scholarly journals Inflammatory Breast Cancer: a model for investigating cluster-based dissemination

2017 ◽  
Author(s):  
Mohit Kumar Jolly ◽  
Marcelo Boareto ◽  
Bisrat G Debeb ◽  
Nicola Aceto ◽  
Mary C Farach-Carson ◽  
...  

AbstractMetastases claim more than 90% of cancer-related patient deaths and are usually seeded by a subset of circulating tumor cells (CTCs) shed off from the primary tumor. In circulation, CTCs are found both as single cells and as clusters of cells. The clusters of CTCs, although many fewer in number, possess much higher metastatic potential as compared to that of individual CTCs. In this review, we highlight recent insights into molecular mechanisms that can enable the formation of these clusters - (a) hybrid epithelial/mesenchymal (E/M) phenotype of cells that couples their ability to migrate and adhere, and (b) intercellular communication that can spatially coordinate the cluster formation and provide survival signals to cancer cells. Building upon these molecular mechanisms, we also offer a possible mechanistic understanding of why clusters are endowed with a higher metastatic potential. Finally, we discuss the highly aggressive Inflammatory Breast Cancer (IBC) as an example of a carcinoma that can metastasize via clusters and corroborates the proposed molecular mechanisms.

2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A799-A799
Author(s):  
Dhiraj Kumar ◽  
Sreeharsha Gurrapu ◽  
Hyunho Han ◽  
Yan Wang ◽  
Seongyeon Bae ◽  
...  

BackgroundLong non-coding RNAs (lncRNAs) are involved in various biological processes and diseases. Malat1 (metastasis-associated lung adenocarcinoma transcript 1), also known as Neat2, is one of the most abundant and highly conserved nuclear lncRNAs. Several studies have shown that the expression of lncRNA Malat1 is associated with metastasis and serving as a predictive marker for various tumor progression. Metastatic relapse often develops years after primary tumor removal as a result of disseminated tumor cells undergoing a period of latency in the target organ.1–4 However, the correlation of tumor intrinsic lncRNA in regulation of tumor dormancy and immune evasion is largely unknown.MethodsUsing an in vivo screening platform for the isolation of genetic entities involved in either dormancy or reactivation of breast cancer tumor cells, we have identified Malat1 as a positive mediator of metastatic reactivation. To functionally uncover the role of Malat1 in metastatic reactivation, we have developed a knock out (KO) model by using paired gRNA CRISPR-Cas9 deletion approach in metastatic breast and other cancer types, including lung, colon and melanoma. As proof of concept we also used inducible knockdown system under in vivo models. To delineate the immune micro-environment, we have used 10X genomics single cell RNA-seq, ChIRP-seq, multi-color flowcytometry, RNA-FISH and immunofluorescence.ResultsOur results reveal that the deletion of Malat1 abrogates the tumorigenic and metastatic potential of these tumors and supports long-term survival without affecting their ploidy, proliferation, and nuclear speckles formation. In contrast, overexpression of Malat1 leads to metastatic reactivation of dormant breast cancer cells. Moreover, the loss of Malat1 in metastatic cells induces dormancy features and inhibits cancer stemness. Our RNA-seq and ChIRP-seq data indicate that Malat1 KO downregulates several immune evasion and stemness associated genes. Strikingly, Malat1 KO cells exhibit metastatic outgrowth when injected in T cells defective mice. Our single-cell RNA-seq cluster analysis and multi-color flow cytometry data show a greater proportion of T cells and reduce Neutrophils infiltration in KO mice which indicate that the immune microenvironment playing an important role in Malat1-dependent immune evasion. Mechanistically, loss of Malat1 is associated with reduced expression of Serpinb6b, which protects the tumor cells from cytotoxic killing by the T cells. Indeed, overexpression of Serpinb6b rescued the metastatic potential of Malat1 KO cells by protecting against cytotoxic T cells.ConclusionsCollectively, our data indicate that targeting this novel cancer-cell-initiated domino effect within the immune system represents a new strategy to inhibit tumor metastatic reactivation.Trial RegistrationN/AEthics ApprovalFor all the animal studies in the present study, the study protocols were approved by the Institutional Animal Care and Use Committee(IACUC) of UT MD Anderson Cancer Center.ConsentN/AReferencesArun G, Diermeier S, Akerman M, et al., Differentiation of mammary tumors and reduction in metastasis upon Malat1 lncRNA loss. Genes Dev 2016 Jan 1;30(1):34–51.Filippo G. Giancotti, mechanisms governing metastatic dormancy and reactivation. Cell 2013 Nov 7;155(4):750–764.Gao H, Chakraborty G, Lee-Lim AP, et al., The BMP inhibitor Coco reactivates breast cancer cells at lung metastatic sites. Cell 2012b;150:764–779.Gao H, Chakraborty G, Lee-Lim AP, et al., Forward genetic screens in mice uncover mediators and suppressors of metastatic reactivation. Proc Natl Acad Sci U S A 2014 Nov 18; 111(46): 16532–16537.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 3904-3904
Author(s):  
Leah A. Marquez-Curtis ◽  
Marcin Wysoczynski ◽  
Mariusz Z. Ratajczak ◽  
Anna Janowska-Wieczorek

Abstract There is increasing evidence that platelets contribute to cancer metastasis, and yet platelet concentrates are frequently transfused to cancer patients to treat thrombocytopenia after chemotherapy. Recently we reported that microvesicles derived from activated platelets (PMV) transfer various surface receptors/adhesion molecules to normal and malignant target cells and modulate their biological responses (Blood2001; 98:3143; Exp Hematol2002; 30:450). In this work, we hypothesized that the interaction of PMV with cancer cells increases their invasive and metastatic potential. PMV were isolated from outdated platelet concentrates and pre-incubated with human breast cancer cell lines (MDA-MB-231, BT-549 and T47D), and the effect of PMV on the invasive/metastatic potential of these cancer cells was evaluated. We determined (i) the transfer of the platelet-derived antigen CD41 to cancer cells and the adhesion of these cells to human umbilical vein endothelial cells (HUVEC), (ii) the expression of matrix metalloproteinases (MMPs) by breast cancer cells and their ability to cross the reconstituted basement membrane Matrigel, (iii) the expression of CXCR4, the cognate receptor of the a-chemokine SDF-1, produced in bone marrow, in these cell lines after incubation with PMV, and (iv) the effects of PMV on the interactions of the tumor cells with stroma. We found that PMV transfer platelet-derived CD41 integrin to the surface of breast cancer cells and promote their adhesion to HUVEC. Preincubation with PMV upregulates the mRNA for MMP-9 and protein secretion in invasive breast cancer cells (MDA-MB-231 and BT-549) and enhances their trans-Matrigel chemoinvasion. PMV also transfer CXCR4 to the surface of the breast cancer cells and stimulate the trans-Matrigel migration of MDA-MB-231 cells towards SDF-1, which was abrogated by AMD3100, a CXCR4 antagonist. Finally we found that PMV increase activation of the latent form of MMP-2 constitutively secreted by fibroblastic cells in co-cultures of tumor cells with bone marrow stroma. Thus, we conclude that PMV may enhance the invasive and metastatic potential of breast cancer cells. Because concentrations of PMV are known to be higher in old platelet concentrates than in fresh ones, we recommend that cancer patients should preferably be transfused with fresh platelet concentrates only.


2011 ◽  
Vol 29 (15_suppl) ◽  
pp. e11133-e11133
Author(s):  
A. Bhattacharyya ◽  
S. Krishnamurthy ◽  
A. Lodhi ◽  
C. Hall ◽  
A. Anderson ◽  
...  

2021 ◽  
Author(s):  
Jingya Bu ◽  
Weiliang Zhong ◽  
Meixian Li ◽  
Shuiqing He ◽  
Mingzhe Zhang ◽  
...  

