Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Annu Rev Immunol ; 36: 695-715, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29490163

ABSTRACT

The unique class of heavy chain-only antibodies, present in Camelidae, can be shrunk to just the variable region of the heavy chain to yield VHHs, also called nanobodies. About one-tenth the size of their full-size counterparts, nanobodies can serve in applications similar to those for conventional antibodies, but they come with a number of signature advantages that find increasing application in biology. They not only function as crystallization chaperones but also can be expressed inside cells as such, or fused to other proteins to perturb the function of their targets, for example, by enforcing their localization or degradation. Their small size also affords advantages when applied in vivo, for example, in imaging applications. Here we review such applications, with particular emphasis on those areas where conventional antibodies would face a more challenging environment.


Subject(s)
Single-Domain Antibodies/genetics , Single-Domain Antibodies/immunology , Animals , Antibody Formation , Cell Surface Display Techniques , Genetic Engineering , Humans , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Single-Domain Antibodies/biosynthesis , Single-Domain Antibodies/therapeutic use , Structure-Activity Relationship
2.
Nat Immunol ; 24(4): 595-603, 2023 04.
Article in English | MEDLINE | ID: mdl-36941400

ABSTRACT

Upon detecting pathogens or cell stress, several NOD-like receptors (NLRs) form inflammasome complexes with the adapter ASC and caspase-1, inducing gasdermin D (GSDMD)-dependent cell death and maturation and release of IL-1ß and IL-18. The triggers and activation mechanisms of several inflammasome-forming sensors are not well understood. Here we show that mitochondrial damage activates the NLRP10 inflammasome, leading to ASC speck formation and caspase-1-dependent cytokine release. While the AIM2 inflammasome can also sense mitochondrial demise by detecting mitochondrial DNA (mtDNA) in the cytosol, NLRP10 monitors mitochondrial integrity in an mtDNA-independent manner, suggesting the recognition of distinct molecular entities displayed by the damaged organelles. NLRP10 is highly expressed in differentiated human keratinocytes, in which it can also assemble an inflammasome. Our study shows that this inflammasome surveils mitochondrial integrity. These findings might also lead to a better understanding of mitochondria-linked inflammatory diseases.


Subject(s)
Cytokines , Inflammasomes , Humans , Inflammasomes/metabolism , Caspase 1/metabolism , Cytokines/metabolism , Cell Death , DNA, Mitochondrial/genetics , Interleukin-1beta/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism
3.
Immunity ; 54(11): 2650-2669.e14, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34592166

ABSTRACT

Longitudinal analyses of the innate immune system, including the earliest time points, are essential to understand the immunopathogenesis and clinical course of coronavirus disease (COVID-19). Here, we performed a detailed characterization of natural killer (NK) cells in 205 patients (403 samples; days 2 to 41 after symptom onset) from four independent cohorts using single-cell transcriptomics and proteomics together with functional studies. We found elevated interferon (IFN)-α plasma levels in early severe COVD-19 alongside increased NK cell expression of IFN-stimulated genes (ISGs) and genes involved in IFN-α signaling, while upregulation of tumor necrosis factor (TNF)-induced genes was observed in moderate diseases. NK cells exert anti-SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) activity but are functionally impaired in severe COVID-19. Further, NK cell dysfunction may be relevant for the development of fibrotic lung disease in severe COVID-19, as NK cells exhibited impaired anti-fibrotic activity. Our study indicates preferential IFN-α and TNF responses in severe and moderate COVID-19, respectively, and associates a prolonged IFN-α-induced NK cell response with poorer disease outcome.


