Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Kidney Int ; 98(4): 897-905, 2020 10.
Article in English | MEDLINE | ID: mdl-32763116

ABSTRACT

Allogeneic transplants elicit dynamic T cell responses that are modulated by positive and negative co-stimulatory receptors. Understanding mechanisms that intrinsically modulate the immune responses to transplants is vital to develop rational treatment for rejection. Here, we have investigated the impact of programed cell death-1 (PD-1) protein, a negative co-stimulatory receptor, on the rejection of MHC incompatible kidney transplants in mice. T cells were found to rapidly infiltrate the kidneys of A/J mice transplanted to C57BL/6 mice, which peaked at six days and decline by day 14. The T cells primarily encircled tubules with limited infiltration of the tubular epithelium. Lipocalin 2 (LCN2), a marker of tubular injury, also peaked in the urine at day six and then declined. Notably, flow cytometry demonstrated that most of the T cells expressed PD-1 (over 90% of CD8 and about 75% of CD4 cells) at day six. Administration of blocking antibody to PD-L1, the ligand for PD-1, before day six increased T cell infiltrates and urinary LCN2, causing terminal acute rejection. In contrast, blocking PD-1/PD-L1 interactions after day six caused only a transient increase in urinary LCN2. Depleting CD4 and CD8 T cells virtually eliminated LCN2 in the urine in support of T cells injuring tubules. Thus, our data indicate that PD-1/PD-L1 interactions are not just related to chronic antigenic stimulation of T cells but are critical for the regulation of acute T cell responses to renal transplants.


Subject(s)
Kidney Transplantation , Programmed Cell Death 1 Receptor , Animals , B7-H1 Antigen , CD8-Positive T-Lymphocytes , Cell Death , Ligands , Mice , Mice, Inbred C57BL
2.
Am J Transplant ; 20(10): 2686-2702, 2020 10.
Article in English | MEDLINE | ID: mdl-32320528

ABSTRACT

HLA donor-specific antibodies (DSAs) binding to vascular endothelial cells of the allograft trigger inflammation, vessel injury, and antibody-mediated rejection (AMR). Accumulation of intragraft-recipient macrophages is a histological characteristic of AMR, which portends worse outcome. HLA class I (HLA I) DSAs enhance monocyte recruitment by activating endothelial cells and engaging FcγRs, but the DSA-activated donor endothelial influence on macrophage differentiation is unknown. In this study, we explored the consequence of DSA-activated endothelium on infiltrating monocyte differentiation. Here we show that cardiac allografts from murine recipients treated with MHC I DSA upregulated genes related to monocyte transmigration and Fc receptor stimulation. Human monocytes co-cultured with HLA I IgG-stimulated primary human endothelium promoted monocyte differentiation into CD68+ CD206+ CD163+ macrophages (M(HLA I IgG)), whereas HLA I F(ab')2 stimulated endothelium solely induced higher CD206 (M(HLA I F(ab')2 )). Both macrophage subtypes exhibited significant changes in discrete cytokines/chemokines and unique gene expression profiles. Cross-comparison of gene transcripts between murine DSA-treated cardiac allografts and human co-cultured macrophages identified overlapping genes. These findings uncover the role of HLA I DSA-activated endothelium in monocyte differentiation, and point to a novel, remodeling phenotype of infiltrating macrophages that may contribute to vascular injury.


Subject(s)
Endothelial Cells , Graft Rejection , Allografts , Animals , Graft Rejection/etiology , HLA Antigens , Humans , Inflammation/etiology , Isoantibodies , Macrophages , Mice , Phenotype , Tissue Donors
3.
Innate Immun ; 23(2): 147-154, 2017 02.
Article in English | MEDLINE | ID: mdl-27913791

ABSTRACT

BMAL1 (brain and muscle ARNT-like protein 1, also known as MOP3 or ARNT3) belongs to the family of the basic helix-loop-helix (bHLH)-PAS domain-containing transcription factors, and is a key component of the molecular oscillator that generates circadian rhythms. Here, we report that BMAL1-deficient cells are significantly more susceptible to infection by two major respiratory viruses of the Paramyxoviridae family, namely RSV and PIV3. Embryonic fibroblasts from Bmal1-/- mice produced nearly 10-fold more progeny virus than their wild type controls. These results were supported by animal studies whereby pulmonary infection of RSV produced a more severe disease and morbidity in Bmal1-/-mice. These results show that BMAL1 can regulate cellular innate immunity against specific RNA viruses.


Subject(s)
ARNTL Transcription Factors/metabolism , Fibroblasts/immunology , Lung/pathology , Parainfluenza Virus 3, Human/immunology , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Viruses/immunology , Respirovirus Infections/immunology , ARNTL Transcription Factors/genetics , Animals , Cell Line , Circadian Clocks/genetics , Fibroblasts/virology , Humans , Immunity, Innate/genetics , Mice , Mice, Knockout , RNA, Small Interfering/genetics
4.
Sci Rep ; 6: 38139, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27905537

ABSTRACT

Pneumonia Virus of Mice (PVM) is the only virus that shares the Pneumovirus genus of the Paramyxoviridae family with Respiratory Syncytial Virus (RSV). A deadly mouse pathogen, PVM has the potential to serve as a robust animal model of RSV infection, since human RSV does not fully replicate the human pathology in mice. Like RSV, PVM also encodes two nonstructural proteins that have been implicated to suppress the IFN pathway, but surprisingly, they exhibit no sequence similarity with their RSV equivalents. The molecular mechanism of PVM NS function, therefore, remains unknown. Here, we show that recombinant PVM NS proteins degrade the mouse counterparts of the IFN pathway components. Proteasomal degradation appears to be mediated by ubiquitination promoted by PVM NS proteins. Interestingly, NS proteins of PVM lowered the levels of several ISG (IFN-stimulated gene) proteins as well. These results provide a molecular foundation for the mechanisms by which PVM efficiently subverts the IFN response of the murine cell. They also reveal that in spite of their high sequence dissimilarity, the two pneumoviral NS proteins are functionally and mechanistically similar.


Subject(s)
Interferons/metabolism , Murine pneumonia virus/metabolism , Viral Nonstructural Proteins/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Interferons/genetics , Metabolic Networks and Pathways/immunology , Mice , Murine pneumonia virus/genetics , Murine pneumonia virus/pathogenicity , Pneumovirus Infections/genetics , Pneumovirus Infections/immunology , Pneumovirus Infections/virology , Proteolysis , Respiratory Syncytial Virus Infections/etiology , Respiratory Syncytial Viruses/genetics , Respiratory Syncytial Viruses/metabolism , Respiratory Syncytial Viruses/pathogenicity , Viral Nonstructural Proteins/genetics
5.
J Virol ; 89(19): 10115-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26178980

ABSTRACT

2'-5'-Oligoadenylate synthetase-like protein (OASL) is an interferon-inducible antiviral protein. Here we describe differential inhibitory activities of human OASL and the two mouse OASL homologs against respiratory syncytial virus (RSV) replication. Interestingly, nonstructural protein 1 (NS1) of RSV promoted proteasome-dependent degradation of specific OASL isoforms. We conclude that OASL acts as a cellular antiviral protein and that RSV NS1 suppresses this function to evade cellular innate immunity and allow virus growth.


Subject(s)
2',5'-Oligoadenylate Synthetase/physiology , Respiratory Syncytial Viruses/physiology , Viral Nonstructural Proteins/physiology , Virus Replication/physiology , 2',5'-Oligoadenylate Synthetase/immunology , Animals , HEK293 Cells , Host-Pathogen Interactions , Humans , Immune Evasion , Immunity, Cellular , Mice , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/genetics , Respiratory Syncytial Viruses/immunology , Viral Nonstructural Proteins/genetics , Virus Replication/immunology
6.
PLoS Pathog ; 11(3): e1004779, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25811886

ABSTRACT

Innate immunity is the first line of defense against microbial insult. The transcription factor, IRF3, is needed by mammalian cells to mount innate immune responses against many microbes, especially viruses. IRF3 remains inactive in the cytoplasm of uninfected cells; upon virus infection, it gets phosphorylated and then translocates to the nucleus, where it binds to the promoters of antiviral genes and induces their expression. Such genes include type I interferons (IFNs) as well as Interferon Stimulated Genes (ISGs). IRF3-/- cells support enhanced replication of many viruses and therefore, the corresponding mice are highly susceptible to viral pathogenesis. Here, we provide evidence for an unexpected pro-microbial role of IRF3: the replication of the protozoan parasite, Toxoplasma gondii, was significantly impaired in IRF3-/- cells. In exploring whether the transcriptional activity of IRF3 was important for its pro-parasitic function, we found that ISGs induced by parasite-activated IRF3 were indeed essential, whereas type I interferons were not important. To delineate the signaling pathway that activates IRF3 in response to parasite infection, we used genetically modified human and mouse cells. The pro-parasitic signaling pathway, which we termed PISA (Parasite-IRF3 Signaling Activation), activated IRF3 without any involvement of the Toll-like receptor or RIG-I-like receptor pathways, thereby ruling out a role of parasite-derived RNA species in activating PISA. Instead, PISA needed the presence of cGAS, STING, TBK1 and IRF3, indicating the necessity of DNA-triggered signaling. To evaluate the physiological significance of our in vitro findings, IRF3-/- mice were challenged with parasite infection and their morbidity and mortality were measured. Unlike WT mice, the IRF3-/- mice did not support replication of the parasite and were resistant to pathogenesis caused by it. Our results revealed a new paradigm in which the antiviral host factor, IRF3, plays a cell-intrinsic pro-parasitic role.


Subject(s)
Interferon Regulatory Factor-3/immunology , Signal Transduction/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , Animals , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/immunology , Gene Knockdown Techniques , HEK293 Cells , Humans , Interferon Regulatory Factor-3/genetics , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Receptors, Immunologic , Signal Transduction/genetics , Toxoplasmosis/genetics
7.
Immunity ; 40(6): 936-48, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24931123

ABSTRACT

Virus infection is sensed in the cytoplasm by retinoic acid-inducible gene I (RIG-I, also known as DDX58), which requires RNA and polyubiquitin binding to induce type I interferon (IFN) and activate cellular innate immunity. We show that the human IFN-inducible oligoadenylate synthetases-like (OASL) protein has antiviral activity and mediates RIG-I activation by mimicking polyubiquitin. Loss of OASL expression reduced RIG-I signaling and enhanced virus replication in human cells. Conversely, OASL expression suppressed replication of a number of viruses in a RIG-I-dependent manner and enhanced RIG-I-mediated IFN induction. OASL interacted and colocalized with RIG-I, and through its C-terminal ubiquitin-like domain specifically enhanced RIG-I signaling. Bone-marrow-derived macrophages from mice deficient for Oasl2 showed that among the two mouse orthologs of human OASL, Oasl2 is functionally similar to human OASL. Our findings show a mechanism by which human OASL contributes to host antiviral responses by enhancing RIG-I activation.


Subject(s)
2',5'-Oligoadenylate Synthetase/immunology , DEAD-box RNA Helicases/immunology , DNA Virus Infections/immunology , Interferon Type I/immunology , RNA Virus Infections/immunology , 2',5'-Oligoadenylate Synthetase/genetics , Animals , DEAD Box Protein 58 , HCT116 Cells , HEK293 Cells , Humans , Immunity, Innate , Interferon Regulatory Factor-7/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Polyubiquitin , Protein Binding/immunology , RNA Interference , RNA, Small Interfering , Receptors, Immunologic , Signal Transduction/immunology , Virus Replication/immunology
8.
J Biol Chem ; 289(22): 15166-78, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24700465

ABSTRACT

Centriole duplication is the process by which two new daughter centrioles are generated from the proximal end of preexisting mother centrioles. Accurate centriole duplication is important for many cellular and physiological events, including cell division and ciliogenesis. Centrosomal protein 4.1-associated protein (CPAP), centrosomal protein of 152 kDa (CEP152), and centrobin are known to be essential for centriole duplication. However, the precise mechanism by which they contribute to centriole duplication is not known. In this study, we show that centrobin interacts with CEP152 and CPAP, and the centrobin-CPAP interaction is critical for centriole duplication. Although depletion of centrobin from cells did not have an effect on the centriolar levels of CEP152, it caused the disappearance of CPAP from both the preexisting and newly formed centrioles. Moreover, exogenous expression of the CPAP-binding fragment of centrobin also caused the disappearance of CPAP from both the preexisting and newly synthesized centrioles, possibly in a dominant negative manner, thereby inhibiting centriole duplication and the PLK4 overexpression-mediated centrosome amplification. Interestingly, exogenous overexpression of CPAP in the centrobin-depleted cells did not restore CPAP localization to the centrioles. However, restoration of centrobin expression in the centrobin-depleted cells led to the reappearance of centriolar CPAP. Hence, we conclude that centrobin-CPAP interaction is critical for the recruitment of CPAP to procentrioles to promote the elongation of daughter centrioles and for the persistence of CPAP on preexisting mother centrioles. Our study indicates that regulation of CPAP levels on the centrioles by centrobin is critical for preserving the normal size, shape, and number of centrioles in the cell.


Subject(s)
Cell Cycle Proteins/metabolism , Centrioles/metabolism , Microtubule-Associated Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Division/physiology , Centrioles/genetics , Cloning, Molecular , Green Fluorescent Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Microtubule-Associated Proteins/genetics , Osteosarcoma , Phosphoproteins/metabolism , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , RNA, Small Interfering/genetics
9.
Cell Res ; 23(8): 1025-42, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23877405

ABSTRACT

The balance between the innate immunity of the host and the ability of a pathogen to evade it strongly influences pathogenesis and virulence. The two nonstructural (NS) proteins, NS1 and NS2, of respiratory syncytial virus (RSV) are critically required for RSV virulence. Together, they strongly suppress the type I interferon (IFN)-mediated innate immunity of the host cells by degrading or inhibiting multiple cellular factors required for either IFN induction or response pathways, including RIG-I, IRF3, IRF7, TBK1 and STAT2. Here, we provide evidence for the existence of a large and heterogeneous degradative complex assembled by the NS proteins, which we named "NS-degradasome" (NSD). The NSD is roughly ∼300-750 kD in size, and its degradative activity was enhanced by the addition of purified mitochondria in vitro. Inside the cell, the majority of the NS proteins and the substrates of the NSD translocated to the mitochondria upon RSV infection. Genetic and pharmacological evidence shows that optimal suppression of innate immunity requires mitochondrial MAVS and mitochondrial motility. Together, we propose a novel paradigm in which the mitochondria, known to be important for the innate immune activation of the host, are also important for viral suppression of the innate immunity.


Subject(s)
Immunity, Innate , Mitochondria/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adenosine Triphosphate/metabolism , Animals , Cell Line , GTP Phosphohydrolases/antagonists & inhibitors , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Hepacivirus/metabolism , Humans , Interferon Type I/metabolism , Mice , Mitochondria/drug effects , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Nocodazole/pharmacology , RNA Interference , RNA, Small Interfering/metabolism , Respiratory Syncytial Viruses/metabolism , STAT2 Transcription Factor/genetics , STAT2 Transcription Factor/metabolism , Substrate Specificity , Viral Nonstructural Proteins/metabolism
10.
J Biol Chem ; 278(33): 30569-77, 2003 Aug 15.
Article in English | MEDLINE | ID: mdl-12773528

ABSTRACT

To explore the interdomain co-operativity during human plasminogen (HPG) activation by streptokinase (SK), we expressed the cDNAs corresponding to each SK domain individually (alpha, beta, and gamma), and also their two-domain combinations, viz. alphabeta and betagamma in Escherichia coli. After purification, alpha and beta showed activator activities of approximately 0.4 and 0.05%, respectively, as compared with that of native SK, measured in the presence of human plasmin, but the bi-domain constructs alphabeta and betagamma showed much higher co-factor activities (3.5 and 0.7% of native SK, respectively). Resonant Mirror-based binding studies showed that the single-domain constructs had significantly lower affinities for "partner" HPG, whereas the affinities of the two-domain constructs were remarkably native-like with regards to both binary-mode as well as ternary mode ("substrate") binding with HPG, suggesting that the vast difference in co-factor activity between the two- and three-domain structures did not arise merely from affinity differences between activator species and HPG. Remarkably, when the co-factor activities of the various constructs were measured with microplasminogen, the nearly 50-fold difference in the co-factor activity between the two- and three-domain SK constructs observed with full-length HPG as substrate was found to be dramatically attenuated, with all three types of constructs now exhibiting a low activity of approximately 1-2% compared to that of SK.HPN and HPG. Thus, the docking of substrate through the catalytic domain at the active site of SK-plasmin(ogen) is capable of engendering, at best, only a minimal level of co-factor activity in SK.HPN. Therefore, apart from conferring additional substrate affinity through kringle-mediated interactions, reported earlier (Dhar et al., 2002; J. Biol. Chem. 277, 13257), selective interactions between all three domains of SK and the kringle domains of substrate vastly accelerate the plasminogen activation reaction to near native levels.


Subject(s)
Fibrinolysin/metabolism , Plasminogen Activators/metabolism , Plasminogen/metabolism , Streptokinase/metabolism , Catalytic Domain , DNA, Complementary , Escherichia coli , Humans , Kringles , Plasminogen Activators/chemistry , Plasminogen Activators/genetics , Protein Structure, Tertiary , Streptokinase/chemistry , Streptokinase/genetics , Substrate Specificity
11.
J Biol Chem ; 277(15): 13257-67, 2002 Apr 12.
Article in English | MEDLINE | ID: mdl-11821385

ABSTRACT

The selective deletion of a discrete surface-exposed epitope (residues 254-262; 250-loop) in the beta domain of streptokinase (SK) significantly decreased the rates of substrate human plasminogen (HPG) activation by the mutant (SK(del254-262)). A kinetic analysis of SK(del254-262) revealed that its low HPG activator activity arose from a 5-6-fold increase in K(m) for HPG as substrate, with little alteration in k(cat) rates. This increase in the K(m) for the macromolecular substrate was proportional to a similar decrease in the binding affinity for substrate HPG as observed in a new resonant mirror-based assay for the real-time kinetic analysis of the docking of substrate HPG onto preformed binary complex. In contrast, studies on the interaction of the two proteins with microplasminogen showed no difference between the rates of activation of microplasminogen under conditions where HPG was activated differentially by nSK and SK(del254-262). The involvement of kringles was further indicated by a hypersusceptibility of the SK(del254-262).plasmin activator complex to epsilon-aminocaproic acid-mediated inhibition of substrate HPG activation in comparison with that of the nSK.plasmin activator complex. Further, ternary binding experiments on the resonant mirror showed that the binding affinity of kringles 1-5 of HPG to SK(del254-262).HPG was reduced by about 3-fold in comparison with that of nSK.HPG . Overall, these observations identify the 250 loop in the beta domain of SK as an important structural determinant of the inordinately stringent substrate specificity of the SK.HPG activator complex and demonstrate that it promotes the binding of substrate HPG to the activator via the kringle(s) during the HPG activation process.


Subject(s)
Kringles , Streptokinase/metabolism , Base Sequence , Biosensing Techniques , DNA Primers , Humans , Hydrolysis , Kinetics , Models, Molecular , Plasminogen/metabolism , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Deletion , Streptokinase/chemistry , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...