Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters











Publication year range
1.
Cancer Biomark ; 41(2): 115-132, 2024.
Article in English | MEDLINE | ID: mdl-39240628

ABSTRACT

BACKGROUND: The molecular system of receptor activator of nuclear factor kappa-ß (RANK) and its ligand (RANKL) plays a role in a variety of physiological and pathological processes. These encompass the regulation of bone metabolism, mammary gland development, immune function, as well as their involvement and tumorigenesis. Nevertheless, limited knowledge exists regarding their function within the tumor microenvironment. METHODS AND RESULTS: We explored the significance of RANK expression in cancer-associated fibroblasts (CAFs) as a prognostic biomarker in early breast cancer patients (BCPs) by immunohistochemistry. Results reveal a significant correlation between high RANK expression in CAFs and an increased risk of metastasis (p= 0.006), shorter metastasis-free survival (MFS) [p= 0.007, OR (95%CI) = 2.290 (1.259-4.156)], and lower overall survival (OS) [p= 0.004, OR (95%CI) = 2.469 (1.343-4.541)]. Upon analyzing the phenotype of CD34(-) CAFs isolated from primary tumors in BCPs, we observed co-expression of RANK with CD105 marker by immunofluorescence and flow cytometry, characteristic of mesenchymal stem/stromal cells (MSCs), suggesting the possible cellular origin. Also RANKL-RANK system increase the OCT-4, SOX-2 and DKK-1 (dickkopf 1) gene expression in CD34(-) CAFs by RT-PCR. Moreover, this system plays a crucial role in the migration of these CD34(-) CAFs. CONCLUSIONS: These results support the clinical relevance of RANK in CAFs and propose its potential as a future therapeutic target in the treatment of early BCPs.


Subject(s)
Biomarkers, Tumor , Breast Neoplasms , Cancer-Associated Fibroblasts , Neoplasm Staging , Receptor Activator of Nuclear Factor-kappa B , Humans , Female , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/genetics , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Receptor Activator of Nuclear Factor-kappa B/metabolism , Prognosis , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Neoplasm Metastasis , Middle Aged , Tumor Microenvironment , RANK Ligand/metabolism , RANK Ligand/genetics , Adult , Aged , Cell Line, Tumor
2.
Cells ; 13(7)2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38606999

ABSTRACT

Cervical cancer (CC) is the fourth leading cancer among women and is one of the principal gynecological malignancies. In the tumor microenvironment, cancer-associated fibroblasts (CAFs) play a crucial role during malignant progression, exhibiting a variety of heterogeneous phenotypes. CAFs express phenotypic markers like fibroblast activation protein (FAP), vimentin, S100A4, α-smooth muscle actin (αSMA), and functional markers such as MMP9. This study aimed to evaluate the protein expression of vimentin, S100A4, αSMA, FAP, and MMP9 in mesenchymal stem cells (MSC)-CAF cells, as well as in cervical cancer samples. MSC cells were stimulated with HeLa and SiHa tumor cell supernatants, followed by protein evaluation and cytokine profile to confirm differentiation towards a CAF phenotype. In addition, automated immunohistochemistry (IHQa) was performed to evaluate the expression of these proteins in CC samples at different stages. Our findings revealed a high expression of FAP in stimulated MSC cells, accompanied by the secretion of pro/anti-inflammatory cytokines. In the other hand, CC samples were observed to have high expression of FAP, vimentin, αSMA, and MMP9. Most importantly, there was a high expression of their activation proteins αSMA and FAP during the different stages. In the early stages, a myofibroblast-like phenotype (CAFs αSMA+ FAP+), and in the late stages a protumoral phenotype (CAF αSMA- FAP+). In summary, FAP has a crucial role in the activation of CAFs during cervical cancer progression.


Subject(s)
Cancer-Associated Fibroblasts , Uterine Cervical Neoplasms , Humans , Female , Cancer-Associated Fibroblasts/metabolism , Vimentin/metabolism , Matrix Metalloproteinase 9/metabolism , Uterine Cervical Neoplasms/metabolism , Neoplastic Processes , Phenotype , Tumor Microenvironment
3.
Clin Transl Oncol ; 26(10): 2395-2417, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38602644

ABSTRACT

The journey of cancer development is a multifaceted and staged process. The array of treatments available for cancer varies significantly, dictated by the disease's type and stage. Cancer-associated fibroblasts (CAFs), prevalent across various cancer types and stages, play a pivotal role in tumor genesis, progression, metastasis, and drug resistance. The strategy of concurrently targeting cancer cells and CAFs holds great promise in cancer therapy. In this review, we focus intently on CAFs, delving into their critical role in cancer's progression. We begin by exploring the origins, classification, and surface markers of CAFs. Following this, we emphasize the key cytokines and signaling pathways involved in the interplay between cancer cells and CAFs and their influence on the tumor immune microenvironment. Additionally, we examine current therapeutic approaches targeting CAFs. This article underscores the multifarious roles of CAFs within the tumor microenvironment and their potential applications in cancer treatment, highlighting their importance as key targets in overcoming drug resistance and enhancing the efficacy of tumor therapies.


Subject(s)
Cancer-Associated Fibroblasts , Neoplasms , Tumor Microenvironment , Humans , Cancer-Associated Fibroblasts/pathology , Cancer-Associated Fibroblasts/metabolism , Neoplasms/pathology , Neoplasms/therapy , Drug Resistance, Neoplasm , Signal Transduction , Cytokines/metabolism , Disease Progression
4.
Pathobiology ; 91(2): 99-107, 2024.
Article in English | MEDLINE | ID: mdl-37369175

ABSTRACT

INTRODUCTION: The characterization of tumor microenvironment (TME) related factors and their impact on tumor progression have attracted much interest. We investigated cancer cells and cancer-associated fibroblasts (CAFs) to evaluate biomarkers that are associated with neoplastic progression, observing them in different interface zones of colorectal cancer. METHODS: On 357 CRC tissue microarrays, using immunohistochemistry, we examined the associations of podoplanin and α-SMA expressed in cancer cells and CAFs and evaluated them in different areas: tumor core, invasive front, tumor budding, tumor-stroma ratio (TSR) scoring, and desmoplastic stroma. RESULTS: CAFs expressing α-SMA were found in more than 90% of the cases. Podoplanin+ was detected in cancer cells and CAFs, with positivities of 38.6% and 70%, respectively. Higher α-SMA+ CAFs and podoplanin+ cancer cells were observed predominantly at the TSR score area: 94.3% and 64.3% of cases, respectively. The status of podoplanin in CAFs+ was higher in the desmoplastic area (71.6%). Stroma-high tumors showed increased expression of α-SMA and podoplanin in comparison with stroma-low tumors. The status of podoplanin in cancer cells was observed in association with lymphatic invasion and distant metastasis. CONCLUSION: The substance of the CRC was composed predominantly of the surrounding stroma-α-SMA+ CAFs. Podoplanin expressed in the prognosticator zones was associated with unfavorable pathological features. The combination of histologic and protein-related biomarkers can result in a tool for the stratification of patients with CRC.


Subject(s)
Cancer-Associated Fibroblasts , Colorectal Neoplasms , Humans , Cancer-Associated Fibroblasts/metabolism , Fibroblasts/pathology , Membrane Proteins/metabolism , Biomarkers , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/pathology , Biomarkers, Tumor/metabolism , Tumor Microenvironment
5.
Technol Cancer Res Treat ; 21: 15330338221131647, 2022.
Article in English | MEDLINE | ID: mdl-36222020

ABSTRACT

Breast cancer is the leading cause of cancer death among women worldwide. In solid tumors, the microenvironment plays a critical role in tumor development, and it has been described a communication between the different cell types that conform the stroma, including fibroblasts, pericytes, adipocytes, immune cells and cancer-associated fibroblasts. Intercellular communication is bidirectional, complex, multifactorial and is mediated by the secretion of molecules and extracellular vesicles. The extracellular vesicles are vesicles limited by two membranes that are secreted by normal and cancer cells into the extracellular space. Extracellular vesicle cargo is complex and includes proteins, miRNAs, DNA and lipids, and their composition is specific to their parent cells. Extracellular vesicles are taken up for neighboring or distant cells. Particularly, extracellular vesicles from breast cancer cells are taken up for fibroblasts and it induces the activation of fibroblasts into cancer-associated fibroblasts. Interestingly, cancer associated fibroblasts release extracellular vesicles that are taken up for breast cancer cells and promote migration, invasion, proliferation, epithelial-mesenchymal transition, changes in metabolism, chemoresistance, evasion of immune system and remodeling of extracellular matrix. In addition, the enrichment of specific cargos in extracellular vesicles of breast cancer patients has been suggested to be used as biomarkers of the disease. Here we review the current literature about the intercommunication between tumor cells and cancer associated fibroblasts through extracellular vesicles in breast cancer.


Subject(s)
Breast Neoplasms , Cancer-Associated Fibroblasts , Extracellular Vesicles , MicroRNAs , Biomarkers , Breast Neoplasms/pathology , Cancer-Associated Fibroblasts/metabolism , Extracellular Vesicles/metabolism , Female , Humans , Lipids , MicroRNAs/genetics , MicroRNAs/metabolism , Tumor Microenvironment
6.
J Exp Med ; 219(6)2022 06 06.
Article in English | MEDLINE | ID: mdl-35583818

ABSTRACT

Excessive collagen deposition by fibroblasts surrounding some tumors has seriously limited the efficacy of checkpoint inhibitor therapies. Chen et al. (2022. J. Exp. Med.https://doi.org/10.1084/jem.20210693) show that IL-17 promotes collagen deposition by cancer-associated fibroblasts, enhancing immune exclusion of tumors, and that targeting IL-17-triggered HIF1α expression can reverse matrix mediated immune exclusion.


Subject(s)
Cancer-Associated Fibroblasts , Interleukin-17 , Neoplasms , Cancer-Associated Fibroblasts/metabolism , Humans , Interleukin-17/metabolism , Neoplasms/metabolism , T-Lymphocytes
7.
Clin Transl Oncol ; 24(5): 757-769, 2022 May.
Article in English | MEDLINE | ID: mdl-34839457

ABSTRACT

Colorectal cancer (CRC) is one of the leading causes of mortality among cancers. Many aspects of this cancer are under investigation to find established markers of diagnosis, prognosis, and also potential drug targets. In this review article, we are going to discuss the possible solution to all these aims by investigating the literature about cancer-associated fibroblasts (CAFs) involved in CRC. Moreover, we are going to review their interaction with the tumor microenvironment (TME) and vitamin D and their role in tumorigenesis and metastasis. Moreover, we are going to expand more on some markers produced by them or related to them including FAP, a-SMA, CXCL12, TGF- ß, POSTN, and ß1-Integrin. Some signaling pathways related to CAFs are as follows: FAK, AKT, activin A, and YAP/TAZ. Some genes related to the CAFs which are found to be possible therapeutic targets include COL3A1, JAM3, AEBP1 and, CAF-derived TGFB3, WNT2, and WNT54.


Subject(s)
Cancer-Associated Fibroblasts , Colorectal Neoplasms , Cancer-Associated Fibroblasts/metabolism , Carboxypeptidases/metabolism , Carcinogenesis/pathology , Colorectal Neoplasms/pathology , Fibroblasts/metabolism , Humans , Prognosis , Repressor Proteins/metabolism , Transforming Growth Factor beta/metabolism , Tumor Microenvironment
8.
Sci Rep ; 11(1): 23881, 2021 12 13.
Article in English | MEDLINE | ID: mdl-34903806

ABSTRACT

Mast cell tumours (MCTs) are the most frequent malignant skin neoplasm in dogs. Due to the difficulty in purifying large numbers of canine neoplastic mast cells, relatively little is known about their properties. A reproducible in vitro model is needed to increase the understanding about the phenotype and functional properties of neoplastic mast cells. In the present study, we describe the establishment of primary cocultures of neoplastic mast cells from canine cutaneous MCTs and cancer-associated fibroblasts. We confirmed the inability of canine neoplastic mast cells to remain viable for long periods in vitro without the addition of growth factors or in vivo passages in mice. Using a transwell system, we observed that mast cell viability was significantly higher when there is cell-to-cell contact in comparison to non-physical contact conditions and that mast cell viability was significantly higher in high-grade than in low-grade derived primary cultures. Moreover, the use of conditioned medium from co-cultured cells led to a significantly higher tumoral mast cell viability when in monoculture. Signalling mechanisms involved in these interactions might be attractive therapeutic targets to block canine MCT progression and deserve more in-depth investigations.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Cell Communication , Dog Diseases/metabolism , Mast Cells/metabolism , Skin Neoplasms/metabolism , Animals , Cancer-Associated Fibroblasts/pathology , Cells, Cultured , Coculture Techniques/methods , Coculture Techniques/veterinary , Dog Diseases/pathology , Dogs , Female , Male , Mast Cells/pathology , Primary Cell Culture/methods , Primary Cell Culture/veterinary , Skin Neoplasms/pathology , Skin Neoplasms/veterinary
9.
Endocrinol Metab (Seoul) ; 35(4): 673-680, 2020 12.
Article in English | MEDLINE | ID: mdl-33161690

ABSTRACT

Thyroid cancer is the most common endocrine malignancy. Although most thyroid cancer patients are successfully treated and have an excellent prognosis, a percentage of these patients will develop aggressive disease and, eventually, progress to anaplastic thyroid cancer. Since most patients with this type of aggressive thyroid carcinoma will die from the disease, new treatment strategies are urgently needed. Tumor cells live in a complex and dynamic tumor microenvironment composed of different types of stromal cells. Cancer-associated fibroblasts (CAFs) are one of the most important cell components in the tumor microenvironment of most solid tumors, including thyroid cancer. CAFs originate mainly from mesenchymal cells and resident fibroblasts that are activated and reprogrammed in response to paracrine factors and cytokines produced and released by tumor cells. Upon reprogramming, which is distinguished by the expression of different marker proteins, CAFs synthesize and secret soluble factors. The secretome of CAFs directly impacts different functions of tumor cells. This bi-directional interplay between CAFs and tumor cells within the tumor microenvironment ends up fostering tumor cancer progression. CAFs are therefore key regulators of tumor progression and represent an under-explored therapeutic target in thyroid cancer.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Thyroid Carcinoma, Anaplastic/metabolism , Thyroid Neoplasms/metabolism , Tumor Microenvironment , Fibroblasts , Humans , Paracrine Communication , Stromal Cells , Thyroid Neoplasms/pathology
10.
J Mol Histol ; 51(6): 675-684, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33000351

