Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
Add more filters










Publication year range
1.
Cell Rep ; 43(2): 113795, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38367238

ABSTRACT

Activation of endosomal Toll-like receptor (TLR) 7, TLR9, and TLR11/12 is a key event in the resistance against the parasite Toxoplasma gondii. Endosomal TLR engagement leads to expression of interleukin (IL)-12 via the myddosome, a protein complex containing MyD88 and IL-1 receptor-associated kinase (IRAK) 4 in addition to IRAK1 or IRAK2. In murine macrophages, IRAK2 is essential for IL-12 production via endosomal TLRs but, surprisingly, Irak2-/- mice are only slightly susceptible to T. gondii infection, similar to Irak1-/- mice. Here, we report that upon T. gondii infection IL-12 production by different cell populations requires either IRAK1 or IRAK2, with conventional dendritic cells (DCs) requiring IRAK1 and monocyte-derived DCs (MO-DCs) requiring IRAK2. In both populations, we identify interferon regulatory factor 5 as the main transcription factor driving the myddosome-dependent IL-12 production during T. gondii infection. Consistent with a redundant role of DCs and MO-DCs, mutations that affect IL-12 production in both cell populations show high susceptibility to infection in vivo.


Subject(s)
Interleukin-1 Receptor-Associated Kinases , Toxoplasmosis , Animals , Mice , Dendritic Cells , Interferon Regulatory Factors/genetics , Interleukin-12
2.
Innate Immun ; 29(8): 186-200, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37828863

ABSTRACT

NOD1 and NOD2 sense small bacterial peptidoglycan fragments, often called muropeptides, that access the cytosol. These muropeptides include iE-DAP and MDP, the minimal agonists for NOD1 and NOD2, respectively. Here, we synthesized and validated alkyne-modified muropeptides, iE-DAP-Alk and MDP-Alk, for use in click-chemistry reactions. While it has long been known that many cell types respond to extracellular exposure to muropeptides, it is unclear how these innate immune activators access their cytosolic innate immune receptors, NOD1 and NOD2. The subcellular trafficking and transport mechanisms by which muropeptides access these cytosolic innate immune receptors are a major gap in our understanding of these critical host responses. The click-chemistry-enabled agonists developed here will be particularly powerful to decipher the underlying cell biology and biochemistry of NOD1 and NOD2 innate immune sensing.


Subject(s)
Nod1 Signaling Adaptor Protein , Receptor Protein-Tyrosine Kinases , Diaminopimelic Acid/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/genetics , Nod2 Signaling Adaptor Protein/metabolism
3.
Immunity ; 56(5): 998-1012.e8, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37116499

ABSTRACT

Cytosolic innate immune sensing is critical for protecting barrier tissues. NOD1 and NOD2 are cytosolic sensors of small peptidoglycan fragments (muropeptides) derived from the bacterial cell wall. These muropeptides enter cells, especially epithelial cells, through unclear mechanisms. We previously implicated SLC46 transporters in muropeptide transport in Drosophila immunity. Here, we focused on Slc46a2, which was highly expressed in mammalian epidermal keratinocytes, and showed that it was critical for the delivery of diaminopimelic acid (DAP)-muropeptides and activation of NOD1 in keratinocytes, whereas the related transporter Slc46a3 was critical for delivering the NOD2 ligand MDP to keratinocytes. In a mouse model, Slc46a2 and Nod1 deficiency strongly suppressed psoriatic inflammation, whereas methotrexate, a commonly used psoriasis therapeutic, inhibited Slc46a2-dependent transport of DAP-muropeptides. Collectively, these studies define SLC46A2 as a transporter of NOD1-activating muropeptides, with critical roles in the skin barrier, and identify this transporter as an important target for anti-inflammatory intervention.


Subject(s)
Dermatitis , Methotrexate , Mice , Animals , Methotrexate/pharmacology , Inflammation , Peptidoglycan/metabolism , Epithelial Cells/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Nod2 Signaling Adaptor Protein/metabolism , Immunity, Innate , Mammals
4.
Neuron ; 111(9): 1381-1390.e6, 2023 05 03.
Article in English | MEDLINE | ID: mdl-36931278

ABSTRACT

GGGGCC repeat expansion in the C9ORF72 gene is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Repeat RNAs can be translated into dipeptide repeat proteins, including poly(GR), whose mechanisms of action remain largely unknown. In an RNA-seq analysis of poly(GR) toxicity in Drosophila, we found that several antimicrobial peptide genes, such as metchnikowin (Mtk), and heat shock protein (Hsp) genes are activated. Mtk knockdown in the fly eye or in all neurons suppresses poly(GR) neurotoxicity. These findings suggest a cell-autonomous role of Mtk in neurodegeneration. Hsp90 knockdown partially rescues both poly(GR) toxicity in flies and neurodegeneration in C9ORF72 motor neurons derived from induced pluripotent stem cells (iPSCs). Topoisomerase II (TopoII) regulates poly(GR)-induced upregulation of Hsp90 and Mtk. TopoII knockdown also suppresses poly(GR) toxicity in Drosophila and improves survival of C9ORF72 iPSC-derived motor neurons. These results suggest potential novel therapeutic targets for C9ORF72-ALS/FTD.


Subject(s)
Amyotrophic Lateral Sclerosis , Frontotemporal Dementia , Animals , Amyotrophic Lateral Sclerosis/genetics , C9orf72 Protein/genetics , C9orf72 Protein/metabolism , Dipeptides/genetics , DNA Repeat Expansion , Down-Regulation , Drosophila/metabolism , Frontotemporal Dementia/genetics , Frontotemporal Dementia/metabolism , Motor Neurons/metabolism
5.
Nanoscale ; 15(7): 3273-3283, 2023 Feb 16.
Article in English | MEDLINE | ID: mdl-36723053

ABSTRACT

Herein, we present disulfide crosslinked dextran/eudragit S-100 nanoparticles (DEEU NPs) (≈55 nm) for colorectal cancer treatment. These redox environment sensitive DEEU NPs are synthesized by simple oxidation of thiolated polymers in air. This approach allows avoiding the use of any additional chemical crosslinker. These DEEU NPs have high encapsulation efficiency for the doxorubicin (DOX) model drug (≈95%). The prepared DEEU NPs are redox sensitive owing to disulfide units and exhibit ≈80% DOX release in the reducing environment of GSH. Additionally, DOX-DEEU NPs display enhanced cytotoxicity for HCT116 cancer cells as compared to free DOX. Annexin V staining results also support the higher anticancer efficiency of DOX-DEEU NPs via induction of apoptosis. In vivo biodistribution results demonstrate that DEEU NPs can remain in the colon region for up to 24 hours. These results indicate that DEEU NPs can act as a promising platform for colorectal cancer treatment.


Subject(s)
Colorectal Neoplasms , Nanoparticles , Humans , Dextrans , Tissue Distribution , Doxorubicin/pharmacology , Drug Delivery Systems/methods , Oxidation-Reduction , Nanoparticles/therapeutic use , Disulfides , Colorectal Neoplasms/drug therapy
6.
Nat Genet ; 54(12): 1933-1945, 2022 12.
Article in English | MEDLINE | ID: mdl-36396707

ABSTRACT

Retrotransposons are one type of mobile genetic element that abundantly reside in the genomes of nearly all animals. Their uncontrolled activation is linked to sterility, cancer and other pathologies, thereby being largely considered detrimental. Here we report that, within a specific time window of development, retrotransposon activation can license the host's immune system for future antiviral responses. We found that the mdg4 (also known as Gypsy) retrotransposon selectively becomes active during metamorphosis at the Drosophila pupal stage. At this stage, mdg4 activation educates the host's innate immune system by inducing the systemic antiviral function of the nuclear factor-κB protein Relish in a dSTING-dependent manner. Consequently, adult flies with mdg4, Relish or dSTING silenced at the pupal stage are unable to clear exogenous viruses and succumb to viral infection. Altogether, our data reveal that hosts can establish a protective antiviral response that endows a long-term benefit in pathogen warfare due to the developmental activation of mobile genetic elements.


Subject(s)
Drosophila , Retroelements , Animals , Retroelements/genetics , Drosophila/genetics
7.
iScience ; 25(9): 104909, 2022 Sep 16.
Article in English | MEDLINE | ID: mdl-36060064

ABSTRACT

Leishmania parasites use elaborate virulence mechanisms to invade and thrive in macrophages. These virulence mechanisms inhibit host cell defense responses and generate a specialized replicative niche, the parasitophorous vacuole. In this work, we performed a genome-wide RNAi screen in Drosophila macrophage-like cells to identify the host factors necessary for Leishmania amazonensis infection. This screen identified 52 conserved genes required specifically for parasite entry, including several components of the SUMOylation machinery. Further studies in mammalian macrophages found that L. amazonensis infection inhibited SUMOylation within infected macrophages and this inhibition enhanced parasitophorous vacuole growth and parasite proliferation through modulation of multiple genes especially ATP6V0D2, which in turn affects CD36 expression and cholesterol levels. Together, these data suggest that parasites actively sabotage host SUMOylation and alter host transcription to improve their intracellular niche and enhance their replication.

8.
Cell Rep ; 40(7): 111225, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35977521

ABSTRACT

Interleukin-1 receptor-associated kinases (IRAKs) -4, -2, and -1 are involved in transducing signals from Toll-like receptors (TLRs) via the adaptor myeloid differentiation primary-response protein 88 (MYD88). How MYD88/IRAK4/2/1 complexes are formed, their redundancies, and potential non-enzymatic roles are subjects of debate. Here, we examine the hierarchical requirements for IRAK proteins in the context of TLR4 activation and confirmed that the kinase activity of IRAK4 is essential for MYD88 signaling. Surprisingly, the IRAK4 scaffold is required for activation of the E3 ubiquitin ligase TNF receptor-associated factor 6 (TRAF6) by both MYD88 and TIR domain-containing adaptor protein inducing IFN-ß (TRIF), a unique adaptation in the TLR4 response. IRAK4 scaffold is, therefore, essential in integrating MYD88 and TRIF in TLR4 signaling.


Subject(s)
Interleukin-1 Receptor-Associated Kinases , Myeloid Differentiation Factor 88 , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Humans , Interleukin-1 Receptor-Associated Kinases/metabolism , Myeloid Differentiation Factor 88/metabolism , Signal Transduction/physiology , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism
9.
Carbohydr Polym ; 294: 119833, 2022 Oct 15.
Article in English | MEDLINE | ID: mdl-35868778

ABSTRACT

Herein, redox responsive chitosan/stearic acid nanoparticles (CSSA NPs) (≈200 nm) are developed for dual drug delivery. These degradable nanoparticles are prepared based on disulfide (SS) crosslinking chemistry avoiding the use of any external crosslinking agent. CSSA NPs are further loaded with both DOX (hydrophilic) and curcumin (hydrophobic) drugs with ≈86 % and ≈82 % encapsulation efficiency respectively. This approach of combining anticancer therapeutics having different mode of anticancer action allows to develop systems for cancer therapy with enhanced efficacy. In vitro drug release experiments clearly exhibit the low leakage of drug under physiological conditions while ≈98 % DOX and ≈96 % curcumin is released after 136 h under GSH reducing conditions. The cytotoxicity experiments against HCT116 cells demonstrate higher cytotoxicity of dual drug loaded CSSA NPs. In vivo biodistribution experiments with c57bl/6j mice confirms the retention of CSSA NPs in the colon area up to 24 h exhibiting their potential for colorectal cancer therapy.


Subject(s)
Chitosan , Colorectal Neoplasms , Curcumin , Nanoparticles , Animals , Chitosan/chemistry , Colorectal Neoplasms/drug therapy , Curcumin/chemistry , Disulfides/chemistry , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Drug Carriers/chemistry , Drug Delivery Systems , Drug Liberation , Mice , Nanoparticles/chemistry , Stearic Acids , Tissue Distribution
10.
12.
Front Immunol ; 11: 1828, 2020.
Article in English | MEDLINE | ID: mdl-32983094

ABSTRACT

The NLRP3 inflammasome is central to host defense and implicated in various inflammatory diseases and conditions. While the favored paradigm of NLRP3 inflammasome activation stipulates a unifying signal intermediate that de-represses NLRP3, this view has not been tested. Further, structures within NLRP3 required for inflammasome activation are poorly defined. Here we demonstrate that while the NLRP3 LRRs are not auto-repressive and are not required for inflammasome activation by all agonists, distinct sequences within the NLRP3 LRRs positively and negatively modulate inflammasome activation by specific ligands. In addition, elements within the HD1/HD2 "hinge" of NLRP3 and the nucleotide-binding domain have contrasting functions depending upon the specific agonists. Further, while NLRP3 1-432 is minimally sufficient for inflammasome activation by all agonists tested, the pyrin, and linker domains (1-134) function cooperatively and are sufficient for inflammasome activation by certain agonists. Conserved cysteines 8 and 108 appear important for inflammasome activation by sterile, but not infectious insults. Our results define common and agonist-specific regions of NLRP3 that likely mediate ligand-specific responses, discount the hypothesis that NLRP3 inflammasome activation has a unified mechanism, and implicate NLRP3 as an integrator of agonist-specific, inflammasome activating signals.


Subject(s)
Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis Regulatory Proteins/immunology , Apoptosis Regulatory Proteins/metabolism , Francisella/physiology , HEK293 Cells , Humans , Inflammasomes/agonists , Inflammasomes/chemistry , Inflammasomes/immunology , Leucine , Ligands , Listeria monocytogenes/physiology , Macrophages/immunology , Macrophages/metabolism , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/agonists , NLR Family, Pyrin Domain-Containing 3 Protein/chemistry , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , NLR Proteins , Pyrin Domain , Repetitive Sequences, Amino Acid
14.
Viruses ; 11(5)2019 05 01.
Article in English | MEDLINE | ID: mdl-31052481

ABSTRACT

The host immune response and virus-encoded immune evasion proteins pose constant, mutual selective pressure on each other. Virally encoded immune evasion proteins also indicate which host pathways must be inhibited to allow for viral replication. Here, we show that IIV-6 is capable of inhibiting the two Drosophila NF-κB signaling pathways, Imd and Toll. Antimicrobial peptide (AMP) gene induction downstream of either pathway is suppressed when cells infected with IIV-6 are also stimulated with Toll or Imd ligands. We find that cleavage of both Imd and Relish, as well as Relish nuclear translocation, three key points in Imd signal transduction, occur in IIV-6 infected cells, indicating that the mechanism of viral inhibition is farther downstream, at the level of Relish promoter binding or transcriptional activation. Additionally, flies co-infected with both IIV-6 and the Gram-negative bacterium, Erwinia carotovora carotovora, succumb to infection more rapidly than flies singly infected with either the virus or the bacterium. These findings demonstrate how pre-existing infections can have a dramatic and negative effect on secondary infections, and establish a Drosophila model to study confection susceptibility.


