scholarly journals TSPO imaging-guided characterization of the immunosuppressive myeloid tumor microenvironment in patients with malignant glioma

2020 ◽  
Vol 22 (7) ◽  
pp. 1030-1043 ◽  
Author(s):  
Bastian Zinnhardt ◽  
Michael Müther ◽  
Wolfgang Roll ◽  
Philipp Backhaus ◽  
Astrid Jeibmann ◽  
...  

Abstract Background Tumor-associated microglia and macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) are potent immunosuppressors in the glioma tumor microenvironment (TME). Their infiltration is associated with tumor grade, progression, and therapy resistance. Specific tools for image-guided analysis of spatiotemporal changes in the immunosuppressive myeloid tumor compartments are missing. We aimed (i) to evaluate the role of fluorodeoxyglucose (18F)DPA-714* (translocator protein [TSPO]) PET-MRI in the assessment of the immunosuppressive TME in glioma patients, and (ii) to cross-correlate imaging findings with in-depth immunophenotyping. Methods To characterize the glioma TME, a mixed collective of 9 glioma patients underwent [18F]DPA-714-PET-MRI in addition to [18F]fluoro-ethyl-tyrosine (FET)-PET-MRI. Image-guided biopsy samples were immunophenotyped by multiparametric flow cytometry and immunohistochemistry. In vitro autoradiography was performed for image validation and assessment of tracer binding specificity. Results We found a strong relationship (r = 0.84, P = 0.009) between the [18F]DPA-714 uptake and the number and activation level of glioma-associated myeloid cells (GAMs). TSPO expression was mainly restricted to human leukocyte antigen D related–positive (HLA-DR+) activated GAMs, particularly to tumor-infiltrating HLA-DR+ MDSCs and TAMs. [18F]DPA-714–positive tissue volumes exceeded [18F]FET-positive volumes and showed a differential spatial distribution. Conclusion [18F]DPA-714-PET may be used to non-invasively image the glioma-associated immunosuppressive TME in vivo. This imaging paradigm may also help to characterize the heterogeneity of the glioma TME with respect to the degree of myeloid cell infiltration at various disease stages. [18F]DPA-714 may also facilitate the development of new image-guided therapies targeting the myeloid-derived TME.

Author(s):  
Mohammad H. Rashid ◽  
Thaiz F. Borin ◽  
Roxan Ara ◽  
Raziye Piranlioglu ◽  
Bhagelu R. Achyut ◽  
...  

AbstractMyeloid-derived suppressor cells (MDSCs) are an indispensable component of the tumor microenvironment (TME), and our perception regarding the role of MDSCs in tumor promotion is attaining extra layer of intricacy in every study. In conjunction with MDSC’s immunosuppressive and anti-tumor immunity, they candidly facilitate tumor growth, differentiation, and metastasis in several ways that yet to be explored. Alike any other cell types, MDSCs also release a tremendous amount of exosomes or nanovesicles of endosomal origin and partake in intercellular communications by dispatching biological macromolecules. There has not been any experimental study done to characterize the role of MDSCs derived exosomes (MDSC exo) in the modulation of TME. In this study, we isolated MDSC exo and demonstrated that they carry a significant amount of proteins that play an indispensable role in tumor growth, invasion, angiogenesis, and immunomodulation. We observed higher yield and more substantial immunosuppressive potential of exosomes isolated from MDSCs in the primary tumor area than those are in the spleen or bone marrow. Our in vitro data suggest that MDSC exo are capable of hyper activating or exhausting CD8 T-cells and induce reactive oxygen species production that elicits activation-induced cell death. We confirmed the depletion of CD8 T-cells in vivo by treating the mice with MDSC exo. We also observed a reduction in pro-inflammatory M1-macrophages in the spleen of those animals. Our results indicate that immunosuppressive and tumor-promoting functions of MDSC are also implemented by MDSC-derived exosomes which would open up a new avenue of MDSC research and MDSC-targeted therapy.


2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi125-vi125
Author(s):  
Tyler Alban ◽  
Defne Bayik ◽  
Balint Otvos ◽  
Matthew Grabowski ◽  
Manmeet Ahluwalia ◽  
...  

Abstract The immunosuppressive microenvironment in glioblastoma (GBM) enables persistent tumor growth and evasion from tumoricidal immune cell recognition. Despite a large accumulation of immune cells in the GBM microenvironment, tumor growth continues, and evidence for potent immunosuppression via myeloid derived suppressor cells (MDSCs) is now emerging. In agreement with these observations, we have recently established that increased MDSCs over time correlates with poor prognosis in GBM, making these cells of interest for therapeutic targeting. In seeking to reduce MDSCs in GBM, we previously identified the cytokine macrophage migration inhibitory factor (MIF) as a possible activator of MDSC function in GBM. Here, using a novel in vitro co-culture system to reproducibly and rapidly create GBM-educated MDSCs, we observed that MIF was essential in the generation of MDSCs and that MDSCs generated via this approach express a repertoire of MIF receptors. CD74 was the primary MIF receptor in monocytic MDSCs (M-MDSC), which penetrate the tumor microenvironment in preclinical models and patient samples. A screen of MIF/CD74 interaction inhibitors revealed that MN-166, a clinically relevant blood brain barrier penetrant drug, which is currently fast tracked for FDA approval, reduced MDSC generation and function in vitro. This effect was specific to M-MDSC subsets expressing CD74, and appeared as reduced downstream pERK signaling and MCP-1 secretion. In vivo, MN-166 was able reduce tumor-infiltrating MDSCs, while conferring a significant increase in survival in the syngeneic glioma model GL261. These data provide proof of concept that M-MDSCs can be targeted in the tumor microenvironment via MN-166 to reduce tumor growth and provide a rationale for future clinical assessment of MN-166 to reduce M-MDSCs in the tumor microenvironment. Ongoing studies are assessing the effects of MDSC inhibition in combination with immune activating approaches, in order to inhibit immune suppression while simultaneously activating the immune system.


2019 ◽  
Vol 216 (9) ◽  
pp. 2150-2169 ◽  
Author(s):  
Jérôme Mastio ◽  
Thomas Condamine ◽  
George Dominguez ◽  
Andrew V. Kossenkov ◽  
Laxminarasimha Donthireddy ◽  
...  

We have identified a precursor that differentiates into granulocytes in vitro and in vivo yet belongs to the monocytic lineage. We have termed these cells monocyte-like precursors of granulocytes (MLPGs). Under steady state conditions, MLPGs were absent in the spleen and barely detectable in the bone marrow (BM). In contrast, these cells significantly expanded in tumor-bearing mice and differentiated to polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs). Selective depletion of monocytic cells had no effect on the number of granulocytes in naive mice but decreased the population of PMN-MDSCs in tumor-bearing mice by 50%. The expansion of MLPGs was found to be controlled by the down-regulation of Rb1, but not IRF8, which is known to regulate the expansion of PMN-MDSCs from classic granulocyte precursors. In cancer patients, putative MLPGs were found within the population of CXCR1+CD15−CD14+HLA-DR−/lo monocytic cells. These findings describe a mechanism of abnormal myelopoiesis in cancer and suggest potential new approaches for selective targeting of MDSCs.


2018 ◽  
Author(s):  
Ελένη Κοντάκη

Οι Ιδιοπαθείς Φλεγμονώδεις Νόσοι του Εντέρου (ΙΦΝΕ) είναι χρόνιες φλεγμονώδεις παθήσεις του πεπτικού σωλήνα, με υποτροπίαζοντα ή προοδευτικό χαρακτήρα που επηρρεάζουν εκατομμύρια ανθρώπων παγκοσμίως. Ο όρος ΙΦΝΕ περιλαμβάνει τη νόσο του Crohn και την ελκώδη κολίτιδα. Η ηλικία εμφάνισης της νόσου τοποθετείται συνήθως στη δεύτερη και τρίτη δεκαετία της ζωής, με την πλειονότητα των ασθενών να εμφανίζουν κλινική πορεία χαρακτηριζόμενη από υφέσεις και εξάρσεις. Αν και η ακριβής αιτιολογία των ΙΦΝΕ δεν έχει πλήρως αποσαφηνιστεί, θεωρείται νόσος πολυπαραγοντική. Φαίνεται ότι υπάρχει γενετική προδιάθεση, με υπεύθυνα συγκεκριμένα γονίδια που πιθανώς είναι ο πλέον σπουδαίος παράγοντας κινδύνου εμφάνισης των ΙΦΝΕ. Επιπλέον, η αλλαγή του μικροβιώματος δημιουργεί δυσβίωση μεταξύ ξενιστή και μικροβιώματος του εντέρου και αποτελεί σημαντικό παράγοντα του ξενιστή για την αιτιολογία της νόσου. Δεδομένα από πειραματικά μοντέλα κολίτιδας έχουν βοηθήσει σημαντικά στην κατανόηση των ανοσολογικών μηχανισμών που εμπλέκονται στην έναρξη και την εξέλιξη της νόσου. Συνολικά, φαίνεται ότι μία διαταραχή στο ανοσολογικό σύστημα του εντερικού βλεννογόνου οδηγεί σε υπερπαραγωγή φλεγμονωδών κυτταροκινών, απελευθέρωση μεσολαβητών οξειδωτικού στρες και διήθηση του εντέρου από φλεγμονώδη κύτταρα με αποτέλεσμα φλεγμονή του εντέρου και ιστική καταστροφή. Τα κατασταλτικά κύτταρα μυελικής προέλευσης (Myeoid-derived suppressor cells- MDSCs) αποτελούν έναν ετερογενή πληθυσμό κυττάρων που περιλαμβάνει μακροφάγα, κοκκιοκύτταρα, δενδριτικά κύτταρα και κύτταρα μυελικής σειράς σε πρώιμα στάδια διαφοροποίησης. Στα ποντίκια, τα MDSCs χαρακτηρίζονται από την έκφραση των Gr-1 και CD11b δεικτών και περιλαμβάνουν δύο υποομάδες κυττάρων: αυτά με μορφολογία παρόμοια με εκείνη των κοκκιοκυττάρων και φαινότυπο CD11b+Ly6G+Ly6Clow και αυτά με μορφολογία παρόμοια με εκείνα των μονοκυττάρων και φαινότυπο CD11b+Ly6GlowLy6C+. Στους ανθρώπους, ο φαινοτυπικός προσδιορισμός των MDSCs συνιστά πρόκληση, λόγω της έλλειψης ενιαίων κριτηρίων. Συνηθέστερα, χαρακτηρίζονται απο την έκφραση του δείκτη CD33 της μυελικής σειράς και την έλλειψη έκφρασης τόσο του μορίου τάξης ΙΙ του μείζονος συμπλέγματος ιστοσυμβατότητας (MHC-ΙΙ), HLA-DR, όσο και άλλων δεικτών που χαρακτηρίζουν τις υπόλοιπες κυτταρικές σειρές του μυελού των οστών. Αν και ο ρόλος των MDSCs έχει κυρίως μελετηθεί σε πειραματικά μοντέλα καιασθενείς με καρκίνο, πρόσφατες μελέτες αναδεικνύουν το ρόλο αυτών σε πληθώρα παθολογικών καταστάσεων, όπως οι λοιμώξεις, η μεταμόσχευση και η αυτοανοσία. Παραδοσιακά, τα εν λόγω κύτταρα θεωρείται ότι καταστέλλουν την ανοσιακή απάντηση μέσω ποικίλων μηχανισμών. Ωστόσο, η πρόσφατη βιβλιογραφία επισημαίνει την πλαστικότητα αυτών των κυττάρων, δεδομένου ότι το εκάστοτε φλεγμονώδες περιβάλλον μπορεί να τους προσδώσει προφλεγμονώδεις ιδιότητες. Αν και οι κατασταλτικές ιδιότητες των MDSCs στις διαμεσολαβούμενες απο Τ κύτταρα ανοσιακές απαντήσεις είναι καλώς καθορισμένες, ο ρόλος αυτών στα αυτοάνοσα νοσήματα, όπως οι ΙΦΝΕ, είναι αμφιλεγόμενος. Πιο συγκεκριμένα, η ανοσοκατασταλτική δράση των MDSCs υποστηρίζεται από πολλές εργασίες που αναδεικνύουν την αύξηση των CD11b+Gr1+ MDSCs σε πειραματικά μοντέλα κολίτιδας. Παρομοίως, αύξηση των κατασταλτικών CD14+HLA-DRlow MDSCs περιγράφεται και στο περιφερικό αίμα ασθενών με ΙΦΝΕ. Ωστόσο, πρόσφατες μελέτες επισημαίνουν τον προφλεγμονώδη ρόλο των κυττάρων της μυελικής σειράς σε πειραματικά μοντέλα κολίτιδας, καθώς αυτόλογη μεταφορά Ly6Chigh κυττάρων εντερικού βλεννογόνου οδηγεί σε διαφοροποίηση των τελευταίων σε κύτταρα που προάγουν τη φλεγμονή του εντέρου. Στην παρούσα διδακτορική διατριβή, το ενδιαφέρον εστιάστηκε στον ανοσορυθμιστικό ρόλο των MDSCs σε πειραματικά μοντέλα κολίτιδας, καθώς και στις ανοσιακές απαντήσεις που διαμεσολαβούνται από τα Τ κύτταρα σε ασθενείς με ΙΦΝΕ. Ο αριθμός των MDSCs (ταυτοποιούμενα ως CD14+HLA-DR-/lowCD33+CD15+ κύτταρα) προσδιορίστηκε στο περιφερικό αίμα ασθενών με ΙΦΝΕ, ενώ η δράση αυτών αξιολογήθηκε με in vitro δοκιμασίες. Το πειραματικό μοντέλο που χρησιμοποιήθηκε ήταν αυτό της χημικά 2,4,6-trinitrobenzene sulfonic acid (TNBS) επαγόμενης κολίτιδας. Τα MDSCs χαρακτηρίστηκαν με τη μέθοδο της κυτταρομετρία ροής. Η in vivo ανοσοκατασταλτική δράση των MDSCs που καλλιεργήθηκαν από μυελό των οστών υγιών ποντικιών (BM-MDSCs) ελέγχθηκε τόσο με δοκιμασίες απαλοιφής όσο και αυτόλογης μεταφοράς. Η μελέτη σε ανθρώπινα δείγματα ασθενών με ΙΦΝΕ, ανέδειξε αύξηση του αριθμού των MDSCs στο περιφερικό αίμα ασθενών με ενεργό νόσο. Παρομοίως, τα MDSCs, ιδιαίτερα ο υποπληθυσμός που χαρακτηρίζεται ως Ly6G+, αυξήθηκε στα περιφερικά λεμφικά όργανα ποντικών με TNBS επαγόμενη κολίτιδα κατά την ενεργό φάση της νόσου. Αντίθετα με τις αρχικές προσδοκίες, η αυτόλογη μεταφορά BM-MDSCs σε ποντίκια με TNBS κολίτιδα απέτυχε να ελέγξει τη φλεγμονή in vivo. Περαιτέρω μελέτη του μηχανισμού δράσης των MDSCs, έδειξε ότι η έκθεση των εν λόγω κυττάρων στο φλεγμονώδες περιβάλλον της κολίτιδας οδηγεί σε αλλαγή του φαινότυπου (μείωση των Gr1high και αύξηση των Gr1low κυττάρων) αυτών, καθώς και σε μειωμένη έκφραση της πρωτεΐνης CCAAT/enhancer-binding protein beta (CEBPβ), η οποία αποτελεί μεταγραφικό παράγοντα κλειδί στην ανοσοκατασταλτική δράση των MDSCs. Σε συμφωνία με τα αποτελέσματα της μελέτης στο πειραματικό μοντέλο κολίτιδας, MDSCs τα οποία απομονώθηκαν από περιφερικό αίμα ασθενών με ενεργό νόσο όχι μόνο δεν κατέστειλαν αλλά αντιθέτως ενίσχυσαν τον πολλαπλασιασμό αυτόλογων CD4+ Τ κυττάρων ex vivo. Συνολικά, τα δεδομένα αυτά αναδεικνύουν μια παράδοξη λειτουργία των MDSCs σε πειραματικά μοντέλα κολίτιδας, καθώς και στην in vitro ανοσιακή απάντηση ασθενών με ΙΦΝΕ. Η περαιτέρω κατανόηση των μηχανισμών που οδηγούν στην απώλεια του βασικού χαρακτηριστικού των MDSCs, που συνίσταται στην καταστολή των ανοσιακών απαντήσεων, θα μπορούσε να υποβοηθήσει στο σχεδιασμό νέων θεραπευτικών στόχων.


2021 ◽  
Vol 12 (1) ◽  
Author(s):  
David S. Fischer ◽  
Meshal Ansari ◽  
Karolin I. Wagner ◽  
Sebastian Jarosch ◽  
Yiqi Huang ◽  
...  

AbstractThe in vivo phenotypic profile of T cells reactive to severe acute respiratory syndrome (SARS)-CoV-2 antigens remains poorly understood. Conventional methods to detect antigen-reactive T cells require in vitro antigenic re-stimulation or highly individualized peptide-human leukocyte antigen (pHLA) multimers. Here, we use single-cell RNA sequencing to identify and profile SARS-CoV-2-reactive T cells from Coronavirus Disease 2019 (COVID-19) patients. To do so, we induce transcriptional shifts by antigenic stimulation in vitro and take advantage of natural T cell receptor (TCR) sequences of clonally expanded T cells as barcodes for ‘reverse phenotyping’. This allows identification of SARS-CoV-2-reactive TCRs and reveals phenotypic effects introduced by antigen-specific stimulation. We characterize transcriptional signatures of currently and previously activated SARS-CoV-2-reactive T cells, and show correspondence with phenotypes of T cells from the respiratory tract of patients with severe disease in the presence or absence of virus in independent cohorts. Reverse phenotyping is a powerful tool to provide an integrated insight into cellular states of SARS-CoV-2-reactive T cells across tissues and activation states.


Author(s):  
Kosuke Sasaki ◽  
Shigetsugu Takano ◽  
Satoshi Tomizawa ◽  
Yoji Miyahara ◽  
Katsunori Furukawa ◽  
...  

Abstract Background Recent studies indicate that complement plays pivotal roles in promoting or suppressing cancer progression. We have previously identified C4b-binding protein α-chain (C4BPA) as a serum biomarker for the early detection of pancreatic ductal adenocarcinoma (PDAC). However, its mechanism of action remains unclear. Here, we elucidated the functional roles of C4BPA in PDAC cells and the tumor microenvironment. Methods We assessed stromal C4BPA, the C4BPA binding partner CD40, and the number of CD8+ tumor-infiltrating lymphocytes in resected human PDAC tissues via immunohistochemical staining. The biological functions of C4BPA were investigated in peripheral blood mononuclear cells (PBMCs) and human PDAC cell lines. Mouse C4BPA (mC4BPA) peptide, which is composed of 30 amino acids from the C-terminus and binds to CD40, was designed for further in vitro and in vivo experiments. In a preclinical experiment, we assessed the efficacy of gemcitabine plus nab-paclitaxel (GnP), dual immune checkpoint blockades (ICBs), and mC4BPA peptide in a mouse orthotopic transplantation model. Results Immunohistochemical analysis revealed that high stromal C4BPA and CD40 was associated with favorable PDAC prognosis (P=0.0005). Stromal C4BPA strongly correlated with the number of CD8+ tumor-infiltrating lymphocytes (P=0.001). In in vitro experiments, flow cytometry revealed that recombinant human C4BPA (rhC4BPA) stimulation increased CD4+ and CD8+ T cell numbers in PBMCs. rhC4BPA also promoted the proliferation of CD40-expressing PDAC cells. By contrast, combined treatment with gemcitabine and rhC4BPA increased PDAC cell apoptosis rate. mC4BPA peptide increased the number of murine T lymphocytes in vitro and the number of CD8+ tumor-infiltrating lymphocytes surrounding PDAC tumors in vivo. In a preclinical study, GnP/ICBs/mC4BPA peptide treatment, but not GnP treatment, led to the accumulation of a greater number of CD8+ T cells in the periphery of PDAC tumors and to greater tumor regression than did control treatment. Conclusions These findings demonstrate that the combination of GnP therapy with C4BPA inhibits PDAC progression by promoting antitumor T cell accumulation in the tumor microenvironment.


Cancers ◽  
2021 ◽  
Vol 13 (16) ◽  
pp. 3959
Author(s):  
Oluwaseun Adebayo Bamodu ◽  
Yuan-Hung Wang ◽  
Chen-Hsun Ho ◽  
Su-Wei Hu ◽  
Chia-Da Lin ◽  
...  

Background: prostate cancer (PCa) is a principal cause of cancer-related morbidity and mortality. Castration resistance and metastasis are clinical challenges and continue to impede therapeutic success, despite diagnostic and therapeutic advances. There are reports of the oncogenic activity of genetic suppressor element (GSE)1 in breast and gastric cancers; however, its role in therapy resistance, metastasis, and susceptibility to disease recurrence in PCa patients remains unclear. Objective: this study investigated the role of aberrantly expressed GSE1 in the metastasis, therapy resistance, relapse, and poor prognosis of advanced PCa. Methods: we used a large cohort of multi-omics data and in vitro, ex vivo, and in vivo assays to investigate the potential effect of altered GSE1 expression on advanced/castration-resistant PCa (CRPC) treatment responses, disease progression, and prognosis. Results: using a multi-cohort approach, we showed that GSE1 is upregulated in PCa, while tumor-associated calcium signal transducer 2 (TACSTD2) is downregulated. Moreover, the direct, but inverse, correlation interaction between GSE1 and TACSTD2 drives metastatic disease, castration resistance, and disease progression and modulates the clinical and immune statuses of patients with PCa. Patients with GSE1highTACSTD2low expression are more prone to recurrence and disease-specific death than their GSE1lowTACSTD2high counterparts. Interestingly, we found that the GSE1–TACSTD2 expression profile is associated with the therapy responses and clinical outcomes in patients with PCa, especially those with metastatic/recurrent disease. Furthermore, we demonstrate that the shRNA-mediated targeting of GSE1 (shGSE1) significantly inhibits cell proliferation and attenuates cell migration and tumorsphere formation in metastatic PC3 and DU145 cell lines, with an associated suppression of VIM, SNAI2, and BCL2 and the concomitant upregulation of TACSTD2 and BAX. Moreover, shGSE1 enhances sensitivity to the antiandrogens abiraterone and enzalutamide in vitro and in vivo. Conclusion: these data provide preclinical evidence of the oncogenic role of dysregulated GSE1–TACSTD2 signaling and show that the molecular or pharmacological targeting of GSE1 is a workable therapeutic strategy for inhibiting androgen-driven oncogenic signals, re-sensitizing CRPC to treatment, and repressing the metastatic/recurrent phenotypes of patients with PCa.


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii214-ii214
Author(s):  
Jenna Minami ◽  
Nicholas Bayley ◽  
Christopher Tse ◽  
Henan Zhu ◽  
Danielle Morrow ◽  
...  

Abstract Metabolic reprogramming is a hallmark of cancer, and malignant cells must acquire metabolic adaptations to fuel neoplastic progression. Mutations or changes in metabolic gene expression can impose nutrient dependencies in tumors, and even in the absence of metabolic defects, cancer cells can become auxotrophic for particular nutrients or metabolic byproducts generated by other cells in the tumor microenvironment (TME). Conventional cell lines do not recapitulate the metabolic heterogeneity of glioblastoma (GBM), while primary cultured cells do not account for the influences of the microenvironment and the blood brain barrier on tumor biology. Additionally, these systems are under strong selective pressure divergent from that in vivo, leading to reduced heterogeneity between cultured tumor cells. Here, we describe a biobank of direct-from-patient derived orthotopic xenografts (GliomaPDOX) and gliomaspheres that reveal a subset of gliomas that, while able to form in vivo, cannot survive in vitro. RNA sequencing of tumors that can form both in vivo and in vitro (termed “TME-Indifferent”) compared to that of tumors that can only form in vivo (termed “TME-Dependent”) revealed transcriptional changes associated with altered nutrient availability, emphasizing the unique metabolic programs impacted by the tumor microenvironment. Furthermore, TME-dependent tumors lack metabolic signatures associated with nutrient biosynthesis, thus indicating a potential dependency of these tumors on scavenging specific nutrients from the extracellular milieu. Collectively, these data emphasize the metabolic heterogeneity within GBM, and reveal a subset of gliomas that lack metabolic plasticity, indicating a potential brain-microenvironment specific metabolic dependency that can be targeted for therapy.


2021 ◽  
Vol 20 ◽  
pp. 153303382199528
Author(s):  
Qing Lv ◽  
Qinghua Xia ◽  
Anshu Li ◽  
Zhiyong Wang

This study was performed to investigate the role of interleukin-1 receptor accessory protein (IL1RAP) in stomach carcinoma in vitro and in vivo, determine whether IL1RAP knockdown could regulate the development of stomach carcinoma, and elucidate the relationship between IL1RAP knockdown and inflammation by tumor microenvironment-related inflammatory factors in stomach carcinoma. We first used TCGA and GEPIA systems to predict the potential function of IL1RAP. Second, western blot and RT-PCR were used to analyze the expression, or mRNA level, of IL1RAP at different tissue or cell lines. Third, the occurrence and development of stomach carcinoma in vitro and in vivo were observed by using IL1RAP knockdown lentivirus. Finally, the inflammation of stomach carcinoma in vitro and in vivo was observed. Results show that in GEPIA and TCGA systems, IL1RAP expression in STAD tumor tissue was higher than normal, and high expression of IL1RAP in STAD patients had a worse prognostic outcome. Besides, GSEA shown IL1RAP was negative correlation of apopopsis, TLR4 and NF-κB signaling pathway. We also predicted that IL1RAP may related to IL-1 s, IL-33, and IL-36 s in STAD. The IL1RAP expression and mRNA level in tumor, or MGC803, cells were increased. Furthermore, IL1RAP knockdown by lentivirus could inhibit stomach carcinoma development in vitro and in vivo through weakening tumor cell proliferation, migration, invasion, therefore reducing tumor volume, weight, and biomarker levels, and increasing apoptotic level. Finally, we found IL1RAP knockdown could increase inflammation of tumor microenvironment-related inflammatory factors of stomach carcinoma, in vitro and in vivo. Our study demonstrates that IL1RAP is possibly able to regulate inflammation and apoptosis in stomach carcinoma. Furthermore, TLR4, NF-κB, IL-1 s, IL-33, and IL-36 s maybe the downstream target factor of IL1RAP in inflammation. These results may provide a new strategy for stomach carcinoma development by regulating inflammation.


Author(s):  
Atsuhito Uneda ◽  
Kazuhiko Kurozumi ◽  
Atsushi Fujimura ◽  
Kentaro Fujii ◽  
Joji Ishida ◽  
...  

AbstractGlioblastoma (GBM) is the most lethal primary brain tumor characterized by significant cellular heterogeneity, namely tumor cells, including GBM stem-like cells (GSCs) and differentiated GBM cells (DGCs), and non-tumor cells such as endothelial cells, vascular pericytes, macrophages, and other types of immune cells. GSCs are essential to drive tumor progression, whereas the biological roles of DGCs are largely unknown. In this study, we focused on the roles of DGCs in the tumor microenvironment. To this end, we extracted DGC-specific signature genes from transcriptomic profiles of matched pairs of in vitro GSC and DGC models. By evaluating the DGC signature using single cell data, we confirmed the presence of cell subpopulations emulated by in vitro culture models within a primary tumor. The DGC signature was correlated with the mesenchymal subtype and a poor prognosis in large GBM cohorts such as The Cancer Genome Atlas and Ivy Glioblastoma Atlas Project. In silico signaling pathway analysis suggested a role of DGCs in macrophage infiltration. Consistent with in silico findings, in vitro DGC models promoted macrophage migration. In vivo, coimplantation of DGCs and GSCs reduced the survival of tumor xenograft-bearing mice and increased macrophage infiltration into tumor tissue compared with transplantation of GSCs alone. DGCs exhibited a significant increase in YAP/TAZ/TEAD activity compared with GSCs. CCN1, a transcriptional target of YAP/TAZ, was selected from the DGC signature as a candidate secreted protein involved in macrophage recruitment. In fact, CCN1 was secreted abundantly from DGCs, but not GSCs. DGCs promoted macrophage migration in vitro and macrophage infiltration into tumor tissue in vivo through secretion of CCN1. Collectively, these results demonstrate that DGCs contribute to GSC-dependent tumor progression by shaping a mesenchymal microenvironment via CCN1-mediated macrophage infiltration. This study provides new insight into the complex GBM microenvironment consisting of heterogeneous cells.


Sign in / Sign up

Export Citation Format

Share Document