Extracellular Vesicles Protect the Neonatal Lung from Hyperoxic Injury Through the Epigenetic and Transcriptomic Reprogramming of Myeloid Cells

Author(s):  
Gareth R. Willis ◽  
Monica Reis ◽  
Ali Hashemi Gheinani ◽  
Angeles Fernandez-Gonzalez ◽  
Elizabeth S. Taglauer ◽  
...  
Endocrinology ◽  
2021 ◽  
Author(s):  
Amy E Baek ◽  
Natalia Krawczynska ◽  
Anasuya Das Gupta ◽  
Svyatoslav Victorovich Dvoretskiy ◽  
Sixian You ◽  
...  

Abstract Cholesterol has been implicated in the clinical progression of breast cancer, a disease that continues to be the most commonly diagnosed cancer in women. Previous work has identified the cholesterol metabolite, 27-hydroxycholesterol (27HC), as a major mediator of the effects of cholesterol on breast tumor growth and progression. 27HC can act as an estrogen receptor (ER) modulator to promote the growth of ERα+ tumors, and a liver x receptor (LXR) ligand in myeloid immune cells to establish an immune-suppressive program. In fact, the metastatic properties of 27HC require the presence of myeloid cells, with neutrophils (PMNs) being essential for the increase in lung metastasis in murine models. In an effort to further elucidate the mechanisms by which 27HC alters breast cancer progression, we made the striking finding that 27HC promoted the secretion of extracellular vesicles (EVs), a diverse assortment of membrane bound particles that include exosomes. The resulting EVs had a size distribution that was skewed slightly larger, compared to EVs generated by treating cells with vehicle. The increase in EV secretion and size was consistent across three different subtypes: primary murine PMNs, RAW264.7 monocytic cells and 4T1 murine mammary cancer cells. Label-free analysis of 27HC-EVs indicated that they had a different metabolite composition to those from vehicle-treated cells. Importantly, 27HC-EVs from primary PMNs promoted tumor growth and metastasis in two different syngeneic models, demonstrating the potential role of 27HC induced EVs in the progression of breast cancer. EVs from PMNs were taken up by cancer cells, macrophages and PMNs, but not T cells. Since EVs did not alter proliferation of cancer cells, it is likely that their pro-tumor effects are mediated through interactions with myeloid cells. Interestingly, RNA-seq analysis of tumors from 27HC-EV treated mice do not display significantly altered transcriptomes, suggesting that the effects of 27HC-EVs occur early on in tumor establishment and growth. Future work will be required to elucidate the mechanisms by which 27HC increases EV secretion, and how these EVs promote breast cancer progression. Collectively however, our data indicate that EV secretion and content can be regulated by a cholesterol metabolite, which may have detrimental effects in terms of disease progression, important findings given the prevalence of both breast cancer and hypercholesterolemia.


2019 ◽  
Vol 79 (18) ◽  
pp. 4715-4728 ◽  
Author(s):  
Viktor Fleming ◽  
Xiaoying Hu ◽  
Céline Weller ◽  
Rebekka Weber ◽  
Christopher Groth ◽  
...  

Cells ◽  
2021 ◽  
Vol 10 (8) ◽  
pp. 2081
Author(s):  
Hendrik Gassmann ◽  
Kira Schneider ◽  
Valentina Evdokimova ◽  
Peter Ruzanov ◽  
Sebastian J. Schober ◽  
...  

Ewing sarcoma (EwS) is an aggressive pediatric cancer of bone and soft tissues characterized by scant T cell infiltration and predominance of immunosuppressive myeloid cells. Given the important roles of extracellular vesicles (EVs) in cancer-host crosstalk, we hypothesized that EVs secreted by EwS tumors target myeloid cells and promote immunosuppressive phenotypes. Here, EVs were purified from EwS and fibroblast cell lines and exhibited characteristics of small EVs, including size (100–170 nm) and exosome markers CD63, CD81, and TSG101. Treatment of healthy donor-derived CD33+ and CD14+ myeloid cells with EwS EVs but not with fibroblast EVs induced pro-inflammatory cytokine release, including IL-6, IL-8, and TNF. Furthermore, EwS EVs impaired differentiation of these cells towards monocytic-derived dendritic cells (moDCs), as evidenced by reduced expression of co-stimulatory molecules CD80, CD86 and HLA-DR. Whole transcriptome analysis revealed activation of gene expression programs associated with immunosuppressive phenotypes and pro-inflammatory responses. Functionally, moDCs differentiated in the presence of EwS EVs inhibited CD4+ and CD8+ T cell proliferation as well as IFNγ release, while inducing secretion of IL-10 and IL-6. Therefore, EwS EVs may promote a local and systemic pro-inflammatory environment and weaken adaptive immunity by impairing the differentiation and function of antigen-presenting cells.


Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 703-703
Author(s):  
Athalia Rachel Pyzer ◽  
Dina Stroopinsky ◽  
Hasan Rajabi ◽  
Abigail J. Washington ◽  
Leandra Cole ◽  
...  

Abstract Myeloid-derived suppressor cells (MDSCs) play a critical role in promoting immune tolerance and disease growth. We have previously shown that MDSCs are expanded in patients with AML and can be induced from healthy donor PBMCs by co-culture with leukemic cells; a mechanism dependent on expression of the MUC1-C oncoprotein. We sought to elucidate the precise mechanism by which MUC1-C signaling mediates the expansion of this immune suppressive population of immature myeloid cells. We have previously demonstrated that AML cells release membrane bound extracellular vesicles, which traffic to co-cultured cells. We hypothesized that AML EVs may mediate the expansion of MDSCs. MOLM-14 and THP-1 AML EVs were isolated using the ExoQuick precipitation technique, and analyzed by flow cytometry, and compared to size standardized beads, demonstrating particles between 200-300nM in diameter. Furthermoreisolated AML EVs were visualised using Transmission Electron Microscopy demonstrating multiple rounded structures measuring 100-200nM in diameter and bound by darkly staining membrane. Subsequently, healthy donor PBMCs were cultured for three days with GFP tagged AML EVs and then quantified for CD33+/HLADR-/CD11b+ MDSCs and HLADR+/CD11c+ antigen presenting myeloid cells by flow cytometry. In the PBMCs co-cultured with EVs, the proportion of MDSCs increased 8-fold, whilst the proportion of HLADR+/CD11c+ antigen presenting myeloid cells decreased by 10 fold (n=3, p<0.05). We subsequently investigated how MUC1-C signaling, necessary for the expansion of MDSCs, might alter AML extracellular vesicles composition. We evaluated AML EVs for the presence of the pro-proliferative oncoprotein c-Myc by immune-blotting, demonstrating that AML cells secrete EVs containing c-Myc, which is abrogated by downregulation of MUC1-C. Furthermore, EVs containing MUC1 and c-myc led to an up-regulation of the c-Myc downstream targets cyclin D2 and cyclin E1 in co-cultured MDSCs, indicating that c-Myc containing EVs may drive MDSC proliferation. Critically, EVs from MUC1-C silenced AML cells failed to elicit this increase in c-Myc and cyclin D2 and E1 expression in EV exposed MDSCs. Interestingly, exposure of MDSCs to AML EVs lead to an increased expression of PD-L1, which was abrogated in EVs from MUC1-C silenced AML cells. We then sought to determine how MUC1 signaling promotes c-Myc signaling in AML. MUC1-C silencing did not alter c-Myc mRNA levels suggesting a post-transcriptional level of regulation. Micro RNAs are small non-encoding RNA molecules involved in post-translational regulation of gene expression. MiR34a, a known p53 inhibitor, has been implicated in regulating the expansion of MDSCs and it is known that tumor cells suppress MiR34a expression as part of their self-protective armoury. Furthermore, MiR34a is a predicted negative regulator of c-Myc, due to a complementary sequence for MiR34a in the c-Myc promoter region. Using qPCR, we have demonstrated that MUC1-C silencing results in increased expression of MiRNA34a. Furthermore, over-expression of MiR34a in AML cells led to a dramatic down-regulation of c-Myc protein expression, and conversely silencing of MiR34a led to a significant upregulation of c-Myc expression, confirming that MiR34a regulates c-Myc expression in AML. To confirm MiR34a as a critical negative regulator of MDSC expansion, MiR34a altered cells were interrogated for their ability to elicit an expansion of MDSCs in co-cultured PBMCs. Overexpression of MiR34a in AML cells partially abrogated their ability to induce MDSCs from co-cultured donor PBMCs. In concert, silencing of MiR34a in MUC1-C silenced AML cells, recapitulated their ability to induce MDSCs in this model. Taken together, this study illustrates a novel role of the MUC1-C and c-Myc oncoproteins in driving MDSC proliferation and MDSC PD-L1 expression. We have demonstrated that AML EVs alter the tumor microenvironment away from antigen presentation capable myeloid cells and towards immature immune suppressive MDSCs. Disclosures Arnason: Gilead: Consultancy. Küfe:Genus Oncology: Equity Ownership. Rosenblatt:Astex: Research Funding; BMS: Research Funding; DCPrime: Research Funding. Avigan:Astex: Research Funding; DCPrime: Research Funding.


2020 ◽  
Vol 21 (17) ◽  
pp. 6319
Author(s):  
Ihor Arkhypov ◽  
Samantha Lasser ◽  
Vera Petrova ◽  
Rebekka Weber ◽  
Christopher Groth ◽  
...  

Extracellular vesicles (EV) can carry proteins, RNA and DNA, thus serving as communication tools between cells. Tumor cells secrete EV, which can be taken up by surrounding cells in the tumor microenvironment as well as by cells in distant organs. Tumor-derived EV (TEV) contain factors induced by tumor-associated hypoxia such as heat shock proteins or a variety of microRNA (miRNA). The interaction of TEV with tumor and host cells can promote cancer angiogenesis, invasion and metastasis. Myeloid cells are widely presented in tissues, comprise the majority of immune cells and play an essential role in immune reactions and tissue remodeling. However, in cancer, the differentiation of myeloid cells and their functions are impaired, resulting in tumor promotion. Such alterations are due to chronic inflammatory conditions associated with cancer and are mediated by the tumor secretome, including TEV. A high capacity of myeloid cells to clear EV from circulation put them in the central position in EV-mediated formation of pre-metastatic niches. The exposure of myeloid cells to TEV could trigger numerous signaling pathways. Progenitors of myeloid cells alter their differentiation upon the contact with TEV, resulting in the generation of myeloid-derived suppressor cells (MDSC), inhibiting anti-tumor function of T and natural killer (NK) cells and promoting thereby tumor progression. Furthermore, TEV can augment MDSC immunosuppressive capacity. Different subsets of mature myeloid cells such as monocytes, macrophages, dendritic cells (DC) and granulocytes take up TEV and acquire a protumorigenic phenotype. However, the delivery of tumor antigens to DC by TEV was shown to enhance their immunostimulatory capacity. The present review will discuss a diverse and complex EV-mediated crosstalk between tumor and myeloid cells in the context of the tumor type, TEV-associated cargo molecules and type of recipient cells.


Cancers ◽  
2020 ◽  
Vol 12 (11) ◽  
pp. 3457
Author(s):  
Alekhya Mazumdar ◽  
Joaquin Urdinez ◽  
Aleksandar Boro ◽  
Matthias J. E. Arlt ◽  
Fabian E. Egli ◽  
...  

The pre-metastatic niche (PMN) is a tumor-driven microenvironment in distant organs that can foster and support the survival and growth of disseminated tumor cells. This facilitates the establishment of secondary lesions that eventually form overt metastasis, the main cause of cancer-related death. In recent years, tumor-derived extracellular-vesicles (EVs) have emerged as potentially key drivers of the PMN. The role of the PMN in osteosarcoma metastasis is poorly understood and the potential contribution of osteosarcoma cell-derived EVs to PMN formation has not been investigated so far. Here, we characterize pulmonary PMN development using the spontaneously metastasizing 143-B xenograft osteosarcoma mouse model. We demonstrate the accumulation of CD11b+ myeloid cells in the pre-metastatic lungs of tumor-bearing mice. We also establish that highly metastatic 143-B and poorly metastatic SAOS-2 osteosarcoma cell-derived EV education in naïve mice can recapitulate the recruitment of myeloid cells to the lungs. Surprisingly, despite EV-induced myeloid cell infiltration in the pre-metastatic lungs, 143-B and SAOS-2 EVs do not contribute towards the 143-B metastatic burden in the context of both spontaneous as well as experimental metastasis in severe-combined immunodeficient (SCID) mice. Taken together, OS-derived EVs alone may not be able to form a functional PMN, and may perhaps require a combination of tumor-secreted factors along with EVs to do so. Additionally, our study gives a valuable insight into the PMN complexity by providing the transcriptomic signature of the premetastatic lungs in an osteosarcoma xenograft model for the first time. In conclusion, identification of regulators of cellular and molecular changes in the pre-metastatic lungs might lead to the development of a combination therapies in the future that interrupt PMN formation and combat osteosarcoma metastasis.


Sign in / Sign up

Export Citation Format

Share Document