Subject(s)
COVID-19/immunology , Interferon-alpha/immunology , Killer Cells, Natural/immunology , SARS-CoV-2/immunology , Tumor Necrosis Factor-alpha/metabolism , Base Sequence , Humans , Immunity, Innate/immunology , Inflammation/immunology , Interferon-alpha/blood , Pulmonary Fibrosis/pathology , RNA-Seq , Severity of Illness Index , Transcriptome/genetics , United Kingdom , United States
4.
Nature ; 629(8013): 893-900, 2024 May.
Article in English | MEDLINE | ID: mdl-38632402

ABSTRACT

The blood-brain barrier (BBB) protects the central nervous system from infections or harmful substances1; its impairment can lead to or exacerbate various diseases of the central nervous system2-4. However, the mechanisms of BBB disruption during infection and inflammatory conditions5,6 remain poorly defined. Here we find that activation of the pore-forming protein GSDMD by the cytosolic lipopolysaccharide (LPS) sensor caspase-11 (refs. 7-9), but not by TLR4-induced cytokines, mediates BBB breakdown in response to circulating LPS or during LPS-induced sepsis. Mice deficient in the LBP-CD14 LPS transfer and internalization pathway10-12 resist BBB disruption. Single-cell RNA-sequencing analysis reveals that brain endothelial cells (bECs), which express high levels of GSDMD, have a prominent response to circulating LPS. LPS acting on bECs primes Casp11 and Cd14 expression and induces GSDMD-mediated plasma membrane permeabilization and pyroptosis in vitro and in mice. Electron microscopy shows that this features ultrastructural changes in the disrupted BBB, including pyroptotic endothelia, abnormal appearance of tight junctions and vasculature detachment from the basement membrane. Comprehensive mouse genetic analyses, combined with a bEC-targeting adeno-associated virus system, establish that GSDMD activation in bECs underlies BBB disruption by LPS. Delivery of active GSDMD into bECs bypasses LPS stimulation and opens the BBB. In CASP4-humanized mice, Gram-negative Klebsiella pneumoniae infection disrupts the BBB; this is blocked by expression of a GSDMD-neutralizing nanobody in bECs. Our findings outline a mechanism for inflammatory BBB breakdown, and suggest potential therapies for diseases of the central nervous system associated with BBB impairment.


Subject(s)
Blood-Brain Barrier , Brain , Endothelial Cells , Gasdermins , Inflammation , Animals , Female , Humans , Male , Mice , Basement Membrane/metabolism , Basement Membrane/ultrastructure , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Blood-Brain Barrier/ultrastructure , Blood-Brain Barrier/virology , Brain/metabolism , Brain/pathology , Brain/ultrastructure , Caspases, Initiator/metabolism , Dependovirus , Endothelial Cells/metabolism , Endothelial Cells/ultrastructure , Gasdermins/antagonists & inhibitors , Gasdermins/metabolism , Inflammation/pathology , Inflammation/metabolism , Klebsiella pneumoniae/physiology , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/blood , Lipopolysaccharides/pharmacology , Mice, Inbred C57BL , Pyroptosis , Sepsis/metabolism , Sepsis/pathology , Sepsis/microbiology , Single-Cell Analysis , Tight Junctions/metabolism , Tight Junctions/ultrastructure
5.
Nat Immunol ; 21(7): 718-719, 2020 07.
Article in English | MEDLINE | ID: mdl-32577009
6.
Immunity ; 47(4): 606-608, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29045891

ABSTRACT

Zanoni et al. (2017) show that the LPS receptor CD14 promotes internalization of oxidized phospholipids, a hallmark of dying cells, triggering hyperactivation of dendritic cells and macrophages. Their findings reveal CD14 as a regulator of responses to infection and damage, relaying context-dependent signals that determine inflammatory cell fate decisions.


Subject(s)
Inflammasomes , Lipopolysaccharides , Friends , Lipopolysaccharide Receptors , Phagocytes
7.
PLoS Biol ; 20(11): e3001351, 2022 11.
Article in English | MEDLINE | ID: mdl-36342970

ABSTRACT

Pyrin is a cytosolic immune sensor that nucleates an inflammasome in response to inhibition of RhoA by bacterial virulence factors, triggering the release of inflammatory cytokines, including IL-1ß. Gain-of-function mutations in the MEFV gene encoding Pyrin cause autoinflammatory disorders, such as familial Mediterranean fever (FMF) and Pyrin-associated autoinflammation with neutrophilic dermatosis (PAAND). To precisely define the role of Pyrin in pathogen detection in human immune cells, we compared initiation and regulation of the Pyrin inflammasome response in monocyte-derived macrophages (hMDM). Unlike human monocytes and murine macrophages, we determined that hMDM failed to activate Pyrin in response to known Pyrin activators Clostridioides difficile (C. difficile) toxins A or B (TcdA or TcdB), as well as the bile acid analogue BAA-473. The Pyrin inflammasome response was enabled in hMDM by prolonged priming with either LPS or type I or II interferons and required an increase in Pyrin expression. Notably, FMF mutations lifted the requirement for prolonged priming for Pyrin activation in hMDM, enabling Pyrin activation in the absence of additional inflammatory signals. Unexpectedly, in the absence of a Pyrin response, we found that TcdB activated the NLRP3 inflammasome in hMDM. These data demonstrate that regulation of Pyrin activation in hMDM diverges from monocytes and highlights its dysregulation in FMF.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Familial Mediterranean Fever , Humans , Mice , Animals , Pyrin/genetics , Pyrin/metabolism , Familial Mediterranean Fever/genetics , Familial Mediterranean Fever/metabolism , Inflammasomes/metabolism , Mutation , Macrophages/metabolism
8.
PLoS Pathog ; 17(6): e1009687, 2021 06.
Article in English | MEDLINE | ID: mdl-34181691

ABSTRACT

COVID-19 outbreak is the biggest threat to human health in recent history. Currently, there are over 1.5 million related deaths and 75 million people infected around the world (as of 22/12/2020). The identification of virulence factors which determine disease susceptibility and severity in different cell types remains an essential challenge. The serine protease TMPRSS2 has been shown to be important for S protein priming and viral entry, however, little is known about its regulation. SPINT2 is a member of the family of Kunitz type serine protease inhibitors and has been shown to inhibit TMPRSS2. Here, we explored the existence of a co-regulation between SPINT2/TMPRSS2 and found a tightly regulated protease/inhibitor expression balance across tissues. We found that SPINT2 negatively correlates with SARS-CoV-2 expression in Calu-3 and Caco-2 cell lines and was down-regulated in secretory cells from COVID-19 patients. We validated our findings using Calu-3 cell lines and observed a strong increase in viral load after SPINT2 knockdown, while overexpression lead to a drastic reduction of the viral load. Additionally, we evaluated the expression of SPINT2 in datasets from comorbid diseases using bulk and scRNA-seq data. We observed its down-regulation in colon, kidney and liver tumors as well as in alpha pancreatic islets cells from diabetes Type 2 patients, which could have implications for the observed comorbidities in COVID-19 patients suffering from chronic diseases.


Subject(s)
COVID-19/metabolism , Membrane Glycoproteins/metabolism , SARS-CoV-2/metabolism , Virus Internalization , A549 Cells , COVID-19/genetics , Caco-2 Cells , Humans , Membrane Glycoproteins/genetics , SARS-CoV-2/genetics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Severity of Illness Index
9.
J Clin Immunol ; 42(2): 325-335, 2022 02.
Article in English | MEDLINE | ID: mdl-34783940

ABSTRACT

PURPOSE: NLRC4-associated autoinflammatory disease (NLRC4-AID) is an autosomal dominant condition presenting with a range of clinical manifestations which can include macrophage activation syndrome (MAS) and severe enterocolitis. We now report the first homozygous mutation in NLRC4 (c.478G > A, p.A160T) causing autoinflammatory disease with immune dysregulation and find that heterozygous carriers in the general population are at increased risk of developing ulcerative colitis. METHODS: Circulating immune cells and inflammatory markers were profiled and historical clinical data interrogated. DNA was extracted and sequenced using standard procedures. Inflammasome activation assays for ASC speck formation, pyroptosis, and IL-1ß/IL-18 secretion confirmed pathogenicity of the mutation in vitro. Genome-wide association of NLRC4 (A160T) with ulcerative colitis was examined using data from the IBD exomes portal. RESULTS: A 60-year-old Brazilian female patient was evaluated for recurrent episodes of systemic inflammation from six months of age. Episodes were characterized by recurrent low-grade fever, chills, oral ulceration, uveitis, arthralgia, and abdominal pain, followed by diarrhea with mucus and variable skin rash. High doses of corticosteroids were somewhat effective in controlling disease and anti-IL-1ß therapy partially controlled symptoms. While on treatment, serum IL-1ß and IL-18 levels remained elevated. Genetic investigations identified a homozygous mutation in NLRC4 (A160T), inherited in a recessive fashion. Increased ASC speck formation and IL-1ß/IL-18 secretion confirmed pathogenicity when NLRC4 (A160T) was analyzed in human cell lines. This allele is significantly enriched in patients with ulcerative colitis: OR 2.546 (95% 1.778-3.644), P = 0.01305. CONCLUSION: NLRC4 (A160T) can either cause recessively inherited autoinflammation and immune dysregulation, or function as a heterozygous risk factor for the development of ulcerative colitis.


Subject(s)
Colitis, Ulcerative , Hereditary Autoinflammatory Diseases , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Calcium-Binding Proteins/genetics , Colitis, Ulcerative/diagnosis , Colitis, Ulcerative/genetics , Female , Genome-Wide Association Study , Humans , Inflammasomes/metabolism , Middle Aged
10.
PLoS Pathog ; 16(8): e1008752, 2020 08.
Article in English | MEDLINE | ID: mdl-32760121

ABSTRACT

Members of the family of pyrin and HIN domain containing (PYHIN) proteins play an emerging role in innate immunity. While absent in melanoma 2 (AIM2) acts a cytosolic sensor of non-self DNA and plays a key role in inflammasome assembly, the γ-interferon-inducible protein 16 (IFI16) restricts retroviral gene expression by sequestering the transcription factor Sp1. Here, we show that the remaining two human PYHIN proteins, i.e. myeloid cell nuclear differentiation antigen (MNDA) and pyrin and HIN domain family member 1 (PYHIN1 or IFIX) share this antiretroviral function of IFI16. On average, knock-down of each of these three nuclear PYHIN proteins increased infectious HIV-1 yield from human macrophages by more than an order of magnitude. Similarly, knock-down of IFI16 strongly increased virus transcription and production in primary CD4+ T cells. The N-terminal pyrin domain (PYD) plus linker region containing a nuclear localization signal (NLS) were generally required and sufficient for Sp1 sequestration and anti-HIV-1 activity of IFI16, MNDA and PYHIN1. Replacement of the linker region of AIM2 by the NLS-containing linker of IFI16 resulted in a predominantly nuclear localization and conferred direct antiviral activity to AIM2 while attenuating its ability to form inflammasomes. The reverse change caused nuclear-to-cytoplasmic relocalization of IFI16 and impaired its antiretroviral activity but did not result in inflammasome assembly. We further show that the Zn-finger domain of Sp1 is critical for the interaction with IFI16 supporting that pyrin domains compete with DNA for Sp1 binding. Finally, we found that human PYHIN proteins also inhibit Hepatitis B virus and simian vacuolating virus 40 as well as the LINE-1 retrotransposon. Altogether, our data show that IFI16, PYHIN1 and MNDA restrict HIV-1 and other viral pathogens by interfering with Sp1-dependent gene expression and support an important role of nuclear PYHIN proteins in innate antiviral immunity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Nucleus/metabolism , HIV Infections/prevention & control , HIV-1/immunology , Macrophages/immunology , Nuclear Proteins/metabolism , Sp1 Transcription Factor/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cell Nucleus/genetics , DNA, Viral/genetics , HEK293 Cells , HIV Infections/immunology , HIV Infections/pathology , HIV Infections/virology , HIV-1/genetics , HIV-1/isolation & purification , Hep G2 Cells , Humans , Immunity, Innate/immunology , Inflammasomes/genetics , Inflammasomes/immunology , Macrophages/metabolism , Macrophages/virology , Nuclear Proteins/genetics , Sp1 Transcription Factor/genetics , Virus Replication
11.
Rheumatology (Oxford) ; 59(9): 2334-2339, 2020 09 01.
Article in English | MEDLINE | ID: mdl-31873740

ABSTRACT

OBJECTIVES: Here we investigated a patient with inflammatory corneal intraepithelial dyskeratosis, mucosal inflammation, tooth abnormalities and, eczema to uncover the genetic and immunological basis of the disease. METHODS: On suspicion of an autoinflammatory condition, Sanger sequencing of nucleotide-binding oligomerization domain-like, leucine-rich repeat pyrin domain containing 1 (NLRP1) was performed and combined with an in vitro inflammasome reconstitution assay to measure caspase-1-mediated IL-1ß cleavage, stimulation of patient peripheral blood mononuclear cells (PBMCs) and whole blood to measure IL-1ß, IL-18 production and quantification of apoptosis-associated speck-like protein containing CARD (ASC) speck formation as a measure of inflammasome activation by flow cytometry. RESULTS: Sanger sequencing revealed a novel mutation (c.175G>C, p.A59P; NM_33004.4) in the inflammasome molecule NLRP1 segregating with disease, although with incomplete penetrance, in three generations. We found that patient PBMCs produced increased IL-1ß in response to inflammatory stimuli, as well as increased constitutive levels of IL-18. Moreover, we demonstrate that expression of the identified NLRP1 A59P variant caused spontaneous IL-1ß cleavage to mature IL-1ß. In addition, patient PBMCs responded to NLRP1 stimulation with increased ASC speck formation as a reflection of elevated inflammasome activity. CONCLUSION: We demonstrate that this novel NLRP1 A59P variant caused increased activation of the NLRP1 inflammasome, resulting in constitutively and inducibly elevated IL-1ß and IL-18 synthesis. We suggest the NLRP1 mutation underlies the pathogenesis of this rare autoinflammatory dyskeratotic disease inherited in an autosomal dominant manner with incomplete penetrance in the patient and within the family for several generations.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Corneal Diseases/genetics , Dyskeratosis Congenita/genetics , Hereditary Autoinflammatory Diseases/genetics , Child, Preschool , Humans , Male , Mutation , NLR Proteins
12.
Proc Natl Acad Sci U S A ; 114(38): 10184-10189, 2017 09 19.
Article in English | MEDLINE | ID: mdl-28874561

ABSTRACT

CD47 is an antiphagocytic ligand broadly expressed on normal and malignant tissues that delivers an inhibitory signal through the receptor signal regulatory protein alpha (SIRPα). Inhibitors of the CD47-SIRPα interaction improve antitumor antibody responses by enhancing antibody-dependent cellular phagocytosis (ADCP) in xenograft models. Endogenous expression of CD47 on a variety of cell types, including erythrocytes, creates a formidable antigen sink that may limit the efficacy of CD47-targeting therapies. We generated a nanobody, A4, that blocks the CD47-SIRPα interaction. A4 synergizes with anti-PD-L1, but not anti-CTLA4, therapy in the syngeneic B16F10 melanoma model. Neither increased dosing nor half-life extension by fusion of A4 to IgG2a Fc (A4Fc) overcame the issue of an antigen sink or, in the case of A4Fc, systemic toxicity. Generation of a B16F10 cell line that secretes the A4 nanobody showed that an enhanced response to several immune therapies requires near-complete blockade of CD47 in the tumor microenvironment. Thus, strategies to localize CD47 blockade to tumors may be particularly valuable for immune therapy.


Subject(s)
CD47 Antigen/antagonists & inhibitors , Immunotherapy/methods , Melanoma, Experimental/therapy , Single-Domain Antibodies/therapeutic use , Anemia/chemically induced , Animals , CD47 Antigen/immunology , Drug Evaluation, Preclinical , Mice, Inbred C57BL , Phagocytosis , Single-Domain Antibodies/immunology , Single-Domain Antibodies/pharmacology , Tumor Microenvironment
13.
J Physiol ; 597(22): 5335-5348, 2019 11.
Article in English | MEDLINE | ID: mdl-31490557

ABSTRACT

Rapid inflammatory responses to cytosolic threats are mediated by inflammasomes - large macromolecular signalling complexes that control the activation of the pro-inflammatory cytokines interleukin (IL)-1ß and IL-18, as well as cell death by pyroptosis. Different inflammasome sensors are activated by diverse direct and indirect signals, and subsequently nucleate the polymerization of the adaptor molecule ASC to form signalling platforms macroscopically observed as ASC specks. Caspase-1 is autocatalytically activated at these sites and subsequently matures pro-inflammatory cytokines and the pore-forming effector molecule gasdermin D. While most molecules and basic assembly principles have been deduced from reductionist experimental systems, we still lack fundamental information on the structure and regulation of these complexes in their physiological environment and in the interplay with other signalling pathways. In this review, novel experimental approaches are proposed, including some that rely on nanobodies and single domain antibodies, to understand inflammasome assembly and regulation in the context of the relevant tissues or cells.


Subject(s)
Inflammasomes/physiology , Signal Transduction/physiology , Animals , Antibodies/metabolism , Humans , Inflammation/metabolism , Inflammation/physiopathology , Single-Domain Antibodies/metabolism
15.
EMBO Rep ; 18(6): 1027-1037, 2017 06.
Article in English | MEDLINE | ID: mdl-28396572

ABSTRACT

The transcription and replication machinery of negative-stranded RNA viruses presents a possible target for interference in the viral life cycle. We demonstrate the validity of this concept through the use of cytosolically expressed single-domain antibody fragments (VHHs) that protect cells from a lytic infection with vesicular stomatitis virus (VSV) by targeting the viral nucleoprotein N. We define the binding sites for two such VHHs, 1004 and 1307, by X-ray crystallography to better understand their inhibitory properties. We found that VHH 1307 competes with the polymerase cofactor P for binding and thus inhibits replication and mRNA transcription, while binding of VHH 1004 likely only affects genome replication. The functional relevance of these epitopes is confirmed by the isolation of escape mutants able to replicate in the presence of the inhibitory VHHs. The escape mutations allow identification of the binding site of a third VHH that presumably competes with P for binding at another site than 1307. Collectively, these binding sites uncover different features on the N protein surface that may be suitable for antiviral intervention.


Subject(s)
Antibodies, Viral/metabolism , Nucleocapsid Proteins/chemistry , Nucleocapsid Proteins/immunology , Single-Domain Antibodies/metabolism , Vesicular stomatitis Indiana virus/physiology , Virus Replication , A549 Cells , Animals , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Binding Sites , Crystallography, X-Ray , DNA Replication , Humans , Mutation , Nucleocapsid Proteins/metabolism , RNA, Viral , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/immunology , Transcription, Genetic , Vesicular stomatitis Indiana virus/drug effects , Vesicular stomatitis Indiana virus/genetics , Vesicular stomatitis Indiana virus/immunology
16.
PLoS Pathog ; 11(10): e1005188, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26431038

ABSTRACT

The ability of phagocytes to clear pathogens is an essential attribute of the innate immune response. The role of signaling lipid molecules such as phosphoinositides is well established, but the role of membrane sphingolipids in phagocytosis is largely unknown. Using a genetic approach and small molecule inhibitors, we show that phagocytosis of Candida albicans requires an intact sphingolipid biosynthetic pathway. Blockade of serine-palmitoyltransferase (SPT) and ceramide synthase-enzymes involved in sphingolipid biosynthesis- by myriocin and fumonisin B1, respectively, impaired phagocytosis by phagocytes. We used CRISPR/Cas9-mediated genome editing to generate Sptlc2-deficient DC2.4 dendritic cells, which lack serine palmitoyl transferase activity. Sptlc2-/- DC2.4 cells exhibited a stark defect in phagocytosis, were unable to bind fungal particles and failed to form a normal phagocytic cup to engulf C. albicans. Supplementing the growth media with GM1, the major ganglioside present at the cell surface, restored phagocytic activity of Sptlc2-/- DC2.4 cells. While overall membrane trafficking and endocytic pathways remained functional, Sptlc2-/- DC2.4 cells express reduced levels of the pattern recognition receptors Dectin-1 and TLR2 at the cell surface. Consistent with the in vitro data, compromised sphingolipid biosynthesis in mice sensitizes the animal to C. albicans infection. Sphingolipid biosynthesis is therefore critical for phagocytosis and in vivo clearance of C. albicans.


Subject(s)
Candida albicans , Candidiasis/immunology , Host-Pathogen Interactions/immunology , Phagocytosis/physiology , Sphingolipids/biosynthesis , Animals , Candida albicans/immunology , Cell Line , Chromatography, Thin Layer , Dendritic Cells/immunology , Dendritic Cells/microbiology , Disease Models, Animal , Flow Cytometry , Gene Knockout Techniques , Humans , Mass Spectrometry , Mice
17.
J Virol ; 89(5): 2792-800, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25540369

ABSTRACT

UNLABELLED: Perturbation of protein-protein interactions relies mostly on genetic approaches or on chemical inhibition. Small RNA viruses, such as influenza A virus, do not easily lend themselves to the former approach, while chemical inhibition requires that the target protein be druggable. A lack of tools thus constrains the functional analysis of influenza virus-encoded proteins. We generated a panel of camelid-derived single-domain antibody fragments (VHHs) against influenza virus nucleoprotein (NP), a viral protein essential for nuclear trafficking and packaging of the influenza virus genome. We show that these VHHs can target NP in living cells and perturb NP's function during infection. Cytosolic expression of NP-specific VHHs (αNP-VHHs) disrupts virus replication at an early stage of the life cycle. Based on their specificity, these VHHs fall into two distinct groups. Both prevent nuclear import of the viral ribonucleoprotein (vRNP) complex without disrupting nuclear import of NP alone. Different stages of the virus life cycle thus rely on distinct nuclear localization motifs of NP. Their molecular characterization may afford new means of intervention in the virus life cycle. IMPORTANCE: Many proteins encoded by RNA viruses are refractory to manipulation due to their essential role in replication. Thus, studying their function and determining how to disrupt said function through pharmaceutical intervention are difficult. We present a novel method based on single-domain-antibody technology that permits specific targeting and disruption of an essential influenza virus protein in the absence of genetic manipulation of influenza virus itself. Characterization of such interactions may help identify new targets for pharmaceutical intervention. This approach can be extended to study proteins encoded by other viral pathogens.


Subject(s)
Cell Nucleus/chemistry , Cell Nucleus/virology , Influenza A virus/physiology , RNA-Binding Proteins/analysis , Single-Domain Antibodies/metabolism , Viral Core Proteins/analysis , Virology/methods , Virus Replication , Animals , Cell Line , Dogs , Gene Expression , Molecular Biology/methods , Nucleocapsid Proteins , Single-Domain Antibodies/genetics
18.
J Biol Chem ; 289(50): 34490-502, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25320092

ABSTRACT

ER-resident proteins destined for degradation are dislocated into the cytosol by components of the ER quality control machinery for proteasomal degradation. Dislocation substrates are ubiquitylated in the cytosol by E2 ubiquitin-conjugating/E3 ligase complexes. UBE2J1 is one of the well-characterized E2 enzymes that participate in this process. However, the physiological function of Ube2j1 is poorly defined. We find that Ube2j1(-/-) mice have reduced viability and fail to thrive early after birth. Male Ube2j1(-/-) mice are sterile due to a defect in late spermatogenesis. Ultrastructural analysis shows that removal of the cytoplasm is incomplete in Ube2j1(-/-) elongating spermatids, compromising the release of mature elongate spermatids into the lumen of the seminiferous tubule. Our findings identify an essential function for the ubiquitin-proteasome-system in spermiogenesis and define a novel, non-redundant physiological function for the dislocation step of ER quality control.


Subject(s)
Spermatogenesis , Ubiquitin-Conjugating Enzymes/metabolism , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Line , Endoplasmic Reticulum/metabolism , Immunoglobulins/metabolism , Infertility, Male/metabolism , Infertility, Male/pathology , Male , Mice , Spermatids/cytology , Spermatids/pathology , Ubiquitin-Conjugating Enzymes/deficiency , Unfolded Protein Response , Up-Regulation
19.
Biophys J ; 105(2): 420-31, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23870263

ABSTRACT

Recent studies have revealed new insights into the endocytosis of vaccinia virus (VACV). However, the mechanism of fusion between viral and cellular membranes remains unknown. We developed a microfluidic device with a cell-trap array for immobilization of individual cells, with which we analyzed the acid-dependent fusion of single virions. VACV particles incorporating enhanced green fluorescent protein (EGFP) and labeled with self-quenching concentrations of R18 membrane dye were used in combination with total internal reflection fluorescence microscopy to measure the kinetics of R18 dequenching and thus single hemifusion events initiated by a fast low-pH trigger. These studies revealed unexpectedly long lag phases between pH change and hemifusion. In addition, we found that EGFP fluorescence in the virus was quenched upon acidification, indicating that protons could access the virus core, possibly through a proton channel. In a fraction of virus particles, EGFP fluorescence was recovered, presumably after fusion-pore formation and exposure of the core to the physiological pH of the host-cell cytosol. Given that virus-encoded cation channels play a crucial role in the life cycle of many viruses and can serve as antiviral drug targets, further investigations into a potential VACV viroporin are justified. Our findings indicate that the microfluidic device described may be highly beneficial to similar studies requiring fast kinetic measurements.


Subject(s)
Protons , Vaccinia virus/metabolism , Virion/metabolism , Virus Internalization , Animals , HeLa Cells , Humans , Hydrogen-Ion Concentration , Ion Transport , Microfluidics , Time Factors
20.
Proc Natl Acad Sci U S A ; 107(20): 9346-51, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20439710

ABSTRACT

To enter host cells, vaccinia virus, a prototype poxvirus, can induce transient macropinocytosis followed by endocytic internalization and penetration through the limiting membrane of pinosomes by membrane fusion. Although mature virions (MVs) of the Western reserve (WR) strain do this in HeLa cells by activating transient plasma membrane blebbing, MVs from the International Health Department-J strain were found to induce rapid formation (and lengthening) of filopodia. When the signaling pathways underlying these responses were compared, differences were observed at the level of Rho GTPases. Key to the filopodial formation was the virus-induced activation of Cdc42, and for the blebbing response the activation of Rac1. In addition, unlike WR, International Health Department-J MVs did not rely on genistein-sensitive tyrosine kinase and PI(3)K activities. Only WR MVs had membrane fusion activity at low pH. Inhibitor profiling showed that MVs from both strains entered cells by macropinocytosis and that this was induced by virion-exposed phosphatidylserine. Both MVs relied on the activation of epidermal growth factor receptor, on serine/threonine kinases, protein kinase C, and p21-activated kinase 1. The results showed that different strains of the same virus can elicit dramatically different responses in host cells during entry, and that different macropinocytic mechanisms are possible in the same cell line through subtle differences in the activating ligand.


Subject(s)
Pinocytosis/physiology , Signal Transduction/physiology , Vaccinia virus/physiology , Virus Internalization , rho GTP-Binding Proteins/metabolism , ErbB Receptors/metabolism , Flow Cytometry , Green Fluorescent Proteins/metabolism , Hydrogen-Ion Concentration , Pseudopodia/virology , RNA, Small Interfering/genetics , Species Specificity , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL