechanism by which RIPK3-initiated necroptosis is essential for host defense against S. pneumoniae.


Subject(s)
Macrophages, Alveolar/immunology , Mitochondria/immunology , Pneumonia, Pneumococcal/immunology , Protein Kinases/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Streptococcus pneumoniae/pathogenicity , Aged , Animals , Calcium Channels/genetics , Calcium Channels/immunology , Case-Control Studies , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Inflammasomes/genetics , Inflammasomes/immunology , Macrophages, Alveolar/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mitochondria/pathology , Mitochondrial Permeability Transition Pore/immunology , Mitochondrial Permeability Transition Pore/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Necroptosis/genetics , Necroptosis/immunology , Pneumonia, Pneumococcal/complications , Pneumonia, Pneumococcal/microbiology , Protein Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction , Streptococcus pneumoniae/immunology
10.
FEBS J ; 288(10): 3186-3196, 2021 05.
Article in English | MEDLINE | ID: mdl-33145969

ABSTRACT

The release of interleukin (IL)-1ß from primary human monocytes in response to extracellular LPS occurs through the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome. In primary monocytes, in response to LPS, NLRP3 inflammasome activation is characterized by an independence of K+ efflux and ASC speck formation and has been termed the 'alternative' pathway. Here, we report that pharmacological inhibition of V-ATPase with bafilomycin A1 exacerbated LPS-induced NLRP3 inflammasome activation in primary human monocytes. Inhibition of V-ATPase in the presence of extracellular LPS led to NLRP3-dependent, K+ efflux-independent, ASC oligomerization and caspase-1 activation. Although V-ATPases are required for lysosomal acidification, we found that acidic lysosomal pH and protease activity were dispensable for this altered response, suggesting that V-ATPase inhibition triggered alternative signalling events. Therefore, V-ATPases may serve additional roles during NLRP3 inflammasome activation in primary human monocytes.


Subject(s)
Inflammasomes/drug effects , Lysosomes/drug effects , Macrolides/pharmacology , Monocytes/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Proton-Translocating ATPases/genetics , Caspase 1/genetics , Caspase 1/immunology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation , Humans , Hydrogen-Ion Concentration , Inflammasomes/immunology , Inflammasomes/metabolism , Inflammation , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Lipopolysaccharides/pharmacology , Lysosomes/immunology , Lysosomes/metabolism , Monocytes/cytology , Monocytes/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/agonists , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Nigericin/pharmacology , Primary Cell Culture , Proton-Translocating ATPases/antagonists & inhibitors , Proton-Translocating ATPases/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Signal Transduction , THP-1 Cells
11.
mBio ; 11(5)2020 10 06.
Article in English | MEDLINE | ID: mdl-33024046

ABSTRACT

Regulated macrophage death has emerged as an important mechanism to defend against intracellular pathogens. However, the importance and consequences of macrophage death during bacterial infection are poorly resolved. This is especially true for the recently described RIPK3-dependent lytic cell death, termed necroptosis. Salmonella enterica serovar Typhimurium is an intracellular pathogen that precisely regulates virulence expression within macrophages to evade and manipulate immune responses, which is a key factor in its ability to cause severe systemic infections. We combined genetic and pharmacological approaches to examine the importance of RIPK3 for S. Typhimurium-induced macrophage death using conditions that recapitulate bacterial gene expression during systemic infection in vivo Our findings indicate that noninvasive S. Typhimurium does not naturally induce macrophage necroptosis but does so in the presence of pan-caspase inhibition. Moreover, our data suggest that RIPK3 induction (following caspase inhibition) does not impact host survival following S. Typhimurium infection, which differs from previous findings based on inert lipopolysaccharide (LPS) injections. Finally, although necroptosis is typically characterized as highly inflammatory, our data suggest that RIPK3 skews the peritoneal myeloid population away from an inflammatory profile to that of a classically noninflammatory profile. Collectively, these data improve our understanding of S. Typhimurium-macrophage interactions, highlight the possibility that purified bacterial components may not accurately recapitulate the complexity of host-pathogen interactions, and reveal a potential and unexpected role for RIPK3 in resolving inflammation.IMPORTANCE Macrophages employ multiple strategies to limit pathogen infection. For example, macrophages may undergo regulated cell death, including RIPK3-dependent necroptosis, as a means of combatting intracellular bacterial pathogens. However, bacteria have evolved mechanisms to evade or exploit immune responses. Salmonella is an intracellular pathogen that avoids and manipulates immune detection within macrophages. We examined the contribution of RIPK3 to Salmonella-induced macrophage death. Our findings indicate that noninvasive Salmonella does not naturally induce necroptosis, but it does so when caspases are inhibited. Moreover, RIPK3 induction (following caspase inhibition) does not impact host survival following Salmonella systemic infection. Finally, our data show that RIPK3 induction results in recruitment of low-inflammatory myeloid cells, which was unexpected, as necroptosis is typically described as highly inflammatory. Collectively, these data improve our understanding of pathogen-macrophage interactions, including outcomes of regulated cell death during infection in vivo, and reveal a potential new role for RIPK3 in resolving inflammation.


Subject(s)
Host-Pathogen Interactions/immunology , Macrophages/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Salmonella Infections, Animal/blood , Animals , Caspase Inhibitors/pharmacology , Caspases/immunology , Inflammasomes , Inflammation , Macrophages/microbiology , Mice , Mice, Inbred C57BL , Necroptosis/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Salmonella Infections, Animal/microbiology , Salmonella typhimurium , Signal Transduction
12.
Front Immunol ; 11: 1718, 2020.
Article in English | MEDLINE | ID: mdl-32849607

ABSTRACT

Receptor interacting protein 1 (RIP1) is an essential sensor of cellular stress, which may respond to apoptosis or cell survival and participate in antiviral pathways. To investigate the roles of fish RIP1 in Singapore grouper iridovirus (SGIV) and red-spotted grouper nervous necrosis virus (RGNNV) infection, a RIP1 homolog from orange-spotted grouper (Epinephelus coioides) (EcRIP1) was cloned and characterized. EcRIP1 encoded a 679 amino acid protein that shares 83.28% identity with that of Perca flavescens and contained a homologous N-terminal kinase (S-TKc) domain, a RIP isotype interaction motif (RHIM), and a C-terminal domain (DD). EcRIP1 was predominantly detected in immune tissues, and its expression was induced by RGNNV or SGIV infection in vitro. Subcellular localization showed that EcRIP1 was distributed in the cytoplasm with point-like uniform and dot-like aggregation forms. Overexpression of EcRIP1 inhibited SGIV and RGNNV replication and positively regulated the expression levels of interferon (IFN) and IFN-stimulated genes and pro-inflammatory factors. EcRIP1 may interact with grouper tumor necrosis factor receptor type 1-associated DEATH domain protein (EcTRADD) to promote SGIV-induced apoptosis, and interact with grouper Toll/interleukin-1 receptor (TIR) domain containing adapter inducing interferon-ß (EcTRIF) and participate in Myeloid Differentiation Factor 88 (MyD88)-independent toll-like receptor (TLR) signaling. EcRIP1 may also interact with grouper tumor necrosis factor receptor-associated factors (TRAFs) as intracellular linker proteins and mediate the signaling of various downstream signaling pathways, including NF-κB and IFN. These results suggest that EcRIP1 may inhibit SGIV and RGNNV infection by regulating apoptosis and various signaling molecules. Our study offers new insights into the regulatory mechanism of RIP1-related signaling, and provides a novel perspective on fish diseases mediated by RIP1.


Subject(s)
Bass/virology , DNA Virus Infections/veterinary , Fish Diseases/virology , Fish Proteins/metabolism , Immunity, Innate , Iridovirus/pathogenicity , Nodaviridae/pathogenicity , RNA Virus Infections/veterinary , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Bass/genetics , Bass/immunology , Bass/metabolism , Cells, Cultured , Cytokines/metabolism , DNA Virus Infections/immunology , DNA Virus Infections/metabolism , DNA Virus Infections/virology , Fish Diseases/genetics , Fish Diseases/immunology , Fish Diseases/metabolism , Fish Proteins/genetics , Fish Proteins/immunology , Host-Pathogen Interactions , Iridovirus/immunology , Nodaviridae/immunology , RNA Virus Infections/immunology , RNA Virus Infections/metabolism , RNA Virus Infections/virology , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Signal Transduction
13.
Dev Comp Immunol ; 109: 103726, 2020 08.
Article in English | MEDLINE | ID: mdl-32376280

ABSTRACT

Receptor-interacting serine/threonine protein kinase 1 (RIPK1) is an important regulator of necroptosis and involved in innate immune response in human and mammal; however, its function in teleost fish mains largely unknown. In this paper, the RIPK1 homologue of black carp (Mylopharyngodon piceus) has been cloned and characterized to explore its role in immunity. Black carp RIPK1 (bcRIPK1) possesses the similar structure to its mammalian counterpart, which has been identified as a cytosolic protein by immunofluorescence staining. Overexpressed bcRIPK1 in host cells led to the decreased transcription of interferon (IFN) and interferon stimulated genes, and exogenous bcRIPK1 in EPC cells led to the decreased transcription of interferon promoters in reporter assay. Our previous study has identified that black carp MAVS (bcMAVS) functions as an antiviral adaptor protein against both grass carp reovirus (GCRV) and spring viremia of carp virus (SVCV). The reporter assay showed that the IFN-inducing ability of bcMAVS was dampened by bcRIPK1 and the plaque assay demonstrated that the antiviral activity of bcMAVS was inhibited by bcRIPK1. The immunofluorescent staining and co-immunoprecipitation identified the interaction between these two molecules. Thus, the data generated in this paper support the conclusion that bcRIPK1 interacts with bcMAVS and negatively regulates bcMAVS-mediated antiviral signaling.


Subject(s)
Carps/immunology , Fish Diseases/immunology , Fish Proteins/immunology , Immunity, Innate/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Amino Acid Sequence , Animals , Carps/genetics , Carps/virology , Fish Diseases/virology , Fish Proteins/classification , Fish Proteins/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Enzymologic , HEK293 Cells , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate/genetics , Interferons/immunology , Interferons/metabolism , Phylogeny , Receptor-Interacting Protein Serine-Threonine Kinases/classification , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Reoviridae/immunology , Reoviridae/physiology , Sequence Homology, Amino Acid , Signal Transduction/immunology
14.
Int Immunopharmacol ; 84: 106546, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32413735

ABSTRACT

Apelin is a neuropeptide that plays an important role in neuronal protection. In this study, we investigated the effects of apelin intracerebroventricular administration on spatial learning and memory-related behaviors, and necroptosis signaling pathways in the hippocampus of streptozotocin (STZ) -injected rats. Apelin treatment was implemented following STZ-induced dementia for 15 days. After conducting a behavioral test (Morris Water Maze), the cellular and molecular aspects were examined to detect the apelin effect on the necroptosis signaling pathway. We demonstrated that STZ administration significantly slowed down the learning capability. However apelin treatment notably reversed this neuroinflammation induced behavioral impairment. Furthermore, molecular investigations showed that apelin treatment reduced the hippocampal RIP1, RIP3, and TNF-α level. Our results suggest that apelin treatment attenuates STZ-induced dementia. This effect may be mediated by inhibition of the necroptosis signaling pathway which seems to be associated with the ability of apelin to reduce central TNF-α level. This data provides evidence of the neuroprotective effect of apelin on STZ-induced learning and memory impairment and characterize some of the underlying mechanisms.


Subject(s)
Apelin/therapeutic use , Memory Disorders/drug therapy , Neuroprotective Agents/therapeutic use , Animals , Apelin/pharmacology , Hippocampus/drug effects , Hippocampus/immunology , Male , Maze Learning/drug effects , Memory Disorders/chemically induced , Necroptosis/drug effects , Neuroprotective Agents/pharmacology , Protein Serine-Threonine Kinases/immunology , Rats, Wistar , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Streptozocin , Tumor Necrosis Factor-alpha/immunology
15.
Front Immunol ; 11: 536, 2020.
Article in English | MEDLINE | ID: mdl-32328060

ABSTRACT

Efficient induction of effector and long-term protective antigen-specific CD8+ T memory response by vaccination is essential to eliminate malignant and pathogen-infected cells. Intracellular infectious bacteria, including Listeria monocytogenes, have been considered potent vectors to carry multiple therapeutic proteins and generate antigen-specific CD8+ T cell responses. Although the role of molecules involved in inflammatory cell death pathways, such as necroptosis (RIPK3-mediated) and pyroptosis (Caspase-1/11-mediated), as effectors of immune response against intracellular bacteria are relatively well understood, their contribution to the adjuvant effect of recombinant bacterial vectors in the context of antigen-specific CD8+ T cell response remained obscure. Therefore, we evaluated the impact of RIPK3 and Caspase-1/11 (Casp-1/11) individual and combined deficiencies on the modulation of antigen-specific CD8+ T cell response during vaccination of mice with ovalbumin-expressing L. monocytogenes (LM-OVA). We observed that Casp-1/11 but not RIPK3 deficiency negatively impacts the capacity of mice to clear LM-OVA. Importantly, both RIPK3 and Casp-1/11 are necessary for optimal LM-OVA-mediated antigen-specific CD8+ T cell response, as measured by in vivo antigen-specific CD8+ T cell proliferation, target cell elimination, and cytokine production. Furthermore, Casp-1/11 and Casp-1/11/RIPK3 combined deficiencies restrict the early initiation of antigen-specific CD8+ T cell memory response. Taken together, our findings demonstrate that RIPK3 and Casp-1/11 influence the quality of CD8+ T cell responses induced by recombinant L. monocytogenes vectors.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Caspase 1/immunology , Caspases, Initiator/immunology , Genetic Vectors/immunology , Listeria monocytogenes/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Animals , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL
16.
Cancer Immunol Res ; 8(5): 710-721, 2020 05.
Article in English | MEDLINE | ID: mdl-32122992

ABSTRACT

Metabolic reprogramming is critical for the polarization and function of tumor-associated macrophages (TAM) and hepatocarcinogenesis, but how this reprogramming occurs is unknown. Here, we showed that receptor-interacting protein kinase 3 (RIPK3), a central factor in necroptosis, is downregulated in hepatocellular carcinoma (HCC)-associated macrophages, which correlated with tumorigenesis and enhanced the accumulation and polarization of M2 TAMs. Mechanistically, RIPK3 deficiency in TAMs reduced reactive oxygen species and significantly inhibited caspase1-mediated cleavage of PPAR. These effects enabled PPAR activation and facilitated fatty acid metabolism, including fatty acid oxidation (FAO), and induced M2 polarization in the tumor microenvironment. RIPK3 upregulation or FAO blockade reversed the immunosuppressive activity of TAMs and dampened HCC tumorigenesis. Our findings provide molecular basis for the regulation of RIPK3-mediated, lipid metabolic reprogramming of TAMs, thus highlighting a potential strategy for targeting the immunometabolism of HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Fatty Acids/metabolism , Liver Neoplasms/pathology , Macrophages/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Tumor Microenvironment/immunology , Animals , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Humans , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Oxidation-Reduction , PPAR gamma/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
17.
J Leukoc Biol ; 107(6): 941-952, 2020 06.
Article in English | MEDLINE | ID: mdl-31985117

ABSTRACT

Receptor interacting protein kinase 1 (RIP1) is a critical effector of inflammatory responses and cell death activation. Cell death pathways regulated by RIP1 include caspase-dependent apoptosis and caspase-independent necroptosis. The kinase activity of RIP1 has been associated with a number of inflammatory, neurodegenerative, and oncogenic diseases. In this study, we use the RIP1 kinase inhibitor GNE684 to demonstrate that RIP1 inhibition can effectively block skin inflammation and immune cell infiltrates in livers of Sharpin mutant (Cpdm; chronic proliferative dermatitis) mice in an interventional setting, after disease onset. On the other hand, genetic inactivation of RIP1 (RIP1 KD) or ablation of RIP3 (RIP3 KO) or MLKL (MLKL KO) did not affect testicular pathology of aging male mice. Likewise, infection with vaccinia virus or with mouse gammaherpesvirus MHV68 resulted in similar viral clearance in wild-type, RIP1 KD, and RIP3 KO mice. In summary, this study highlights the benefits of inhibiting RIP1 in skin inflammation, as opposed to its lack of relevance for testicular longevity and the response to certain viral infections.


Subject(s)
Dermatitis/genetics , Herpesviridae Infections/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Skin/immunology , Vaccinia/genetics , Animals , Chronic Disease , Dermatitis/immunology , Dermatitis/pathology , Dermatitis/virology , Disease Models, Animal , Gammaherpesvirinae/immunology , Gammaherpesvirinae/pathogenicity , Gene Expression Regulation , Herpesviridae Infections/pathology , Herpesviridae Infections/virology , Inflammation , Liver/immunology , Liver/pathology , Liver/virology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Kinase Inhibitors/pharmacology , Protein Kinases/deficiency , Protein Kinases/genetics , Protein Kinases/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Signal Transduction , Skin/pathology , Skin/virology , Testis/immunology , Testis/pathology , Testis/virology , Vaccinia/immunology , Vaccinia/pathology , Vaccinia/virology , Vaccinia virus/immunology , Vaccinia virus/pathogenicity , Virus Replication/immunology
18.
Semin Cell Dev Biol ; 99: 86-95, 2020 03.
Article in English | MEDLINE | ID: mdl-29738881

ABSTRACT

The Receptor-interacting protein kinase Homotypic Interaction Motif (RHIM) is an amino acid sequence that mediates multiple protein:protein interactions in the mammalian programmed cell death pathway known as necroptosis. At least one key RHIM-based complex has been shown to have a functional amyloid fibril structure, which provides a stable hetero-oligomeric platform for downstream signaling. RHIMs and related motifs are present in immunity-related proteins across nature, from viruses to fungi to metazoans. Necroptosis is a hallmark feature of cellular clearance of infection. For this reason, numerous pathogens, including viruses and bacteria, have developed varied methods to modulate necroptosis, focusing on inhibiting RHIM:RHIM interactions, and thus their downstream cell death effects. This review will discuss current understanding of RHIM:RHIM interactions in normal cellular activation of necroptosis, from a structural and cell biology perspective. It will compare the mechanisms by which pathogens subvert these interactions in order to maintain their replicative and infective cycles and consider the similarities between RHIMs and other functional amyloid-forming proteins associated with cell death and innate immunity. It will discuss the implications of the heteromeric nature and structure of RHIM-based amyloid complexes in the context of other functional amyloids.


Subject(s)
Apoptosis/immunology , Necroptosis/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Animals , Humans , Immunity, Innate/immunology , Protein Binding , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
19.
Nat Microbiol ; 5(2): 331-342, 2020 02.
Article in English | MEDLINE | ID: mdl-31844296

ABSTRACT

Viruses manipulate cellular signalling by inducing the degradation of crucial signal transducers, usually via the ubiquitin-proteasome pathway. Here, we show that the murine cytomegalovirus (Murid herpesvirus 1) M45 protein induces the degradation of two cellular signalling proteins, the nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) essential modulator (NEMO) and the receptor-interacting protein kinase 1 (RIPK1), via a different mechanism: it induces their sequestration as insoluble protein aggregates and subsequently facilitates their degradation by autophagy. Aggregation of target proteins requires a distinct sequence motif in M45, which we termed 'induced protein aggregation motif'. In a second step, M45 recruits the retromer component vacuolar protein sorting 26B (VPS26B) and the microtubule-associated protein light chain 3 (LC3)-interacting adaptor protein TBC1D5 to facilitate degradation of aggregates by selective autophagy. The induced protein aggregation motif is conserved in M45-homologous proteins of several human herpesviruses, including herpes simplex virus, Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus, but is only partially conserved in the human cytomegalovirus UL45 protein. We further show that the HSV-1 ICP6 protein induces RIPK1 aggregation and degradation in a similar fashion to M45. These data suggest that induced protein aggregation combined with selective autophagy of aggregates (aggrephagy) represents a conserved viral immune-evasion mechanism.


Subject(s)
Herpesviridae/immunology , Intracellular Signaling Peptides and Proteins/immunology , Receptor-Interacting Protein Serine-Threonine Kinases/immunology , Animals , Autophagy/immunology , Autophagy-Related Protein 5/deficiency , Autophagy-Related Protein 5/genetics , Cells, Cultured , HEK293 Cells , Herpesviridae/metabolism , Herpesviridae/pathogenicity , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/metabolism , Herpesvirus 1, Human/pathogenicity , Host Microbial Interactions/immunology , Humans , Immune Evasion , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Muromegalovirus/immunology , Muromegalovirus/metabolism , Muromegalovirus/pathogenicity , Protein Aggregates/immunology , Proteolysis , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Ribonucleotide Reductases/genetics , Ribonucleotide Reductases/immunology , Ribonucleotide Reductases/metabolism , Viral Proteins/genetics , Viral Proteins/immunology , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...