Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Biol Cell ; : e2400031, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38724262

ABSTRACT

Neutrophils, major regulator of innate immunity have recently emerged as key components of the tumor microenvironment. The role of neutrophils in cancer has been linked to their ability to form neutrophil extracellular traps (NETs), structures composed of decondensed DNA decorated with enzymes that are released into the extracellular space. Here, we discuss the pivotal roles of NETs, in influencing responses to chemotherapy and its severe adverse effect. Highlighting recent insights, we discuss the dual nature of NETs in the context of chemotherapy treatment, examining their potential to either counteract or enhance treatment outcomes. Strategic targeting of NETs emerges as a promising avenue for determining combination therapies that could help counteracting resistance or enhancing chemotherapy efficacy as well as limiting complications due to this type of treatment.

2.
Trends Cancer ; 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38664080

ABSTRACT

Neutrophils, major regulators of innate immunity, have recently emerged as key components of the tumor microenvironment. The role of neutrophils in cancer has been linked to their ability to form neutrophil extracellular traps (NETs), structures composed of decondensed DNA decorated with enzymes that are released into the extracellular space. Here, we discuss the pivotal roles of NETs in influencing responses to anticancer therapies such as chemotherapy, radiotherapy, immunotherapy, and targeted therapy. Highlighting recent insights, we delve into the dual nature of NETs in the context of anticancer treatments, examining their potential to either counteract or enhance treatment outcomes. Strategic targeting of NETs may be a promising avenue for crafting combination therapies to counteract resistance or enhance anticancer treatments' efficacy.

3.
J Clin Invest ; 134(5)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38426501

ABSTRACT

Chemotherapy, which primarily acts on cancer cells, can influence the tumor microenvironment and the recruitment and behavior of stromal cells. In this issue of the JCI, Li et al. explored the potent anticancer effect of the combination of a glutaminase inhibitor (CB-839) and 5-FU against PIK3CA-mutant colorectal cancer tumors. This chemotherapy treatment strongly induced the recruitment of neutrophils that formed neutrophil extracellular traps in cancer, which actively killed cancer cells by inducing apoptosis. This study substantially advances our understanding of the multifaceted role of neutrophils and NETs in the outcome of anticancer treatment.


Subject(s)
Colorectal Neoplasms , Extracellular Traps , Humans , Neutrophils , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Apoptosis , Tumor Microenvironment
4.
Cancer Cell ; 42(3): 474-486.e12, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-38402610

ABSTRACT

Chronic stress is associated with increased risk of metastasis and poor survival in cancer patients, yet the reasons are unclear. We show that chronic stress increases lung metastasis from disseminated cancer cells 2- to 4-fold in mice. Chronic stress significantly alters the lung microenvironment, with fibronectin accumulation, reduced T cell infiltration, and increased neutrophil infiltration. Depleting neutrophils abolishes stress-induced metastasis. Chronic stress shifts normal circadian rhythm of neutrophils and causes increased neutrophil extracellular trap (NET) formation via glucocorticoid release. In mice with neutrophil-specific glucocorticoid receptor deletion, chronic stress fails to increase NETs and metastasis. Furthermore, digesting NETs with DNase I prevents chronic stress-induced metastasis. Together, our data show that glucocorticoids released during chronic stress cause NET formation and establish a metastasis-promoting microenvironment. Therefore, NETs could be targets for preventing metastatic recurrence in cancer patients, many of whom will experience chronic stress due to their disease.


