Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Immunity ; 36(6): 933-46, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22749352

ABSTRACT

The mitochondrial protein MAVS (also known as IPS-1, VISA, and CARDIF) interacts with RIG-I-like receptors (RLRs) to induce type I interferon (IFN-I). NLRX1 is a mitochondrial nucleotide-binding, leucine-rich repeats (NLR)-containing protein that attenuates MAVS-RLR signaling. Using Nlrx1(-/-) cells, we confirmed that NLRX1 attenuated IFN-I production, but additionally promoted autophagy during viral infection. This dual function of NLRX1 paralleled the previously described functions of the autophagy-related proteins Atg5-Atg12, but NLRX1 did not associate with Atg5-Atg12. High-throughput quantitative mass spectrometry and endogenous protein-protein interaction revealed an NLRX1-interacting partner, mitochondrial Tu translation elongation factor (TUFM). TUFM interacted with Atg5-Atg12 and Atg16L1 and has similar functions as NLRX1 by inhibiting RLR-induced IFN-I but promoting autophagy. In the absence of NLRX1, increased IFN-I and decreased autophagy provide an advantage for host defense against vesicular stomatitis virus. This study establishes a link between an NLR protein and the viral-induced autophagic machinery via an intermediary partner, TUFM.


Subject(s)
Autophagy/physiology , Interferon Type I/biosynthesis , Mitochondrial Proteins/physiology , Peptide Elongation Factor Tu/physiology , Adaptor Proteins, Signal Transducing/physiology , Amino Acid Sequence , Animals , Autophagy-Related Protein 12 , Autophagy-Related Protein 5 , Autophagy-Related Proteins , Carrier Proteins/physiology , Cytokines/biosynthesis , Cytokines/genetics , DEAD Box Protein 58 , DEAD-box RNA Helicases/physiology , Fibroblasts/metabolism , Gene Expression Regulation/immunology , HEK293 Cells , Humans , Interferon Type I/genetics , Macrophages, Peritoneal/cytology , Macrophages, Peritoneal/immunology , Mice , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/physiology , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Molecular Sequence Data , Multiprotein Complexes/physiology , Peptide Elongation Factor Tu/chemistry , Protein Interaction Mapping , Proteins/physiology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/physiology , Sequence Alignment , Sequence Homology, Amino Acid , Specific Pathogen-Free Organisms , Vesiculovirus/physiology
2.
Immunity ; 34(6): 854-65, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21703540

ABSTRACT

The nucleotide-binding domain and leucine-rich-repeat-containing (NLR) proteins regulate innate immunity. Although the positive regulatory impact of NLRs is clear, their inhibitory roles are not well defined. We showed that Nlrx1(-/-) mice exhibited increased expression of antiviral signaling molecules IFN-ß, STAT2, OAS1, and IL-6 after influenza virus infection. Consistent with increased inflammation, Nlrx1(-/-) mice exhibited marked morbidity and histopathology. Infection of these mice with an influenza strain that carries a mutated NS-1 protein, which normally prevents IFN induction by interaction with RNA and the intracellular RNA sensor RIG-I, further exacerbated IL-6 and type I IFN signaling. NLRX1 also weakened cytokine responses to the 2009 H1N1 pandemic influenza virus in human cells. Mechanistically, Nlrx1 deletion led to constitutive interaction of MAVS and RIG-I. Additionally, an inhibitory function is identified for NLRX1 during LPS activation of macrophages where the MAVS-RIG-I pathway was not involved. NLRX1 interacts with TRAF6 and inhibits NF-κB activation. Thus, NLRX1 functions as a checkpoint of overzealous inflammation.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Mitochondrial Proteins/immunology , Orthomyxoviridae Infections/immunology , Signal Transduction , Adaptor Proteins, Signal Transducing/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cells, Cultured , Interferon-beta/biosynthesis , Interferon-beta/immunology , Interleukin-6/biosynthesis , Interleukin-6/immunology , Macrophages/immunology , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Mice, Knockout , Mitochondrial Proteins/deficiency , NF-kappa B/immunology , NF-kappa B/metabolism , Nerve Tissue Proteins/immunology , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface , TNF Receptor-Associated Factor 6/immunology , TNF Receptor-Associated Factor 6/metabolism
3.
Immunity ; 30(4): 556-65, 2009 Apr 17.
Article in English | MEDLINE | ID: mdl-19362020

ABSTRACT

The nucleotide-binding domain and leucine-rich-repeat-containing (NLR) family of pattern-recognition molecules mediate host immunity to various pathogenic stimuli. However, in vivo evidence for the involvement of NLR proteins in viral sensing has not been widely investigated and remains controversial. As a test of the physiologic role of the NLR molecule NLRP3 during RNA viral infection, we explored the in vivo role of NLRP3 inflammasome components during influenza virus infection. Mice lacking Nlrp3, Pycard, or caspase-1, but not Nlrc4, exhibited dramatically increased mortality and a reduced immune response after exposure to the influenza virus. Utilizing analogs of dsRNA (poly(I:C)) and ssRNA (ssRNA40), we demonstrated that an NLRP3-mediated response could be activated by RNA species. Mechanistically, NLRP3 inflammasome activation by the influenza virus was dependent on lysosomal maturation and reactive oxygen species (ROS). Inhibition of ROS induction eliminated IL-1beta production in animals during influenza infection. Together, these data place the NLRP3 inflammasome as an essential component in host defense against influenza infection through the sensing of viral RNA.


Subject(s)
Carrier Proteins/physiology , Exosomes/immunology , Immunity, Innate , Influenza A virus/immunology , Orthomyxoviridae Infections/immunology , RNA, Viral , Virus Diseases/immunology , Animals , Carrier Proteins/genetics , Cell Line , Humans , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Influenza, Human/immunology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein
4.
Immunity ; 28(6): 735-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18549796

ABSTRACT

Mitochondrial antiviral immunity involves the detection of viral RNA by intracellular pattern-recognition receptors (PRRs) belonging to the RIG-I-like helicase family. The convergence of these and other signaling molecules to the outer mitochondrial membrane results in the rapid induction of antiviral cytokines including type-1 interferon. Here, we discuss recent studies describing new molecules implicated in the regulation of this antiviral response.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cytokines/metabolism , Mitochondria/metabolism , RNA Helicases/metabolism , RNA, Viral/metabolism , Receptors, Pattern Recognition/metabolism , Signal Transduction , Viruses/immunology , Adaptor Proteins, Signal Transducing/immunology , Animals , Humans , Interferon Type I/immunology , Interferon Type I/metabolism , Mitochondria/immunology , Toll-Like Receptors/metabolism
5.
Mol Cell ; 35(1): 128-35, 2009 Jul 10.
Article in English | MEDLINE | ID: mdl-19595722

ABSTRACT

DNA recombination and repair pathways require structure-specific endonucleases to process DNA structures that include forks, flaps, and Holliday junctions. Previously, we determined that the Drosophila MEI-9-ERCC1 endonuclease interacts with the MUS312 protein to produce meiotic crossovers, and that MUS312 has a MEI-9-independent role in interstrand crosslink (ICL) repair. The importance of MUS312 to pathways crucial for maintaining genomic stability in Drosophila prompted us to search for orthologs in other organisms. Based on sequence, expression pattern, conserved protein-protein interactions, and ICL repair function, we determined that the mammalian ortholog of MUS312 is BTBD12. Orthology between these proteins and S. cerevisiae Slx4 helped identify a conserved interaction with a second structure-specific endonuclease, SLX1. Genetic and biochemical evidence described here and in related papers suggest that MUS312 and BTBD12 direct Holliday junction resolution by at least two distinct endonucleases in different recombination and repair contexts.


Subject(s)
DNA Repair , Drosophila Proteins/metabolism , Endodeoxyribonucleases/metabolism , Endonucleases/metabolism , Recombinases/metabolism , Recombination, Genetic , Amino Acid Sequence , Animals , Brain/abnormalities , Brain/metabolism , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Endodeoxyribonucleases/genetics , Endonucleases/genetics , Female , Gene Expression Profiling , HeLa Cells , Humans , Male , Mice , Molecular Sequence Data , Mutation , Protein Binding , RNA, Small Interfering/genetics , Recombinases/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Sequence Homology, Amino Acid , Transfection , Two-Hybrid System Techniques
6.
Nature ; 451(7178): 573-7, 2008 Jan 31.
Article in English | MEDLINE | ID: mdl-18200010

ABSTRACT

The RIG-like helicase (RLH) family of intracellular receptors detect viral nucleic acid and signal through the mitochondrial antiviral signalling adaptor MAVS (also known as Cardif, VISA and IPS-1) during a viral infection. MAVS activation leads to the rapid production of antiviral cytokines, including type 1 interferons. Although MAVS is vital to antiviral immunity, its regulation from within the mitochondria remains unknown. Here we describe human NLRX1, a highly conserved nucleotide-binding domain (NBD)- and leucine-rich-repeat (LRR)-containing family member (known as NLR) that localizes to the mitochondrial outer membrane and interacts with MAVS. Expression of NLRX1 results in the potent inhibition of RLH- and MAVS-mediated interferon-beta promoter activity and in the disruption of virus-induced RLH-MAVS interactions. Depletion of NLRX1 with small interference RNA promotes virus-induced type I interferon production and decreases viral replication. This work identifies NLRX1 as a check against mitochondrial antiviral responses and represents an intersection of three ancient cellular processes: NLR signalling, intracellular virus detection and the use of mitochondria as a platform for anti-pathogen signalling. This represents a conceptual advance, in that NLRX1 is a modulator of pathogen-associated molecular pattern receptors rather than a receptor, and identifies a key therapeutic target for enhancing antiviral responses.


Subject(s)
Mitochondria/immunology , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Viruses/immunology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Cloning, Molecular , Computational Biology , Humans , Interferon-beta/biosynthesis , Interferon-beta/genetics , Interferon-beta/metabolism , Mice , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/genetics , NF-kappa B/metabolism , Protein Binding , Protein Transport , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Virus Replication
7.
J Biol Chem ; 286(22): 19605-16, 2011 Jun 03.
Article in English | MEDLINE | ID: mdl-21487011

ABSTRACT

ASC/PYCARD is a common adaptor for a diverse set of inflammasomes that activate caspase-1, most prominently the NLR-based inflammasome. Mounting evidence indicates that ASC and these NLRs also elicit non-overlapping functions, but the molecular basis for this difference is unclear. To address this, we performed microarray and network analysis of ASC shRNA knockdown cells. In pathogen-infected cells, an ASC-dependent interactome is centered on the mitogen-activated protein kinase (MAPK) ERK and on multiple chemokines. ASC did not affect the expression of MAPK but affected its phosphorylation by pathogens and Toll-like receptor agonists via suppression of the dual-specificity phosphatase, DUSP10/MKP5. Chemokine induction, DUSP function, and MAPK phosphorylation were independent of caspase-1 and IL-1ß. MAPK activation by pathogen was abrogated in Asc(-/-) but not Nlrp3(-/-), Nlrc4(-/-), or Casp1(-/-) macrophages. These results demonstrate a function for ASC that is distinct from the inflammasome in modulating MAPK activity and chemokine expression and further identify DUSP10 as a novel ASC target.


