CCR1-Mediated Formation of Liver Microenvironment for Cancer Metastasis

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. 4102-4102
Author(s):  
Hideyo Hirai ◽  
Teruaki Fujishita ◽  
Kazuki Kurimoto ◽  
Mitinori Saitou ◽  
Taira Maekawa ◽  
...  

Abstract Accumulating evidence suggests that bone marrow-derived myeloid cells constitute the major components of cancer microenvironment and play significant roles in the progression of cancer. We have previously shown that myeloid cells expressing CCR1, a chemokine receptor, are recruited to the microenvironment of disseminated colon cancer cells in the liver (PNAS 2010). These CCR1+ cells promote metastatic colonization of cancer cells in the liver through producing metalloproteinases MMP9 and MMP2. However, the cell lineage and precise characteristics of these myeloid cells were largely unknown. The aim of this study is to characterize the cell types among these myeloid cells expressing CCR1 and/or MMPs. In order to characterize the CCR1-expressing myeloid cells accumulating at liver metastatic foci, we have established bacterial artificial chromosome (BAC)-based transgenic mouse lines in which membrane-targeted Venus fluorescent protein (mVenus) was expressed under the control of Ccr1cis-regulatory elements. A flow cytometric analysis of adult transgenic mouse bone marrow cells showed that expression of the reporter mVenus fluorescence was restricted to the CD11b+ Gr-1+ cells and the level of endogenous Ccr1 mRNA correlated well with that of mVenus fluorescence, suggesting that expression of the reporter precisely reflected that of the endogenous CCR1. Inoculation of CCL9-expressing mouse colon cancer cell line CMT93 into the spleen of syngeneic hosts causes efficient metastatic colonization of the cancer cells in the liver. Employing this model, we monitored the behavior of the Ccr1-mVenus+ myeloid cells in the cancer metastasis microenvironment, and found that CD45+ CD11b+Ccr1-mVenus+ cells purified from liver metastatic foci were mostly neutrophils. In contrast, within CD45+ CD11b+Ccr1-mVenus– fraction, we identified eosinophils, monocytes and fibrocytes. Notably, the major cell types in the metastatic foci were monocytes and fibrocytes, with the latter producing abundant collagen. Chronologically, accumulation of neutrophils and monocytes preceded that of eosinophils and fibrocytes. In Ccr1−/− mice, early accumulation of Ccr1-mVenus+ Gr-1+ cells was significantly compromised and later recruitment of fibrocytes to the metastatic foci was reduced. Antibody-mediated depletion of neutrophils caused a significant reduction in the fibrocyte number at the metastatic foci. These results suggest that CCR1 plays critical roles in early accumulation of neutrophils and subsequent recruitment of fibrocytes. After having identified four types of myeloid cells in the metastatic foci, we determined the expression levels of CCR1, MMP9 and MMP2 mRNAs in each purified population, and found that CCR1 and MMP9 are expressed preferentially by neutrophils, whereas MMP2 is expressed exclusively by monocytes/fibrocytes. Considering the chronological difference in the appearance of myeloid cells in liver metastatic foci, these results suggest that production of MMP9 and MMP2 in succession by different myeloid cells is necessary for disseminated colon cancer cells to colonize in the liver. In summary, these results suggests that CCR1 is required for recruitment of neutrophils in the early phase of colon cancer dissemination and the subsequent accumulation of fibrocytes, and that MMP9 and MMP2 are produced by neutrophils and fibrocytes, respectively, in a successive manner during the formation of liver metastasis. Further investigation of these cancer-driven host reactions in human colon cancer dissemination may lead to effective preventive measures against cancer metastasis. Disclosures No relevant conflicts of interest to declare.

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Jin Won Park ◽  
Kyung-Ho Jung ◽  
Jin Hee Lee ◽  
Seung Hwan Moon ◽  
Young Seok Cho ◽  
...  

AbstractCD44 is a cell-surface glycoprotein involved in cell–cell interaction, adhesion, and migration. CD44 is found on colon cancer cells and on immune cells. Previous studies of 89Zr PET imaging of CD44 have relied on an anti-human antibody (Ab), which can influence biodistribution in murine models. In this study, we used an Ab that cross-reacts with both human and mouse origin CD44 of all isoforms to unveil the type of leukocyte responsible for high splenic anti-CD44 uptake and investigate how its regulation can influence tumor immuno-PET. The Ab was site-specifically labeled with 89Zr-deferoxamine on cysteine residues. 89Zr-anti-CD44 demonstrated high-specific binding to HT29 human colon cancer cells and monocytic cells that showed CD44 expression. When 89Zr-anti-CD44 was administered to Balb/C nude mice, there was remarkably high splenic uptake but low SNU-C5 tumor uptake (1.2 ± 0.7%ID/g). Among cells isolated from Balb/C mouse spleen, there was greater CD44 expression on CD11b positive myeloid cells than lymphocytes. In cultured monocytic and macrophage cells, LPS stimulation upregulated CD44 expression and increased 89Zr-anti-CD44 binding. Similarly, normal Balb/C mice that underwent lipopolysaccharide (LPS) stimulation showed a significant upregulation of CD44 expression on splenic myeloid cells. Furthermore, LPS treatment stimulated a 2.44-fold increase of 89Zr-anti-CD44 accumulation in the spleen, which was attributable to splenic myeloid cells. Finally, in Balb/C nude mice bearing HT29 tumors, we injected 89Zr-anti-CD44 with greater Ab doses to reduce binding to splenic cells. The results showed lower spleen uptake and improved tumor uptake (2.9 ± 1.3%ID/g) with a total of 300 μg of Ab dose, and further reduction of spleen uptake and greater tumor uptake (5.7 ± 0.0%ID/g) with 700 μg Ab dose. Thus, using an 89Zr labeled Ab that cross-reacts with both human and mouse CD44, we demonstrate that CD44 immuno-PET has the capacity to monitor CD44 regulation on splenic myeloid cells and may also be useful for imaging colon tumors.