Subject(s)
Extracellular Traps , Lung Neoplasms , Humans , Animals , Mice , Neutrophils/pathology , Lung Neoplasms/pathology , Lung/pathology , Tumor Microenvironment
6.
Cancer Cell ; 41(4): 757-775.e10, 2023 04 10.
Article in English | MEDLINE | ID: mdl-37037615

ABSTRACT

Metastasis is the major cause of cancer death, and the development of therapy resistance is common. The tumor microenvironment can confer chemotherapy resistance (chemoresistance), but little is known about how specific host cells influence therapy outcome. We show that chemotherapy induces neutrophil recruitment and neutrophil extracellular trap (NET) formation, which reduces therapy response in mouse models of breast cancer lung metastasis. We reveal that chemotherapy-treated cancer cells secrete IL-1ß, which in turn triggers NET formation. Two NET-associated proteins are required to induce chemoresistance: integrin-αvß1, which traps latent TGF-ß, and matrix metalloproteinase 9, which cleaves and activates the trapped latent TGF-ß. TGF-ß activation causes cancer cells to undergo epithelial-to-mesenchymal transition and correlates with chemoresistance. Our work demonstrates that NETs regulate the activities of neighboring cells by trapping and activating cytokines and suggests that chemoresistance in the metastatic setting can be reduced or prevented by targeting the IL-1ß-NET-TGF-ß axis.


Subject(s)
Breast Neoplasms , Drug Resistance, Neoplasm , Extracellular Traps , Lung Neoplasms , Neutrophils , Tumor Microenvironment , Neutrophils/metabolism , Neutrophils/pathology , Humans , Animals , Mice , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Neoplasm Metastasis , Extracellular Traps/metabolism , Inflammation/pathology
7.
Cell ; 186(7): 1432-1447.e17, 2023 03 30.
Article in English | MEDLINE | ID: mdl-37001503

ABSTRACT

Cancer immunotherapies, including adoptive T cell transfer, can be ineffective because tumors evolve to display antigen-loss-variant clones. Therapies that activate multiple branches of the immune system may eliminate escape variants. Here, we show that melanoma-specific CD4+ T cell therapy in combination with OX40 co-stimulation or CTLA-4 blockade can eradicate melanomas containing antigen escape variants. As expected, early on-target recognition of melanoma antigens by tumor-specific CD4+ T cells was required. Surprisingly, complete tumor eradication was dependent on neutrophils and partly dependent on inducible nitric oxide synthase. In support of these findings, extensive neutrophil activation was observed in mouse tumors and in biopsies of melanoma patients treated with immune checkpoint blockade. Transcriptomic and flow cytometry analyses revealed a distinct anti-tumorigenic neutrophil subset present in treated mice. Our findings uncover an interplay between T cells mediating the initial anti-tumor immune response and neutrophils mediating the destruction of tumor antigen loss variants.


Subject(s)
Melanoma , T-Lymphocytes , Mice , Animals , T-Lymphocytes/pathology , Neutrophils/pathology , Antigenic Drift and Shift , Immunotherapy , CTLA-4 Antigen
8.
Cancer Res ; 82(9): 1774-1788, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35502542

ABSTRACT

Fibroblastic reticular cells (FRC) are immunologically specialized myofibroblasts that control the elasticity of the lymph node, in part through their contractile properties. Swelling of tumor-draining lymph nodes is a hallmark of lymphophilic cancers such as cutaneous melanoma. Melanoma displays high intratumoral heterogeneity with the coexistence of melanoma cells with variable differentiation phenotypes from melanocytic to dedifferentiated states. Factors secreted by melanoma cells promote premetastatic lymph node reprograming and tumor spreading. Elucidating the impact of the melanoma secretome on FRC could help identify approaches to prevent metastasis. Here we show that melanocytic and dedifferentiated melanoma cells differentially impact the FRC contractile phenotype. Factors secreted by dedifferentiated cells, but not by melanocytic cells, strongly inhibited actomyosin-dependent contractile forces of FRC by decreasing the activity of the RHOA-RHO-kinase (ROCK) pathway and the mechano-responsive transcriptional coactivator Yes1 associated transcriptional regulator (YAP). Transcriptional profiling and biochemical analyses indicated that actomyosin cytoskeleton relaxation in FRC is driven by inhibition of the JAK1-STAT3 pathway. This FRC relaxation was associated with increased FRC proliferation and activation and with elevated tumor invasion in vitro. The secretome of dedifferentiated melanoma cells also modulated the biomechanical properties of distant lymph node in premetastatic mouse models. Finally, IL1 produced by dedifferentiated cells was involved in the inhibition of FRC contractility. These data highlight the role of the JAK1-STAT3 and YAP pathways in spontaneous contractility of resting FRC. They also suggest that dedifferentiated melanoma cells specifically target FRC biomechanical properties to favor tumor spreading in the premetastatic lymph node niche. Targeting this remote communication could be an effective strategy to prevent metastatic spread of the disease. SIGNIFICANCE: Communication between dedifferentiated melanoma cells and lymph node fibroblasts reprograms the biomechanical properties of the premetastatic lymph node niche to promote tumor invasion. See related commentary by Lund, p. 1692.


Subject(s)
Melanoma , Skin Neoplasms , Actomyosin/metabolism , Animals , Fibroblasts/metabolism , Humans , Interleukin-1 , Janus Kinase 1/metabolism , Lymph Nodes/pathology , Melanoma/pathology , Mice , STAT3 Transcription Factor/metabolism , Skin Neoplasms/pathology
9.
Science ; 361(6409)2018 09 28.
Article in English | MEDLINE | ID: mdl-30262472

ABSTRACT

Cancer cells from a primary tumor can disseminate to other tissues, remaining dormant and clinically undetectable for many years. Little is known about the cues that cause these dormant cells to awaken, resume proliferating, and develop into metastases. Studying mouse models, we found that sustained lung inflammation caused by tobacco smoke exposure or nasal instillation of lipopolysaccharide converted disseminated, dormant cancer cells to aggressively growing metastases. Sustained inflammation induced the formation of neutrophil extracellular traps (NETs), and these were required for awakening dormant cancer. Mechanistic analysis revealed that two NET-associated proteases, neutrophil elastase and matrix metalloproteinase 9, sequentially cleaved laminin. The proteolytically remodeled laminin induced proliferation of dormant cancer cells by activating integrin α3ß1 signaling. Antibodies against NET-remodeled laminin prevented awakening of dormant cells. Therapies aimed at preventing dormant cell awakening could potentially prolong the survival of cancer patients.


Subject(s)
Carcinogenesis/metabolism , Extracellular Traps/enzymology , Lamins/metabolism , Lung Neoplasms/pathology , Neutrophils/enzymology , Pneumonia/pathology , Animals , DNA/metabolism , Humans , Inflammation/chemically induced , Inflammation/microbiology , Integrin alpha3beta1/metabolism , Leukocyte Elastase/metabolism , Lipopolysaccharides , Lung/pathology , MCF-7 Cells , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/pathology , Pneumonia/chemically induced , Pneumonia/microbiology , Pneumonia, Bacterial/etiology , Pneumonia, Bacterial/pathology , Protein-Arginine Deiminase Type 4 , Protein-Arginine Deiminases/antagonists & inhibitors , Protein-Arginine Deiminases/metabolism , Proteolysis , Rats , Signal Transduction , Smoking , Nicotiana
10.
Science ; 360(6394)2018 06 15.
Article in English | MEDLINE | ID: mdl-29773669

ABSTRACT

The majority of patients with pancreatic ductal adenocarcinoma (PDA) develop metastatic disease after resection of their primary tumor. We found that livers from patients and mice with PDA harbor single disseminated cancer cells (DCCs) lacking expression of cytokeratin 19 (CK19) and major histocompatibility complex class I (MHCI). We created a mouse model to determine how these DCCs develop. Intraportal injection of immunogenic PDA cells into preimmunized mice seeded livers only with single, nonreplicating DCCs that were CK19- and MHCI- The DCCs exhibited an endoplasmic reticulum (ER) stress response but paradoxically lacked both inositol-requiring enzyme 1α activation and expression of the spliced form of transcription factor XBP1 (XBP1s). Inducible expression of XBP1s in DCCs, in combination with T cell depletion, stimulated the outgrowth of macrometastatic lesions that expressed CK19 and MHCI. Thus, unresolved ER stress enables DCCs to escape immunity and establish latent metastases.


Subject(s)
Carcinoma, Pancreatic Ductal/secondary , Endoplasmic Reticulum Stress/immunology , Liver Neoplasms/secondary , Pancreatic Neoplasms/pathology , Tumor Escape , Animals , Carcinoma, Pancreatic Ductal/immunology , Endoribonucleases/genetics , Endoribonucleases/metabolism , Genes, MHC Class I , Genetic Engineering , Humans , Keratin-19/metabolism , Liver Neoplasms/immunology , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/secondary , Pancreatic Neoplasms/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes/immunology , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
11.
Dev Cell ; 43(4): 379-380, 2017 11 20.
Article in English | MEDLINE | ID: mdl-29161586

ABSTRACT

Neutrophil extracellular traps (NETs) are critical for the clearance of large pathogens and are also implicated in thrombosis, autoimmunity, and cancer. In this issue of Developmental Cell, Amulic et al. (2017) show that the terminally differentiated, non-cycling neutrophils repurpose cell-cycle proteins and pathways to form NETs.


Subject(s)
Autoimmunity/immunology , Cell Cycle/physiology , Cyclins/metabolism , Proliferating Cell Nuclear Antigen/immunology , Extracellular Traps/metabolism , Humans , Neutrophil Activation/immunology , Thrombosis/immunology , Thrombosis/metabolism
12.
Oncotarget ; 8(1): 1304-1320, 2017 Jan 03.
Article in English | MEDLINE | ID: mdl-27901489

ABSTRACT

Acto-myosin contractility in carcinoma-associated fibroblasts leads to assembly of the tumor extracellular matrix. The pro-inflammatory cytokine LIF governs fibroblast activation in cancer by regulating the myosin light chain 2 activity. So far, however, how LIF mediates cytoskeleton contractility remains unknown. Using phenotypic screening assays based on knock-down of LIF-dependent genes in fibroblasts, we identified the glycoprotein ICAM-1 as a crucial regulator of stroma fibroblast proinvasive matrix remodeling. We demonstrate that the membrane-bound ICAM-1 isoform is necessary and sufficient to promote inflammation-dependent extracellular matrix contraction, which favors cancer cell invasion. Indeed, ICAM-1 mediates generation of acto-myosin contractility downstream of the Src kinases in stromal fibroblasts. Moreover, acto-myosin contractility regulates ICAM-1 expression by establishing a positive feedback signaling. Thus, targeting stromal ICAM-1 might constitute a possible therapeutic mean to counteract tumor cell invasion and dissemination.


Subject(s)
Actomyosin/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Intercellular Adhesion Molecule-1/metabolism , Actomyosin/genetics , Cells, Cultured , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Head and Neck Neoplasms/genetics , Humans , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Signal Transduction , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Microenvironment
13.
Nat Commun ; 6: 10204, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26667266

ABSTRACT

Carcinoma-associated fibroblasts (CAF) mediate the onset of a proinvasive tumour microenvironment. The proinflammatory cytokine LIF reprograms fibroblasts into a proinvasive phenotype, which promotes extracellular matrix remodelling and collective invasion of cancer cells. Here we unveil that exposure to LIF initiates an epigenetic switch leading to the constitutive activation of JAK1/STAT3 signalling, which results in sustained proinvasive activity of CAF. Mechanistically, p300-histone acetyltransferase acetylates STAT3, which, in turn, upregulates and activates the DNMT3b DNA methyltransferase. DNMT3b methylates CpG sites of the SHP-1 phosphatase promoter, which abrogates SHP-1 expression, and results in constitutive phosphorylation of JAK1. Sustained JAK1/STAT3 signalling is maintained by DNA methyltransferase DNMT1. Consistently, in human lung and head and neck carcinomas, STAT3 acetylation and phosphorylation are inversely correlated with SHP-1 expression. Combined inhibition of DNMT activities and JAK signalling, in vitro and in vivo, results in long-term reversion of CAF-associated proinvasive activity and restoration of the wild-type fibroblast phenotype.