Subject(s)
Drosophila Proteins/immunology , Drosophila melanogaster/immunology , Drosophila melanogaster/virology , Iridovirus/physiology , Toll-Like Receptors/immunology , Animals , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Host-Pathogen Interactions , Immunity, Innate , Iridovirus/genetics , Toll-Like Receptors/genetics , Transcription Factors/genetics , Transcription Factors/immunology , Virus Replication
15.
Insect Biochem Mol Biol ; 108: 16-23, 2019 05.
Article in English | MEDLINE | ID: mdl-30857831

ABSTRACT

Fruit flies elicit effective defense responses against numerous microbes. The responses against Gram-negative bacteria are mediated by the Imd pathway, an evolutionarily conserved NF-κB pathway recognizing meso-diaminopimelic acid (DAP)-type peptidoglycan from bacterial cell walls. Several reviews already provide a detailed view of ligand recognition and signal transduction during Imd signaling, but the formation and regulation of the signaling complex immediately downstream of the peptidoglycan-sensing receptors is still elusive. In this review, we focus on the formation of the Imd amyloidal signaling center and post-translational modifications in the assembly and disassembly of the Imd signaling complex.


Subject(s)
Amyloid/metabolism , Drosophila/metabolism , NF-kappa B/metabolism , Amyloid/immunology , Animals , Drosophila/immunology , Drosophila Proteins/metabolism , Immunity, Innate , Protein Processing, Post-Translational , Receptors, Cell Surface/metabolism , Signal Transduction
16.
Cell Rep ; 25(8): 2110-2120.e3, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30463009

ABSTRACT

Macroautophagy and cell death both contribute to innate immunity, but little is known about how these processes integrate. Drosophila larval salivary glands require autophagy for developmentally programmed cell death, and innate immune signaling factors increase in these dying cells. Here, we show that the nuclear factor κB (NF-κB) factor Relish, a component of the immune deficiency (Imd) pathway, is required for salivary gland degradation. Surprisingly, of the classic Imd pathway components, only Relish and the PGRP receptors were involved in salivary gland degradation. Significantly, Relish controls salivary gland degradation by regulating autophagy but not caspases. In addition, expression of either Relish or PGRP-LC causes premature autophagy induction and subsequent gland degradation. Relish controls autophagy by regulating the expression of Atg1, a core component and activator of the autophagy pathway. Together these findings demonstrate that a NF-κB pathway regulates autophagy during developmentally programmed cell death.


Subject(s)
Autophagy-Related Protein-1 Homolog/metabolism , Autophagy , Drosophila Proteins/metabolism , Drosophila melanogaster/cytology , Drosophila melanogaster/metabolism , Transcription Factors/metabolism , Animals , Apoptosis , Carrier Proteins/metabolism , Caspases/metabolism , Drosophila Proteins/chemistry , Drosophila melanogaster/genetics , Gene Expression Regulation , Salivary Glands/cytology , Transcription Factors/chemistry
17.
Immunity ; 49(2): 225-234.e4, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30119996

ABSTRACT

Antiviral immunity in Drosophila involves RNA interference and poorly characterized inducible responses. Here, we showed that two components of the IMD pathway, the kinase dIKKß and the transcription factor Relish, were required to control infection by two picorna-like viruses. We identified a set of genes induced by viral infection and regulated by dIKKß and Relish, which included an ortholog of STING. We showed that dSTING participated in the control of infection by picorna-like viruses, acting upstream of dIKKß to regulate expression of Nazo, an antiviral factor. Our data reveal an antiviral function for STING in an animal model devoid of interferons and suggest an evolutionarily ancient role for this molecule in antiviral immunity.


Subject(s)
Drosophila Proteins/metabolism , Drosophila melanogaster/immunology , Drosophila melanogaster/virology , I-kappa B Kinase/metabolism , Membrane Proteins/metabolism , Peptide Initiation Factors/metabolism , Picornaviridae Infections/immunology , Animals , Cell Line , Dicistroviridae/immunology , Drosophila Proteins/genetics , I-kappa B Kinase/genetics , Membrane Proteins/genetics , Peptide Initiation Factors/genetics , RNA Interference , Transcription Factors/metabolism
18.
BMC Biol ; 16(1): 60, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29855367

