scholarly journals Nicastrin haploinsufficiency alters expression of type-I interferon-stimulated genes in two immortalized human cell lines

2018 ◽  
Author(s):  
Li Cao ◽  
David J. Morales-Heil ◽  
Elisha D. O. Roberson

A.AbstractBackgroundHidradenitis suppurativa (HS) is a chronic skin disease. The symptoms can be severe, and include intensely painful nodules and abscesses in apocrine-gland rich inverse skin, such as the buttocks, under the arms, and the groin. Autosomal dominant forms of HS exist, but are rare. Some of these kindred have heterozygous loss-of-function rare variants in the γ-secretase complex component nicastrin (NCSTN).ObjectivesWe wanted to know what effect NCSTN haploinsufficiency has on human keratinocytes to assess potential mechanisms for lesion development.MethodsWe knocked down nicastrin using an shRNA construct in both a keratinocyte cell line (HEK001) and an embryonic kidney cell line (HEK293). We assessed differential gene expression using RNA microarray. We also generated a NCSTN heterozygous deletion in the HEK293 line using CRISPR/Cas9 genome-editing and assessed NFKB activity in this line using a luciferase reporter.ResultsThe keratinocyte NCSTN knockdown cell line demonstrated significantly increased expression of genes related to the type-I interferon response pathway when compared to controls. Both HEK001 and HEK293 knockdowns demonstrated evidence for altered growth. We observed a small, but significant increase in NFKB signaling in response to TNF treatment a HEK293 line genome-edited for reduced NCSTN.ConclusionsOur data suggest a role for increased keratinocyte inflammatory responsiveness in familial HS. Confirming this phenotype, and characterizing additional effects in different cell types, will require study beyond cell lines in primary cells and tissues.

Blood ◽  
2017 ◽  
Vol 130 (Suppl_1) ◽  
pp. 782-782
Author(s):  
Takuji Yamauchi ◽  
Takeshi Masuda ◽  
Matthew C. Canver ◽  
Michael Seiler ◽  
Mohammad Shboul ◽  
...  

Abstract Genome-wide knockout screening employing CRISPR-Cas9 genome-editing is a powerful tool for functional genomics. However, identifying actionable targets for cancer therapy has been challenging due, in part, to the complex genetic background of cell lines used for screening. To overcome this obstacle, we generated two acute myeloid leukemia (AML) lines from mouse leukemia models based on activity of the leukemia oncogenes CALM/AF10 or MLL/AF9. Both lines exhibited a normal karyotype and intact Tp53 activity. Using these lines, we performed genome-wide CRISPR-Cas9 screening, followed by a second screen in vivo . We then selected genes meeting the following criteria: 1) they encoded a protein with an available inhibitor or 2) their germline mutation loss-of-function phenotype was relatively moderate based on the literature or the human exome-sequencing database. We excluded genes with a well-defined function in leukemogenesis as well as those encoding components of basal cellular machineries. Among genes significantly depleted in our primary screen was the mRNA decapping enzyme scavenger (Dcps), which encodes a mRNA 5' cap binding protein implicated in mRNA decay. Read counts for each Dcps-targeted single-guide RNA (sgRNA) significantly decreased in vitro (AML cell lines) and in vivo (mouse AML model). A negative selection CRISPR-Cas9 mutagenesis scan of all Dcps coding exons revealed that the C-terminal Dcps domain, namely aa 230-240, plays a critical role in AML survival. RG3039, an orally active quinazoline derivative, is a DCPS inhibitor that was originally developed to treat spinal muscular atrophy (SMA) and has been judged safe in a phase I trial in healthy volunteers (Van Meerbeke JP et al. Hum Mol Genet. 2013). We validated RG3039 binding to DCPS protein in AML cells via a cellular thermal shift assay (CETSA). We then assessed anti-leukemia effects of RG3039 by treating human AML lines with RG3039 and generating growth curves. That analysis showed that RG3039 had dose-dependent anti-proliferative effects. RG3039 treatment induced cell cycle arrest and apoptosis, revealed by EdU incorporation assay and Annexin V stain, respectively. Since DCPS protein was predominantly nuclear in human primary AML cells, we hypothesized that DCPS primarily functions in that compartment rather than in the cytoplasmic mRNA 3' end decay pathway. To search for nuclear proteins potentially interacting with DCPS, we undertook immunoprecipitation with an anti-DCPS antibody of lysates of AML cells followed by mass spectrometry analysis. Among highly significant interactors, we identified components of pre-mRNA processing machineries including spliceosomes. To assess whether DCPS inhibition would impair pre-mRNA splicing, we treated AML cells with RG3039 and performed RNA-sequencing to determine potential transcriptome-wide splicing changes. Alternative 5' splice site selection was most frequently observed in RG3039-treated cells, and most aberrant splicing events involved the first exon. Bioinformatic prediction analysis revealed that approximately 40% of the aberrantly-spliced genes were NMD (nonsense-mediated mRNA decay)-sensitive. Gene Set Enrichment Analysis identified gene signatures representing a type-I interferon response, reminiscent of that observed in RNAi-treated cells, in RG3039-treated AML cells. We next explored effects of DCPS deficiency on normal hematopoiesis in humans. To do so, we examined peripheral blood counts of three children harboring germline homozygous loss-of-function mutations as well as heterozygous relatives in a family reported previously (Ng et al. Hum Mol Genet 2015). They exhibited normal blood counts, indicating that DCPS is dispensable for steady-state hematopoiesis in humans. Finally, to investigate potential anti-leukemia effects of RG3039 in vivo, we tested RG3039 efficacy using patient-derived xenograft (PDX) AML models established from three human AML lines. RG3039 exhibited anti-leukemic activity, as evidenced by the lower leukemia burden in PB and BM of RG3039-treated mice. They survived significantly longer than vehicle-treated mice, indicative of therapeutic efficacy of RG3039 monotherapy against AML in vivo. In summary, our findings shed a new light on a pre-mRNA metabolic pathway regulated by DCPS. They also identify DCPS as a novel target for AML therapy and suggest potential "repurposing" of RG3039 as an anti-leukemia drug. Disclosures Seiler: H3 Biomedicine, Inc.: Employment. Orkin: Epizyme Inc.: Consultancy; Bioverativ: Consultancy. Buonamici: H3 Biomedicine Inc.: Employment.


We have cloned a set of seven cDNAs corresponding to mRNAs induced by type I interferon in hum an cells, and are using these probes to characterize the response to interferon in detail. We have studied the kinetics of induction of each m RN A in three different hum an cell lines, the sensitivity of induction to cycloheximide, the effect of w ithdraw ing interferon and the response of a receptor-positive interferon-resistant cell line.


2020 ◽  
Vol 40 (5) ◽  
Author(s):  
Jianli Shi ◽  
Ping Feng ◽  
Tingting Gu

Abstract Background: Influenza A virus (IAV) has greatly affected public health in recent decades. Accumulating data indicated that host microRNAs (miRNAs) were related to IAV replication. The present study mainly focused on the effects of microRNA-21-3p (miR-21-3p) on H5N1 replication. Methods: The levels of miR-21-3p, virus structural factors (matrix 1 (M1), nucleoprotein (NP)), type I interferon (IFN) response markers (IFN-β, IFN-α), IFN-stimulated genes (protein kinase R (PKR), myxovirus resistance A (MxA), 2′-5′-oligoadenylate synthetase 2 (OAS)), and fibroblast growth factor 2 (FGF2) were measured by quantitative real-time polymerase chain reaction (qRT-PCR). The protein levels of M1, NP, and FGF2 were tested by Western blot assay. The virus titer was assessed by tissue culture infective dose 50% (TCID50) assay. The dual-luciferase reporter assay and ribonucleic acid (RNA) immunoprecipitation (RIP) assay were used to verify the interaction between miR-21-3p and FGF2. Results: MiR-21-3p was reduced in H5N1-infected patients and A549 cells. MiR-21-3p overexpression facilitated the levels of M1, NP, TCID50 value, and reduced the levels of IFN-β, IFN-α, PKR, MxA, and OAS in H5N1-infected A549 cells. FGF2 was verified as a direct target of miR-21-3p. The introduction of FGF2 counteracted miR-21-3p-mediated decrease in the levels of M1, NP, and TCID50 value, as well as reduction in the levels of IFN-β, IFN-α, PKR, MxA, and OAS in H5N1-infected A549 cells. Conclusion: MiR-21-3p down-regulated FGF2 expression to accelerate H5N1 replication and confine IFN response.


2021 ◽  
Author(s):  
Da-Yuan Chen ◽  
Nazimuddin Khan ◽  
Brianna J. Close ◽  
Raghuveera K. Goel ◽  
Benjamin Blum ◽  
...  

SARS-CoV-2 can infect multiple organs, including lung, intestine, kidney, heart, liver, and brain. The molecular details of how the virus navigates through diverse cellular environments and establishes replication are poorly defined. Here, we generated a panel of phenotypically diverse, SARS-CoV-2-infectable human cell lines representing different body organs and performed longitudinal survey of cellular proteins and pathways broadly affected by the virus. This revealed universal inhibition of interferon signaling across cell types following SARS-CoV-2 infection. We performed systematic analyses of the JAK-STAT pathway in a broad range of cellular systems, including immortalized cells and primary-like cardiomyocytes, and found that SARS-CoV-2 targeted the proximal pathway components, including Janus kinase 1 (JAK1), tyrosine kinase 2 (Tyk2), and the interferon receptor subunit 1 (IFNAR1), resulting in cellular desensitization to type I IFN. Detailed mechanistic investigation of IFNAR1 showed that the protein underwent ubiquitination upon SARS-CoV-2 infection. Furthermore, chemical Inhibition of JAK kinases enhanced infection of stem cell-derived cultures, indicating that the virus benefits from inhibiting the JAK-STAT pathway. These findings suggest that the suppression of interferon signaling is a mechanism widely used by the virus to evade antiviral innate immunity, and that targeting the viral mediators of immune evasion may help block virus replication in patients with COVID-19. IMPORTANCE SARS-CoV-2 can infect various organs in the human body, but the molecular interface between the virus and these organs remains unexplored. In this study, we generated a panel of highly infectable human cell lines originating from various body organs and employed these cells to identify cellular processes commonly or distinctly disrupted by SARS-CoV-2 in different cell types. One among the universally impaired processes was interferon signaling. Systematic analysis of this pathway in diverse culture systems showed that SARS-CoV-2 targets the proximal JAK-STAT pathway components, destabilizes the type I interferon receptor though ubiquitination, and consequently renders the infected cells resistant to type I interferon. These findings illuminate how SARS-CoV-2 can continue to propagate in different tissues even in the presence of a disseminated innate immune response.


2020 ◽  
Vol 117 (32) ◽  
pp. 19475-19486
Author(s):  
Carina Elsner ◽  
Aparna Ponnurangam ◽  
Julia Kazmierski ◽  
Thomas Zillinger ◽  
Jenny Jansen ◽  
...  

The DNA sensor cGAS catalyzes the production of the cyclic dinucleotide cGAMP, resulting in type I interferon responses. We addressed the functionality of cGAS-mediated DNA sensing in human and murine T cells. Activated primary CD4+T cells expressed cGAS and responded to plasmid DNA by upregulation of ISGs and release of bioactive interferon. In mouse T cells, cGAS KO ablated sensing of plasmid DNA, and TREX1 KO enabled cells to sense short immunostimulatory DNA. Expression ofIFIT1andMX2was downregulated and upregulated in cGAS KO and TREX1 KO T cell lines, respectively, compared to parental cells. Despite their intact cGAS sensing pathway, human CD4+T cells failed to mount a reverse transcriptase (RT) inhibitor-sensitive immune response following HIV-1 infection. In contrast, infection of human T cells with HSV-1 that is functionally deficient for the cGAS antagonist pUL41 (HSV-1ΔUL41N) resulted in a cGAS-dependent type I interferon response. In accordance with our results in primary CD4+T cells, plasmid challenge or HSV-1ΔUL41N inoculation of T cell lines provoked an entirely cGAS-dependent type I interferon response, including IRF3 phosphorylation and expression of ISGs. In contrast, no RT-dependent interferon response was detected following transduction of T cell lines with VSV-G-pseudotyped lentiviral or gammaretroviral particles. Together, T cells are capable to raise a cGAS-dependent cell-intrinsic response to both plasmid DNA challenge or inoculation with HSV-1ΔUL41N. However, HIV-1 infection does not appear to trigger cGAS-mediated sensing of viral DNA in T cells, possibly by revealing viral DNA of insufficient quantity, length, and/or accessibility to cGAS.


2007 ◽  
Vol 9 (2) ◽  
pp. 122-135 ◽  
Author(s):  
Outi Rautsi ◽  
Saara Lehmusvaara ◽  
Tuula Salonen ◽  
Katja Häkkinen ◽  
Maarit Sillanpää ◽  
...  

Author(s):  
Ruben S. A. Goedegebuure ◽  
Esther A. Kleibeuker ◽  
Francesca M. Buffa ◽  
Kitty C. M. Castricum ◽  
Syed Haider ◽  
...  

Abstract Background Improvement of radiotherapy efficacy requires better insight in the dynamic responses that occur during irradiation. Here, we aimed to identify the molecular responses that are triggered during clinically applied fractionated irradiation. Methods Gene expression analysis was performed by RNAseq or microarray analysis of cancer cells or xenograft tumors, respectively, subjected to 3–5 weeks of 5 × 2 Gy/week. Validation of altered gene expression was performed by qPCR and/or ELISA in multiple cancer cell lines as well as in pre- and on-treatment biopsies from esophageal cancer patients (NCT02072720). Targeted protein inhibition and CRISPR/Cas-induced gene knockout was used to analyze the role of type I interferons and cGAS/STING signaling pathway in the molecular and cellular response to fractionated irradiation. Results Gene expression analysis identified type I interferon signaling as the most significantly enriched biological process induced during fractionated irradiation. The commonality of this response was confirmed in all irradiated cell lines, the xenograft tumors and in biopsies from esophageal cancer patients. Time-course analyses demonstrated a peak in interferon-stimulated gene (ISG) expression within 2–3 weeks of treatment. The response was accompanied by a variable induction of predominantly interferon-beta and/or -lambda, but blocking these interferons did not affect ISG expression induction. The same was true for targeted inhibition of the upstream regulatory STING protein while knockout of STING expression only delayed the ISG expression induction. Conclusions Collectively, the presented data show that clinically applied fractionated low-dose irradiation can induce a delayed type I interferon response that occurs independently of interferon expression or STING signaling. These findings have implications for current efforts that aim to target the type I interferon response for cancer treatment.


2014 ◽  
Vol 89 (2) ◽  
pp. 1156-1167 ◽  
Author(s):  
Joeri Kint ◽  
Marcela Fernandez-Gutierrez ◽  
Helena J. Maier ◽  
Paul Britton ◽  
Martijn A. Langereis ◽  
...  

ABSTRACTCoronaviruses from both theAlphacoronavirusandBetacoronavirusgenera interfere with the type I interferon (IFN) response in various ways, ensuring the limited activation of the IFN response in most cell types. Of the gammacoronaviruses that mainly infect birds, little is known about the activation of the host immune response. We show that the prototypicalGammacoronavirus, infectious bronchitis virus (IBV), induces a delayed activation of the IFN response in primary renal cells, tracheal epithelial cells, and a chicken cell line. In fact,Ifnβexpression is delayed with respect to the peak of viral replication and the accompanying accumulation of double-stranded RNA (dsRNA). In addition, we demonstrate that MDA5 is the primary sensor forGammacoronavirusinfections in chicken cells. Furthermore, we provide evidence that accessory proteins 3a and 3b of IBV modulate the response at the transcriptional and translational levels. Finally, we show that, despite the lack of activation of the IFN response during the early phase of IBV infection, the signaling of nonself dsRNA through both MDA5 and TLR3 remains intact in IBV-infected cells. Taken together, this study provides the first comprehensive analysis of host-virus interactions of aGammacoronaviruswith avian innate immune responses.IMPORTANCEOur results demonstrate that IBV has evolved multiple strategies to avoid the activation of the type I interferon response. Taken together, the present study closes a gap in the understanding of host-IBV interaction and paves the way for further characterization of the mechanisms underlying immune evasion strategies as well as the pathogenesis of gammacoronaviruses.


Sign in / Sign up

Export Citation Format

Share Document