Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Int J Mol Sci ; 24(10)2023 May 12.
Article in English | MEDLINE | ID: mdl-37240029

ABSTRACT

Despite advances in treatment for metastatic melanoma patients, patients with liver metastasis have an unfavorable prognosis. A better understanding of the development of liver metastasis is needed. The multifunctional cytokine Transforming Growth Factor ß (TGF-ß) plays various roles in melanoma tumors and metastasis, affecting both tumor cells and cells from the surrounding tumor microenvironment. To study the role of TGF-ß in melanoma liver metastasis, we created a model to activate or repress the TGF-ß receptor pathway in vitro and in vivo in an inducible manner. For this, we engineered B16F10 melanoma cells to have inducible ectopic expression of a constitutively active (ca) or kinase-inactive (ki) TGF-ß receptor I, also termed activin receptor-like kinase (ALK5). In vitro, stimulation with TGF-ß signaling and ectopic caALK5 expression reduced B16F10 cell proliferation and migration. Contrasting results were found in vivo; sustained caALK5 expression in B16F10 cells in vivo increased the metastatic outgrowth in liver. Blocking microenvironmental TGF-ß did not affect metastatic liver outgrowth of both control and caALK5 expressing B16F10 cells. Upon characterizing the tumor microenvironment of control and caALk5 expressing B16F10 tumors, we observed reduced (cytotoxic) T cell presence and infiltration, as well as an increase in bone marrow-derived macrophages in caALK5 expressing B16F10 tumors. This suggests that caALK5 expression in B16F10 cells induces changes in the tumor microenvironment. A comparison of newly synthesized secreted proteins upon caALK5 expression by B16F10 cells revealed increased secretion of matrix remodeling proteins. Our results show that TGF-ß receptor activation in B16F10 melanoma cells can increase metastatic outgrowth in liver in vivo, possibly through remodeling of the tumor microenvironment leading to altered infiltration of immune cells. These results provide insights in the role of TGF-ß signaling in B16F10 liver metastasis and could have implications regarding the use of TGF-ß inhibitors for the treatment of melanoma patients with liver metastasis.


Subject(s)
Liver Neoplasms , Melanoma , Humans , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/metabolism , Cytokines , Transforming Growth Factor beta/metabolism , Cell Line, Tumor , Tumor Microenvironment
2.
Front Oncol ; 12: 797453, 2022.
Article in English | MEDLINE | ID: mdl-35756604

ABSTRACT

Cervical cancer is the fourth most common cancer in women worldwide. Squamous cell carcinoma (SCC) and adenocarcinoma (AC) are the most common histological types, with AC patients having worse prognosis. Over the last two decades, incidence rates of AC have increased, highlighting the importance of further understanding AC tumorigenesis, and the need to investigate new treatment options. The cytokine TGF-ß functions as a tumour suppressor in healthy tissue. However, in tumour cells this suppressive function can be overcome. Therefore there is an increasing interest in using TGF-ß inhibitors in the treatment of cancer. Here, we hypothesize that TGF-ß plays a different role in SCC and AC. Analysis of RNA-seq data from the TCGA, using a TGF-ß response signature, resulted in separate clustering of the two subtypes. We further investigated the expression of TGF-ß-signalling related proteins (TßR1/2, SMAD4, pSMAD2, PAI-1, αvß6 and MMP2/9) in a cohort of 62 AC patients. Low TßR2 and SMAD4 expression was associated with worse survival in AC patients and interestingly, high PAI-1 and αvß6 expression was also correlated with worse survival. Similar correlations of TßR2, PAI-1 and αvß6 with clinical parameters were found in previously reported SCC analyses. However, when comparing expression levels between SCC and AC patient samples, pSMAD2, SMAD4, PAI-1 and αvß6 showed lower expression in AC compared to SCC. Because of the low expression of core TßR1/2, (p-)SMAD2 and SMAD4 proteins and the correlation with worse prognosis, TGF-ß pathway most likely leads to tumour inhibitory effects in AC and therefore the use of TGF-ß inhibitors would not be recommended. However, given the correlation of PAI-1 and αvß6 with poor prognosis, the use of TGF- ß inhibitors might be of interest in SCC and in the subsets of AC patients with high expression of these TGF-ß associated proteins.

3.
Mol Cancer Res ; 20(10): 1516-1531, 2022 10 04.
Article in English | MEDLINE | ID: mdl-35731212

ABSTRACT

Patients with bladder cancer often have a poor prognosis due to the highly invasive and metastatic characteristics of bladder cancer cells. Epithelial-to-mesenchymal transition (EMT) has been causally linked to bladder cancer invasion. The E3 ubiquitin ligase, tumor necrosis factor receptor-associated factor 4 (TRAF4) has been implicated as a tumor promoter in a wide range of cancers. In contrast, here we show that low TRAF4 expression is associated with poor overall survival in patients with bladder cancer. We show that the TRAF4 gene is epigenetically silenced and that ERK mediates TRAF4 phosphorylation, resulting in lower TRAF4 protein levels in bladder cancer cells. In addition, we demonstrate that TRAF4 is inversely correlated with an EMT gene signature/protein marker expression. Functionally, by manipulating TRAF4 expression, we show that TRAF4 regulates EMT genes and epithelial and invasive properties in bladder cancer cells. Transcriptomic analysis of dysregulated TRAF4 expression in bladder cancer cell lines revealed that high TRAF4 expression enhances the bone morphogenetic protein (BMP)/SMAD and inhibits the NF-κB signaling pathway. Mechanistically, we show that TRAF4 targets the E3 ubiquitin ligase SMURF1, a negative regulator of BMP/SMAD signaling, for proteasomal degradation in bladder cancer cells. This was corroborated in patient samples where TRAF4 positively correlates with phospho-SMAD1/5, and negatively correlates with phospho-NFκb-p65. Lastly, we show that genetic and pharmacologic inhibition of SMURF1 inhibits the migration of aggressive mesenchymal bladder cancer cells. IMPLICATIONS: Our findings identify E3 ubiquitin ligase TRAF4 as a potential therapeutic target or biomarker for bladder cancer progression.


Subject(s)
TNF Receptor-Associated Factor 4 , Urinary Bladder Neoplasms , Bone Morphogenetic Proteins/metabolism , Carcinogens , Humans , NF-kappa B/metabolism , Signal Transduction , TNF Receptor-Associated Factor 4/genetics , TNF Receptor-Associated Factor 4/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Urinary Bladder Neoplasms/genetics
4.
Cancers (Basel) ; 14(10)2022 May 19.
Article in English | MEDLINE | ID: mdl-35626109

ABSTRACT

Transforming growth factor-ß (TGF-ß) signaling is tightly controlled in duration and intensity during embryonic development and in the adult to maintain tissue homeostasis. To visualize the TGF-ß/SMAD3 signaling kinetics, we developed a dynamic TGF-ß/SMAD3 transcriptional fluorescent reporter using multimerized SMAD3/4 binding elements driving the expression of a quickly folded and highly unstable GFP protein. We demonstrate the specificity and sensitivity of this reporter and its wide application to monitor dynamic TGF-ß/SMAD3 transcriptional responses in both 2D and 3D systems in vitro, as well as in vivo, using live-cell and intravital imaging. Using this reporter in B16F10 cells, we observed single cell heterogeneity in response to TGF-ß challenge, which can be categorized into early, late, and non-responders. Because of its broad application potential, this reporter allows for new discoveries into how TGF-ß/SMAD3-dependent transcriptional dynamics are affected during multistep and reversible biological processes.

5.
Methods Mol Biol ; 2488: 47-65, 2022.
Article in English | MEDLINE | ID: mdl-35347682

ABSTRACT

Epithelial to mesenchymal transition (EMT) is crucial during embryonic development, tissue fibrosis, and cancer progression. Epithelial cells that display a cobblestone-like morphology can undergo a switch to mesenchymal-like phenotype, displaying an elongated spindle shape or a fibroblast-like morphology. EMT is characterized by timely and reversible alterations of molecular and cellular processes. The changes include loss of epithelial and gain of mesenchymal marker expression, loss of polarity, increased cell migratory and invasive properties. Epithelial cells can progress unevenly during this transition and attain hybrid E/M states or metastable EMT states, referred to as epithelial cell plasticity. To gain a deeper insight into the mechanism of EMT, understanding the dynamic aspects of this process is essential. One of the most prominent factors to induce EMT is the cytokine transforming growth factor-ß (TGF-ß). This chapter discusses molecular and cellular techniques to monitor TGF-ß-induced signaling and EMT changes in normal and cancer cell lines. These methods include measuring the TGF-ß-induced activation of its intracellular SMAD effectors proteins and changes in epithelial/mesenchymal marker expression and localization. Moreover, we describe assays of cell migration and dynamic reorganization of the actin cytoskeleton and stress filaments that are frequently part of the TGF-ß-induced EMT cellular response.


Subject(s)
Epithelial-Mesenchymal Transition , Transforming Growth Factor beta , Epithelial Cells , Epithelial-Mesenchymal Transition/genetics , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology
6.
NPJ Breast Cancer ; 7(1): 140, 2021 Oct 27.
Article in English | MEDLINE | ID: mdl-34707097

ABSTRACT

Reactivation of dormant cancer cells can lead to cancer relapse, metastasis, and patient death. Dormancy is a nonproliferative state and is linked to late relapse and death. No targeted therapy is currently available to eliminate dormant cells, highlighting the need for a deeper understanding and reliable models. Here, we thoroughly characterize the dormant D2.OR and ZR-75-1, and proliferative D2A1 breast cancer cell line models in vivo and/or in vitro, and assess if there is overlap between a dormant and a senescent phenotype. We show that D2.OR but not D2A1 cells become dormant in the liver of an immunocompetent model. In vitro, we show that D2.OR and ZR-75-1 cells in response to a 3D environment or serum-free conditions are growth-arrested in G1, of which a subpopulation resides in a 4NG1 state. The dormancy state is reversible and not associated with a senescence phenotype. This will aid future research on breast cancer dormancy.

7.
Curr Protoc ; 1(5): e116, 2021 May.
Article in English | MEDLINE | ID: mdl-33961349

ABSTRACT

The liver is a frequent site of cancer metastasis, but current treatment options for cancer patients with liver metastasis are limited, resulting in poor prognosis. Colonization of the liver by cancer cells is a multistep and temporally controlled process. Investigating this process in biological relevant settings in a dynamic manner may lead to new therapeutic avenues. Experimental mouse models of liver metastasis combined with high-resolution microscopy methods can facilitate study of the mechanisms that underlie the outgrowth of cancer cells in the liver. Intravital imaging can provide information on the behavior of tumor cells in their biological setting, in time frames of hours to days. In this unit, we describe the experimental induction of liver metastasis through administration of cancer cells into mice via mesenteric vein injection. The behavior of these injected cells can then be studied using intravital imaging by surgical exposure or through an abdominal imaging window. The approach is described for use with an upright multiphoton microscope, making it widely applicable. © 2021 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Inducing liver metastasis through mesenteric vein injection Basic Protocol 2: Short-term imaging of tumor cells in mouse liver Basic Protocol 3: Long-term imaging of tumor cells in mouse liver using an abdominal imaging window.


Subject(s)
Intravital Microscopy , Liver Neoplasms , Animals , Diagnostic Imaging , Disease Models, Animal , Humans , Mice
8.
Cancer Discov ; 11(3): 678-695, 2021 03.
Article in English | MEDLINE | ID: mdl-33203734

ABSTRACT

Circulating tumor cells (CTC) are shed by cancer into the bloodstream, where a viable subset overcomes oxidative stress to initiate metastasis. We show that single CTCs from patients with melanoma coordinately upregulate lipogenesis and iron homeostasis pathways. These are correlated with both intrinsic and acquired resistance to BRAF inhibitors across clonal cultures of BRAF-mutant CTCs. The lipogenesis regulator SREBP2 directly induces transcription of the iron carrier Transferrin (TF), reducing intracellular iron pools, reactive oxygen species, and lipid peroxidation, thereby conferring resistance to inducers of ferroptosis. Knockdown of endogenous TF impairs tumor formation by melanoma CTCs, and their tumorigenic defects are partially rescued by the lipophilic antioxidants ferrostatin-1 and vitamin E. In a prospective melanoma cohort, presence of CTCs with high lipogenic and iron metabolic RNA signatures is correlated with adverse clinical outcome, irrespective of treatment regimen. Thus, SREBP2-driven iron homeostatic pathways contribute to cancer progression, drug resistance, and metastasis. SIGNIFICANCE: Through single-cell analysis of primary and cultured melanoma CTCs, we have uncovered intrinsic cancer cell heterogeneity within lipogenic and iron homeostatic pathways that modulates resistance to BRAF inhibitors and to ferroptosis inducers. Activation of these pathways within CTCs is correlated with adverse clinical outcome, pointing to therapeutic opportunities.This article is highlighted in the In This Issue feature, p. 521.


Subject(s)
Ferroptosis/genetics , Lipogenesis/genetics , Melanoma/genetics , Melanoma/metabolism , Neoplastic Cells, Circulating/metabolism , Sterol Regulatory Element Binding Protein 2/genetics , Transferrin/metabolism , Biomarkers, Tumor , Cells, Cultured , Disease Susceptibility , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Melanoma/pathology , Mutation , Neoplastic Cells, Circulating/pathology , Signal Transduction , Single-Cell Analysis , Sterol Regulatory Element Binding Protein 2/metabolism
9.
Clin Transl Med ; 10(7): e160, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33252863

ABSTRACT

The presence of liver metastases drastically worsens the prognosis of cancer patients. The liver is the second most prevalent metastatic site in cancer patients, but systemic therapeutic opportunities that target liver metastases are still limited. To aid the discovery of novel treatment options for metastatic liver disease, we provide insight into the cellular and molecular steps required for liver colonization. For successful colonization in the liver, adaptation of tumor cells and surrounding stroma is essential. This includes the formation of a pre-metastatic niche, the creation of a fibrotic and immune suppressive environment, angiogenesis, and adaptation of tumor cells. We illustrate that transforming growth factor ß (TGF-ß) is a central cytokine in all these processes. At last, we devise that future research should focus on TGF-ß inhibitory strategies, especially in combination with immunotherapy. This promising systemic treatment strategy has potential to eliminate distant metastases as the efficacy of immunotherapy will be enhanced.

10.
Cell ; 177(7): 1903-1914.e14, 2019 06 13.
Article in English | MEDLINE | ID: mdl-31031007

ABSTRACT

Xenograft cell transplantation into immunodeficient mice has become the gold standard for assessing pre-clinical efficacy of cancer drugs, yet direct visualization of single-cell phenotypes is difficult. Here, we report an optically-clear prkdc-/-, il2rga-/- zebrafish that lacks adaptive and natural killer immune cells, can engraft a wide array of human cancers at 37°C, and permits the dynamic visualization of single engrafted cells. For example, photoconversion cell-lineage tracing identified migratory and proliferative cell states in human rhabdomyosarcoma, a pediatric cancer of muscle. Additional experiments identified the preclinical efficacy of combination olaparib PARP inhibitor and temozolomide DNA-damaging agent as an effective therapy for rhabdomyosarcoma and visualized therapeutic responses using a four-color FUCCI cell-cycle fluorescent reporter. These experiments identified that combination treatment arrested rhabdomyosarcoma cells in the G2 cell cycle prior to induction of apoptosis. Finally, patient-derived xenografts could be engrafted into our model, opening new avenues for developing personalized therapeutic approaches in the future.


