Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
J Cell Sci ; 133(24)2020 12 22.
Article in English | MEDLINE | ID: mdl-33443095

ABSTRACT

The tissue microenvironment supports normal tissue function and regulates the behaviour of parenchymal cells. Tumour cell behaviour, on the other hand, diverges significantly from that of their normal counterparts, rendering the microenvironment hostile to tumour cells. To overcome this problem, tumours can co-opt and remodel the microenvironment to facilitate their growth and spread. This involves modifying both the biochemistry and the biophysics of the normal microenvironment to produce a tumour microenvironment. In this Cell Science at a Glance article and accompanying poster, we outline the key processes by which epithelial tumours influence the establishment of the tumour microenvironment. As the microenvironment is populated by genetically normal cells, we discuss how controlling the microenvironment is both a significant challenge and a key vulnerability for tumours. Finally, we review how new insights into tumour-microenvironment interactions has led to the current consensus on how these processes may be targeted as novel anti-cancer therapies.


Subject(s)
Neoplasms , Tumor Microenvironment , Humans , Neoplasms/genetics
2.
J Proteome Res ; 19(10): 4093-4103, 2020 10 02.
Article in English | MEDLINE | ID: mdl-32870688

ABSTRACT

Tumorigenesis involves a complex interplay between genetically modified cancer cells and their adjacent normal tissue, the stroma. We used an established breast cancer mouse model to investigate this inter-relationship. Conditional activation of Rho-associated protein kinase (ROCK) in a model of mammary tumorigenesis enhances tumor growth and progression by educating the stroma and enhancing the production and remodeling of the extracellular matrix. We used peptide matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) to quantify the proteomic changes occurring within tumors and their stroma in their regular spatial context. Peptides were ranked according to their ability to discriminate between the two groups, using a receiver operating characteristic tool. Peptides were identified by liquid chromatography tandem mass spectrometry, and protein expression was validated by quantitative immunofluorescence using an independent set of tumor samples. We have identified and validated four key proteins upregulated in ROCK-activated mammary tumors relative to those expressing kinase-dead ROCK, namely, collagen I, α-SMA, Rab14, and tubulin-ß4. Rab14 and tubulin-ß4 are expressed within tumor cells, whereas collagen I is localized within the stroma. α-SMA is predominantly localized within the stroma but is also expressed at higher levels in the epithelia of ROCK-activated tumors. High expression of COL1A, the gene encoding the pro-α 1 chain of collagen, correlates with cancer progression in two human breast cancer genomic data sets, and high expression of COL1A and ACTA2 (the gene encoding α-SMA) are associated with a low survival probability (COLIA, p = 0.00013; ACTA2, p = 0.0076) in estrogen receptor-negative breast cancer patients. To investigate whether ROCK-activated tumor cells cause stromal cancer-associated fibroblasts (CAFs) to upregulate expression of collagen I and α-SMA, we treated CAFs with medium conditioned by primary mammary tumor cells in which ROCK had been activated. This led to abundant production of both proteins in CAFs, clearly highlighting the inter-relationship between tumor cells and CAFs and identifying CAFs as the potential source of high levels of collagen 1 and α-SMA and associated enhancement of tissue stiffness. Our research emphasizes the capacity of MALDI-MSI to quantitatively assess tumor-stroma inter-relationships and to identify potential prognostic factors for cancer progression in human patients, using sophisticated mouse cancer models.


Subject(s)
Cancer-Associated Fibroblasts , Proteomics , Animals , Extracellular Matrix , Fibroblasts , Humans , Mice , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , rab GTP-Binding Proteins
3.
Mol Cancer ; 16(1): 19, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28137279

ABSTRACT

BACKGROUND: Breast cancer is the major cause of cancer-related mortality in women. It is thought that quiescent stem-like cells within solid tumors are responsible for cancer maintenance, progression and eventual metastasis. We recently reported that the chemokine receptor CCR7, a multi-functional regulator of breast cancer, maintains the stem-like cell population. METHODS: This study used a combination of molecular and cellular assays on primary mammary tumor cells from the MMTV-PyMT transgenic mouse with or without CCR7 to examine the signaling crosstalk between CCR7 and Notch pathways. RESULTS: We show for the first time that CCR7 functionally intersects with the Notch signaling pathway to regulate mammary cancer stem-like cells. In this cell subpopulation, CCR7 stimulation activated the Notch signaling pathway, and deletion of CCR7 significantly reduced the levels of activated cleaved Notch1. Moreover, blocking Notch activity prevented specific ligand-induced signaling of CCR7 and augmentation of mammary cancer stem-like cell function. CONCLUSION: Crosstalk between CCR7 and Notch1 promotes stemness in mammary cancer cells and may ultimately potentiate mammary tumor progression. Therefore, dual targeting of both the CCR7 receptor and Notch1 signaling axes may be a potential therapeutic avenue to specifically inhibit the functions of breast cancer stem cells.


