Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Immunol ; 209(5): 907-915, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35922063

ABSTRACT

The PD-L1/2-PD-1 immune checkpoint is essential for the proper induction of peripheral tolerance and limits autoimmunity, whereas tumor cells exploit their expression to promote immune evasion. Many different cell types express PD-L1/2, either constitutively or upon stimulation, but the factors driving this expression are often poorly defined. In this study, using genome-wide CRISPR activation screening, we identified three factors that upregulate PD-L1 expression: GATA2, MBD6, and transcription cofactor vestigial-like protein 3 (VGLL3). VGLL3 acts as a transcriptional regulator, and its expression induced PD-L1 in many different cell types. Conversely, loss of VGLL3 impaired IFN-γ-induced PD-L1/2 expression in human keratinocytes. Mechanistically, by performing a second screen to identify proteins acting in concert with VGLL3, we found that VGLL3 forms a complex with TEAD1 and RUNX1/3 to drive expression of PD-L1/2. Collectively, our work identified a new transcriptional complex controlling PD-L1/2 expression and suggests that VGLL3, in addition to its known role in the expression of proinflammatory genes, can balance inflammation by upregulating the anti-inflammatory factors PD-L1 and PD-L2.


Subject(s)
B7-H1 Antigen , Programmed Cell Death 1 Receptor , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Core Binding Factor Alpha 2 Subunit/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Immune Evasion , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Programmed Cell Death 1 Receptor/genetics , TEA Domain Transcription Factors , Transcription Factors/genetics
2.
N Engl J Med ; 382(3): 256-265, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31940699

ABSTRACT

Deficiency of ubiquitin-specific peptidase 18 (USP18) is a severe type I interferonopathy. USP18 down-regulates type I interferon signaling by blocking the access of Janus-associated kinase 1 (JAK1) to the type I interferon receptor. The absence of USP18 results in unmitigated interferon-mediated inflammation and is lethal during the perinatal period. We describe a neonate who presented with hydrocephalus, necrotizing cellulitis, systemic inflammation, and respiratory failure. Exome sequencing identified a homozygous mutation at an essential splice site on USP18. The encoded protein was expressed but devoid of negative regulatory ability. Treatment with ruxolitinib was followed by a prompt and sustained recovery. (Funded by King Saud University and others.).


Subject(s)
Hereditary Autoinflammatory Diseases/drug therapy , Interferons/metabolism , Interleukins/metabolism , Janus Kinase 1/antagonists & inhibitors , Janus Kinase Inhibitors/therapeutic use , Loss of Function Mutation , Pyrazoles/therapeutic use , Ubiquitin Thiolesterase/deficiency , Homozygote , Humans , Hydrocephalus/genetics , Infant, Newborn , Male , Nitriles , Pyrimidines , Receptors, Interferon/metabolism , Remission Induction , Shock, Septic/genetics , Signal Transduction/genetics , Ubiquitin Thiolesterase/genetics , Exome Sequencing
3.
J Infect Dis ; 221(6): 890-894, 2020 03 02.
Article in English | MEDLINE | ID: mdl-31637432

ABSTRACT

ISG15-deficient humans exhibit permanent, low-level expression of antiviral effectors that safely protect them from various viruses. Because the murine ISG15 axis functions differently, we identified animal models that recapitulate the human condition for the development of ISG15-targeting broad-spectrum antivirals. Canine, porcine, and rhesus macaque ISG15, such as human ISG15, stabilize USP18, a potent inhibitor of type I interferon (IFN)-I. Type I Interferon-primed ISG15-knockout porcine and rhesus cells demonstrate enhanced ISG expression and protection against vesicular stomatitis Indiana virus infection compared with wild type. Collectively, we unveil the interspecies diversity of the ability of ISG15/USP18 axis to control IFN-I signaling and reveal the therapeutic potential of ISG15-deficient porcine and rhesus models.


Subject(s)
Antiviral Agents/pharmacology , Animals , Cells, Cultured , Cytokines/metabolism , Gene Deletion , Gene Expression Regulation, Enzymologic/drug effects , Humans , Macaca mulatta , Phylogeny , Species Specificity , Swine , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Ubiquitins/metabolism
4.
Nat Commun ; 15(1): 5179, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898037

ABSTRACT

Viral genetic diversity presents significant challenges in developing antivirals with broad-spectrum activity and high barriers to resistance. Here we report development of proteolysis targeting chimeras (PROTACs) targeting the dengue virus envelope (E) protein through coupling of known E fusion inhibitors to ligands of the CRL4CRBN E3 ubiquitin ligase. The resulting small molecules block viral entry through inhibition of E-mediated membrane fusion and interfere with viral particle production by depleting intracellular E in infected Huh 7.5 cells. This activity is retained in the presence of point mutations previously shown to confer partial resistance to the parental inhibitors due to decreased inhibitor-binding. The E PROTACs also exhibit broadened spectrum of activity compared to the parental E inhibitors against a panel of mosquito-borne flaviviruses. These findings encourage further exploration of targeted protein degradation as a differentiated and potentially advantageous modality for development of broad-spectrum direct-acting antivirals.


Subject(s)
Antiviral Agents , Dengue Virus , Flavivirus , Proteolysis , Virus Internalization , Humans , Proteolysis/drug effects , Animals , Antiviral Agents/pharmacology , Flavivirus/drug effects , Flavivirus/genetics , Flavivirus/metabolism , Virus Internalization/drug effects , Dengue Virus/drug effects , Dengue Virus/physiology , Dengue Virus/genetics , Culicidae/virology , Ubiquitin-Protein Ligases/metabolism , Viral Envelope Proteins/metabolism , Cell Line
5.
Nat Commun ; 15(1): 6726, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39112502

ABSTRACT

Arteriviruses infect a variety of mammalian hosts, but the receptors used by these viruses to enter cells are poorly understood. We identified the neonatal Fc receptor (FcRn) as an important pro-viral host factor via comparative genome-wide CRISPR-knockout screens with multiple arteriviruses. Using a panel of cell lines and divergent arteriviruses, we demonstrate that FcRn is required for the entry step of arterivirus infection and serves as a molecular barrier to arterivirus cross-species infection. We also show that FcRn synergizes with another known arterivirus entry factor, CD163, to mediate arterivirus entry. Overexpression of FcRn and CD163 sensitizes non-permissive cells to infection and enables the culture of fastidious arteriviruses. Treatment of multiple cell lines with a pre-clinical anti-FcRn monoclonal antibody blocked infection and rescued cells from arterivirus-induced death. Altogether, this study identifies FcRn as a novel pan-arterivirus receptor, with implications for arterivirus emergence, cross-species infection, and host-directed pan-arterivirus countermeasure development.


Subject(s)
Histocompatibility Antigens Class I , Receptors, Fc , Receptors, Virus , Receptors, Fc/metabolism , Receptors, Fc/genetics , Humans , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/genetics , Animals , Receptors, Virus/metabolism , Receptors, Virus/genetics , Cell Line , Virus Internalization , Antigens, CD/metabolism , Antigens, CD/genetics , Receptors, Cell Surface/metabolism , Receptors, Cell Surface/genetics , HEK293 Cells
6.
J Mol Med (Berl) ; 100(9): 1253-1265, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35538149

ABSTRACT

Many autoimmune diseases exhibit a strikingly increased prevalence in females, with primary Sjögren's syndrome (pSS) being the most female-predominant example. However, the molecular basis underlying the female-bias in pSS remains elusive. To address this knowledge gap, we performed genome-wide, allele-specific profiling of minor salivary gland-derived mesenchymal stromal cells (MSCs) from pSS patients and control subjects, and detected major differences in the regulation of X-linked genes. In control female MSCs, X-linked genes were expressed from both paternal and maternal X chromosomes with a median paternal ratio of ~ 0.5. However, in pSS female MSCs, X-linked genes exhibited preferential expression from one of the two X chromosomes. Concomitantly, pSS MSCs showed decrease in XIST levels and reorganization of H3K27me3+ foci in the nucleus. Moreover, the HLA-locus-expressed miRNA miR6891-5p was decreased in pSS MSCs. miR6891-5p inhibition in control MSCs caused XIST dysregulation, ectopic silencing, and allelic skewing. Allelic skewing was accompanied by the mislocation of protein products encoded by the skewed genes, which was recapitulated by XIST and miR6891-5p disruption in control MSCs. Our data reveal X skewing as a molecular hallmark of pSS and highlight the importance of restoring X-chromosomal allelic balance for pSS treatment. KEY MESSAGES: X-linked genes exhibit skewing in primary Sjögren's syndrome (pSS). X skewing in pSS associates with alterations in H3K27me3 deposition. pSS MSCs show decreased levels of miR6891-5p, a HLA-expressed miRNA. miR6891-5p inhibition causes H3K27me3 dysregulation and allelic skewing.


Subject(s)
Genes, X-Linked , MicroRNAs , Sjogren's Syndrome , Female , Histones/genetics , Humans , MicroRNAs/genetics , Sjogren's Syndrome/genetics
7.
J Exp Med ; 217(5)2020 05 04.
Article in English | MEDLINE | ID: mdl-32092142

ABSTRACT

Type I interferonopathies are monogenic disorders characterized by enhanced type I interferon (IFN-I) cytokine activity. Inherited USP18 and ISG15 deficiencies underlie type I interferonopathies by preventing the regulation of late responses to IFN-I. Specifically, USP18, being stabilized by ISG15, sterically hinders JAK1 from binding to the IFNAR2 subunit of the IFN-I receptor. We report an infant who died of autoinflammation due to a homozygous missense mutation (R148Q) in STAT2. The variant is a gain of function (GOF) for induction of the late, but not early, response to IFN-I. Surprisingly, the mutation does not enhance the intrinsic activity of the STAT2-containing transcriptional complex responsible for IFN-I-stimulated gene induction. Rather, the STAT2 R148Q variant is a GOF because it fails to appropriately traffic USP18 to IFNAR2, thereby preventing USP18 from negatively regulating responses to IFN-I. Homozygosity for STAT2 R148Q represents a novel molecular and clinical phenocopy of inherited USP18 deficiency, which, together with inherited ISG15 deficiency, defines a group of type I interferonopathies characterized by an impaired regulation of late cellular responses to IFN-I.


Subject(s)
Gain of Function Mutation/genetics , Interferon Type I/metabolism , STAT2 Transcription Factor/genetics , Ubiquitin Thiolesterase/deficiency , Amino Acid Sequence , Base Sequence , Cell Line , Female , Gene Expression Regulation , Homozygote , Humans , Infant, Newborn , Male , Pedigree , Phenotype , Protein Domains , STAT2 Transcription Factor/chemistry , Ubiquitin Thiolesterase/genetics , Exome Sequencing
8.
Cell Rep ; 31(6): 107633, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32402279

ABSTRACT

Most monogenic disorders have a primary clinical presentation. Inherited ISG15 deficiency, however, has manifested with two distinct presentations to date: susceptibility to mycobacterial disease and intracranial calcifications from hypomorphic interferon-II (IFN-II) production and excessive IFN-I response, respectively. Accordingly, these patients were managed for their infectious and neurologic complications. Herein, we describe five new patients with six novel ISG15 mutations presenting with skin lesions who were managed for dermatologic disease. Cellularly, we denote striking specificity to the IFN-I response, which was previously assumed to be universal. In peripheral blood, myeloid cells display the most robust IFN-I signatures. In the affected skin, IFN-I signaling is observed in the keratinocytes of the epidermis, endothelia, and the monocytes and macrophages of the dermis. These findings define the specific cells causing circulating and dermatologic inflammation and expand the clinical spectrum of ISG15 deficiency to dermatologic presentations as a third phenotype co-dominant to the infectious and neurologic manifestations.


Subject(s)
Cytokines/deficiency , Interferon Type I/immunology , Skin/pathology , Ubiquitins/deficiency , Alleles , Case-Control Studies , Child , Child, Preschool , Cytokines/genetics , Cytokines/immunology , Dermatitis/genetics , Dermatitis/immunology , Dermatitis/pathology , Female , HEK293 Cells , Humans , Infant , Male , Mutation , Myeloid Cells/immunology , Myeloid Cells/pathology , Necrosis , Pedigree , Ubiquitins/genetics , Ubiquitins/immunology
9.
Biomaterials ; 108: 35-43, 2016 11.
Article in English | MEDLINE | ID: mdl-27619238

ABSTRACT

Nanoparticles are regularly used as contrast agents in bioimaging. Unlike other agents such as composite materials, nanoparticles can also be used for treating as well as imaging disease. Here we synthesized lanthanide functionalized gold nanoparticles that can be used for both imaging and therapy in vivo. That is a multifunctional nanoplatform was developed based on a simple and versatile method, by incorporating 10-nm gold nanoparticles and lanthanide ions (Gd(3+) and Yb(3+)), denoted as LnAu nanoparticles hereby. The LnAu nanoparticles were then surface-modified using a PEGylated amphiphilic polymer (C18MH-mPEG), and the resulting PEG modified LnAu nanoparticles (PEG-LnAu) display good monodispersion in water and good solubility in biological media. Due to the low toxicity in vitro and in vivo (as determined by a cell viability assay and histological and serum biochemistry analysis), the PEG-LnAu nanoparticles can be successfully applied to in vivo magnetic resonance imaging (MRI), in vivo computed tomography (CT) imaging and photothermal therapy (PTT) for tumor-bearing mice. Therefore, the present work developed an easy yet powerful strategy to combine lanthanide ions and gold nanoparticles to a unified nanoplatform for integrating bioimaging and therapy.


Subject(s)
Magnetic Resonance Imaging/methods , Metal Nanoparticles/therapeutic use , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/therapy , Phototherapy/methods , Theranostic Nanomedicine/methods , Tomography, X-Ray Computed/methods , Animals , Cell Line, Tumor , Coated Materials, Biocompatible/chemistry , Contrast Media/chemical synthesis , Gold/chemistry , Gold/therapeutic use , HeLa Cells , Humans , Lanthanoid Series Elements/chemistry , Lanthanoid Series Elements/therapeutic use , Metal Nanoparticles/chemistry , Metal Nanoparticles/ultrastructure , Mice , Polyethylene Glycols/chemistry , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL