Systemic infection with Candida albicans in breast tumor bearing mice: Cytokines dysregulation and induction of regulatory T cells

2019 ◽  
Vol 29 (1) ◽  
pp. 49-55 ◽  
Author(s):  
N. Ahmadi ◽  
A. Ahmadi ◽  
E. Kheirali ◽  
M. Hossein Yadegari ◽  
M. Bayat ◽  
...  
2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A679-A679
Author(s):  
Ying Zheng ◽  
Andriana Lebid ◽  
Andrew Pardoll ◽  
Juan Fu ◽  
Chirag Patel ◽  
...  

BackgroundActivins, members of the transforming growth factor-ß (TGF-ß) superfamily, were isolated and identified in endocrine system, and have been widely studied in endocrine-related cancers,1 2 but not substantially in the context of immune system and endocrine-unrelated cancers.3–5 It has been reported that upon binding to the receptors, activins cause the intracellular recruitment and phosphorylation of smad proteins, which mediate the expression of Foxp3.6–9 Therefore, we hypothesized that the blockade of the interaction of activins and their receptors will inhibit the activins-mediated Foxp3 induction in CD4+ T cells, thus modify the immune suppressive tumor microenvironment and achieve the goal of cancer immunotherapy.MethodsELISA (enzyme-linked immunosorbent assay) has been performed to determine the plasma level of Activin A in tumor-bearing mice and cancer patients. In vitro iTreg (induced regulatory T cells) differentiation has been done to naïve CD4+ cells isolated from wild type mice in the presence or absence of Activin A, and the percentage of Foxp3+ cells was demonstrated by flow cytometric analysis. qRT-PCR analysis has been conducted to determine the mRNA level of activin receptor isotypes in the immune subpopulations sorted from Foxp3-YFP mice. In the end, in vivo subcutaneous transplanted tumor studies have been done to evaluate the anti-tumor therapeutic effects of activin-receptor 1c blockade.ResultsWe show here that tumor-bearing mice had elevated Activin A levels, which correlated directly with tumor burden. Likewise, cancer patients had elevated plasma Activin A compared to healthy controls. Importantly, our in vitro studies suggested that Activin A promoted differentiation of conventional CD4+ cells into Foxp3-expressing induced Tregs, especially when TGF-ß was limited. Database and qRT-PCR analysis of sorted major immune cell subsets in mice revealed that activin receptor 1C (Acvr1c) was uniquely expressed on Tregs and was highly upregulated during iTreg differentiation. Mice deficient in Acvr1c were more resistant to cancer progression compared to wild type mice. This phenotype correlated with reduced expression of the FoxP3 transcription factor in CD4+ cells. Similar phenomena were observed when we treated the mice with anti-Acvr1c antibody after tumor inoculation. This anti-tumor therapeutic effect was more significant when anti-Acvr1c antibody was administrated in combination with anti-PD-1 antibody.ConclusionsBlocking Activin A signaling through its receptor 1c is a promising and disease-specific strategy for preventing the accumulation of immunosuppressive iTregs in cancer. Hence it represents a potential candidate for cancer immunotherapy.AcknowledgementsThis research is supported by the Bloomberg-Kimmel Institute (Immunometabolism Program & Immune Modulation Program), the Melanoma Research Alliance, the NIH (RO1AI099300, RO1AI089830, and R01AI137046), and The DoD (PC130767).ReferencesRisbridger GP, Schmitt JF, Robertson DM. Activins and inhibins in endocrine and other tumors. Endocr Rev 2001;22(6):836–858.Cui X, et al. Perspectives of small molecule inhibitors of activin receptor-like kinase in anti-tumor treatment and stem cell differentiation (Review). Mol Med Rep 2019;19(6):5053–5062.Michael IP, et al. ALK7 signaling manifests a homeostatic tissue barrier that is abrogated during tumorigenesis and metastasis. Dev Cell 2019;49(3):409–424.Wu B, et al. The TGF-ß superfamily cytokine Activin-A is induced during autoimmune neuroinflammation and drives pathogenic Th17 cell differentiation. Immunity 2021;54(2):308–323.Antsiferova M, et al. Activin promotes skin carcinogenesis by attraction and reprogramming of macrophages. MBO Mol Med 2017;9(1):27–45.Tsuchida K, et al. Activin isoforms signal through type I receptor serine/threonine kinase ALK7. Mol Cell Endocrinol 2004;220(1–2):59–65.Khalil AM, et al. Differential binding activity of TGF-ß family proteins to select TGF-ß receptors. J Pharmacol Exp Ther 2016;358(3):423–430.Huber S, et al. Activin a promotes the TGF-beta-induced conversion of CD4+CD25- T cells into Foxp3+ induced regulatory T cells. J Immunol 2009;182(8):4633–4640.Iizuka-Koga M, et al. Induction and maintenance of regulatory T cells by transcription factors and epigenetic modifications. J Autoimmun 2017;83:113–121.Ethics ApprovalAll animal experiments were performed under protocols approved by the Johns Hopkins University Institutional Animal Care and Use Committee (IACUC).


Blood ◽  
2008 ◽  
Vol 111 (4) ◽  
pp. 2112-2121 ◽  
Author(s):  
Paria Mirmonsef ◽  
Gladys Tan ◽  
Gang Zhou ◽  
Tricia Morino ◽  
Kimberly Noonan ◽  
...  

Immune reconstitution of autologous hematopoietic stem-cell transplant recipients with the progeny of mature T cells in the graft leads to profound changes in the emerging functional T-cell repertoire. In the steady state, the host is frequently tolerant to tumor antigens, reflecting dominant suppression of naive and effector T cells by regulatory T cells (Tregs). We examined the relative frequency and function of these 3 components within the tumor-specific T-cell compartment during immune reconstitution. Grafts from tumor-bearing donors exerted a significant antitumor effect in irradiated, syngeneic tumor-bearing recipients. This was associated with dramatic clonal expansion and interferon-γ (IFNγ) production by previously tolerant tumor-specific T cells. While donor-derived Tregs expanded in recipients, they did not inhibit the antigen-driven expansion of effector T cells in the early posttransplantation period. Indeed, the repopulation of tumor-specific effector T cells significantly exceeded that of Tregs, the expansion of which was limited by IL-2 availability. Although the intrinsic suppressive capacity of Tregs remained intact, their diminished frequency was insufficient to suppress effector cell function. These findings provide an explanation for the reversal of tolerance leading to tumor rejection in transplant recipients and likely contribute to the efficacy of adoptive T-cell therapies in lymphopenic hosts.


Immunity ◽  
2011 ◽  
Vol 34 (3) ◽  
pp. 422-434 ◽  
Author(s):  
Pushpa Pandiyan ◽  
Heather R. Conti ◽  
Lixin Zheng ◽  
Alanna C. Peterson ◽  
Douglas R. Mathern ◽  
...  

2004 ◽  
Vol 4 (14) ◽  
pp. 1785-1793 ◽  
Author(s):  
Shigeto Hontsu ◽  
Hiroyuki Yoneyama ◽  
Satoshi Ueha ◽  
Yuya Terashima ◽  
Masahiro Kitabatake ◽  
...  

2004 ◽  
Vol 39 (6) ◽  
pp. 941-946 ◽  
Author(s):  
Michael Nicholl ◽  
Andrew Lodge ◽  
Ian Brown ◽  
Sonia L. Sugg

2012 ◽  
Vol 14 (3) ◽  
pp. 328-333 ◽  
Author(s):  
Min Ho Jeong ◽  
Kwang Mo Yang ◽  
Yoo Jin Choi ◽  
Sung Dae Kim ◽  
Young Hyun Yoo ◽  
...  

2020 ◽  
Vol 2020 ◽  
pp. 1-21
Author(s):  
Teng Zhang ◽  
Fangfang Duan ◽  
Danhua Su ◽  
Long Ma ◽  
Jiezuan Yang ◽  
...  

To study the homogeneity and heterogeneity of CD4+CD25+ T cells receptor β-chain complementarity determining region 3 (TCR β CDR3) repertoires in breast tumor tissues, lung metastatic tissues, and spleens from 4T1 tumor-bearing BALB/c mice. We used high-throughput sequencing to analyze the characteristics and changes of CD4+CD25+ TCR β CDR3 repertoires among tumor tissues, lung metastatic tissues, and spleens. The diversity of the CD4+CD25+ TCR β CDR3 repertoires in breast tumor tissue was similar to that of lung metastatic tissues and less pronounced than that of spleen tissues. Breast tumor tissues and lung metastatic tissues had a greater number of high-frequency CDR3 sequences and intermediate-frequency CDR3 sequences than those of spleens. The proportion of unique productive CDR3 sequences in breast tumor tissues and lung metastatic tissues was significantly greater than that in the spleens. The diversity and frequency of the CDR3 repertoires remained homogeneous in breast tumors and lung metastatic tissues and showed great heterogeneity in the spleens, which suggested that the breast tissues and lung metastatic tissues have characteristics of CD4+CD25+ T cells that relate to the tumor microenvironment. However, the number and characteristics of overlapping CDR3 sequences suggested that there were some different CD4+CD25+ T cells in tumors and in the circulatory immune system. The study may be used to further explore the characteristics of the CDR3 repertoires and determine the source of the CD4+CD25+ T cells in the breast cancer microenvironment.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 2177-2177
Author(s):  
Todd W. Kelley ◽  
Olga Efimova ◽  
Jonathan Schumacher ◽  
Philippe Szankasi

Abstract Abstract 2177 Background: A reliable phenotypic marker to distinguish peripherally induced CD4+FOXP3+ regulatory T cells (iTregs) from natural thymus-derived CD4+FOXP3+ regulatory T cells (nTregs) ex vivo would aid in the understanding of Treg biology and may accelerate the design of immune modulating therapies. As an example, if Tregs in the tumor microenvironment were shown to be predominantly induced, rather than thymus-derived, then strategies designed to prevent their accumulation would be influenced accordingly. Recently, the transcription factor Helios was identified as a potential marker expressed by nTregs but not iTregs. Such a marker would allow for identification of origin (peripheral versus thymic) in otherwise phenotypically identical T cells without more laborious methods that have been proposed such as analysis of FOXP3 methylation patterns. Given these findings, we undertook a study of Helios expression patterns in in vitro induced and natural Tregs as well as Tregs in the solid tumor and lymphoma microenvironments. Methods: CD4+FOXP3- conventional T cells (Tc) were isolated from the spleens of wild type C57BL/6 or FOXP3-GFP transgenic mice and treated with TGF-beta (10ng/mL) and low dose IL-2 under conditions of CD3 stimulation or both CD3 and CD28 co-stimulation for 5 days to induce Tregs. The resulting cells were analyzed by flow cytometry for Helios and FOXP3 expression. In some experiments, Tc were treated with TGF-beta and all-trans retinoic acid (ATRA) for five days and analyzed for Helios, FOXP3, CD39 and CD73. Treg suppression assays were performed by co-plating Tregs with CFSE-labeled Tc at 1:4 ratios (Treg:Tc) under conditions of CD3/CD28 stimulation for 3 days. Suppression of Tc proliferation was then quantified by CFSE flow cytometry. In some experiments, C57BL/6 mice were inoculated subcutaneously with syngeneic melanoma cells (B16F10 cells). Similarly, BALB/c mice were inoculated with syngeneic B cell lymphoma cells (A20 cells). Mice were euthanized when tumors reached 2cm in size and single cell tumor suspensions were created. Tumor infiltrating CD4+ T cells were subsequently evaluated by flow cytometry. Results: Tregs induced by TGF-beta in the context of CD3 stimulation alone were nearly uniformly Helios negative while those induced by TGF-beta during CD3 and CD28 co-stimulation were predominantly Helios positive. TGF-beta treatment was more effective at inducing Tregs under conditions of CD3 and CD28 co-stimulation. The addition of ATRA to cultures further increased the number of resultant iTregs as compared to identical conditions lacking ATRA. Although more numerous, ATRA induced Tregs demonstrated decreased Helios expression, decreased suppressive function, identical expression of the ectonucleotidase CD39 but higher levels of the ectonucleotidase CD73 compared to Tregs induced without ATRA. Melanoma and B cell lymphoma infiltrating Tregs expressed high levels of Helios. Helios expression by tumor infiltrating Tregs was greater than in Tregs from the spleens of tumor-bearing mice and from spleens of non-tumor bearing control mice. Conclusions: 1. Helios expression by iTregs is dependent upon stimulation conditions and is increased by CD28 stimulation but inhibited by ATRA treatment. 2. Helios should not be used as a marker for Treg origin. 3. Tumor infiltrating Tregs are nearly uniformly Helios positive and express Helios at higher levels than peripheral circulating Tregs. Disclosures: No relevant conflicts of interest to declare.


Sign in / Sign up

Export Citation Format

Share Document