Subject(s)
Mammary Neoplasms, Experimental/metabolism , Neoplastic Stem Cells/metabolism , Receptor, Notch1/metabolism , Receptors, CCR7/genetics , Animals , Female , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/virology , Mice , Mice, Transgenic , Receptor, Notch1/genetics , Receptors, CCR7/metabolism , Signal Transduction , Tumor Cells, Cultured
4.
Genesis ; 54(12): 636-646, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27775859

ABSTRACT

The serine/threonine kinases ROCK1 and ROCK2 are central mediators of actomyosin contractile force generation that act downstream of the RhoA small GTP-binding protein. As a result, they have key roles in regulating cell morphology and proliferation, and have been implicated in numerous pathological conditions and diseases including hypertension and cancer. Here we describe the generation of a gene-targeted mouse line that enables CRE-inducible expression of a conditionally-active fusion between the ROCK2 kinase domain and the hormone-binding domain of a mutated estrogen receptor (ROCK2:ER). This two-stage system of regulation allows for tissue-selective expression of the ROCK2:ER fusion protein, which then requires administration of estrogen analogues such as tamoxifen or 4-hydroxytamoxifen to elicit kinase activity. This conditional gain-of-function system was validated in multiple tissues by crossing with mice expressing CRE recombinase under the transcriptional control of cytokeratin14 (K14), murine mammary tumor virus (MMTV) or cytochrome P450 Cyp1A1 (Ah) promoters, driving appropriate expression in the epidermis, mammary or intestinal epithelia respectively. Given the interest in ROCK signaling in normal physiology and disease, this mouse line will facilitate research into the consequences of ROCK activation that could be used to complement conditional knockout models. Birth Defects Research (Part A) 106:636-646, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Estrogen Receptor alpha/genetics , Recombinant Fusion Proteins/genetics , rho-Associated Kinases/genetics , Animals , Cytochrome P-450 CYP1A1/genetics , Epidermis/metabolism , Estrogen Receptor alpha/biosynthesis , Female , Gene Expression Regulation, Developmental/drug effects , Humans , Integrases/genetics , Intestinal Mucosa/metabolism , Mammary Glands, Animal/metabolism , Mice , Organ Specificity/genetics , Promoter Regions, Genetic/genetics , Recombinant Fusion Proteins/biosynthesis , Signal Transduction/drug effects , Tamoxifen/administration & dosage , rho-Associated Kinases/biosynthesis
5.
Mol Cancer ; 14: 115, 2015 Jun 06.
Article in English | MEDLINE | ID: mdl-26047945

ABSTRACT

BACKGROUND: The expression of the chemokine receptor CCR6 has been previously correlated with higher grades and stages of breast cancer and decreased relapse-free survival. Also, its cognate chemokine ligand CCL20 has been reported to induce proliferation of cultured human breast epithelial cells. METHODS: To establish if CCR6 plays a functional role in mammary tumorigenesis, a bigenic MMTV-PyMT CCR6-null mouse was generated and mammary tumor development was assessed. Levels of tumor-infiltrating immune cells within tumor-bearing mammary glands from MMTV-PyMT Ccr6 (WT) and Ccr6 (-/-) mice were also analyzed. RESULTS: Deletion of CCR6 delayed tumor onset, significantly reduced the extent of initial hyperplastic outgrowth, and decreased tumor incidence in PyMT transgenic mice. CCR6 was then shown to promote the recruitment of pro-tumorigenic macrophages to the tumor site, facilitating the onset of neoplasia. CONCLUSIONS: This study delineated for the first time a role for CCR6 in the development of breast cancer, and demonstrated a critical function for this receptor in maintaining the pro-tumorigenic cancer microenvironment.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , Macrophages/metabolism , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Experimental/metabolism , Mammary Tumor Virus, Mouse/metabolism , Receptors, CCR6/metabolism , Animals , Carcinogenesis/pathology , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Deletion , Macrophages/pathology , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Experimental/pathology , Mice, Inbred C57BL , Mice, Transgenic , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Tumor Microenvironment
6.
J Mammary Gland Biol Neoplasia ; 19(2): 203-11, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24997735

