Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Viruses ; 13(10)2021 10 08.
Article in English | MEDLINE | ID: mdl-34696459

ABSTRACT

Zika virus (ZIKV) is a mosquito-borne flavivirus, and its infection may cause severe neurodegenerative diseases. The outbreak of ZIKV in 2015 in South America has caused severe human congenital and neurologic disorders. Thus, it is vitally important to determine the inner mechanism of ZIKV infection. Here, our data suggested that the ubiquitin-specific peptidase 38 (USP38) played an important role in host resistance to ZIKV infection, during which ZIKV infection did not affect USP38 expression. Mechanistically, USP38 bound to the ZIKV envelope (E) protein through its C-terminal domain and attenuated its K48-linked and K63-linked polyubiquitination, thereby repressed the infection of ZIKV. In addition, we found that the deubiquitinase activity of USP38 was essential to inhibit ZIKV infection, and the mutant that lacked the deubiquitinase activity of USP38 lost the ability to inhibit infection. In conclusion, we found a novel host protein USP38 against ZIKV infection, and this may represent a potential therapeutic target for the treatment and prevention of ZIKV infection.


Subject(s)
Ubiquitin-Specific Proteases/pharmacology , Ubiquitination , Zika Virus Infection/drug therapy , Zika Virus/drug effects , A549 Cells , HeLa Cells , Humans , Receptor, EphB2 , Ubiquitin-Specific Proteases/metabolism , Viral Envelope Proteins/drug effects , Zika Virus Infection/virology
2.
Virulence ; 12(1): 1795-1807, 2021 12.
Article in English | MEDLINE | ID: mdl-34282707

ABSTRACT

Zika virus (ZIKV) infection can cause severe neurological disorders, including Guillain-Barre syndrome and meningoencephalitis in adults and microcephaly in fetuses. Here, we reveal that laminin receptor 1 (LAMR1) is a novel host resistance factor against ZIKV infection. Mechanistically, we found that LAMR1 binds to ZIKV envelope (E) protein via its intracellular region and attenuates E protein ubiquitination through recruiting the deubiquitinase eukaryotic translation initiation factor 3 subunit 5 (EIF3S5). We further found that the conserved G282 residue of E protein is essential for its interaction with LAMR1. Moreover, a G282A substitution abolished the binding of E protein to LAMR1 and inhibited LAMR1-mediated E protein deubiquitination. Together, our results indicated that LAMR1 represses ZIKV infection through binding to E protein and attenuating its ubiquitination.


Subject(s)
Receptors, Laminin/metabolism , Ribosomal Proteins/metabolism , Ubiquitination , Viral Envelope Proteins/chemistry , Zika Virus Infection , Humans , Zika Virus
3.
BMC Biol ; 18(1): 182, 2020 11 26.
Article in English | MEDLINE | ID: mdl-33243234

ABSTRACT

BACKGROUND: Extracellular adenosine triphosphate (ATP), a key danger-associated molecular pattern (DAMP) molecule, is released to the extracellular medium during inflammation by injured parenchymal cells, dying leukocytes, and activated platelets. ATP directly activates the plasma membrane channel P2X7 receptor (P2X7R), leading to an intracellular influx of K+, a key trigger inducing NLRP3 inflammasome activation. However, the mechanism underlying P2X7R-mediated activation of NLRP3 inflammasome is poorly understood, and additional molecular mediators have not been identified. Here, we demonstrate that Paxillin is the molecule connecting the P2X7 receptor and NLRP3 inflammasome through protein interactions. RESULTS: We show a distinct mechanism by which Paxillin promotes ATP-induced activation of the P2X7 receptor and NLRP3 inflammasome. Extracellular ATP induces Paxillin phosphorylation and then facilitates Paxillin-NLRP3 interaction. Interestingly, Paxillin enhances NLRP3 deubiquitination and activates NLRP3 inflammasome upon ATP treatment and K+ efflux. Moreover, we demonstrated that USP13 is a key enzyme for Paxillin-mediated NLRP3 deubiquitination upon ATP treatment. Notably, extracellular ATP promotes Paxillin and NLRP3 migration from the cytosol to the plasma membrane and facilitates P2X7R-Paxillin interaction and PaxillinNLRP3 association, resulting in the formation of the P2X7R-Paxillin-NLRP3 complex. Functionally, Paxillin is essential for ATP-induced NLRP3 inflammasome activation in mouse BMDMs and BMDCs as well as in human PBMCs and THP-1-differentiated macrophages. CONCLUSIONS: We have identified paxillin as a mediator of NLRP3 inflammasome activation. Paxillin plays key roles in ATP-induced activation of the P2X7 receptor and NLRP3 inflammasome by facilitating the formation of the P2X7R-Paxillin-NLRP3 complex.


Subject(s)
Adenosine Triphosphate/metabolism , Inflammasomes/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Paxillin/genetics , Receptors, Purinergic P2X7/genetics , Animals , HEK293 Cells , HeLa Cells , Humans , Inflammasomes/metabolism , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Paxillin/metabolism , Receptors, Purinergic P2X7/metabolism
4.
Front Immunol ; 11: 51, 2020.
Article in English | MEDLINE | ID: mdl-32117232

ABSTRACT

During host-virus co-evolution, cells develop innate immune systems to inhibit virus invasion, while viruses employ strategies to suppress immune responses and maintain infection. Here, we reveal that Zika virus (ZIKV), a re-emerging arbovirus causing public concerns and devastating complications, restricts host immune responses through a distinct mechanism. ZIKV nonstructural protein 5 (NS5) interacts with the host retinoic acid-inducible gene I (RIG-I), an essential signaling molecule for defending pathogen infections. NS5 subsequently represses K63-linked polyubiquitination of RIG-I, attenuates the phosphorylation and nuclear translocation of interferon regulatory factor 3 (IRF3), and inhibits the expression and production of interferon-ß (IFN-ß), thereby restricting the RIG-I signaling pathway. Interestingly, we demonstrate that the methyltransferase (MTase) domain of NS5 is required for the repression of RIG-I ubiquitination, IRF3 activation, and IFN-ß production. Detailed studies further reveal that the conservative active site D146 of NS5 is critical for the suppression of the RIG-I signaling. Therefore, we uncover an essential role of NS5 conservative site D146 in ZIKV-mediated repression of innate immune system, illustrate a distinct mechanism by which ZIKV evades host immune responses, and discover a potential target for anti-viral infection.


Subject(s)
DEAD Box Protein 58/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Viral Nonstructural Proteins/metabolism , Zika Virus Infection/metabolism , Zika Virus/metabolism , Active Transport, Cell Nucleus , Binding Sites , Cell Nucleus/metabolism , Host-Pathogen Interactions , Humans , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Phosphorylation , Protein Binding , Ubiquitination , Zika Virus/physiology , Zika Virus Infection/virology
5.
PLoS Pathog ; 16(3): e1008335, 2020 03.
Article in English | MEDLINE | ID: mdl-32187211

ABSTRACT

One of the fundamental reactions of the innate immune responses to pathogen infection is the release of pro-inflammatory cytokines, including IL-1ß, processed by the NLRP3 inflammasome. The stimulator of interferon genes (STING) has the essential roles in innate immune response against pathogen infections. Here we reveal a distinct mechanism by which STING regulates the NLRP3 inflammasome activation, IL-1ß secretion, and inflammatory responses in human cell lines, mice primary cells, and mice. Interestingly, upon HSV-1 infection and cytosolic DNA stimulation, STING binds to NLRP3 and promotes the inflammasome activation through two approaches. First, STING recruits NLRP3 and facilitates NLRP3 localization in the endoplasmic reticulum, thereby facilitating the inflammasome formation. Second, STING interacts with NLRP3 and attenuates K48- and K63-linked polyubiquitination of NLRP3, thereby promoting the inflammasome activation. Collectively, we demonstrate that the cGAS-STING-NLRP3 signaling is essential for host defense against HSV-1 infection.


Subject(s)
Endoplasmic Reticulum/immunology , Herpes Simplex/immunology , Inflammasomes/immunology , Membrane Proteins/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Animals , Endoplasmic Reticulum/metabolism , Herpes Simplex/genetics , Herpes Simplex/metabolism , Herpes Simplex/virology , Herpesvirus 1, Human/genetics , Herpesvirus 1, Human/physiology , Humans , Immunity, Innate , Inflammasomes/genetics , Inflammasomes/metabolism , Macrophages/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Protein Binding , Protein Transport
6.
Nano Lett ; 19(4): 2215-2222, 2019 04 10.
Article in English | MEDLINE | ID: mdl-30543300

ABSTRACT

Zika virus (ZIKV) has emerged as a global health threat due to its unexpected causal link to devastating neurological disorders such as fetal microcephaly; however, to date, no approved vaccine or specific treatment is available for ZIKV infection. Here we develop a biomimetic nanodecoy (ND) that can trap ZIKV, divert ZIKV away from its intended targets, and inhibit ZIKV infection. The ND, which is composed of a gelatin nanoparticle core camouflaged by mosquito medium host cell membranes, effectively adsorbs ZIKV and inhibits ZIKV replication in ZIKV-susceptible cells. Using a mouse model, we demonstrate that NDs significantly attenuate the ZIKV-induced inflammatory responses and degenerative changes and thus improve the survival rate of ZIKV-challenged mice. Moreover, by trapping ZIKV, NDs successfully prevent ZIKV from passing through physiologic barriers into the fetal brain and thereby mitigate ZIKV-induced fetal microcephaly in pregnant mice. We anticipate that this study will provide new insights into the development of safe and effective protection against ZIKV and various other viruses that threaten public health.


Subject(s)
Microcephaly/prevention & control , Nanoparticles/administration & dosage , Zika Virus Infection/prevention & control , Zika Virus/drug effects , Animals , Biomimetics/methods , Cell Membrane/drug effects , Cell Membrane/virology , Culicidae/drug effects , Culicidae/virology , Disease Models, Animal , Female , Fetus , Gelatin/administration & dosage , Gelatin/chemistry , Humans , Mice , Microcephaly/pathology , Microcephaly/virology , Nanoparticles/chemistry , Pregnancy , Zika Virus/pathogenicity , Zika Virus Infection/pathology , Zika Virus Infection/virology
7.
Nat Commun ; 9(1): 106, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29317641

ABSTRACT

Zika virus (ZIKV) infection is a public health emergency and host innate immunity is essential for the control of virus infection. The NLRP3 inflammasome plays a key role in host innate immune responses by activating caspase-1 to facilitate interleukin-1ß (IL-1ß) secretion. Here we report that ZIKV stimulates IL-1ß secretion in infected patients, human PBMCs and macrophages, mice, and mice BMDCs. The knockdown of NLRP3 in cells and knockout of NLRP3 in mice inhibit ZIKV-mediated IL-1ß secretion, indicating an essential role for NLRP3 in ZIKV-induced IL-1ß activation. Moreover, ZIKV NS5 protein is required for NLRP3 activation and IL-1ß secretion by binding with NLRP3 to facilitate the inflammasome complex assembly. Finally, ZIKV infection in mice activates IL-1ß secretion, leading to inflammatory responses in the mice brain, spleen, liver, and kidney. Thus we reveal a mechanism by which ZIKV induces inflammatory responses by facilitating NLRP3 inflammasome complex assembly and IL-1ß activation.


Subject(s)
Inflammasomes/immunology , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Zika Virus Infection/immunology , Zika Virus/immunology , Aedes , Animals , Brain/immunology , Brain/pathology , Caspase 1/genetics , Caspase 1/immunology , Cell Line , Chlorocebus aethiops , Enzyme Activation/genetics , Enzyme Activation/immunology , HEK293 Cells , Humans , Immunity, Innate/immunology , Interleukin-1beta/blood , Kidney/immunology , Kidney/pathology , L Cells , Liver/immunology , Liver/pathology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Neutrophils/immunology , Spleen/immunology , Spleen/pathology , THP-1 Cells , Vero Cells , Viral Nonstructural Proteins/immunology , Zika Virus Infection/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...