Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Methods Enzymol ; 667: 183-227, 2022.
Article in English | MEDLINE | ID: mdl-35525542

ABSTRACT

Pseudokinase domains are found throughout the kingdoms of life and serve myriad roles in cell signaling. These domains, which resemble protein kinases but are catalytically-deficient, have been described principally as protein interaction domains. Broadly, pseudokinases have been reported to function as: allosteric regulators of conventional enzymes; scaffolds to nucleate assembly and/or localization of signaling complexes; molecular switches; or competitors of signaling complex assembly. From detailed structural and biochemical studies of individual pseudokinases, a picture of how they mediate protein interactions is beginning to emerge. Many such studies have relied on recombinant protein production in insect cells, where endogenous chaperones and modifying enzymes favor bona fide folding of pseudokinases. Here, we describe methods for co-expression of pseudokinases and their interactors in insect cells, as exemplified by the MLKL pseudokinase, which is the terminal effector in the necroptosis cell death pathway, and its upstream regulator kinase RIPK3. These methods are broadly applicable to co-expression of other pseudokinases with their interaction partners from bacmids using the baculovirus-insect cell expression system.


Subject(s)
Baculoviridae , Receptor-Interacting Protein Serine-Threonine Kinases , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Insecta , Necroptosis , Phosphorylation , Protein Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
2.
Nat Commun ; 12(1): 6783, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34811356

ABSTRACT

The ancestral origins of the lytic cell death mode, necroptosis, lie in host defense. However, the dysregulation of necroptosis in inflammatory diseases has led to widespread interest in targeting the pathway therapeutically. This mode of cell death is executed by the terminal effector, the MLKL pseudokinase, which is licensed to kill following phosphorylation by its upstream regulator, RIPK3 kinase. The precise molecular details underlying MLKL activation are still emerging and, intriguingly, appear to mechanistically-diverge between species. Here, we report the structure of the human RIPK3 kinase domain alone and in complex with the MLKL pseudokinase. These structures reveal how human RIPK3 structurally differs from its mouse counterpart, and how human RIPK3 maintains MLKL in an inactive conformation prior to induction of necroptosis. Residues within the RIPK3:MLKL C-lobe interface are crucial to complex assembly and necroptotic signaling in human cells, thereby rationalizing the strict species specificity governing RIPK3 activation of MLKL.


Subject(s)
Cell Death/physiology , Necroptosis/physiology , Protein Kinases/chemistry , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Cell Death/genetics , HT29 Cells , Humans , Mice , Necroptosis/genetics , Phosphorylation , Protein Conformation , Protein Interaction Domains and Motifs , Protein Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Recombinant Proteins , Signal Transduction
3.
Biomed Pharmacother ; 142: 112082, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34449307

ABSTRACT

RIPK1 is a protein kinase that simultaneously regulates inflammation, apoptosis, and necroptosis. It is thought that RIPK1 has separate functions through its scaffold structure and kinase domains. Moreover, different post-translational modifications in RIPK1 play distinct or even opposing roles. Under different conditions, in different cells and species, and/or upon exposure to different stimuli, infections, and substrates, RIPK1 activation can lead to diverse results. Despite continuous research, many of the conclusions that have been drawn regarding the complex interactions of RIPK1 are controversial. This review is based on an examination and analysis of recent studies on the RIPK1 structure, post-translational modifications, and activation conditions, which can affect its functions. Finally, because of the diverse functions of RIPK1 and their relevance to the pathogenesis of many diseases, we briefly introduce the roles of RIPK1 in inflammatory and autoimmune diseases and the prospects of its use in future diagnostics and treatments.


Subject(s)
Inflammation/pathology , Protein Processing, Post-Translational , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis/physiology , Autoimmune Diseases/physiopathology , Humans , Necroptosis/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry
4.
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
5.
Proc Natl Acad Sci U S A ; 118(14)2021 04 06.
Article in English | MEDLINE | ID: mdl-33790016

ABSTRACT

Receptor-interacting protein kinases 3 (RIPK3), a central node in necroptosis, polymerizes in response to the upstream signals and then activates its downstream mediator to induce cell death. The active polymeric form of RIPK3 has been indicated as the form of amyloid fibrils assembled via its RIP homotypic interaction motif (RHIM). In this study, we combine cryogenic electron microscopy and solid-state NMR to determine the amyloid fibril structure of RIPK3 RHIM-containing C-terminal domain (CTD). The structure reveals a single protofilament composed of the RHIM domain. RHIM forms three ß-strands (referred to as strands 1 through 3) folding into an S shape, a distinct fold from that in complex with RIPK1. The consensus tetrapeptide VQVG of RHIM forms strand 2, which zips up strands 1 and 3 via heterozipper-like interfaces. Notably, the RIPK3-CTD fibril, as a physiological fibril, exhibits distinctive assembly compared with pathological fibrils. It has an exceptionally small fibril core and twists in both handedness with the smallest pitch known so far. These traits may contribute to a favorable spatial arrangement of RIPK3 kinase domain for efficient phosphorylation.


Subject(s)
Amyloid/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Amino Acid Motifs , Amyloid/metabolism , Cryoelectron Microscopy , Humans , Necroptosis , Phosphorylation , Protein Domains , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism
6.
Nat Commun ; 12(1): 2211, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33850121

ABSTRACT

Phosphorylation of the MLKL pseudokinase by the RIPK3 kinase leads to MLKL oligomerization, translocation to, and permeabilization of, the plasma membrane to induce necroptotic cell death. The precise choreography of MLKL activation remains incompletely understood. Here, we report Monobodies, synthetic binding proteins, that bind the pseudokinase domain of MLKL within human cells and their crystal structures in complex with the human MLKL pseudokinase domain. While Monobody-32 constitutively binds the MLKL hinge region, Monobody-27 binds MLKL via an epitope that overlaps the RIPK3 binding site and is only exposed after phosphorylated MLKL disengages from RIPK3 following necroptotic stimulation. The crystal structures identified two distinct conformations of the MLKL pseudokinase domain, supporting the idea that a conformational transition accompanies MLKL disengagement from RIPK3. These studies provide further evidence that MLKL undergoes a large conformational change upon activation, and identify MLKL disengagement from RIPK3 as a key regulatory step in the necroptosis pathway.


Subject(s)
Cell Death/physiology , Necroptosis/physiology , Protein Kinases/chemistry , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Binding Sites , Cell Membrane , Crystallography, X-Ray , HT29 Cells , Humans , Mice , Molecular Conformation , Molecular Dynamics Simulation , Mutation , Phosphorylation , Protein Conformation , Protein Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Recombinant Proteins , Sequence Alignment , U937 Cells
7.
Nat Commun ; 12(1): 1627, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33712586

ABSTRACT

RIPK3 amyloid complex plays crucial roles during TNF-induced necroptosis and in response to immune defense in both human and mouse. Here, we have structurally characterized mouse RIPK3 homogeneous self-assembly using solid-state NMR, revealing a well-ordered N-shaped amyloid core structure featured with 3 parallel in-register ß-sheets. This structure differs from previously published human RIPK1/RIPK3 hetero-amyloid complex structure, which adopted a serpentine fold. Functional studies indicate both RIPK1-RIPK3 binding and RIPK3 amyloid formation are essential but not sufficient for TNF-induced necroptosis. The structural integrity of RIPK3 fibril with three ß-strands is necessary for signaling. Molecular dynamics simulations with a mouse RIPK1/RIPK3 model indicate that the hetero-amyloid is less stable when adopting the RIPK3 fibril conformation, suggesting a structural transformation of RIPK3 from RIPK1-RIPK3 binding to RIPK3 amyloid formation. This structural transformation would provide the missing link connecting RIPK1-RIPK3 binding to RIPK3 homo-oligomer formation in the signal transduction.


Subject(s)
Amyloid/metabolism , Amyloid/ultrastructure , Necroptosis/physiology , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Benzothiazoles , Cell Survival , Drosophila , Herpesviridae , Humans , Mice , Molecular Dynamics Simulation , Necroptosis/genetics , Protein Conformation , Rats , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Sequence Alignment , Sequence Analysis, Protein , Signal Transduction
8.
Nat Commun ; 12(1): 819, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33547302

ABSTRACT

Regulated cell death is essential in development and cellular homeostasis. Multi-protein platforms, including the Death-Inducing Signaling Complex (DISC), co-ordinate cell fate via a core FADD:Caspase-8 complex and its regulatory partners, such as the cell death inhibitor c-FLIP. Here, using electron microscopy, we visualize full-length procaspase-8 in complex with FADD. Our structural analysis now reveals how the FADD-nucleated tandem death effector domain (tDED) helical filament is required to orientate the procaspase-8 catalytic domains, enabling their activation via anti-parallel dimerization. Strikingly, recruitment of c-FLIPS into this complex inhibits Caspase-8 activity by altering tDED triple helix architecture, resulting in steric hindrance of the canonical tDED Type I binding site. This prevents both Caspase-8 catalytic domain assembly and tDED helical filament elongation. Our findings reveal how the plasticity, composition and architecture of the core FADD:Caspase-8 complex critically defines life/death decisions not only via the DISC, but across multiple key signaling platforms including TNF complex II, the ripoptosome, and RIPK1/RIPK3 necrosome.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/chemistry , Caspase 8/chemistry , Fas-Associated Death Domain Protein/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Catalytic Domain , Cloning, Molecular , Cryoelectron Microscopy , Death Domain Receptor Signaling Adaptor Proteins/chemistry , Death Domain Receptor Signaling Adaptor Proteins/genetics , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Isoforms/chemistry , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Multimerization , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Regulated Cell Death/genetics , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
9.
Nature ; 587(7832): 133-138, 2020 11.
Article in English | MEDLINE | ID: mdl-32968279

ABSTRACT

Cell death in human diseases is often a consequence of disrupted cellular homeostasis. If cell death is prevented without restoring cellular homeostasis, it may lead to a persistent dysfunctional and pathological state. Although mechanisms of cell death have been thoroughly investigated1-3, it remains unclear how homeostasis can be restored after inhibition of cell death. Here we identify TRADD4-6, an adaptor protein, as a direct regulator of both cellular homeostasis and apoptosis. TRADD modulates cellular homeostasis by inhibiting K63-linked ubiquitination of beclin 1 mediated by TRAF2, cIAP1 and cIAP2, thereby reducing autophagy. TRADD deficiency inhibits RIPK1-dependent extrinsic apoptosis and proteasomal stress-induced intrinsic apoptosis. We also show that the small molecules ICCB-19 and Apt-1 bind to a pocket on the N-terminal TRAF2-binding domain of TRADD (TRADD-N), which interacts with the C-terminal domain (TRADD-C) and TRAF2 to modulate the ubiquitination of RIPK1 and beclin 1. Inhibition of TRADD by ICCB-19 or Apt-1 blocks apoptosis and restores cellular homeostasis by activating autophagy in cells with accumulated mutant tau, α-synuclein, or huntingtin. Treatment with Apt-1 restored proteostasis and inhibited cell death in a mouse model of proteinopathy induced by mutant tau(P301S). We conclude that pharmacological targeting of TRADD may represent a promising strategy for inhibiting cell death and restoring homeostasis to treat human diseases.


Subject(s)
Apoptosis/drug effects , Homeostasis/drug effects , TNF Receptor-Associated Death Domain Protein/antagonists & inhibitors , TNF Receptor-Associated Death Domain Protein/metabolism , Animals , Autophagy/drug effects , Baculoviral IAP Repeat-Containing 3 Protein/metabolism , Beclin-1/chemistry , Beclin-1/metabolism , Bortezomib/antagonists & inhibitors , Bortezomib/pharmacology , Cell Line , Humans , Huntingtin Protein/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Male , Mice , Models, Molecular , Neurofibrillary Tangles/metabolism , Proteome/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , TNF Receptor-Associated Death Domain Protein/chemistry , TNF Receptor-Associated Death Domain Protein/deficiency , TNF Receptor-Associated Factor 2/metabolism , Ubiquitination , alpha-Synuclein/metabolism , tau Proteins/metabolism
10.
Eur J Med Chem ; 201: 112337, 2020 Sep 01.
Article in English | MEDLINE | ID: mdl-32659605

ABSTRACT

With the aim to develop new chemical tools based on simplified natural metabolites to help deciphering the molecular mechanism of necroptosis, simplified benzazole fragments including 2-aminobenzimidazole and the 2-aminobenzothiazole analogs were prepared during the synthesis of the marine benzosceptrin B. Conpounds inhibiting the RIPK1 protein kinase were discovered. A library of 54 synthetic analogs were prepared and evaluated through a phenotypic screen using the inhibition of the necrotic cell death induced by TNF-α in human Jurkat T cells deficient for the FADD protein. This article reports the design, synthesis and biological evaluation of a series of 2-aminobenzazoles on the necroptotic cell death through the inhibition of RIPK1 protein kinase. The 2-aminobenzimidazole and 2-aminobenzothiazole platforms presented herein can serve as novel chemical tools to study the molecular regulation of necroptosis and further develop lead drug candidates for chronic pathologies involving necroptosis.


Subject(s)
Imidazoles/pharmacology , Necroptosis/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Binding Sites , Drug Design , Fas-Associated Death Domain Protein/deficiency , Humans , Imidazoles/chemical synthesis , Imidazoles/metabolism , Jurkat Cells , Molecular Docking Simulation , Molecular Structure , Protein Binding , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/metabolism , Pyrroles/chemical synthesis , Pyrroles/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Structure-Activity Relationship
11.
Nat Commun ; 11(1): 3060, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32561735

ABSTRACT

The MLKL pseudokinase is the terminal effector in the necroptosis cell death pathway. Phosphorylation by its upstream regulator, RIPK3, triggers MLKL's conversion from a dormant cytoplasmic protein into oligomers that translocate to, and permeabilize, the plasma membrane to kill cells. The precise mechanisms underlying these processes are incompletely understood, and were proposed to differ between mouse and human cells. Here, we examine the divergence of activation mechanisms among nine vertebrate MLKL orthologues, revealing remarkable specificity of mouse and human RIPK3 for MLKL orthologues. Pig MLKL can restore necroptotic signaling in human cells; while horse and pig, but not rat, MLKL can reconstitute the mouse pathway. This selectivity can be rationalized from the distinct conformations observed in the crystal structures of horse and rat MLKL pseudokinase domains. These studies identify important differences in necroptotic signaling between species, and suggest that, more broadly, divergent regulatory mechanisms may exist among orthologous pseudoenzymes.


Subject(s)
Protein Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Animals , Chickens , Crystallography, X-Ray , Cytoplasm/enzymology , HEK293 Cells , Horses , Humans , Mice , Necroptosis , Necrosis/metabolism , Phosphorylation , Protein Conformation , Rats , Signal Transduction , Smegmamorpha , Swine , U937 Cells , Xenopus
12.
FEBS Lett ; 594(14): 2294-2302, 2020 07.
Article in English | MEDLINE | ID: mdl-32412649

ABSTRACT

Receptor-interacting serine/threonine-protein kinase 3 (RIPK3) is a central protein in necroptosis with great potential as a target for treating necroptosis-associated diseases, such as Crohn's disease. However, blockade of RIPK3 kinase activity leads to unexpected RIPK3-initiated apoptosis. Herein, we found that PP2, a known SRC inhibitor, inhibits TNF-α-induced necroptosis without initiating apoptosis. Further investigation showed that PP2 acts as an inhibitor of not only SRC but also RIPK3. PP2 does not disturb the integrity of the RIPK1-RIPK3-mixed lineage kinase domain-like pseudokinase (MLKL) necroptosome or the autophosphorylation of RIPK3 at T231/S232 but disrupts RIPK3 oligomerization, thereby impairing the phosphorylation and oligomerization of MLKL. These results demonstrate the essential role of RIPK3 oligomerization in necroptosis and suggest a potential RIPK3 oligomerization-targeting strategy for therapeutic development.


Subject(s)
Apoptosis , Necroptosis/drug effects , Protein Multimerization/drug effects , Pyrimidines/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor , Mice , Phosphorylation/drug effects , Tumor Necrosis Factor-alpha/pharmacology , src-Family Kinases/antagonists & inhibitors
13.
Trends Cell Biol ; 30(3): 189-200, 2020 03.
Article in English | MEDLINE | ID: mdl-31959328

ABSTRACT

The serine/threonine kinase RIPK1 has emerged as a crucial component of the inflammatory response activated downstream of several immune receptors, where it paradoxically functions as a scaffold to protect the cell from death or instead as an active kinase to promote the killing of the cell. While RIPK1 kinase-dependent cell death has revealed its physiological importance in the context of microbial infection, aberrant activation of RIPK1 is also demonstrated to promote cell death-driven inflammatory pathologies, highlighting the importance of fundamentally understanding proper RIPK1 regulation. Recent advances in the field demonstrated the crucial role of phosphorylation in the fine-tuning of RIPK1 activation and, additionally, question the exact mechanism by which RIPK1 enzymatic activity transmits the death signal.


Subject(s)
Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Animals , Cell Death , Humans , Models, Biological , Phosphorylation , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Ubiquitination
14.
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
15.
Int J Mol Med ; 44(3): 771-786, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31198981

ABSTRACT

Necroptosis is a type of programmed cell death with necrotic morphology, occurring in a variety of biological processes, including inflammation, immune response, embryonic development and metabolic abnormalities. The current nomenclature defines necroptosis as cell death mediated by signal transduction from receptor­interacting serine/threonine kinase (RIP) 1 to RIP3 (hereafter called RIP1/RIP3). However, RIP3­dependent cell death would be a more precise definition of necroptosis. RIP3 is indispensable for necroptosis, while RIP1 is not consistently involved in the signal transduction. Notably, deletion of RIP1 even promotes RIP3­mediated necroptosis under certain conditions. Necroptosis was previously thought as an alternate process of cell death in case of apoptosis inhibition. Currently, necroptosis is recognized to serve a pivotal role in regulating various physiological processes. Of note, it mediates a variety of human diseases, such as ischemic brain injury, immune system disorders and cancer. Targeting and inhibiting necroptosis, therefore, has the potential to be used for therapeutic purposes. To date, research has elucidated the suppression of RIP1/RIP3 via effective inhibitors and highlighted their potential application in disease therapy. The present review focused on the molecular mechanisms of RIP1/RIP3­mediated necroptosis, explored the functions of RIP1/RIP3 in necroptosis, discussed their potential as a novel therapeutic target for disease therapy, and provided valuable suggestions for further study in this field.


Subject(s)
Disease Susceptibility , Multifactorial Inheritance , Necroptosis/genetics , Nuclear Pore Complex Proteins/genetics , RNA-Binding Proteins/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Animals , Biomarkers , Gene Expression Regulation/drug effects , Humans , Molecular Targeted Therapy , Necroptosis/drug effects , Nuclear Pore Complex Proteins/antagonists & inhibitors , Nuclear Pore Complex Proteins/chemistry , Nuclear Pore Complex Proteins/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Structure-Activity Relationship
16.
Nat Commun ; 10(1): 1729, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30988283

ABSTRACT

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


Subject(s)
Apoptosis , Models, Immunological , Receptor-Interacting Protein Serine-Threonine Kinases/physiology , Animals , Caspase 8/genetics , Caspase 8/metabolism , Caspase 8/physiology , Cell Line , I-kappa B Kinase/metabolism , I-kappa B Kinase/physiology , Immunity/physiology , Mice , Phosphorylation , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Serine/chemistry , Serine/metabolism , Yersinia , Yersinia Infections/immunology
17.
Trends Biochem Sci ; 44(1): 53-63, 2019 01.
Article in English | MEDLINE | ID: mdl-30509860

ABSTRACT

The recent implication of the cell death pathway, necroptosis, in innate immunity and a range of human pathologies has led to intense interest in the underlying molecular mechanism. Unlike the better-understood apoptosis pathway, necroptosis is a caspase-independent pathway that leads to cell lysis and release of immunogens downstream of death receptor and Toll-like receptor (TLR) ligation. Here we review the role of recent structural studies of the core machinery of the pathway, the protein kinases receptor-interacting protein kinase (RIPK)1 and RIPK3, and the terminal effector, the pseudokinase mixed lineage kinase domain-like protein (MLKL), in shaping our mechanistic understanding of necroptotic signaling. Structural studies have played a key role in establishing models that describe MLKL's transition from a dormant monomer to a killer oligomer and revealing important interspecies differences.


Subject(s)
Cell Death , Protein Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Humans , Protein Conformation , Protein Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry
18.
J Med Chem ; 61(24): 11398-11414, 2018 12 27.
Article in English | MEDLINE | ID: mdl-30480444

ABSTRACT

We herein report the structural optimization and structure-activity relationship studies of 5-(2,3-dihydro-1 H-indol-5-yl)-7 H-pyrrolo[2,3- d]pyrimidin-4-amine derivatives as a new class of receptor-interacting protein kinase 1 (RIPK1) inhibitors. Among all obtained RIPK1 inhibitors, 1-(5-{4-amino-7-ethyl-7 H-pyrrolo[2,3- d]pyrimidin-5-yl}-2,3-dihydro-1 H-indol-1-yl)-2-[3-(trifluoromethoxy)phenyl]ethan-1-one (22b) is the most active one. This compound potently inhibited RIPK1 with a binding affinity ( KD) of 0.004 µM and an enzymatic IC50 value of 0.011 µM and also showed good kinase selectivity. It could efficiently protect cells from necroptosis and attenuate the necrotic cell death of vascular endothelial cells induced by tumor cells both in vitro and in vivo. Importantly, compound 22b exhibited excellent antimetastasis activity in the experimental B16 melanoma lung metastasis model. It also displayed favorable pharmacokinetic properties. Collectively, 22b could be a promising agent for preventing tumor metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Lung Neoplasms/secondary , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Receptor-Interacting Protein Serine-Threonine Kinases/antagonists & inhibitors , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Dose-Response Relationship, Drug , HT29 Cells , Human Umbilical Vein Endothelial Cells , Humans , Lung Neoplasms/drug therapy , Male , Melanoma/pathology , Mice, Inbred C57BL , Molecular Docking Simulation , Phosphorylation/drug effects , Protein Kinases/metabolism , Rats, Sprague-Dawley , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Structure-Activity Relationship , Xenograft Model Antitumor Assays
19.
Cell Physiol Biochem ; 51(1): 46-62, 2018.
Article in English | MEDLINE | ID: mdl-30439713

ABSTRACT

BACKGROUND/AIMS: This study was developed to investigate a potential therapeutic method for myocardial ischemia/reperfusion injury involving the promotion of miR-24-3p expression. METHODS: Microarray analysis was used to screen differentially expressed genes in a myocardial ischemia/reperfusion (I/R) injury mouse model. Gene set enrichment analysis was utilized to determine vital signaling pathways. Targeting verification was conducted with a luciferase reporter assay. Myocardial I/R injury was developed in mice, and the expression levels of RIPK1 and miR-24-3p were investigated by qRT-PCR and Western blot. Hemodynamic parameters and the activity of serum myocardial enzymes were measured to evaluate cardiac function. Infarct area was observed through HE and TTC staining. Myocardial cell apoptosis was examined by TUNEL staining and caspase-3 activity analysis. RESULTS: RIPK1 was an upregulated mRNA found by microarray analysis and a verified target of the downregulated miRNA miR-24-3p. The upregulation of RIPK1 (1.8-fold) and the downregulation of miR-24-3p (0.3-fold) were confirmed in I/R mice. RIPK1 led to impaired cardiac function indexes, increased infarct area and cell apoptosis, while miR-24-3p could reverse the injury by regulating RIPK1. The TNF signaling pathway was proven to be involved in myocardial I/R injury through the detection of the dysregulation of related proteins. CONCLUSION: In conclusion, RIPK1 was upregulated and miR-24-3p was downregulated in a myocardial I/R injury mouse model. RIPK1 could aggravate myocardial I/R injury via the TNF signaling pathway, while miR-24-3p could suppress RIPK1 and therefore exert cardioprotective effects in myocardial I/R injury.


Subject(s)
MicroRNAs/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , 3' Untranslated Regions , Animals , Antagomirs/metabolism , Cluster Analysis , Creatine Kinase, MB Form/blood , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Oligonucleotide Array Sequence Analysis , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction , Tumor Necrosis Factors/metabolism , Ventricular Function, Left/physiology
20.
Immunity ; 49(5): 873-885.e7, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30366765

ABSTRACT

Receptor interacting protein 2 (RIP2) plays a role in sensing intracellular pathogens, but its function in T cells is unclear. We show that RIP2 deficiency in CD4+ T cells resulted in chronic and severe interleukin-17A-mediated inflammation during Chlamydia pneumoniae lung infection, increased T helper 17 (Th17) cell formation in lungs of infected mice, accelerated atherosclerosis, and more severe experimental autoimmune encephalomyelitis. While RIP2 deficiency resulted in reduced conventional Th17 cell differentiation, it led to significantly enhanced differentiation of pathogenic (p)Th17 cells, which was dependent on RORα transcription factor and interleukin-1 but independent of nucleotide oligomerization domain (NOD) 1 and 2. Overexpression of RIP2 resulted in suppression of pTh17 cell differentiation, an effect mediated by its CARD domain, and phenocopied by a cell-permeable RIP2 CARD peptide. Our data suggest that RIP2 has a T cell-intrinsic role in determining the balance between homeostatic and pathogenic Th17 cell responses.


Subject(s)
Cell Differentiation/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Th17 Cells/cytology , Th17 Cells/metabolism , Animals , Atherosclerosis , Biomarkers , Caspase Activation and Recruitment Domain , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/mortality , Gene Expression , Immunophenotyping , Inflammation/genetics , Inflammation/metabolism , Interleukin-17/biosynthesis , Interleukin-1beta , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Receptor-Interacting Protein Serine-Threonine Kinases/chemistry , Receptor-Interacting Protein Serine-Threonine Kinases/deficiency , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...