Subject(s)
Animals, Genetically Modified/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Muscle Neoplasms , Rhabdomyosarcoma , Zebrafish/metabolism , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/immunology , Female , Heterografts , Humans , K562 Cells , Male , Muscle Neoplasms/drug therapy , Muscle Neoplasms/immunology , Muscle Neoplasms/metabolism , Muscle Neoplasms/pathology , Neoplasm Transplantation , Phthalazines/pharmacology , Piperazines/pharmacology , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/immunology , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/pathology , Temozolomide/pharmacology , Xenograft Model Antitumor Assays , Zebrafish/genetics , Zebrafish/immunology
11.
Ann Surg ; 266(5): 765-771, 2017 11.
Article in English | MEDLINE | ID: mdl-28742689

ABSTRACT

OBJECTIVES: To investigate the relevance of lymphangiogenic gene expression in primary and liver metastasis of colorectal cancer (CRC) and identify determinants of lymphatic invasion. BACKGROUND: Lymphatic development promoting vascular endothelial growth factor C (VEGFC) is associated with poor outcome in primary CRC. For colorectal liver metastasis (CRLM), intrahepatic lymph invasion and lymph node metastasis are poor prognostic factors. Exact biological factors promoting lymphatic involvement remain elusive, just as the association with molecular subtypes of CRC. METHODS: We designed a lymphangiogenic gene set (VEGFC, Nrp-2, PDPN, LYVE-1, MRC1, CCL-21) and applied it to large datasets of CRC. Gene expression of the lymphangiogenic signature was assessed in resected CRLM specimens by Rt-QPCR. In vitro experiments were performed with colon cancer cell line Colo320 (high Nrp-2 expression) and human dermal microvascular lymphatic endothelial cells (LECs). RESULTS: Lymphangiogenic gene expression was associated with poor prognosis in both primary and liver metastasis of CRC. CRLM with high expression of consensus molecular subtype-4 identifier genes also exhibited high lymphangiogenic gene expression. Lymph node recurrence following CRLM resection was associated with high expression of VEGFC and Nrp-2. Blocking Nrp-2 significantly reduced invasion of Colo320 cells through an LEC monolayer. CONCLUSIONS: Lymphangiogenic gene expression is correlated with worse prognosis and consensus molecular subtype-4 in both primary and liver metastatic CRC. VEGFC and Nrp-2 expression may be predictive of lymph node involvement in recurrence after resection of CRLM. Nrp-2, expressed on both tumor and LECs, may have a mechanistic role in lymphatic invasion and is a potential novel target in CRC.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/pathology , Hepatectomy , Liver Neoplasms/metabolism , Liver Neoplasms/surgery , Lymph Nodes/pathology , Lymphangiogenesis/genetics , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Hepatectomy/methods , Humans , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Lymphatic Metastasis , Prognosis , Real-Time Polymerase Chain Reaction
12.
Gut ; 66(6): 1106-1115, 2017 06.
Article in English | MEDLINE | ID: mdl-27670374