Stem Cells ◽  
2007 ◽  
Vol 25 (2) ◽  
pp. 385-391 ◽  
Author(s):  
Yasushi Koike ◽  
Yasushi Adachi ◽  
Yasuhiro Suzuki ◽  
Masayoshi Iwasaki ◽  
Naoko Koike-Kiriyama ◽  
...  

2009 ◽  
Vol 136 (5) ◽  
pp. A-9-A-10
Author(s):  
Jan Cerny ◽  
Hanchen Li ◽  
Calin Stoicov ◽  
Xueli Fan ◽  
Jian Hua Liu ◽  
...  

2021 ◽  
Vol 11 (10) ◽  
pp. 1918-1923
Author(s):  
Xinfa Zhang ◽  
Cheng Han

This study aims to investigate whether bone marrow mesenchymal stem cell (BMSC) exosomes (BMSC-exos) affects the progression of colon cancer. Ultracentrifugation was used to extract and collect BMSC-exos which were assessed under electron microscope and by flow cytometry. The BMSCs were divided into two groups: control group treated with α-MEM basal medium and experimental group with exosomes (10 μg/ml). Exos were extracted from BMSCs and co-cultured with colon cancer cells, followed by analysis of cell viability by CCK-8 assay and GLUT3 mRNA and protein expression by RT-qPCR and Western blot. The electron microscope analysis indicated that the primary BMSCs showed a long spindle shape with a negative expression of antigen CD34 and positive antigen CD90. Importantly, exos inhibited the viability of colon cancer cells HCT116 and decreased the expression of GLUT3, suggesting that exos might increase the colon cancer cell apoptosis. In conclusion, BMSC-exos inhibit cell progression in colon cancer and might be served as a promising biomarker.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A917-A917
Author(s):  
Niamh Leonard ◽  
Oliver Treacy ◽  
Hannah Egan ◽  
Grace O’Malley ◽  
Elena Tomas Bort ◽  
...  

BackgroundColorectal cancer is the fourth most common occurring cancer and despite new treatment options it remains the third leading cause of cancer related deaths worldwide.1 Of the four Consensus Molecular Subtypes (CMS), the mesenchymal stromal rich CMS4 tumours are shown to have the worst disease free progression survival. However the role mesenchymal stromal cells (MSC) play in the tumour immune microenvironment has yet to be fully elucidated. Understanding the complex communication in this stromal cell rich multicellular environment is challenging but may reveal novel targets for the treatment of colorectal cancer patients.MethodsTumour cell secretome (TCS) was generated from colon cancer cells with/without the addition of TNF-a, an inflammatory stimulus using both human and mouse cell lines. MSCs were then conditioned with the TCS and inflammatory TCS and changes in surface and secreted immunomodulatory molecules were assessed using RNA-seq, flow cytometry and ELISA analysis. Macrophage antigen processing and migration following co-culture with the TCS conditioned MSCs was observed using DQ-ova and transwell experiments. A Gelatin Methacryloyl hydrogel, 3D triple culture systems was established to study the role of MSC in the colon tumour immune microenvironment. HCT116 colon cancer cell line with THP1 monocytic cell line and primary bone marrow derived MSCs were embedded in the hydrogel and incubated for 10days, changing the media on Day 8 with/without the addition of TNF-a. Cell proliferation viability and protein secretion were assessed from the 3D CRC system.ResultsBioplex analysis revealed secretion of potent chemokines and cytokines from the cancer cells. This inflammatory TSC resulted in increased expression of cell surface MSC immunomodulatory markers PD-L1 and CD47 and a variety of secreted molecules. These conditioned MSCs reduced macrophage-mediated antigen processing and increase monocyte migration. A triple culture 3D model of CRC was successfully developed, and while the addition of MSC to the system did not alter spheroid size they increased the release of potent chemokines(CCL2, CXCL12), cytokines (IL-6, IL8) and growth factors (GM-CSF) from the culture system.ConclusionsThe inflammatory tumour cell secretome can alter MSC surface expression and secretion of a variety of immunomodulatory makers. These tumour conditioned MSCs can alter innate immune cell antigen processing and migration. When MSCs are combined in 3D with monocytes and colon cancer cells the MSC significantly alter the secretion of immune modulating and tumour promoting factors from the culture system. Targeting MSC immune suppression in the colon tumour microenvironment could be a novel therapeutic target.Ethics ApprovalHuman MSC (hMSC) were isolated from the bone marrow of three healthy volunteers at Galway University Hospital under an ethically approved protocol (NUIG Research Ethics Committee, Ref: 08/May/14) according to a standardized procedure.ReferenceRawla P, Sunkara T, Barsouk A. Epidemiology of colorectal cancer: incidence, mortality, survival, and risk factors. Prz Gastroenterol 2019;14(2):89-103.


Nanoscale ◽  
2019 ◽  
Vol 11 (37) ◽  
pp. 17357-17367 ◽  
Author(s):  
Alina Sigaeva ◽  
Aryan Morita ◽  
Simon R. Hemelaar ◽  
R. Schirhagl

While some cell types readily ingest nanoparticles, others just don't. We report that, for certain cells, the uptake can be enhanced if the particles are administered from the basolateral side or if the cells are treated with trypsin-EDTA.


Sign in / Sign up

Export Citation Format

Share Document