Subject(s)
Carcinogenesis/metabolism , Epigenomics , Fibroblasts/physiology , Neoplasms/metabolism , Animals , Antibodies, Neutralizing , Carcinogenesis/genetics , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/metabolism , RNA Interference
14.
Mol Cell ; 59(4): 664-76, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-26236014

ABSTRACT

The most critical stage in initiation of melanoma metastasis is the radial to vertical growth transition, yet the triggers of this transition remain elusive. We suggest that the microenvironment drives melanoma metastasis independently of mutation acquisition. Here we examined the changes in microenvironment that occur during melanoma radial growth. We show that direct contact of melanoma cells with the remote epidermal layer triggers vertical invasion via Notch signaling activation, the latter serving to inhibit MITF function. Briefly, within the native Notch ligand-free microenvironment, MITF, the melanocyte lineage master regulator, binds and represses miR-222/221 promoter in an RBPJK-dependent manner. However, when radial growth brings melanoma cells into contact with distal differentiated keratinocytes that express Notch ligands, the activated Notch intracellular domain impairs MITF binding to miR-222/221 promoter. This de-repression of miR-222/221 expression triggers initiation of invasion. Our findings may direct melanoma prevention opportunities via targeting specific microenvironments.


Subject(s)
Keratinocytes/physiology , Melanoma, Experimental/secondary , Microphthalmia-Associated Transcription Factor/metabolism , Skin Neoplasms/pathology , Animals , Base Sequence , Binding Sites , Cell Communication , Cell Line, Tumor , Coculture Techniques , Gene Expression Regulation, Neoplastic , Melanoma, Experimental/metabolism , Mice, Inbred NOD , Mice, SCID , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Invasiveness , Neoplasm Transplantation , Promoter Regions, Genetic , RNA Interference , Receptors, Notch/metabolism , Signal Transduction , Skin Neoplasms/metabolism
15.
Oncotarget ; 6(28): 24636-48, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26284589

ABSTRACT

BASIGIN/CD147/EMMPRIN is a multifunctional transmembrane glycoprotein strongly expressed in tumours. BASIGIN controls tumour metabolism, particularly glycolysis by facilitating lactic acid export through the two monocarboxylate transporters MCT1 and hypoxia-inducible MCT4. However, before being recognized as a co-carrier of MCTs, BASIGIN was described as an inducer of extracellular matrix metalloproteases (MMPs). Early on, a model emerged in which, tumour cells use the extracellular domain of BASIGIN to recognize and stimulate neighbouring fibroblasts to produce MMPs. However, this model has remained hypothetical since a direct link between BASIGIN and MMPs production has not yet been clearly established. To validate the BASIGIN/MMP hypothesis, we developed BASIGIN knockouts in three human tumour cell lines derived from glioma, colon, and lung adenocarcinoma. By using co-culture experiments of either human or mouse fibroblasts and tumour cell lines we showed, contrary to what has been abundantly published, that the disruption of BASIGIN in tumour cells and in MEFs has no action on the production of MMPs. Our findings do not support the notion that the pro-tumoural action of BASIGIN is mediated via induction of MMPs. Therefore, we propose that to date, the strongest pro-tumoural action of BASIGIN is mediated through the control of fermentative glycolysis.


Subject(s)
Basigin/metabolism , Fibroblasts/enzymology , Gene Knockout Techniques , Matrix Metalloproteinases/metabolism , Monocarboxylic Acid Transporters/metabolism , Muscle Proteins/metabolism , Neoplasms/enzymology , Neoplasms/metabolism , Symporters/metabolism , Animals , Basigin/genetics , Cell Line, Tumor , Coculture Techniques , Culture Media, Conditioned/metabolism , Down-Regulation , Endonucleases/metabolism , Gene Expression Regulation, Neoplastic , Humans , Matrix Metalloproteinases/genetics , Mice , Neoplasms/genetics , Paracrine Communication , Signal Transduction , Transfection , Zinc Fingers
16.
Nat Commun ; 5: 4255, 2014 Jun 25.
Article in English | MEDLINE | ID: mdl-24963846

ABSTRACT

Rounded-amoeboid cancer cells use actomyosin contractility driven by Rho-ROCK and JAK-STAT3 to migrate efficiently. It has been suggested that rounded-amoeboid cancer cells do not require matrix metalloproteinases (MMPs) to invade. Here we compare MMP levels in rounded-amoeboid and elongated-mesenchymal melanoma cells. Surprisingly, we find that rounded-amoeboid melanoma cells secrete higher levels of several MMPs, including collagenase MMP-13 and gelatinase MMP-9. As a result, rounded-amoeboid melanoma cells degrade collagen I more efficiently than elongated-mesenchymal cells. Furthermore, using a non-catalytic mechanism, MMP-9 promotes rounded-amoeboid 3D migration through regulation of actomyosin contractility via CD44 receptor. MMP-9 is upregulated in a panel of rounded-amoeboid compared with elongated-mesenchymal melanoma cell lines and its levels are controlled by ROCK-JAK-STAT3 signalling. MMP-9 expression increases during melanoma progression and it is particularly prominent in the invasive fronts of lesions, correlating with cell roundness. Therefore, rounded-amoeboid cells use both catalytic and non-catalytic activities of MMPs for invasion.


Subject(s)
Actomyosin/metabolism , Cell Movement , Janus Kinases/metabolism , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 9/metabolism , Melanoma/metabolism , STAT3 Transcription Factor/metabolism , rho-Associated Kinases/metabolism , Cell Line, Tumor , Humans , Melanoma/pathology , Neoplasm Invasiveness , Signal Transduction
17.
Med Sci (Paris) ; 30(4): 391-7, 2014 Apr.
Article in French | MEDLINE | ID: mdl-24801033

ABSTRACT

Cellular and molecular crosstalks between cancer and non-cancer tumor-associated cells result in tumor growth and metastatic spreading. During carcinoma development, tumor cells secrete signaling molecules that influence the surrounding non-cancer cells, which, in return, favor tumor cell growth, survival, migration and metastasis. Carcinoma-associated fibroblasts (CAF) are the most abundant population of non-cancer cells found in tumors, and their presence is often associated with poor clinical prognosis. Here, we summarize the pro-carcinogenic roles of CAF cells during carcinogenesis, with a specific focus on their abilities to drive cancer cell-dependent pro-invasive extracellular matrix remodeling.


Subject(s)
Fibroblasts , Neoplasm Invasiveness/pathology , Neoplasms/pathology , Tumor Microenvironment , Humans
18.
Cell Rep ; 7(5): 1664-1678, 2014 Jun 12.
Article in English | MEDLINE | ID: mdl-24857661

ABSTRACT

Signaling crosstalk between tumor cells and fibroblasts confers proinvasive properties to the tumor microenvironment. Here, we identify leukemia inhibitory factor (LIF) as a tumor promoter that mediates proinvasive activation of stromal fibroblasts independent of alpha-smooth muscle actin (α-SMA) expression. We demonstrate that a pulse of transforming growth factor ß (TGF-ß) establishes stable proinvasive fibroblast activation by inducing LIF production in both fibroblasts and tumor cells. In fibroblasts, LIF mediates TGF-ß-dependent actomyosin contractility and extracellular matrix remodeling, which results in collective carcinoma cell invasion in vitro and in vivo. Accordingly, carcinomas from multiple origins and melanomas display strong LIF upregulation, which correlates with dense collagen fiber organization, cancer cell collective invasion, and poor clinical outcome. Blockade of JAK activity by Ruxolitinib (JAK inhibitor) counteracts fibroblast-dependent carcinoma cell invasion in vitro and in vivo. These findings establish LIF as a proinvasive fibroblast producer independent of α-SMA and may open novel therapeutic perspectives for patients with aggressive primary tumors.


Subject(s)
Carcinogenesis/metabolism , Carcinoma/metabolism , Fibroblasts/metabolism , Leukemia Inhibitory Factor/metabolism , Melanoma/metabolism , Tumor Microenvironment , Actins/genetics , Actins/metabolism , Actomyosin/metabolism , Animals , Carcinogenesis/pathology , Carcinoma/pathology , Cell Line, Tumor , Cell Movement , Extracellular Matrix/metabolism , Female , Fibroblasts/drug effects , Humans , Janus Kinases/antagonists & inhibitors , Leukemia Inhibitory Factor/genetics , Melanoma/pathology , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Nitriles , Pyrazoles/pharmacology , Pyrimidines , Signal Transduction , Transforming Growth Factor beta/pharmacology , Up-Regulation
19.
Methods Mol Biol ; 961: 243-52, 2013.
Article in English | MEDLINE | ID: mdl-23325648

ABSTRACT

Cancer cell invasion and dissemination from primary tumors are complex multistep mechanisms which remain poorly understood. It is now clear that cancer cells can adapt their mode of invasion to the signalling provided by the surrounding stroma. Single and collective cancer cell invasion are the two invasion features most currently observed and described by pathologists. Here we describe a three-dimensional organotypic assay that allows the study of squamous cell carcinoma cell collective invasion induced by the carcinoma associated fibroblasts. This model preserves the relationship between epithelial and mesenchymal cells, which are observed in vivo, and allows to decipher the molecular and cellular mechanisms involving the tumor and its stromal microenvironment. This three-dimensional model of invasion provides an invaluable tool to gain major insights in the understanding of tumor cell dissemination.


Subject(s)
Carcinoma, Squamous Cell/pathology , Coculture Techniques/methods , Fibroblasts/pathology , Keratinocytes/pathology , Neoplasm Invasiveness/pathology , Skin/pathology , Cell Line, Tumor , Cells, Cultured , Coculture Techniques/instrumentation , Humans , Models, Biological , Tissue Culture Techniques/instrumentation , Tissue Culture Techniques/methods , Tumor Microenvironment
20.
Cancer Cell ; 20(2): 229-45, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21840487

ABSTRACT

Proinflammatory cytokines are frequently observed in the tumor microenvironment, and chronic inflammation is involved in cancer initiation and progression. We show that cytokine signaling through the receptor subunit GP130-IL6ST and the kinase JAK1 generates actomyosin contractility through Rho-kinase dependent signaling. This pathway generates contractile force in stromal fibroblasts to remodel the extracellular matrix to create tracks for collective migration of squamous carcinoma cells and provides the high levels of actomyosin contractility required for migration of individual melanoma cells in the rounded, "amoeboid" mode. Thus, cytokine signaling can generate actomyosin contractility in both stroma and tumor cells. Strikingly, actomyosin contractility itself positively modulates activity of the transcription factor STAT3 downstream of JAK1, demonstrating positive feedback within the signaling network.


Subject(s)
Actomyosin/metabolism , Janus Kinase 1/metabolism , Neoplasms/metabolism , Signal Transduction , Stromal Cells/metabolism , rho-Associated Kinases/metabolism , Cell Movement , Humans , Melanoma/metabolism , Melanoma/pathology , Neoplasms/pathology , STAT3 Transcription Factor/metabolism , Stromal Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...