Abstract Background: As a tumor metastasis suppressor, tetraspanin CD82 is reduced in many malignant tumors and often affects the composition of tumor microenvironment by changing the heterogeneity of cell membrane. EGFR or c-Met signaling pathway can regulate the metastasis ability of tumor cells and participate in the formation of tetraspanin web. The study of CD82 palmitoylation modification and metabolic pathway of tumor related molecules in tumor cells is still incomplete. This article focuses on studying the expression and distribution of EGFR and c-Met in cancer cells as well as related metabolic pathways and their molecular mechanisms after studying different palmitoylation site mutations.Methods: Western blot and immunofluorescence methods were used to detect the distribution of EGFR in breast cancer MDA-MB-231 cells after different CD82 palmitoylation site mutations. Then use the immunoprecipitation method to determine the interaction relationship between the molecules and the molecular mechanism.Results: We found that when CD82 combined with palmitoylation mutation at Cys5+Cys74 can enhance the internalization of EGFR, but has no effect on the expression and location of c-Met. When CD82 is combined with palmitoylation mutation at the Cys5+Cys74 site, with the assistance of tubulin, EGFR is internalized and strengthened by direct binding to CD82 and a large number of localizations on the recycling endosome. By forming the EGFR/CD82/Rab11a/FIP2 complex, it is metabolized through the circulation pathway, and re-expression of EGFR and CD82 on the cell membrane.Conclusions: From our results, we can demonatrate that CD82 palmitoylation mutation can change the distribution of EGFR in breast cancer cells, which may provide new ideas for breast cancer treatment.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2859-2859
Author(s):  
Gyeongsin Park ◽  
Hae-Jung Kim ◽  
Bo-Bae Park ◽  
Woo-Chan Park ◽  
Kyo-Young Lee ◽  
...  

Abstract Cancer stem cells are subpopulations of cancer cells with potential to seed and develop new cancer mass causing recurrence and/or metastasis, but phenotypic characteristics of such cell populations remains uncertain. As an initial approach to identify heterogeneity in cancer mass, we performed comparative study using matched pairs of primary breast cancer masses and their immediate metastatic counterparts in regional LN. First, in the gene expression study of primary tumor mass and their LN counterpart using cDNA microarray, about 100 genes showed differential expression between the two, but with variations depending on cases (3 Exp). In addition, when fresh individually matched surgical block from primary mass and LN mass were each inoculated into NOD/SCID mice, higher growth of LN-derived mass was observed supporting distinction between the two (2 Exp). In subsequent search for phenotypic difference between the two, 99 primary-LN tissue pairs with individual matching were immunostained for CD24, a newly proposed marker for cancer stem cell, and analysed by semiquantitative scoring (0, 1+, 2+, 3+, 4+). Among 73 cases with positive staining for CD24, higher level of CD24 expression in LN mass compared to its matched primary tumor tissue was observed in 38 cases (52.1%), while the opposite was for only 7 cases (9.6%). Furthermore, high level expression (4+) of CD24 was observed more frequently in LN tumor tissues (63.0%, 46/73) than in their primary tumor counterparts (26%, 19/73)( P = 0.000). These results support the notion that cancer cells in tumor mass are clonally heterogeneous and suggest that CD24 expression could be related to characteristic of metastatic cancer stem cells.


Author(s):  
Samad Beheshtirouy ◽  
Farhad Mirzaei ◽  
Shirin Eyvazi ◽  
Vahideh Tarhriz

: Breast cancer is a heterogeneous malignancy which is the second cause of mortality among women in the world. Increasing the resistance to anti-cancer drugs in breast cancer cells persuades researchers to search the novel therapies approaches for the treatment of the malignancy. Among the novel methods, therapeutic peptides which target and disrupt tumor cells have been of great interest. Therapeutic peptides are short amino acids monomer chains with high specificity to bind and modulate a protein interaction of interest. Several advantages of peptides such as specific binding on tumor cells surface, low molecular weight and low toxicity on normal cells make the peptides as an appealing therapeutic agents against solid tumors, particularly breast cancer. Also, National Institutes of Health (NIH) describes therapeutic peptides as suitable candidate for the treatment of drug-resistant breast cancer. In this review, we attempt to review the different therapeutic peptides against breast cancer cells which can be used in treatment and diagnosis of the malignancy. Meanwhile, we presented an overview of peptide vaccines which have been developed for the treatment of breast cancer.


Sign in / Sign up

Export Citation Format

Share Document