ABSTRACT

BACKGROUND AND AIM: Colorectal cancer (CRC) remains one of the leading causes of cancer-related death. Novel therapeutics are urgently needed, especially for tumours with activating mutations in KRAS (∼40%). Here we investigated the role of RAF1 in CRC, as a potential, novel target. METHODS: Colonosphere cultures were established from human tumour specimens obtained from patients who underwent colon or liver resection for primary or metastatic adenocarcinoma. The role of RAF1 was tested by generating knockdowns (KDs) using three independent shRNA constructs or by using RAF1-kinase inhibitor GW5074. Clone-initiating and tumour-initiating capacities were assessed by single-cell cloning and injecting CRC cells into immune-deficient mice. Expression of tight junction (TJ) proteins, localisation of polarity proteins and activation of MEK-ERK pathway was analysed by western blot, immunohistochemistry and immunofluorescence. RESULTS: KD or pharmacological inhibition of RAF1 significantly decreased clone-forming and tumour-forming capacity of all CRC cultures tested, including KRAS-mutants. This was not due to cytotoxicity but, at least in part, to differentiation of tumour cells into goblet-like cells. Inhibition of RAF1-kinase activity restored apicobasal polarity and the formation of TJs in vitro and in vivo, without reducing MEK-ERK phosphorylation. MEK-inhibition failed to restore polarity and TJs. Moreover, RAF1-impaired tumours were characterised by normalised tissue architecture. CONCLUSIONS: RAF1 plays a critical role in maintaining the transformed phenotype of CRC cells, including those with mutated KRAS. The effects of RAF1 are kinase-dependent, but MEK-independent. Despite the lack of activating mutations in RAF1, its kinase domain is an attractive therapeutic target for CRC.


Subject(s)
Adenocarcinoma/genetics , Colorectal Neoplasms/genetics , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/genetics , Adenocarcinoma/drug therapy , Animals , Cell Differentiation/drug effects , Cell Polarity/drug effects , Cell Polarity/genetics , Colorectal Neoplasms/drug therapy , Gene Expression , Gene Knockdown Techniques , Humans , Indoles/pharmacology , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/metabolism , Mice , Neoplasm Transplantation , Phenols/pharmacology , Phosphorylation , Primary Cell Culture , Protein Kinase Inhibitors/pharmacology , RNA, Small Interfering/genetics , Tight Junctions , Tumor Cells, Cultured
13.
J Exp Med ; 213(12): 2575-2589, 2016 11 14.
Article in English | MEDLINE | ID: mdl-27810924

ABSTRACT

Cell transplantation into immunodeficient mice has revolutionized our understanding of regeneration, stem cell self-renewal, and cancer; yet models for direct imaging of engrafted cells has been limited. Here, we characterize zebrafish with mutations in recombination activating gene 2 (rag2), DNA-dependent protein kinase (prkdc), and janus kinase 3 (jak3). Histology, RNA sequencing, and single-cell transcriptional profiling of blood showed that rag2 hypomorphic mutant zebrafish lack T cells, whereas prkdc deficiency results in loss of mature T and B cells and jak3 in T and putative Natural Killer cells. Although all mutant lines engraft fluorescently labeled normal and malignant cells, only the prkdc mutant fish reproduced as homozygotes and also survived injury after cell transplantation. Engraftment into optically clear casper, prkdc-mutant zebrafish facilitated dynamic live cell imaging of muscle regeneration, repopulation of muscle stem cells within their endogenous niche, and muscle fiber fusion at single-cell resolution. Serial imaging approaches also uncovered stochasticity in fluorescently labeled leukemia regrowth after competitive cell transplantation into prkdc mutant fish, providing refined models to assess clonal dominance and progression in the zebrafish. Our experiments provide an optimized and facile transplantation model, the casper, prkdc mutant zebrafish, for efficient engraftment and direct visualization of fluorescently labeled normal and malignant cells at single-cell resolution.


Subject(s)
DNA-Activated Protein Kinase/deficiency , Imaging, Three-Dimensional/methods , Neoplasm Transplantation , Optical Phenomena , Single-Cell Analysis/methods , Zebrafish/metabolism , Anemia/pathology , Animals , Base Sequence , Clone Cells , DNA-Activated Protein Kinase/metabolism , Disease Models, Animal , Gamma Rays , Homozygote , Humans , Immunocompromised Host/radiation effects , Luminescent Proteins/metabolism , Muscle Cells/pathology , Muscle Cells/radiation effects , Mutation/genetics , Proto-Oncogene Proteins c-myc/metabolism , Regeneration/radiation effects , Transplantation, Homologous , V(D)J Recombination/genetics , Zebrafish Proteins/metabolism , Red Fluorescent Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...