immune response genes
Recently Published Documents


TOTAL DOCUMENTS

291
(FIVE YEARS 52)

H-INDEX

36
(FIVE YEARS 3)

EMBO Reports ◽  
2021 ◽  
Author(s):  
Meetali Singh ◽  
Maxime Chazal ◽  
Piergiuseppe Quarato ◽  
Loan Bourdon ◽  
Christophe Malabat ◽  
...  

2021 ◽  
Vol 17 (11) ◽  
pp. e1010100
Author(s):  
Marine J. Petit ◽  
Matthew W. Kenaston ◽  
Oanh H. Pham ◽  
Ariana A. Nagainis ◽  
Adam T. Fishburn ◽  
...  

Dengue virus (DENV) disruption of the innate immune response is critical to establish infection. DENV non-structural protein 5 (NS5) plays a central role in this disruption, such as antagonism of STAT2. We recently found that DENV serotype 2 (DENV2) NS5 interacts with Polymerase associated factor 1 complex (PAF1C). The primary members of PAF1C are PAF1, LEO1, CTR9, and CDC73. This nuclear complex is an emerging player in the immune response. It promotes the expression of many genes, including genes related to the antiviral, antimicrobial and inflammatory responses, through close association with the chromatin of these genes. Our previous work demonstrated that NS5 antagonizes PAF1C recruitment to immune response genes. However, it remains unknown if NS5 antagonism of PAF1C is complementary to its antagonism of STAT2. Here, we show that knockout of PAF1 enhances DENV2 infectious virion production. By comparing gene expression profiles in PAF1 and STAT2 knockout cells, we find that PAF1 is necessary to express immune response genes that are STAT2-independent. Finally, we mapped the viral determinants for the NS5-PAF1C protein interaction. We found that NS5 nuclear localization and the C-terminal region of the methyltransferase domain are required for its interaction with PAF1C. Mutation of these regions rescued the expression of PAF1-dependent immune response genes that are antagonized by NS5. In sum, our results support a role for PAF1C in restricting DENV2 replication that NS5 antagonizes through its protein interaction with PAF1C.


2021 ◽  
Vol 12 ◽  
Author(s):  
Peik M. A. Brundin ◽  
Britt-Marie Landgren ◽  
Peter Fjällström ◽  
Mohamed M. Shamekh ◽  
Jan-Åke Gustafsson ◽  
...  

Sex hormones are known to interact with the immune system on multiple levels but information on the types of sex hormone receptors (SHR) and their expression levels in immune cells is scarce. Estrogen, testosterone and progesterone are all considered to interact with the immune system through their respective cell receptors (ERα and ERβ including the splice variant ERβ2, AR and PGR). In this study expression levels of SHR genes in peripheral blood mononuclear cells (PBMCs) and cell subsets (CD4+ and CD8+ T-cells, CD56+ NK-cells, CD14+ monocytes and CD19+ B-cells) were analyzed using standard manual qPCR or a qPCR array (TLDA). Nine healthy individuals including men (n = 2), premenopausal (Pre-MP, n = 5) and postmenopausal (post-MP, n = 2) women were sampled for PBMCs which were separated to cell subsets using FACS. Ten Pre-MP women were longitudinally sampled for total PBMCs at different phases of the menstrual cycle. We found that ERα was most abundant and, unexpectedly, that ERβ2 was the dominant ERβ variant in several FACS sorted cell subsets. In total PBMCs, SHR (ERα, ERβ1, ERβ2, and AR) expression did not fluctuate according to the phase of the menstrual cycle and PGR was not expressed. However, several immune response genes (GATA3, IFNG, IL1B, LTA, NFKB1, PDCD1, STAT3, STAT5A, TBX21, TGFB1, TNFA) were more expressed during the ovulatory and mid-luteal phases. Sex hormone levels did not correlate significantly with gene expression of SHR or immune response genes, but sex hormone-binding globulin (SHBG), a steroid hormone transporting protein, was positively correlated to expression of ERβ1 gene. This study provides new insights in the distribution of ERs in immune cells. Furthermore, expression patterns of several immune response genes differ significantly between phases of the menstrual cycle, supporting a role for sex hormones in the immune response.


2021 ◽  
Author(s):  
Titto Augustine ◽  
Peter John ◽  
Tyler Friedman ◽  
Jeeshan Jiffry ◽  
Hillary Guzik ◽  
...  

Background: Microsatellite stable (MSS) colorectal cancer (CRC) represents ~85% of all CRCs. These tumors are poorly immunogenic and largely resistant to immunotherapy, necessitating a need to develop new immune enhancing strategies. Oncolytic reovirus has a high propensity to replicate in KRAS mutant tumors which account for ~50% of MSS CRCs. Current study explores the ability of reovirus to potentiate the effect of immune checkpoint inhibition in MSS CRC. Methods: Effectiveness of reovirus infection was quantified through MTT assay for cell viability, and expression of immune-response genes by flow cytometry, RT-qPCR, and microarray. Computational analysis of differentially expressed genes was performed by TAC, DAVID and STRING. Combinatorial approach using anti-PD-1 monoclonal antibody was assessed in ex vivo and in vivo models. Live-cell imaging, tumor volume and survival were measured for quantification of anti-tumor activity. Expression of pattern recognition receptors (PRRs), cell surface and activation markers of immune cells, and PD-1/PD-L1 axis were studied using multi-color flow cytometry, immunoblotting, immunohistochemistry, and immunofluorescence. Results: Reovirus infection exerted growth arrest and expression of immune-response genes in CRCs cell lines in a KRAS-dependent manner. However, microsatellite instability, rather than KRAS status determined immune-repose pathways, functionalities and biological processes post-reovirus infection. Furthermore, reovirus significantly enhanced the anti-tumor activity of anti-human PD-1 [nivolumab] treatment in MSS CRC cell lines ex vivo. Similarly, reovirus increased the activity of anti-mouse PD-1 treatment in the CT26 [MSS, KRASMut], but not the MC38 [MSI, KRASWt] syngeneic mouse model of CRC. Combinatorial treatment has reduced the proliferative index, increased apoptosis and differentially altered PD-L1/PD-1 signaling among CT26 and MC38 tumors. Activation of innate immune system and expression of PRRs and antigen presentation markers were observed under reovirus and anti-PD-1 treatment that additionally reduced immunosuppressive macrophages. This led to an increase in T cell subsets, increase in effector T cell activation, and decrease in exhaustion markers specifically within CT26 microenvironment. Conclusion: The current study systematically evaluates immune characteristics and immune microenvironment of CRC under reovirus/anti-PD-1 combination treatment that proves increased effectiveness among MSS compared to MSI CRCs. This is a promising regimen warranting translation into clinical trials.


2021 ◽  
Author(s):  
Shawndra Wibisono ◽  
Phillip Wibisono ◽  
Jingru Sun

We have previously demonstrated that OCTR-1, an octopamine G protein-couple receptor, functions in the sensory neurons ASH to suppress the innate immune response in Caenorhabditis elegans by inhibiting the expression of immune genes (Sun et al. 2011 Science 332:729-732). Here we discover that OCTR-1 also regulates temperature effects on lifespan in C. elegans. At the normal growth temperature 20⁰C, octr-1(ok371) mutant animals have similar lifespan to wild-type N2 animals. However, at higher temperature 25⁰C, octr-1(ok371) mutants live significant longer than wild-type N2 animals. These results suggest that OCTR-1 may mediate temperature effects on lifespan. Furthermore, we found the OCTR-1-expressing ASH chemosensory neurons are involved in the OCTR-1-mediated regulation on longevity. However, interestingly, the thermosensory AFD neurons do not play a role in this regulation at 25⁰C. RNA-seq data analysis showed that 63 immune response genes were significantly down-regulated in octr-1(ok371) mutants relative to wild-type animals at 25⁰C. We further demonstrated that inactivation of several most-downregulated genes by RNA interference in wild-type N2 animals significantly extended their lifespan, similar to the phenotype of octr-1(ok371) animals. These observations suggest a new molecular regulation mechanism that downregulation of immune genes extends the lifespan of C. elegans, which is opposite to the general belief that an increase in defense immunity extends lifespan.


2021 ◽  
Vol 35 (S1) ◽  
Author(s):  
Anton Musabirov ◽  
Y.A. Menchits ◽  
A.V. Guz ◽  
L.A. Lebedeva ◽  
Z.M. Kachaev ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document