ABSTRACT

Cancer stem cells are believed to be a subset of heterogeneous tumour cells responsible for tumour initiation, growth, local invasion, and metastasis. In breast cancer, numerous factors have been implicated in regulation of cancer stem cells, but there is still a paucity of information regarding precise molecular and cellular mechanisms guiding their pathobiology. Components of both the adaptive and the innate immune system have been shown to play a crucial role in supporting breast cancer growth and spread, and recently some immune mediators, both molecules and cells, have been reported to influence breast cancer stem cell biology. This review summarises a small, pioneering body of evidence for the potentially important function of the "immuniche" in maintaining and supporting breast cancer stem cells.


Subject(s)
Breast Neoplasms/immunology , Immune System/immunology , Mammary Neoplasms, Animal/immunology , Neoplastic Stem Cells/immunology , Animals , Female , Humans
7.
Article in English | MEDLINE | ID: mdl-38979935

ABSTRACT

A key characteristic of cancer cells is their ability to induce changes in their microenvironment that render it permissive to tumor growth, invasion and metastasis. Indeed, these changes are required for tumor progression. Consequently, the tumor microenvironment is emerging as a key source of new targets against cancer, with novel therapies aimed at reversing tumor-promoting changes, reinstating a tumor-hostile microenvironment and suppressing disease progression. RHO-ROCK signaling, and consequent tension within the cellular actomyosin cytoskeleton, regulates a paracrine signaling cascade that establishes a tumor-promoting microenvironment. Here, we show that consistent with our observations in breast cancer, enhanced ROCK activity and consequent production of CRELD2 is associated with the recruitment and tumor-promoting polarization of cancer-associated fibroblasts in cutaneous squamous cell carcinoma. Our observations provide support for the notion that the role of RHO-ROCK signaling in establishing a tumor-promoting microenvironment may be conserved across patients and potentially also different cancer types.

8.
Life Sci Alliance ; 6(10)2023 10.
Article in English | MEDLINE | ID: mdl-37536977

ABSTRACT

Epithelial-mesenchymal transition is essential for tissue patterning and organization. It involves both regulation of cell motility and alterations in the composition and organization of the ECM-a complex environment of proteoglycans and fibrous proteins essential for tissue homeostasis, signaling in response to chemical and biomechanical stimuli, and is often dysregulated under conditions such as cancer, fibrosis, and chronic wounds. Here, we demonstrate that basonuclin-2 (BNC2), a mesenchymal-expressed gene, that is, strongly associated with cancer and developmental defects across genome-wide association studies, is a novel regulator of ECM composition and degradation. We find that at endogenous levels, BNC2 controls the expression of specific collagens, matrix metalloproteases, and other matrisomal components in breast cancer cells, and in fibroblasts that are primarily responsible for the production and processing of the ECM within the tumour microenvironment. In so doing, BNC2 modulates the motile and invasive properties of cancers, which likely explains the association of high BNC2 expression with increasing cancer grade and poor patient prognosis.


Subject(s)
DNA-Binding Proteins , Genome-Wide Association Study , Neoplasms , Humans , Collagen/metabolism , Epithelial-Mesenchymal Transition/genetics , Extracellular Matrix/metabolism , Neoplasms/metabolism , Tumor Microenvironment/genetics , DNA-Binding Proteins/metabolism
9.
Adv Sci (Weinh) ; 9(21): e2103332, 2022 07.
Article in English | MEDLINE | ID: mdl-35611998

ABSTRACT

To fully investigate cellular responses to stimuli and perturbations within tissues, it is essential to replicate the complex molecular interactions within the local microenvironment of cellular niches. Here, the authors introduce Alginate-based tissue engineering (ALTEN), a biomimetic tissue platform that allows ex vivo analysis of explanted tissue biopsies. This method preserves the original characteristics of the source tissue's cellular milieu, allowing multiple and diverse cell types to be maintained over an extended period of time. As a result, ALTEN enables rapid and faithful characterization of perturbations across specific cell types within a tissue. Importantly, using single-cell genomics, this approach provides integrated cellular responses at the resolution of individual cells. ALTEN is a powerful tool for the analysis of cellular responses upon exposure to cytotoxic agents and immunomodulators. Additionally, ALTEN's scalability using automated microfluidic devices for tissue encapsulation and subsequent transport, to enable centralized high-throughput analysis of samples gathered by large-scale multicenter studies, is shown.


Subject(s)
Lab-On-A-Chip Devices , Tissue Engineering , Alginates , Biomimetics , Cell Communication , Tissue Engineering/methods
10.
Small GTPases ; 11(5): 354-370, 2020 09.
Article in English | MEDLINE | ID: mdl-29455593

ABSTRACT

The ability to rapidly respond to applied force underpins cell/tissue homeostasis. This response is mediated by mechanotransduction pathways that regulate remodeling and tension of the actomyosin cytoskeleton to counterbalance external forces. Enhanced extracellular matrix tension hyper-activates mechanotransduction and characterizes diseased states such as cancer, but is also required for normal epidermal regeneration. While the impact of extracellular matrix tension on signaling and cell biology are well appreciated, that of acute compressive force is under-studied. We show here that acute compressive force applied to cells and tissues in a native 3-dimensional context elevates RHOA-GTP levels and increases regulatory myosin phosphorylation, actomyosin contractility and tension via ROCK. In consequence, cell proliferation was increased, as was the expression of regulators of epithelial-mesenchymal transition. Pharmacological inhibition of ROCK abrogated myosin phosphorylation, but not RHOA activation. Our results strongly suggest that acute compressive stress impairs cellular homeostasis in a RHO/ROCK-dependent manner, with implications for disease states such as cancer.


Subject(s)
rho-Associated Kinases/metabolism , Actomyosin/metabolism , Cells, Cultured , HEK293 Cells , Humans , Signal Transduction , Stress, Physiological , rhoA GTP-Binding Protein/metabolism
11.
J Exp Med ; 217(6)2020 06 01.
Article in English | MEDLINE | ID: mdl-32289156

ABSTRACT

Current immunotherapies involving CD8+ T cell responses show remarkable promise, but their efficacy in many solid tumors is limited, in part due to the low frequency of tumor-specific T cells in the tumor microenvironment (TME). Here, we identified a role for host atypical chemokine receptor 4 (ACKR4) in controlling intratumor T cell accumulation and activation. In the absence of ACKR4, an increase in intratumor CD8+ T cells inhibited tumor growth, and nonhematopoietic ACKR4 expression was critical. We show that ACKR4 inhibited CD103+ dendritic cell retention in tumors through regulation of the intratumor abundance of CCL21. In addition, preclinical studies indicate that ACKR4 and CCL21 are potential therapeutic targets to enhance responsiveness to immune checkpoint blockade or T cell costimulation.


Subject(s)
Chemokine CCL21/metabolism , Immunity , Neoplasms/immunology , Receptors, CCR/metabolism , Animals , Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Dendritic Cells/immunology , Disease Models, Animal , Humans , Immune Checkpoint Inhibitors/pharmacology , Integrin alpha Chains/metabolism , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplasms/genetics , Stromal Cells/metabolism , Survival Analysis
12.
Small GTPases ; 7(3): 139-46, 2016 07 02.
Article in English | MEDLINE | ID: mdl-27168253

ABSTRACT

The mechanical properties of the ECM strongly influence the behavior of all cell types within a given tissue. Increased matrix tension promotes epithelial cell proliferation by engaging mitogenic mechanotransduction signaling including the Salvador/Warts/Hippo, PI 3-kinase, Rho, Wnt and MAP kinase pathways. The Rho signaling pathways in particular are capable of increasing intra-cellular tension by elevating the production and contractility of the actomyosin cytoskeleton, which counteracts tension changes within the matrix in a process termed mechano-reciprocity. We have discovered that Rho-ROCK signaling increases the production of ECM through paracrine signaling between the epithelium and fibroblasts and also the remodeling of the ECM by regulating focal adhesion dynamics in fibroblasts. These two phenomena together cause increased ECM tension. Enhanced mechano-reciprocity results in ever-increasing intra- and extra-cellular tension in a vicious cycle that promotes cell proliferation and tumor progression. These insights reveal that inhibiting mechano-reciprocity, reducing ECM tension and targeting cancer-associated fibroblasts in a coordinated fashion has potential as cancer therapy.


Subject(s)
Mechanical Phenomena , Signal Transduction , rho-Associated Kinases/metabolism , Animals , Homeostasis , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL