Human Norovirus Triggers Primary B Cell Immune Activation In Vitro.
Human norovirus (HNoV) is a global health and socioeconomic burden,estimated to infect every individual at least five times during their lifetime. The underlying mechanism for the potential lack of long-term immune protection from HNoV infections is not understood and prompted us to investigate HNoV susceptibility of primary human B cells and its functional impact. Primary B cells isolated from whole blood were infected with HNoV-positive stool samples and harvested at 3??days postinfection (dpi) to assess the viral RNA yield by reverse transcriptase quantitative PCR (RT-qPCR). A 3- to 18-fold increase in the HNoV RNA yield was observed in 50 to 60% of donors. Infection was further confirmed in B cells derived from splenic and lymph node biopsy specimens. Next,we characterized infection of whole-blood-derived B cells by flow cytometry in specific functional B cell subsets (naive CD27- IgD+,memory-switched CD27+ IgD-,memory-unswitched CD27+ IgD+,and double-negative CD27- IgD- cells). While the susceptibilities of the subsets were similar,changes in the B cell subset distribution upon infection were observed,which were also noted after treatment with HNoV virus-like particles and the predicted recombinant NS1 protein. Importantly,primary B cell stimulation with the predicted recombinant NS1 protein triggered B cell activation and induced metabolic changes. These data demonstrate that primary B cells are susceptible to HNoV infection and suggest that the NS1 protein can alter B cell activation and metabolism in vitro,which could have implications for viral pathogenesis and immune responses in vivo. IMPORTANCE Human norovirus (HNoV) is the most prevalent causative agent of gastroenteritis worldwide. Infection results in a self-limiting disease that can become chronic and severe in the immunocompromised,the elderly,and infants. There are currently no approved therapeutic and preventative strategies to limit the health and socioeconomic burdens associated with HNoV infections. Moreover,HNoV does not elicit lifelong immunity as repeat infections are common,presenting a challenge for vaccine development. Given the importance of B cells for humoral immunity,we investigated the susceptibility and impact of HNoV infection on human B cells. We found that HNoV replicates in human primary B cells derived from blood,spleen,and lymph node specimens,while the nonstructural protein NS1 can activate B cells. Because of the secreted nature of NS1,we put forward the hypothesis that HNoV infection can modulate bystander B cell function with potential impacts on systemic immune responses.
View Publication
文献
K. Joannou et al. (may 2022)
Journal of immunology (Baltimore,Md. : 1950) 208 9 2131--2140
Thymocyte Maturation and Emigration in Adult Mice.
Several unique waves of ?? T cells are generated solely in the fetal/neonatal thymus,whereas additional ?? T cell subsets are generated in adults. One intriguing feature of ?? T cell development is the coordination of differentiation and acquisition of effector function within the fetal thymus; however,it is less clear whether this paradigm holds true in adult animals. In this study,we investigated the relationship between maturation and thymic export of adult-derived ?? thymocytes in mice. In the Rag2pGFP model,immature (CD24+) ?? thymocytes expressed high levels of GFP whereas only a minority of mature (CD24-) ?? thymocytes were GFP+ Similarly,most peripheral GFP+ ?? T cells were immature. Analysis of ?? recent thymic emigrants (RTEs) indicated that most ?? T cell RTEs were CD24+ and GFP+,and adoptive transfer experiments demonstrated that immature ?? thymocytes can mature outside the thymus. Mature ?? T cells largely did not recirculate to the thymus from the periphery; rather,a population of mature ?? thymocytes that produced IFN-? or IL-17 remained resident in the thymus for at least 60 d. These data support the existence of two populations of ?? T cell RTEs in adult mice: a majority subset that is immature and matures in the periphery after thymic emigration,and a minority subset that completes maturation within the thymus prior to emigration. Additionally,we identified a heterogeneous population of resident ?? thymocytes of unknown functional importance. Collectively,these data shed light on the generation of the ?? T cell compartment in adult mice.
View Publication
文献
A. Revenko et al. (apr 2022)
Journal for immunotherapy of cancer 10 4
Direct targeting of FOXP3 in Tregs with AZD8701, a novel antisense oligonucleotide to relieve immunosuppression in cancer.
BACKGROUND The Regulatory T cell (Treg) lineage is defined by the transcription factor FOXP3,which controls immune-suppressive gene expression profiles. Tregs are often recruited in high frequencies to the tumor microenvironment where they can suppress antitumor immunity. We hypothesized that pharmacological inhibition of FOXP3 by systemically delivered,unformulated constrained ethyl-modified antisense oligonucleotides could modulate the activity of Tregs and augment antitumor immunity providing therapeutic benefit in cancer models and potentially in man. METHODS We have identified murine Foxp3 antisense oligonucleotides (ASOs) and clinical candidate human FOXP3 ASO AZD8701. Pharmacology and biological effects of FOXP3 inhibitors on Treg function and antitumor immunity were tested in cultured Tregs and mouse syngeneic tumor models. Experiments were controlled by vehicle and non-targeting control ASO groups as well as by use of multiple independent FOXP3 ASOs. Statistical significance of biological effects was evaluated by one or two-way analysis of variance with multiple comparisons. RESULTS AZD8701 demonstrated a dose-dependent knockdown of FOXP3 in primary Tregs,reduction of suppressive function and efficient target downregulation in humanized mice at clinically relevant doses. Surrogate murine FOXP3 ASO,which efficiently downregulated Foxp3 messenger RNA and protein levels in primary Tregs,reduced Treg suppressive function in immune suppression assays in vitro. FOXP3 ASO promoted more than 70% reduction in FOXP3 levels in Tregs in vitro and in vivo,strongly modulated Treg effector molecules (eg,ICOS,CTLA-4,CD25 and 4-1BB),and augmented CD8+ T cell activation and produced antitumor activity in syngeneic tumor models. The combination of FOXP3 ASOs with immune checkpoint blockade further enhanced antitumor efficacy. CONCLUSIONS Antisense inhibitors of FOXP3 offer a promising novel cancer immunotherapy approach. AZD8701 is being developed clinically as a first-in-class FOXP3 inhibitor for the treatment of cancer currently in Ph1a/b clinical trial (NCT04504669).
View Publication
文献
A. M. Chinn et al. ( 2022)
Frontiers in pharmacology 13 833832
PDE4B Is a Homeostatic Regulator of Cyclic AMP in Dendritic Cells.
Chronic decreases in the second messenger cyclic AMP (cAMP) occur in numerous settings,but how cells compensate for such decreases is unknown. We have used a unique system-murine dendritic cells (DCs) with a DC-selective depletion of the heterotrimeric GTP binding protein G$\alpha$s-to address this issue. These mice spontaneously develop Th2-allergic asthma and their DCs have persistently lower cAMP levels. We found that phosphodiesterase 4B (PDE4B) is the primary phosphodiesterase expressed in DCs and that its expression is preferentially decreased in G$\alpha$s-depleted DCs. PDE4B expression is dynamic,falling and rising in a protein kinase A-dependent manner with decreased and increased cAMP concentrations,respectively. Treatment of DCs that drive enhanced Th2 immunity with a PDE4B inhibitor ameliorated DC-induced helper T cell response. We conclude that PDE4B is a homeostatic regulator of cellular cAMP concentrations in DCs and may be a target for treating Th2-allergic asthma and other settings with low cellular cAMP concentrations.
View Publication
文献
D. J. Friedman et al. (apr 2022)
Journal of immunology (Baltimore,Md. : 1950) 208 8 1845--1850
Cutting Edge: Enhanced Antitumor Immunity in ST8Sia6 Knockout Mice.
Inhibitory receptors have a critical role in the regulation of immunity. Siglecs are a family of primarily inhibitory receptors expressed by immune cells that recognize specific sialic acid modifications on cell surface glycans. Many tumors have increased sialic acid incorporation. Overexpression of the sialyltransferase ST8Sia6 on tumors led to altered immune responses and increased tumor growth. In this study,we examined the role of ST8Sia6 on immune cells in regulating antitumor immunity. ST8Sia6 knockout mice had an enhanced immune response to tumors. The loss of ST8Sia6 promoted an enhanced intratumoral activation of macrophages and dendritic cells,including upregulation of CD40. Intratumoral regulatory T cells exhibited a more inflammatory phenotype in ST8Sia6 knockout mice. Using adoptive transfer studies,the change in regulatory T cell phenotype was not cell intrinsic and depended on the loss of ST8Sia6 expression in APCs. Thus,ST8Sia6 generates ligands for Siglecs that dampen antitumor immunity.
View Publication
文献
X. Guan et al. (jun 2022)
Nature 606 7915 791--796
Androgen receptor activity in T cells limits checkpoint blockade efficacy.
Immune checkpoint blockade has revolutionized the field of oncology,inducing durable anti-tumour immunity in solid tumours. In patients with advanced prostate cancer,immunotherapy treatments have largely failed1-5. Androgen deprivation therapy is classically administered in these patients to inhibit tumour cell growth,and we postulated that this therapy also affects tumour-associated T cells. Here we demonstrate that androgen receptor (AR) blockade sensitizes tumour-bearing hosts to effective checkpoint blockade by directly enhancing CD8 T cell function. Inhibition of AR activity in CD8 T cells prevented T cell exhaustion and improved responsiveness to PD-1 targeted therapy via increased IFN$\gamma$ expression. AR bound directly to Ifng and eviction of AR with a small molecule significantly increased cytokine production in CD8 T cells. Together,our findings establish that T cell intrinsic AR activity represses IFN$\gamma$ expression and represents a novel mechanism of immunotherapy resistance.
View Publication
文献
P. Li et al. (mar 2022)
Journal for immunotherapy of cancer 10 3
1$\alpha$,25(OH)2D3 reverses exhaustion and enhances antitumor immunity of human cytotoxic T cells.
BACKGROUND Epidemiological surveys have revealed that low serum vitamin D level was correlated with increased risk of tumors. Dysfunctional T cells in patients with tumor are characterized as exhausted with high levels of immune checkpoint receptors (ICRs). However,whether the reduced level of vitamin D in patients with cancer correlates with cytotoxic T-cell exhaustion is unknown. METHODS Periphery blood samples from 172 patients with non-small cell lung cancer (NSCLC) were prospectively collected. Patients with NSCLC received one course of intravenous docetaxel (75 mg/m2) followed by treatment with or without rocaltrol at a dose of 0.5-2.0 µg/day for total of 3 weeks. We performed phenotypical and functional analysis of T-cell through flow cytometry. Vitamin D receptor (VDR) knockout and overexpression CD8+ and V$\delta$2+ T cells were constructed using Cas9-gRNA targeted and overexpressing approaches to identify 1$\alpha$,25(OH)2D3/VDR-mediated transcription regulation for ICRs or antitumor activity in T cells. RESULTS We show that serum level of vitamin D is negatively correlated with expression of programmed cell death-1 (PD-1),T-cell immunoreceptor with Ig and ITIM domains (TIGIT),and T-cell immunoglobulin and mucin-domain containing-3 (Tim-3),but positively correlated with CD28 expression on CD8+ and V$\gamma$9V$\delta$2+ T cells in patients with NSCLC. 1$\alpha$,25(OH)2D3,the active form of vitamin D,promotes the nuclear translocation of VDR,which binds to the promoter region of Pdcd1,Tim3,and Tigit genes and inhibits their expression. Besides,1$\alpha$,25(OH)2D3 pretreatment also promotes the methylation of CpG island in the promoter region of the Pdcd1 gene and increases H3K27 acetylation at the promoter region of the Cd28 gene,which leads to surface PD-1 downregulation and CD28 upregulation,respectively. We further reveal that VDR-mediated Ca2+ influx enhanced expression of Th1 cytokines via T-cell receptor activation. Functionally,1$\alpha$,25(OH)2D3 pretreated CD8+ T cells or V$\gamma$9V$\delta$2+ T cells showed increased Th1 cytokine production and enhanced antitumor immunity. Finally,oral 1$\alpha$,25(OH)2D3 could also decrease expression of PD-1,Tim-3,TIGIT and increase expression of CD28,resulting in cytokine production (associated with antitumor immunity) by cytotoxic T cells of patients with NSCLC. CONCLUSIONS Our findings uncover the pleiotropic effects of 1$\alpha$,25(OH)2D3 in rescuing the exhausted phenotype of human cytotoxic T cells in patients with tumor and in promoting their antitumor immunity. TRIAL REGISTRATION NUMBER ChiCTR2100051135.
View Publication
文献
R. Bitsch et al. (mar 2022)
Journal for immunotherapy of cancer 10 3
STAT3 inhibitor Napabucasin abrogates MDSC immunosuppressive capacity and prolongs survival of melanoma-bearing mice.
BACKGROUND Myeloid-derived suppressor cells (MDSCs) represent a negative prognostic factor in malignant melanoma. These cells are generated under chronic inflammatory conditions typical of cancer. The transcription factor signal transducer and activator of transcription 3 (STAT3) orchestrates MDSC accumulation and acquisition of immunosuppressive properties. Here we studied STAT3 inhibition by Napabucasin as a way to block MDSC accumulation and activity and its potential to treat malignant melanoma. METHODS In vitro generated murine MDSC and primary MDSC from melanoma-bearing mice were used to investigate the effects of Napabucasin on MDSC in vitro. The RET transgenic mouse model of malignant melanoma was used to examine Napabucasin therapy efficiency and its underlying mechanisms in vivo. Furthermore,STAT3 activation and its correlation with survival were explored in MDSC from 19 patients with malignant melanoma and human in vitro generated monocytic myeloid-derived suppressor cell (M-MDSC) were used to evaluate the effects of Napabucasin. RESULTS Napabucasin was able to abrogate the capacity of murine MDSC to suppress CD8+ T-cell proliferation. The STAT3 inhibitor induced apoptosis in murine MDSC,significantly increased expression of molecules associated with antigen processing and presentation,as well as slightly decreased expression of immunosuppressive factors on these cells. RET transgenic mice treated with Napabucasin showed prolonged survival accompanied by a strong accumulation of tumor-infiltrating antigen-presenting cells and activation of CD8+ and CD4+ T cells. Interestingly,patients with malignant melanoma with high expression of activated STAT3 in circulating M-MDSC showed significantly worse progression-free survival (PFS) than patients with low levels of activated STAT3. In addition,Napabucasin was able to abrogate suppressive capacity of human in vitro generated M-MDSC. CONCLUSION Our findings demonstrate that STAT3 inhibitor Napabucasin completely abrogated the immunosuppressive capacity of murine MDSC and human M-MDSC and improved melanoma-bearing mouse survival. Moreover,patients with malignant melanoma with high expression levels of activated STAT3 in M-MDSC displayed shorter PFS,indicating its role as a promising therapeutic target in patients with malignant melanoma and a predictive marker for their clinical outcome.
View Publication
文献
Y. Shen et al. (mar 2022)
Journal for immunotherapy of cancer 10 3
Cancer cell-intrinsic resistance to BiTE therapy is mediated by loss of CD58 costimulation and modulation of the extrinsic apoptotic pathway.
BACKGROUND Bispecific T-cell engager (BiTE) molecules induce redirected lysis of cancer cells by T cells and are an emerging modality for solid tumor immunotherapy. While signs of clinical activity have been demonstrated,efficacy of T-cell engagers (TCEs) in solid tumors settings,molecular determinants of response,and underlying mechanisms of resistance to BiTE therapy require more investigation. METHODS To uncover cancer cell-intrinsic genetic modifiers of TCE-mediated cytotoxicity,we performed genome-wide CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) loss-of-function and CRISPRa (CRISPR activation) gain-of-function screens using TCEs against two distinct tumor-associated antigens (TAAs). By using in vitro T-cell cytotoxicity assays and in vivo efficacy studies,we validated the roles of two common pathways identified in our screen,T-cell costimulation pathway and apoptosis pathway,as key modifiers of BiTE activity. RESULTS Our genetic screens uncovered TAAs-independent cancer cell-intrinsic genes with functions in autophagy,T-cell costimulation,the apoptosis pathway,chromatin remodeling,and cytokine signaling that altered responsiveness to BiTE-mediated killing. Notably,loss of CD58 (the ligand of the CD2 T-cell costimulatory receptor),a gene frequently altered in cancer,led to decreased TCE-mediated cytotoxicity,T-cell activation and antitumor efficacy in vitro and in vivo. Moreover,the effects of CD58 loss were synergistically compounded by concurrent loss of CD80/CD86 (ligands for the CD28 T-cell costimulatory receptor),whereas joint CD2 and CD28 costimulation additively enhanced TCE-mediated killing,indicating non-redundant costimulatory mechanisms between the two pathways. Additionally,loss of CFLAR (Caspase-8 and FADD Like Apoptosis Regulator),BCL2L1,and BID (BH3 Interacting Domain Death Agonist) induced profound changes in sensitivity to TCEs,indicating that key regulators of apoptosis,which are frequently altered in cancer,impact tumor responsiveness to BiTE therapy. CONCLUSIONS This study demonstrates that genetic alterations central to carcinogenesis and commonly detected in cancer samples lead to significant modulation of BiTE antitumor activity in vitro and in vivo,findings with relevance for a better understanding of patient responses to BiTE therapy and novel combinations that enhance TCE efficacy.
View Publication
文献
A. Goral et al. ( 2022)
Frontiers in immunology 13 781364
A Specific CD44lo CD25lo Subpopulation of Regulatory T Cells Inhibits Anti-Leukemic Immune Response and Promotes the Progression in a Mouse Model of Chronic Lymphocytic Leukemia.
Regulatory T cells (Tregs) are capable of inhibiting the proliferation,activation and function of T cells and play an important role in impeding the immune response to cancer. In chronic lymphocytic leukemia (CLL) a dysfunctional immune response and elevated percentage of effector-like phenotype Tregs have been described. In this study,using the Eµ-TCL1 mouse model of CLL,we evaluated the changes in the Tregs phenotype and their expansion at different stages of leukemia progression. Importantly,we show that Tregs depletion in DEREG mice triggered the expansion of new anti-leukemic cytotoxic T cell clones leading to leukemia eradication. In TCL1 leukemia-bearing mice we identified and characterized a specific Tregs subpopulation,the phenotype of which suggests its role in the formation of an immunosuppressive microenvironment,supportive for leukemia survival and proliferation. This observation was also confirmed by the gene expression profile analysis of these TCL1-specific Tregs. The obtained data on Tregs are consistent with those described so far,however,above all show that the changes in the Tregs phenotype described in CLL result from the formation of a specific,described in this study Tregs subpopulation. In addition,functional tests revealed the ability of Tregs to inhibit T cells that recognize model antigens expressed by leukemic cells. Moreover,inhibition of Tregs with a MALT1 inhibitor provided a therapeutic benefit,both as monotherapy and also when combined with an immune checkpoint inhibitor. Altogether,activation of Tregs appears to be crucial for CLL progression.
View Publication
文献
Y. N. Yoon et al. (mar 2022)
Journal for immunotherapy of cancer 10 3
PI3K$\delta$/$\gamma$ inhibitor BR101801 extrinsically potentiates effector CD8+ T cell-dependent antitumor immunity and abscopal effect after local irradiation.
BACKGROUND Radiotherapy enhances antitumor immunity. However,it also induces immunosuppressive responses,which are major hurdles for an effective treatment. Thus,targeting the immunosuppressive tumor microenvironment is essential for enhancing the antitumor immunity after radiotherapy. Retrospective studies show that a blockade of PI3K$\delta$ and/or $\gamma$,which are abundant in leukocytes,exhibits antitumor immune response by attenuating activity of immune suppressive cells,however,the single blockade of PI3K$\delta$ or $\gamma$ is not sufficient to completely eliminate solid tumor. METHODS We used BR101801,PI3K$\delta$/$\gamma$ inhibitor in the CT-26 syngeneic mouse model with a subcutaneously implanted tumor. BR101801 was administered daily,and the target tumor site was locally irradiated. We monitored the tumor growth regularly and evaluated the immunological changes using flow cytometry,ELISpot,and transcriptional analysis. RESULTS This study showed that BR101801 combined with irradiation promotes systemic antitumor immunity and abscopal response by attenuating the activity of immune suppressive cells in the CT-26 tumor model. BR101801 combined with irradiation systemically reduced the proliferation of regulatory T cells (Tregs) and enhanced the number of tumor-specific CD8$\alpha$+ T cells in the tumor microenvironment,thereby leading to tumor regression. Furthermore,the high ratio of CD8$\alpha$+ T cells to Tregs was maintained for 14 days after irradiation,resulting in remote tumor regression in metastatic lesions,the so-called abscopal effect. Moreover,our transcriptomic analysis showed that BR101801 combined with irradiation promoted the immune-stimulatory tumor microenvironment,suggesting that the combined therapy converts immunologically cold tumors into hot one. CONCLUSIONS Our data suggest the first evidence that PI3K$\delta$/$\gamma$ inhibition combined with irradiation promotes systemic antitumor immunity against solid tumors,providing the preclinical result of the potential use of PI3K$\delta$/$\gamma$ inhibitor as an immune-regulatory radiosensitizer.
View Publication
文献
E. Vamva et al. (mar 2022)
STAR protocols 3 1 101228
An optimized measles virus glycoprotein-pseudotyped lentiviral vector production system to promote efficient transduction of human primary B cells.
Measles virus envelope pseudotyped LV (MV-LV) can achieve high B cell transduction rates (up to 50%),but suffers from low titers. To overcome current limitations,we developed an optimized MV-LV production protocol that achieved consistent B cell transduction efficiency up to 75%. We detail this protocol along with analytical assays to assess the results of MV-LV mediated B cell transduction,including flow cytometry for B cell phenotypic characterization and measurement of transduction efficiency,and ddPCR for VCN analysis.
View Publication