Subject(s)
Chemokines/biosynthesis , Cytoskeletal Proteins/metabolism , Dual-Specificity Phosphatases/metabolism , Inflammasomes/metabolism , Macrophages/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Phosphatases/metabolism , Animals , Apoptosis Regulatory Proteins , CARD Signaling Adaptor Proteins , Cell Line , Chemokines/genetics , Cytoskeletal Proteins/genetics , Dual-Specificity Phosphatases/genetics , Enzyme Activation/physiology , Gene Knockdown Techniques , Humans , Inflammasomes/genetics , Macrophages/cytology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/genetics , Mitogen-Activated Protein Kinase Phosphatases/genetics
8.
J Immunol ; 185(8): 4515-9, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20861349

ABSTRACT

Nucleotide-binding domain leucine-rich repeat (NLR) proteins are regulators of inflammation and immunity. Although first described 8 y ago, a physiologic role for NLRP12 has remained elusive until now. We find that murine Nlrp12, an NLR linked to atopic dermatitis and hereditary periodic fever in humans, is prominently expressed in dendritic cells (DCs) and neutrophils. Nlrp12-deficient mice exhibit attenuated inflammatory responses in two models of contact hypersensitivity that exhibit features of allergic dermatitis. This cannot be attributed to defective Ag processing/presentation, inflammasome activation, or measurable changes in other inflammatory cytokines. Rather, Nlrp12(-/-) DCs display a significantly reduced capacity to migrate to draining lymph nodes. Both DCs and neutrophils fail to respond to chemokines in vitro. These findings indicate that NLRP12 is important in maintaining neutrophils and peripheral DCs in a migration-competent state.


Subject(s)
Chemotaxis, Leukocyte/immunology , Dendritic Cells/immunology , Dermatitis, Contact/immunology , Intracellular Signaling Peptides and Proteins/immunology , Myeloid Cells/immunology , Animals , Dendritic Cells/metabolism , Dermatitis, Contact/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , Polymerase Chain Reaction
9.
J Immunol ; 182(4): 2395-404, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19201894

ABSTRACT

Periodontal disease is a chronic inflammatory disorder that leads to the destruction of tooth-supporting tissue and affects 10-20 million people in the U.S. alone. The oral pathogen Porphyromonas gingivalis causes inflammatory host response leading to periodontal and other secondary inflammatory diseases. To identify molecular components that control host response to P. gingivalis in humans, roles for the NLR (NBD-LRR) protein, NLRP3 (cryopyrin, NALP3), and its adaptor apoptotic speck protein containing a C-terminal caspase recruitment domain (ASC) were studied. P. gingivalis strain A7436 induces cell death in THP1 monocytic cells and in human primary peripheral blood macrophages. This process is ASC and NLRP3 dependent and can be replicated by P. gingivalis LPS and Escherichia coli. P. gingivalis-induced cell death is caspase and IL-1 independent and exhibits morphological features consistent with necrosis including loss of membrane integrity and release of cellular content. Intriguingly, P. gingivalis-induced cell death is accompanied by the formation of ASC aggregation specks, a process not previously described during microbial infection. ASC specks are observed in P. gingivalis-infected primary human mononuclear cells and are dependent on NLRP3. This work shows that P. gingivalis causes ASC- and NLRP3-dependent necrosis, accompanied by ASC speck formation.


Subject(s)
Bacteroidaceae Infections/metabolism , Carrier Proteins/metabolism , Cytoskeletal Proteins/metabolism , Macrophages/microbiology , Monocytes/microbiology , Necrosis/metabolism , Bacteroidaceae Infections/immunology , Bacteroidaceae Infections/pathology , Blotting, Western , CARD Signaling Adaptor Proteins , Carrier Proteins/immunology , Cytoskeletal Proteins/immunology , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Humans , Macrophages/immunology , Macrophages/metabolism , Microscopy, Electron, Transmission , Monocytes/immunology , Monocytes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Necrosis/immunology , Necrosis/microbiology , Porphyromonas gingivalis , Reverse Transcriptase Polymerase Chain Reaction
10.
Proc Natl Acad Sci U S A ; 105(35): 13015-20, 2008 Sep 02.
Article in English | MEDLINE | ID: mdl-18728195

ABSTRACT

The semaphorin and plexin family of ligand and receptor proteins provides important axon guidance cues required for development. Recent studies have expanded the role of semaphorins and plexins in the regulation of cardiac, circulatory and immune system function. Within the immune system, semaphorins and plexins regulate cell-cell interactions through a complex network of receptor and ligand pairs. Immune cells at different stages of development often express multiple semaphorins and plexins, leading to multivariate interactions, involving more than one ligand and receptor within each functional group. Because of this complexity, the significance of semaphorin and plexin regulation on individual immune cell types has yet to be fully appreciated. In this work, we examined the regulation of T cells by semaphorin 6D. Both in vitro and in vivo T cell stimulation enhanced semaphorin 6D expression. However, semaphorin 6D was only expressed by a majority of T cells during the late phases of activation. Consequently, the targeted disruption of semaphorin 6D receptor-ligand interactions inhibited T cell proliferation at late but not early phases of activation. This proliferation defect was associated with reduced linker of activated T cells protein phosphorylation, which may reflect semaphorin 6D regulation of c-Abl kinase activity. Semaphorin 6D disruption also inhibited expression of CD127, which is required during the multiphase antigen-presenting cell and T cell interactions leading to selection of long-lived lymphocytes. This work reveals a role for semaphorin 6D as a regulator of the late phase of primary immune responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunity/immunology , Semaphorins/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , Cell Proliferation , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Semaphorins/antagonists & inhibitors , Signal Transduction
11.
Nat Commun ; 12(1): 1222, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33619272

ABSTRACT

Programmed death-ligand 1 is a glycoprotein expressed on antigen presenting cells, hepatocytes, and tumors which upon interaction with programmed death-1, results in inhibition of antigen-specific T cell responses. Here, we report a mechanism of inhibiting programmed death-ligand 1 through small molecule-induced dimerization and internalization. This represents a mechanism of checkpoint inhibition, which differentiates from anti-programmed death-ligand 1 antibodies which function through molecular disruption of the programmed death 1 interaction. Testing of programmed death ligand 1 small molecule inhibition in a humanized mouse model of colorectal cancer results in a significant reduction in tumor size and promotes T cell proliferation. In addition, antigen-specific T and B cell responses from patients with chronic hepatitis B infection are significantly elevated upon programmed death ligand 1 small molecule inhibitor treatment. Taken together, these data identify a mechanism of small molecule-induced programmed death ligand 1 internalization with potential therapeutic implications in oncology and chronic viral infections.


Subject(s)
B7-H1 Antigen/metabolism , Endocytosis , Immune Checkpoint Inhibitors/pharmacology , Small Molecule Libraries/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antiviral Agents/pharmacology , CHO Cells , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Cricetulus , Disease Models, Animal , Female , Hepatitis B virus/drug effects , Humans , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/metabolism , Protein Multimerization/drug effects , Small Molecule Libraries/chemistry
12.
13.
Sci Rep ; 10(1): 1835, 2020 02 04.
Article in English | MEDLINE | ID: mdl-32020034

ABSTRACT

Chronic hepatitis B (CHB) infection functional cure is defined as sustained loss of HBsAg and several therapeutic strategies are in clinical development designed to pharmacologically reduce serum HBsAg, break immune tolerance, and increase functional cure rates. However, little is known about pre-treatment HBsAg levels as an indicator of HBV immune potential. Here, we compared the phenotypes and HBV-specific response of lymphocytes in CHB patients stratified by serum HBsAg levels <500 (HBslo) or >50,000 IU/ml (HBshi) using immunological assays (flow cytometry, ICS, ELISPOT). HBshi patients had significantly higher expression of inhibitory PD-1 on CD4+ T cells, particularly among TEMRA subset, and higher FcRL5 expression on B cells. Upon HBcAg(core) or HBsAg(env)-stimulation, 85% and 60% of HBslo patients had IFNγ+TNFα+ and IFNγ+ IL2+ CD4+ T cell responses respectively, in comparison to 33% and 13% of HBshi patients. Checkpoint blockade with αPD-1 improved HBV-specific CD4+ T cell function only in HBslo patients. HBsAg-specific antibody-secreting cells (ASCs) response was not different between these groups, yet αPD-1 treatment resulted in significantly higher fold change in ASCs among patients with HBsAg <100 IU/ml compared to patients with HBsAg >5,000 IU/ml. Thus, serum HBsAg correlates with inhibitory receptor expression, HBV-specific CD4+ T cell responses, and augmentation by checkpoint blockade.


Subject(s)
B-Lymphocytes/immunology , Hepatitis B Surface Antigens/blood , Hepatitis B virus/immunology , Hepatitis B, Chronic/immunology , T-Lymphocytes/immunology , Biomarkers/blood , Flow Cytometry , Hepatitis B, Chronic/blood , Humans , Programmed Cell Death 1 Receptor/metabolism
14.
J Med Chem ; 57(5): 2091-106, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-23944386

ABSTRACT

Hepatitis C virus (HCV) assembles many host cellular proteins into unique membranous replication structures as a prerequisite for viral replication, and PI4KIIIα is an essential component of these replication organelles. RNA interference of PI4KIIIα results in a breakdown of this replication complex and cessation of HCV replication in Huh-7 cells. PI4KIIIα is a lipid kinase that interacts with the HCV nonstructural 5A protein (NS5A) and enriches the HCV replication complex with its product, phosphoinositol 4-phosphate (PI4P). Elevated levels of PI4P at the endoplasmic reticulum have been linked to HCV infection in the liver of HCV infected patients. We investigated if small molecule inhibitors of PI4KIIIα could inhibit HCV replication in vitro. The synthesis and structure-activity relationships associated with the biological inhibition of PI4KIIIα and HCV replication are described. These efforts led directly to identification of quinazolinone 28 that displays high selectivity for PI4KIIIα and potently inhibits HCV replication in vitro.


Subject(s)
1-Phosphatidylinositol 4-Kinase/antagonists & inhibitors , Antiviral Agents/pharmacology , Enzyme Inhibitors/pharmacology , Hepacivirus/drug effects , Animals , Antiviral Agents/chemistry , Drug Discovery , Enzyme Inhibitors/chemistry , Hepacivirus/enzymology , Hepacivirus/physiology , Magnetic Resonance Spectroscopy , Mass Spectrometry , Rats , Structure-Activity Relationship , Virus Replication/drug effects
15.
Methods Mol Biol ; 1031: 65-70, 2013.
Article in English | MEDLINE | ID: mdl-23824888

ABSTRACT

Mouse embryonic fibroblasts (MEFs) are commonly utilized as a primary cell culture model and have several advantages over other types of ex vivo-derived cells. However, the successful generation of MEFs is time consuming and requires a certain level of mouse expertise to successfully complete. Thus, primary ear-derived fibroblasts offer an acceptable alternative to MEFs. Fibroblasts derived from the pinna of adult mice are easily attainable with minimal skill, proliferate rapidly, and are easy to manipulate. Likewise, because they are derived from adult mice, other organs can be concurrently harvested for the isolation of additional types of primary cells. Similar to MEFs, ear fibroblasts are an excellent ex vivo model system to study mechanisms associated with virus infection and produce a diverse array of inflammatory mediators, such as cytokines and interferon. Here, we describe a highly versatile and simple method for the derivation, maintenance, and viral challenge of primary ear-derived fibroblasts from mice.


Subject(s)
Ear , Fibroblasts/cytology , Primary Cell Culture/methods , Animals , Cell Culture Techniques/methods , Mice
16.
Methods Mol Biol ; 629: 141-58, 2010.
Article in English | MEDLINE | ID: mdl-20387148

ABSTRACT

Shortly after the cellular mechanism of RNA interference (RNAi) was first described, scientists began using this powerful technique to study gene function. This included designing better methods for the successful delivery of small interfering RNAs (siRNAs) and short hairpin RNAs (shRNAs) into mammalian cells. While the simplest method for RNAi is the cytosolic delivery of siRNA oligonucleotides, this technique is limited to cells capable of transfection and is primarily utilized during transient in vitro studies. The introduction of shRNA into mammalian cells through infection with viral vectors allows for stable integration of shRNA and long-term knockdown of the targeted gene; however, several challenges exist with the implementation of this technology. Here we describe some well-tested protocols which should increase the chances of successful design, delivery, and assessment of gene knockdown by shRNA. We provide suggestions for designing shRNA targets and controls, a protocol for sequencing through the secondary structure of the shRNA hairpin structure, and protocols for packaging and delivery of shRNA lentiviral particles. Using real-time PCR and functional assays we demonstrate the successful knockdown of ASC, an inflammatory adaptor molecule. These studies demonstrate the practicality of including two shRNAs with different efficacies of knockdown to provide an additional level of control and to verify dose dependency of functional effects. Along with the methods described here, as new techniques and algorithms are designed in the future, shRNA is likely to include further promising application and continue to be a critical component of gene discovery.


Subject(s)
Gene Knockdown Techniques/methods , RNA, Small Interfering/chemical synthesis , RNA, Small Interfering/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Adhesion , Cell Line , Enzyme-Linked Immunosorbent Assay , Humans , Interleukin-1beta/metabolism , Lentivirus/physiology , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, RNA , Transduction, Genetic , Virus Assembly
17.
PLoS One ; 4(5): e5466, 2009.
Article in English | MEDLINE | ID: mdl-19404494

ABSTRACT

BACKGROUND: Host responses to viral infection include both immune activation and programmed cell death. The mitochondrial antiviral signaling adaptor, MAVS (IPS-1, VISA or Cardif) is critical for host defenses to viral infection by inducing type-1 interferons (IFN-I), however its role in virus-induced apoptotic responses has not been elucidated. PRINCIPAL FINDINGS: We show that MAVS causes apoptosis independent of its function in initiating IFN-I production. MAVS-induced cell death requires mitochondrial localization, is caspase dependent, and displays hallmarks of apoptosis. Furthermore, MAVS(-/-) fibroblasts are resistant to Sendai virus-induced apoptosis. A functional screen identifies the hepatitis C virus NS3/4A and the Severe Acute Respiratory Syndrome coronavirus (SARS-CoV) nonstructural protein (NSP15) as inhibitors of MAVS-induced apoptosis, possibly as a method of immune evasion. SIGNIFICANCE: This study describes a novel role for MAVS in controlling viral infections through the induction of apoptosis, and identifies viral proteins which inhibit this host response.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Apoptosis/physiology , Host-Pathogen Interactions/physiology , Viral Proteins/physiology , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis/immunology , Base Sequence , Caspases/metabolism , Cell Line , Cells, Cultured , Endoribonucleases , Host-Pathogen Interactions/immunology , Humans , Interferon Regulatory Factor-3/antagonists & inhibitors , Interferon Regulatory Factor-3/genetics , Interferon Type I/biosynthesis , Mice , Mice, Knockout , Mitochondria/metabolism , Models, Biological , NF-kappa B/metabolism , Protein Structure, Tertiary , RNA, Small Interfering/genetics , RNA-Dependent RNA Polymerase/physiology , Sendai virus/pathogenicity , Viral Nonstructural Proteins/physiology
18.
Mol Cell Biol ; 28(5): 1841-50, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18160710

ABSTRACT

The recently discovered nucleotide binding domain-leucine rich repeat (NLR) gene family is conserved from plants to mammals, and several members are associated with human autoinflammatory or immunodeficiency disorders. This family is defined by a central nucleotide binding domain that contains the highly conserved Walker A and Walker B motifs. Although the nucleotide binding domain is a defining feature of this family, it has not been extensively studied in its purified form. In this report, we show that purified Monarch-1/NLRP12, an NLR protein that negatively regulates NF-kappaB signaling, specifically binds ATP and exhibits ATP hydrolysis activity. Intact Walker A/B motifs are required for this activity. These motifs are also required for Monarch-1 to undergo self-oligomerization, Toll-like receptor- or CD40L-activated association with NF-kappaB-inducing kinase (NIK) and interleukin-1 receptor-associated kinase 1 (IRAK-1), degradation of NIK, and inhibition of IRAK-1 phosphorylation. The stable expression of a Walker A/B mutant in THP-1 monocytes results in increased production of proinflammatory cytokines and chemokines to an extent comparable to that in cells in which Monarch-1 is silenced via short hairpin RNA. The results of this study are consistent with a model wherein ATP binding regulates the anti-inflammatory activity of Monarch-1.


Subject(s)
Adenosine Triphosphate/metabolism , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Adenosine Triphosphate/analysis , Amino Acid Motifs , Amino Acid Sequence , CD40 Antigens/pharmacology , Cell Line , Chemokines/analysis , Cytokines/analysis , DNA, Complementary/genetics , Enzyme Activation , Escherichia coli/genetics , Hemagglutinins/metabolism , Humans , Interleukin-1 Receptor-Associated Kinases/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/isolation & purification , Kidney/cytology , Molecular Sequence Data , Monocytes/drug effects , Mutation , Precipitin Tests , Protein Structure, Tertiary , Recombinant Fusion Proteins/metabolism , Restriction Mapping , Time Factors , Transfection
19.
J Immunol ; 178(3): 1256-60, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17237370

ABSTRACT

CATERPILLER (NOD, NBD-LRR) proteins are rapidly emerging as important mediators of innate and adaptive immunity. Among these, Monarch-1 operates as a novel attenuating factor of inflammation by suppressing inflammatory responses in activated monocytes. However, the molecular mechanisms by which Monarch-1 performs this important function are not well understood. In this report, we show that Monarch-1 inhibits CD40-mediated activation of NF-kappaB via the non-canonical pathway in human monocytes. This inhibition stems from the ability of Monarch-1 to associate with and induce proteasome-mediated degradation of NF-kappaB inducing kinase. Congruently, silencing Monarch-1 with shRNA enhances the expression of p52-dependent chemokines.


Subject(s)
Chemokines/biosynthesis , Intracellular Signaling Peptides and Proteins/physiology , Monocytes/metabolism , NF-kappa B p52 Subunit/metabolism , NF-kappa B/metabolism , CD40 Antigens/antagonists & inhibitors , CD40 Antigens/metabolism , Humans , Inflammation , Proteasome Endopeptidase Complex/metabolism , Protein Serine-Threonine Kinases/metabolism , NF-kappaB-Inducing Kinase
20.
Exp Cell Res ; 313(1): 65-76, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-17069796

ABSTRACT

Analysis of lung cancer response to chemotherapeutic agents showed the accumulation of a Taxol-induced protein that reacted with an anti-phospho-MEK1/2 antibody. Mass spectroscopy identified the protein as nucleophosmin/B23 (NPM), a multifunctional protein with diverse roles: ribosome biosynthesis, p53 regulation, nuclear-cytoplasmic shuttling, and centrosome duplication. Our work demonstrates that following cellular exposure to mitosis-arresting agents, NPM is phosphorylated and its chromatographic property is altered, suggesting changes in function during mitosis. To determine the functional relevance of NPM, its expression in tumor cells was reduced by siRNA. Cells with reduced NPM were treated with Taxol followed by microarray profiling accompanied by gene/protein pathway analyses. These studies demonstrate several expected and unexpected consequences of NPM depletion. The predominant downstream effectors of NPM are genes involved in cell proliferation, cancer, and the cell cycle. In congruence with its role in cancer, NPM is over-expressed in primary malignant lung cancer tissues. We also demonstrate a role for NPM in the expression of genes encoding SET (TAF1beta) and the histone methylase SET8. Additionally, we show that NPM is required for a previously unobserved G2/M upregulation of TAF1A, which encodes the rDNA transcription factor TAF(I)48. These results demonstrate multi-faceted functions of NPM that can affect cancer cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Nuclear Proteins/metabolism , Paclitaxel/pharmacology , Base Sequence , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin/drug effects , Chromatin/genetics , DNA, Neoplasm/genetics , DNA, Ribosomal/genetics , Gene Expression/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Mitosis/drug effects , Mitosis/genetics , Multiprotein Complexes , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Nucleophosmin , Phosphorylation , RNA, Small Interfering/genetics , Transcription, Genetic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL