Osteosarcoma cell-derived exosomes affect tumor microenvironment by specific packaging of microRNAs

2019 ◽  
Vol 41 (5) ◽  
pp. 666-677 ◽  
Author(s):  
Lavinia Raimondi ◽  
Angela De Luca ◽  
Alessia Gallo ◽  
Viviana Costa ◽  
Giovanna Russelli ◽  
...  

Abstract Bone microenvironment provides growth and survival signals essential for osteosarcoma (OS) initiation and progression. OS cells regulate communications inside tumor microenvironment through different ways and, among all, tumor-derived exosomes support cancer progression and metastasis. To define the contribution of OS-derived exosomes inside the microenvironment, we investigated the effects induced in bone remodeling mechanism and tumor angiogenesis. We demonstrated that exosomes promoted osteoclasts differentiation and bone resorption activity. Furthermore, exosomes potentiated tube formation of endothelial cells and increased angiogenic markers expression. We therefore investigated the micro RNA (miRNA) cargo from exosomes and their parental cells by performing small RNA sequencing through NGS Illumina platform. Hierarchical clustering highlighted a unique molecular profile of exosomal miRNA; bioinformatic analysis by DIANA-mirPath revealed that miRNAs identified take part in various biological processes and carcinogenesis. Among these miRNAs, some were already known for their involvement in the tumor microenvironment establishment, as miR-148a and miR-21-5p. Enforced expression of miR-148a and miR-21-5p in Raw264.7 and hTert immortalized umbilical vein endothelial cells recapitulated the effects induced by exosomes. Overall, our study highlighted the importance of OS exosomes in tumor microenvironment also by a specific packaging of miRNAs.

2021 ◽  
Vol 7 ◽  
Author(s):  
Qian Qian Guo ◽  
Jing Gao ◽  
Xiao Wei Wang ◽  
Xian Lun Yin ◽  
Shu Cui Zhang ◽  
...  

Numerous miRNAs have been detected in mitochondria, which play important roles in many physiological and pathophysiological processes. However, the dynamic changes of miRNA distribution in mitochondria and their mechanisms in reactive oxygen species (ROS)-induced endothelial injury remain unclear. Therefore, miRNA levels in whole cells and mitochondria of H2O2-treated endothelial cells were analyzed by small RNA sequencing in the present study. The results showed that H2O2 significantly reduced the relative mitochondrial distribution of dozens of miRNAs in human umbilical vein endothelial cells (HUVECs). Among the high-abundance miRNAs, miR-301a-3p has the most significant changes in the redistribution between cytosol and mitochondria confirmed by absolute quantitative polymerase chain reaction (qPCR). To unravel the mechanism of miR-301a-3p distribution in mitochondria, RNA pull-down followed by label-free quantitative proteomic analysis was performed, and RNA-binding protein Musashi RNA binding protein 2 (MSI2) was found to specifically bind to miR-301a-3p. Western blotting and immunofluorescence colocalization assay showed that MSI2 was located in mitochondria of various cell types. H2O2 significantly downregulated MSI2 expression in whole endothelial cells, promoted the distribution of MSI2 in cytosol and decreased its distribution in the mitochondria. Moreover, overexpression of MSI2 increased the mitochondrial distribution of miR-301a-3p, whereas inhibition of MSI2 decreased its distribution in mitochondria. Thus, MSI2 might be responsible for the distribution of miR-301a-3p between cytosol and mitochondria in endothelial cells. Our findings revealed for the first time that MSI2 was involved in the regulation of miRNA distribution in mitochondria and provided valuable insight into the mechanism of mitochondrial distribution of miRNAs.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 1055-1055
Author(s):  
Antonella Zucchetto ◽  
Dania Benedetti ◽  
Riccardo Bomben ◽  
Claudio Tripodo ◽  
Fleur Bossi ◽  
...  

Abstract CD38, a negative prognostic marker for patients with CLL, has been demonstrated to be a key molecule in the interactions occurring in the context of tumor microenvironment, mediating both survival and migratory signals for CLL cells. By taking advantage of gene expression profiling studies (GEP) comparing 11 CD38pos (CD38>30%) and 15 CD38neg (CD38<10%) CLLs, we identified as over-expressed in CD38pos CLL cells: i) genes for the two C-C chemokines CCL3 and CCL4 (median-log difference, MLD-CCL3= 3.5; MLD-CCL4=4.4); real-time quantitative PCR (RTQ-PCR) of selected cases confirmed GEP results; ii) the gene for CD49d (MLD=4.4); a high correlation between CD38 and CD49d protein expression, also characterizing the CLL series of the present study, has been reported previously. In vitro experiments, performed on purified tumor cells from additional 11 CD38pos CLL cases cultured for 14 (t14) and 24 (t24) hours in the presence of either the agonist anti-CD38 monoclonal antibody (mAb) IB4 or the non-agonistic anti-CD38 mAb IB6 as control, demonstrated upregulation of CCL3/CCL4 transcripts at t14 (CCL3: mean fold increase=18, p=0.041; CCL4: mean fold increase=13.8, p=0.005), as assessed by RTQ-PCR, and an increased release of CCL3/CCL4 proteins at t24 (CCL3: mean =0.9 ng/mL, mean fold increase=14, p=0.003; CCL4: mean =1.7 ng/mL, mean fold increase=49, p=0.01), as assessed by ELISA. Consistently, immunohistochemistry (IHC) analysis performed in bone marrow biopsies (BMB) from 20 CLL patients (10 CD38pos and 10 CD38neg cases) showed detectable levels of CCL3 in 8 cases, all but one belonging to the CD38pos group (p=0.02). Expression of the CCL3/CCL4 specific receptors CCR1 and CCR5 was examined by flow cytometry in peripheral blood cell subpopulations from 30 CLL (12 CD38pos and 18 CD38neg). Irrespectively of CD38 expression by CLL cells, monocytes showed the highest expression levels for CCR1 and, although at a lesser extent, CCR5. Consistently, CCL3 was able to attract CLL-derived monocytes by in-vitro chemotaxis experiments, and a higher number of infiltrating CD68pos macrophages were found in BMB of CD38pos compared to CD38neg CLLs (p=0.016). In parallel experiments, conditioned media (CM) from CCL3-stimulated macrophages were collected; these CM were able to induce expression of the CD49d-ligand VCAM in human umbilical vein endothelial cells (HUVEC) and human microvascular endothelial cells (ADMEC). As shown by ELISA, TNFalpha was among the cytokines contained in macrophage-CM. This citokine was likely responsible for VCAM up-regulation by HUVEC and ADMEC, as suggested by TNFalpha neutralization experiments leading to a suppression of VCAM-1 induction in endothelial cell models. Again, IHC analysis of CLL BMB showed a meshwork of VCAM-1-positive cells more prominent in the context of lymphoid infiltrates of CD38pos, as compared to CD38neg cases (p=0.002). To verify whether CD49d engagement through VCAM-1 could enhance the protection against spontaneous apoptosis of CLL cells in vitro, we cultured purified CD38pos/CD49dpos CLL cells from 5 cases onto VCAM-1-transfected L cells or mock-transfected L cells. Results demonstrated a substantial improvement in cell viability after CD49d engagement: as high as 70%±25 cells were viable after 10 days of culture on L-VCAM cells compared to 50%±25 in control conditions (p=0.009). Altogether, these results identify molecules involved in a functional cross-talk between CD38/CD49d-expressing CLL and cells of the tumor microenvironment. This interplay may eventually affect survival and recirculation of tumor cells via the CD49d/VCAM pair.


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 5464-5464
Author(s):  
Vi dya Bodempudi ◽  
Olivia Anna Dudek ◽  
Kaoru Terai ◽  
Arkadiusz Zbigniew Dudek

Abstract Background: Angiogenesis is a process involved in the formation of new vessels, and is essential for tumor growth and metastases. Angiogenic factors are critical for the initiation of angiogenesis and maintenance of the vascular network. VG5Q is a newly discovered protein that acts as a potent angiogenic factor in promoting angiogenesis and has also been implicated in the vascular pathophysiology of Klippel-Trenaunay syndrome. However, little is known about its role in cancer progression. Method: In this study, VG5Q expression was screened using RT-PCR and Western blotting in the following cancer cell lines: breast (MCF-7), renal (Caki, ACHN), mesothelioma (H2373, H2596), ovarian (OVCAR5), lung (H226, A549) and pancreatic (Panc-1, MiaPaCa). Human umbilical vein endothelial cells (HUVEC) and blood outgrowth endothelial cells (BOEC) were also screened. Results: All cancer cell lines expressed high levels of VG5Q that were comparable to the levels expressed in endothelial cells. The only exception was MiaPaCa cells. In order to understand the biology of VG5Q in cancer cells, we used siRNA knockdown of VG5Q expression in the mesothelioma cell lines H2373 and H2596. Gene-specific silencing of VG5Q resulted in markedly decreased proliferation, invasion, and soft-agar colony formation in these cells. Furthermore, VG5Q silencing increased apoptosis as seen by an increase in annexin levels. Conclusion: VG5Q silencing has profound effects on cancer cell biology, suggesting the hypothesis that the role of VG5Q may not be limited to angiogenic signaling in endothelial cells.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. e14014-e14014
Author(s):  
Maria Grazia Cerrito ◽  
Davide Pelizzoni ◽  
Marco De Giorgi ◽  
Nunzio Digiacomo ◽  
Marialuisa Lavitrano ◽  
...  

e14014 Background: TNBC represents an important clinical challenge because of poor prognosis. One of the emerging strategy to achieve disease control while reducing toxicity is metronomic chemotherapy (mCHT) which targets the endothelial cells (ECs) and inhibits the tumor growth. mVRL is a promising option in patients (pts) with metastatic breast cancer (MBC), resulting in a median PFS of 7.7 months and median OS of 15.9. To better explain the effect of mVRL we studied the effects of metronomic doses of VRL in in vitro models and compared them with standard doses of the same drug. Methods: Cell viability and cytotoxicity assays were performed on TNBC cancer cells (MDA-MB-231) and Human Umbilical Vein Endothelial Cells (HUVEC). Cell lines were exposed to different concentration (0,01nM-1mM) of VRL for 4 and 96 h. To simulate the metronomic dosing schedule, we replaced the drug-enriched medium every 24h, while to simulate the conventional administration protocol (sCHT) cells were exposed to VRL for 4h, then the medium was changed and replaced with fresh medium without drug every 24 h. The IC50was calculated by non-linear regression fit of the mean values of data obtained in triplicate experiments. Results: A significant anti-proliferative activity was observed on both HUVEC and MDA cells treated with VRL in mCHT as compared to sCHT protocol (see Table). These lower drug concentrations did not have remarkable effects on cell death. Conversely, the higher dose utilized in sCHT produced important cell death in MDA as well as in HUVEC, even if in vivo, the higher dose of drug inducing the largest apoptosis of cancer cells also affectd healthy proliferating cells causing toxicity. Our findings suggest that mCHT inhibited the proliferation of both endothelial and tumour cells and can block cancer progression with minor side effects. Conclusions: This study provides the proof-of-concept that metronomic doses of VRL, but not the standard ones, are able to inhibit, at the same concentration, both the ECs and the TNBC cells. The clinical trial TEMPO-BREAST, which compares metronomic vs standard VRL, is ongoing in MBC pts. [Table: see text]


2021 ◽  
Author(s):  
Ke Xu ◽  
Kai Fang ◽  
Yueping Zhan ◽  
Yuqian Wang ◽  
Chengqi Wu ◽  
...  

Abstract Background Anti-angiogenesis therapy has increasingly become an important strategy for the treatment of colorectal cancer. Recent studies have shown that tumor microenvironment (TME) promotes tumour angiogenesis. Bufalin is an active compound whose anti-tumor efficacy has been proven by previous studies. However, there are very few studies on the anti-angiogenic effects of bufalin. Methods Herein, human umbilical vein endothelial cells (HUVEC) tube formation, migration and adhesion test were used to assess angiogenesis in vitro. Western blot and quantitative PCR were used to detect relevant protein levels and the expressions of mRNAs. Subcutaneous xenograft tumor model and hepatic metastasis model in mice were established to investigate the influence of bufalin on angiogenesis-mediated by TME in vivo. Results We found that the angiogenesis mediated by tumor microenvironment cells was significantly inhibited in the present of bufalin. The results demonstrated that the pro-angiogenic gene in HUVEC such as VEGF, PDGFA, E-selectin and P-selectin were downregulated by bufalin, and the downregulation was regulated by inhibiting the STAT3 pathway. Overexpression STAT3 could reverse the inhibitory effect of bufalin on angiogenesis. What is more, few reduction of angiogenesis when bufalin directly acted on tumor microenvironment cells. Conclusion Our findings demonstrate that bufalin suppresses tumour microenvironment-mediated angiogenesis by inhibiting the STAT3 signaling pathway of vascular endothelial cells, which reveals that bufalin may be used as a new anti-angiogenic adjuvant therapy medicine in the treatment of colorectal cancer.


2019 ◽  
pp. 1-7

IMPORTANCIA DEL MICROAMBIENTE TUMORAL EN LA PROGRESIÓN DEL CÁNCER DE MAMA IMPORTANCE OF TUMOR MICROENVIRONMENT IN BREAST CANCER PROGRESSION Julio E. Valdivia-Silvaa, Eduardo García-Zepedab DOI: https://doi.org/10.33017/RevECIPeru2008.0005/ RESUMEN El microambiente tumoral, en el cáncer de mama y otros de estirpe epitelial, es un tejido complejo que comprende diferentes tipos celulares que incluyen las células tumorales, fibroblastos, células endoteliales, y leucocitos infiltrantes. Citocinas, quimiocinas, factores de crecimiento y proteasas son moléculas claves que controlan la comunicación autocrina y paracrina entre estas células individuales. Bajo algunas circunstancias, dichas moléculas pueden orquestar respuestas del hospedero contra el tumor, pero contradictoriamente existe evidencia que demuestra un rol paradójico que contribuye al crecimiento y progresión de la neoplasia además de inmunosupresión local. Adicionalmente, la progresión del cáncer de mama está asociada con una robusta neovascularización. Es claro que las células “normales” asociadas al tumor, como las inmunes, endoteliales y del estroma, conspiran con las cancerosas en promover este proceso. En ésta revisión enfocamos algunas de las acciones de citocinas inflamatorias y otras moléculas del microambiente tumoral sobre el comportamiento invasivo y metastásico del carcinoma mamario. Una mayor comprensión de estos tipos celulares y constituyentes moleculares del microambiente pueden ser usados en el diseño de terapias más efectivas contra el cáncer. Palabras clave: cáncer de mama, microambiente tumoral, metástasis, inflamación. ABSTRACT The epithelial tumour microenvironment is a complex tissue comprising variable numbers of tumour cells, fibroblasts, endothelial cells and infiltrating leucocytes. Cytokines, chemokines, growth factors, and proteases are key molecules controlling autocrine or paracrine communications within and between these individual cell types. Under some circumstances, endogenous cytokines may orchestrate host responses against the tumour, but there is increasing evidence that the cytokine network contributes to tumour growth, progression and host immuno-suppression. In addition, breast cancer progression is associated with and dependent upon robust neovascularization. It is becoming clear that tumour-associated „normal‟ cells, such as immune/inflammatory cells, endothelial cells and stromal cells, conspire with cancer cells in promoting this process. In this review we outline some of the actions of endogenous inflammatory cytokines and other molecules in tumor microenvironment over metastatic and invasive behavior of the breast carcinoma. A better understanding of these various cellular and molecular constituents of breast cancer microenvironment may be useful in designing more effective therapies. Keywords: breast cancer, tumour microenvironment, metastasis, inflammation.


2021 ◽  
Author(s):  
Swati Srivast ◽  
Iti Garg ◽  
Yamini Singh ◽  
Ramesh Meena ◽  
Anju A Hembrom ◽  
...  

Abstract Outbreak of COVID-19 pandemic in December 2019 affected millions of people globally. After substantial research, there is no specific and reliable biomarker available till date. Present study was designed to identify specific biomarkers to predict COVID-19 severity and tool for formulating treatment. A small cohort of subjects (n=43) were enrolled and categorized in four study groups; Dead (n=16), Severe (n=10) and Moderate (n=7) patients and healthy controls (n=10). Small RNA sequencing was done on Illumina platform after isolation of microRNA from peripheral blood. Differential expression (DE) of miRNA (patients groups compared to control) revealed 118 down-regulated and 103 up-regulated known miRNAs with fold change (FC) expression ≥2 folds and p≤0.05. DE miRNAs were then subjected to functional enrichment and network analysis. Bioinformatic analysis resulted in 31 miRNAs (24 Down-regulated; 7 up-regulated) significantly associated with COVID-19 having AUC>0.8 obtained from ROC curve. Seventeen out of 31 DE miRNAs have been linked to COVID-19 in previous studies. Three miRNAs, hsa-miR-147b-5p and hsa-miR-107 (down-regulated) and hsa-miR-1299 (up-regulated) showed significant unique DE in Dead patients. Another set of 4 miRNAs, hsa-miR-224-5p (down-regulated) and hsa-miR-4659b-3p, hsa-miR-495-3p and hsa-miR-335-3p were differentially up-regulated uniquely in Severe patients. Members of three miRNA families, hsa-miR-20, hsa-miR-32 and hsa-miR-548 were significantly down-regulated in all patients group in comparison to healthy controls. Thus a distinct miRNA expression profile was observed in Dead, Severe and Moderate COVID-19 patients. Present study suggests a panel of miRNAs which identified in COVID-19 patients and could be utilized as potential diagnostic biomarkers for predicting COVID-19 severity.


2020 ◽  
Vol 9 (11) ◽  
pp. 3529
Author(s):  
Mihnea P. Dragomir ◽  
Vlad Moisoiu ◽  
Roxana Manaila ◽  
Barbara Pardini ◽  
Erik Knutsen ◽  
...  

One of the limitations of cancer research has been the restricted focus on tumor cells and the omission of other non-malignant cells that are constitutive elements of this systemic disease. Current research is focused on the bidirectional communication between tumor cells and other components of the tumor microenvironment (TME), such as immune and endothelial cells, and nerves. A major success of this bidirectional approach has been the development of immunotherapy. Recently, a more complex landscape involving a multi-lateral communication between the non-malignant components of the TME started to emerge. A prime example is the interplay between immune and endothelial cells, which led to the approval of anti-vascular endothelial growth factor-therapy combined with immune checkpoint inhibitors and classical chemotherapy in non-small cell lung cancer. Hence, a paradigm shift approach is to characterize the crosstalk between different non-malignant components of the TME and understand their role in tumorigenesis. In this perspective, we discuss the interplay between nerves and immune cells within the TME. In particular, we focus on exosomes and microRNAs as a systemic, rapid and dynamic communication channel between tumor cells, nerves and immune cells contributing to cancer progression. Finally, we discuss how combinatorial therapies blocking this tumorigenic cross-talk could lead to improved outcomes for cancer patients.


2020 ◽  
Vol 21 (7) ◽  
pp. 2606 ◽  
Author(s):  
Oskar Ciesielski ◽  
Marta Biesiekierska ◽  
Baptiste Panthu ◽  
Varvara Vialichka ◽  
Luciano Pirola ◽  
...  

Tumors require a constant supply of nutrients to grow which are provided through tumor blood vessels. To metastasize, tumors need a route to enter circulation, that route is also provided by tumor blood vessels. Thus, angiogenesis is necessary for both tumor progression and metastasis. Angiogenesis is tightly regulated by a balance of angiogenic and antiangiogenic factors. Angiogenic factors of the vascular endothelial growth factor (VEGF) family lead to the activation of endothelial cells, proliferation, and neovascularization. Significant VEGF-A upregulation is commonly observed in cancer cells, also due to hypoxic conditions, and activates endothelial cells (ECs) by paracrine signaling stimulating cell migration and proliferation, resulting in tumor-dependent angiogenesis. Conversely, antiangiogenic factors inhibit angiogenesis by suppressing ECs activation. One of the best-known anti-angiogenic factors is thrombospondin-1 (TSP-1). In pathological angiogenesis, the balance shifts towards the proangiogenic factors and an angiogenic switch that promotes tumor angiogenesis. Here, we review the current literature supporting the notion of the existence of two different endothelial lineages: normal endothelial cells (NECs), representing the physiological form of vascular endothelium, and tumor endothelial cells (TECs), which are strongly promoted by the tumor microenvironment and are biologically different from NECs. The angiogenic switch would be also important for the explanation of the differences between NECs and TECs, as angiogenic factors, cytokines and growth factors secreted into the tumor microenvironment may cause genetic instability. In this review, we focus on the epigenetic differences between the two endothelial lineages, which provide a possible window for pharmacological targeting of TECs.


Sign in / Sign up

Export Citation Format

Share Document