Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Signal ; 17(820): eabg8145, 2024 Jan 23.
Article in English | MEDLINE | ID: mdl-38261657

ABSTRACT

Inflammasomes are multiprotein complexes that drive inflammation and contribute to protective immunity against pathogens and immune pathology in autoinflammatory diseases. Inflammasomes assemble when an inflammasome scaffold protein senses an activating signal and forms a signaling platform with the inflammasome adaptor protein ASC. The NLRP subfamily of NOD-like receptors (NLRs) includes inflammasome nucleators (such as NLRP3) and also NLRP12, which is genetically linked to familial autoinflammatory disorders that resemble diseases caused by gain-of-function NLRP3 mutants that generate a hyperactive NLRP3 inflammasome. We performed a screen to identify ASC inflammasome-nucleating proteins among NLRs that have the canonical pyrin-NACHT-LRR domain structure. Only NLRP3 and NLRP6 could initiate ASC polymerization to form "specks," and NLRP12 failed to nucleate ASC polymerization. However, wild-type NLRP12 inhibited ASC inflammasome assembly induced by wild-type and gain-of-function mutant NLRP3, an effect not seen with disease-associated NLRP12 mutants. The capacity of NLRP12 to suppress NLRP3 inflammasome assembly was limited to human NLRP3 and was not observed for wild-type murine NLRP3. Furthermore, peripheral blood mononuclear cells from patients with an NLRP12 mutant-associated inflammatory disorder produced increased amounts of the inflammatory cytokine IL-1ß in response to NLRP3 stimulation. Thus, our findings provide insights into NLRP12 biology and suggest that NLRP3 inhibitors in clinical trials for NLRP3-driven diseases may also be effective in treating NLRP12-associated autoinflammatory diseases.


Subject(s)
Hereditary Autoinflammatory Diseases , Inflammasomes , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing , Intracellular Signaling Peptides and Proteins , Leukocytes, Mononuclear , NLR Family, Pyrin Domain-Containing 3 Protein , Syndrome
2.
Emerg Microbes Infect ; 10(1): 2326-2339, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34821529

ABSTRACT

ABSTRACTMelioidosis is a serious infectious disease endemic in Southeast Asia, Northern Australia and has been increasingly reported in other tropical and subtropical regions in the world. Percutaneous inoculation through cuts and wounds on the skin is one of the major modes of natural transmission. Despite cuts in skin being a major route of entry, very little is known about how the causative bacterium Burkholderia pseudomallei initiates an infection at the skin and the disease manifestation at the skin known as cutaneous melioidosis. One key issue is the lack of suitable and relevant infection models. Employing an in vitro 2D keratinocyte cell culture, a 3D skin equivalent fibroblast-keratinocyte co-culture and ex vivo organ culture from human skin, we developed infection models utilizing surrogate model organism Burkholderia thailandensis to investigate Burkholderia-skin interactions. Collectively, these models show that the bacterial infection was largely limited at the wound's edge. Infection impedes wound closure, triggers inflammasome activation and cellular extrusion in the keratinocytes as a potential way to control bacterial infectious load at the skin. However, extensive infection over time could result in the epidermal layer being sloughed off, potentially contributing to formation of skin lesions.


Subject(s)
Burkholderia pseudomallei/physiology , Burkholderia/physiology , Epidermis/microbiology , Inflammasomes/metabolism , Keratinocytes/microbiology , Melioidosis/microbiology , Skin/microbiology , Wounds and Injuries/microbiology , Cells, Cultured , Epidermis/metabolism , Humans , Keratinocytes/metabolism , Melioidosis/metabolism , Melioidosis/pathology , Models, Biological , Skin/metabolism , Skin/pathology , Wounds and Injuries/metabolism , Wounds and Injuries/pathology
3.
Nat Commun ; 12(1): 188, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33420028

ABSTRACT

Nod-like receptor (NLR) proteins activate pyroptotic cell death and IL-1 driven inflammation by assembling and activating the inflammasome complex. Closely related sensor proteins NLRP1 and CARD8 undergo unique auto-proteolysis-dependent activation and are implicated in auto-inflammatory diseases; however, their mechanisms of activation are not understood. Here we report the structural basis of how the activating domains (FIINDUPA-CARD) of NLRP1 and CARD8 self-oligomerize to assemble distinct inflammasome complexes. Recombinant FIINDUPA-CARD of NLRP1 forms a two-layered filament, with an inner core of oligomerized CARD surrounded by an outer ring of FIINDUPA. Biochemically, self-assembled NLRP1-CARD filaments are sufficient to drive ASC speck formation in cultured human cells-a process that is greatly enhanced by NLRP1-FIINDUPA which forms oligomers in vitro. The cryo-EM structures of NLRP1-CARD and CARD8-CARD filaments, solved here at 3.7 Å, uncover unique structural features that enable NLRP1 and CARD8 to discriminate between ASC and pro-caspase-1. In summary, our findings provide structural insight into the mechanisms of activation for human NLRP1 and CARD8 and reveal how highly specific signaling can be achieved by heterotypic CARD interactions within the inflammasome complexes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , CARD Signaling Adaptor Proteins/metabolism , Inflammasomes/chemistry , Inflammasomes/metabolism , Neoplasm Proteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/genetics , CARD Signaling Adaptor Proteins/genetics , Caspase 1/metabolism , Cryoelectron Microscopy , HEK293 Cells , Humans , Inflammasomes/genetics , Inflammation , Molecular Docking Simulation , Mutation , NLR Proteins , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Signal Transduction
4.
Science ; 370(6521)2020 12 04.
Article in English | MEDLINE | ID: mdl-33093214

ABSTRACT

Immune sensor proteins are critical to the function of the human innate immune system. The full repertoire of cognate triggers for human immune sensors is not fully understood. Here, we report that human NACHT, LRR, and PYD domains-containing protein 1 (NLRP1) is activated by 3C proteases (3Cpros) of enteroviruses, such as human rhinovirus (HRV). 3Cpros directly cleave human NLRP1 at a single site between Glu130 and Gly131 This cleavage triggers N-glycine-mediated degradation of the autoinhibitory NLRP1 N-terminal fragment via the cullinZER1/ZYG11B complex, which liberates the activating C-terminal fragment. Infection of primary human airway epithelial cells by live human HRV triggers NLRP1-dependent inflammasome activation and interleukin-18 secretion. Our findings establish 3Cpros as a pathogen-derived trigger for the human NLRP1 inflammasome and suggest that NLRP1 may contribute to inflammatory diseases of the airway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Cysteine Endopeptidases/metabolism , Immunity, Innate , Inflammasomes/metabolism , Respiratory Mucosa/virology , Rhinovirus/enzymology , Viral Proteins/metabolism , 3C Viral Proteases , Adaptor Proteins, Signal Transducing/chemistry , Apoptosis Regulatory Proteins/chemistry , Cell Cycle Proteins/metabolism , Cullin Proteins/metabolism , Glutamine/chemistry , Glutamine/metabolism , Glycine/chemistry , Glycine/metabolism , HEK293 Cells , HeLa Cells , Humans , Interleukin-18/metabolism , NLR Proteins , Proteolysis
5.
Nat Commun ; 9(1): 4993, 2018 11 26.
Article in English | MEDLINE | ID: mdl-30478312

ABSTRACT

Signals arising from bacterial infections are detected by pathogen recognition receptors (PRRs) and are transduced by specialized adapter proteins in mammalian cells. The Receptor-interacting-serine/threonine-protein kinase 2 (RIPK2 or RIP2) is such an adapter protein that is critical for signal propagation of the Nucleotide-binding-oligomerization-domain-containing proteins 1/2 (NOD1 and NOD2). Dysregulation of this signaling pathway leads to defects in bacterial detection and in some cases autoimmune diseases. Here, we show that the Caspase-activation-and-recruitment-domain (CARD) of RIP2 (RIP2-CARD) forms oligomeric structures upon stimulation by either NOD1-CARD or NOD2-2CARD. We reconstitute this complex, termed the RIPosome in vitro and solve the cryo-EM filament structure of the active RIP2-CARD complex at 4.1 Å resolution. The structure suggests potential mechanisms by which CARD domains from NOD1 and NOD2 initiate the oligomerization process of RIP2-CARD. Together with structure guided mutagenesis experiments at the CARD-CARD interfaces, we demonstrate molecular mechanisms how RIP2 is activated and self-propagating such signal.


Subject(s)
Receptor-Interacting Protein Serine-Threonine Kinase 2/chemistry , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Signal Transduction , Amino Acid Sequence , CARD Signaling Adaptor Proteins/chemistry , CARD Signaling Adaptor Proteins/metabolism , Cryoelectron Microscopy , HEK293 Cells , Humans , Models, Molecular , Protein Binding , Protein Domains , Protein Multimerization , Receptor-Interacting Protein Serine-Threonine Kinase 2/ultrastructure , Recombinant Proteins/metabolism , Structure-Activity Relationship
6.
J Biol Chem ; 293(49): 18864-18878, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30291141

ABSTRACT

The inflammasome is a critical molecular complex that activates interleukin-1 driven inflammation in response to pathogen- and danger-associated signals. Germline mutations in the inflammasome sensor NLRP1 cause Mendelian systemic autoimmunity and skin cancer susceptibility, but its endogenous regulation remains less understood. Here we use a proteomics screen to uncover dipeptidyl dipeptidase DPP9 as a novel interacting partner with human NLRP1 and a related inflammasome regulator, CARD8. DPP9 functions as an endogenous inhibitor of NLRP1 inflammasome in diverse primary cell types from human and mice. DPP8/9 inhibition via small molecule drugs and CRISPR/Cas9-mediated genetic deletion specifically activate the human NLRP1 inflammasome, leading to ASC speck formation, pyroptotic cell death, and secretion of cleaved interleukin-1ß. Mechanistically, DPP9 interacts with a unique autoproteolytic domain (Function to Find Domain (FIIND)) found in NLRP1 and CARD8. This scaffolding function of DPP9 and its catalytic activity act synergistically to maintain NLRP1 in its inactive state and repress downstream inflammasome activation. We further identified a single patient-derived germline missense mutation in the NLRP1 FIIND domain that abrogates DPP9 binding, leading to inflammasome hyperactivation seen in the Mendelian autoinflammatory disease Autoinflammation with Arthritis and Dyskeratosis. These results unite recent findings on the regulation of murine Nlrp1b by Dpp8/9 and uncover a new regulatory mechanism for the NLRP1 inflammasome in primary human cells. Our results further suggest that DPP9 could be a multifunctional inflammasome regulator involved in human autoinflammatory diseases.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/metabolism , Inflammasomes/metabolism , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Boronic Acids/pharmacology , CARD Signaling Adaptor Proteins/metabolism , Dipeptides/pharmacology , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Germ-Line Mutation , HEK293 Cells , Humans , Inflammation/genetics , Mutation, Missense , NLR Proteins , Neoplasm Proteins/metabolism , Protein Binding , Protein Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...