ABSTRACT

BACKGROUND: Drosophila is a powerful model for the study of factors modulating innate immunity. This study examines the effect of water-loss dehydration on innate immune responsiveness in the Drosophila renal system (Malpighian tubules; MTs), and how this leads to elevated host defense and contributes to immunosenescence. RESULTS: A short period of desiccation-elevated peptidoglycan recognition protein-LC (PGRP-LC) expression in MTs, increased antimicrobial peptide (AMP) gene induction, and protected animals from bacterial infection. We show that desiccation increased ecdysone synthesis in MTs, while inhibition of ecdysone synthesis or ecdysone receptor expression, specifically within MTs, prevented induction of PGRP-LC and reduced protection from bacterial infection. Additionally, aged flies are constitutively water-stressed and have elevated levels of ecdysone and PGRP-LC. Conversely, adults aged at high relative humidity show less water loss and have reduced expression of PGRP-LC and AMPs. CONCLUSIONS: The Drosophila renal system is an important contributor to host defense and can modulate immune responses in an organ autonomous manner, responding to environmental changes such as desiccation. Desiccation primes immune responsiveness by elevating PGRP-LC expression specifically in MTs. In response to desiccation, ecdysone is produced in MTs and acts in a paracrine fashion to increase PGRP-LC expression, immune responsiveness, and improve host defense. This activity of the renal system may contribute to the immunosenescence observed in Drosophila.


Subject(s)
Bacterial Infections/immunology , Carrier Proteins/metabolism , Dehydration/immunology , Drosophila Proteins/metabolism , Drosophila melanogaster/immunology , Ecdysone/metabolism , Immunity, Innate , Malpighian Tubules/immunology , Animals , Drosophila melanogaster/microbiology , Models, Animal , Receptors, Steroid/metabolism , Signal Transduction
19.
PLoS Pathog ; 14(5): e1007020, 2018 05.
Article in English | MEDLINE | ID: mdl-29746571

ABSTRACT

The fruit fly Drosophila melanogaster is a powerful model system for the study of innate immunity in vector insects as well as mammals. For vector insects, it is particularly important to understand all aspects of their antiviral immune defenses, which could eventually be harnessed to control the transmission of human pathogenic viruses. The immune responses controlling RNA viruses in insects have been extensively studied, but the response to DNA virus infections is poorly characterized. Here, we report that infection of Drosophila with the DNA virus Invertebrate iridescent Virus 6 (IIV-6) triggers JAK-STAT signaling and the robust expression of the Turandots, a gene family encoding small secreted proteins. To drive JAK-STAT signaling, IIV-6 infection more immediately induced expression of the unpaireds, a family of IL-6-related cytokine genes, via a pathway that required one of the three Drosophila p38 homologs, p38b. In fact, both Stat92E and p38b were required for the survival of IIV-6 infected flies. In addition, in vitro induction of the unpaireds required an NADPH-oxidase, and in vivo studies demonstrated Nox was required for induction of TotA. These results argue that ROS production, triggered by IIV-6 infection, leads to p38b activation and unpaired expression, and subsequent JAK-STAT signaling, which ultimately protects the fly from IIV-6 infection.


Subject(s)
Drosophila melanogaster/immunology , Drosophila melanogaster/virology , Iridovirus/pathogenicity , Signal Transduction/immunology , Animals , Animals, Genetically Modified , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunity, Innate , Iridovirus/immunology , Janus Kinases/genetics , Janus Kinases/metabolism , Mitogen-Activated Protein Kinase 11/genetics , Mitogen-Activated Protein Kinase 11/metabolism , Reactive Oxygen Species/metabolism , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Bio Protoc ; 7(23)2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29276726

ABSTRACT

This protocol describes how to generate and harvest antibody-free L. amazonensis amastigotes, and how to infect adult Drosophila melanogaster with these parasites. This model recapitulates key aspects of the interactions between Leishmania amastigotes and animal phagocytes.

SELECTION OF CITATIONS
SEARCH DETAIL
...