ABSTRACT

The purpose of this study was to evaluate the expression of Hedgehog (HH) signaling molecules (SHH and GLI-1) by cancer-associated fibroblasts (CAF) in oral squamous cell carcinoma (OSCC). Immunohistochemistry was used to detect molecular HH signaling and CAF-related protein expression, including α-SMA and S100A4, in 70 samples of human OSCC. The colocalization of α-SMA and S100A4 with SHH was also evaluated by double-staining. In vitro study was performed using primary normal oral fibroblast (NOF) and CAF through immunofluorescence and Western Blot for CAF-proteins, SHH, and GLI-1. Forty-five cases (64.28%) were positive for α-SMA exclusively in tumor stroma, and S100A4 was identified in the cytoplasm of CAFs in 94.28% (n = 66) of the cases. With respect to stromal cells, 64 (91.43%) OSCC cases were positive for SHH, and 31 were positive for GLI-1 (44.29%); positive correlations were found between SHH and α-SMA (p < 0.0001, φ = 0.51), as well as between SHH and S100A4 (p = 0.087, φ = 0.94). Protein expression of SHH and GLI-1 was observed in primary CAFs and NOFs. Although SHH was found to be localized in the cellular cytoplasm of both cell types, GLI-1 was present only in the nuclei of CAF. Our results indicate that CAFs are not only potential sources of HH ligands in tumor stroma, but may also respond to HH signaling through nuclear GLI-1 activation. We further observed that elevated SHH expression by OSCC cells was associated with higher CAF density, reinforcing the chemoattractant role played by these molecules.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Hedgehog Proteins/metabolism , Mouth Neoplasms/metabolism , Signal Transduction , Biomarkers , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/genetics , Humans , Immunohistochemistry , Ligands , Mouth Neoplasms/etiology , Mouth Neoplasms/pathology , Protein Binding , Protein Transport , Stromal Cells/metabolism , Stromal Cells/pathology , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism
11.
Biochim Biophys Acta Proteins Proteom ; 1868(12): 140525, 2020 12.
Article in English | MEDLINE | ID: mdl-32866629

ABSTRACT

The signaling events triggered by soluble mediators released from both transformed and stromal cells shape the phenotype of tumoral cells and have significant implications in cancer development and progression. In this study we performed an in vitro heterotypic signaling assays by evaluating the proteome diversity of human dermal fibroblasts after stimulation with the conditioned media obtained from malignant melanoma cells. In addition, we also evaluated the changes in the proteome of melanoma cells after stimulation with their own conditioned media as well as with the conditioned medium from melanoma-stimulated fibroblasts. Our results revealed a clear rearrangement in the proteome of stromal and malignant cells upon crosstalk of soluble mediators. The main proteome signature of fibroblasts stimulated with melanoma conditioned medium was related to protein synthesis, which indicates that this process might be an early response of stromal cells. In addition, the conditioned medium derived from 'primed' stromal cells (melanoma-stimulated fibroblasts) was more effective in altering the functional phenotype (cell migration) of malignant cells than the conditioned medium from non-stimulated fibroblasts. Collectively, self- and cross-stimulation may play a key role in shaping the tumor microenvironment and enable tumoral cells to succeed in the process of melanoma progression and metastasis. Although the proteome landscape of cells participating in such a heterotypic signaling represents a snapshot of a highly dynamic state, understanding the diversity of proteins and enriched biological pathways resulting from stimulated cell states may allow for targeting specific cell regulatory motifs involved in melanoma progression and metastasis.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Dermis/cytology , Melanoma/metabolism , Proteome , Signal Transduction , Adaptation, Physiological , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Cell Movement/genetics , Computational Biology/methods , Culture Media, Conditioned , Humans , Melanoma/genetics , Melanoma/pathology , Phenotype , Proteomics/methods , Tumor Microenvironment
12.
Clin Transl Oncol ; 22(9): 1591-1602, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32056128

ABSTRACT

BACKGROUND: Tumor metastasis is a terrifying characteristic of cancer. Numerous studies have been conducted to overcome metastasis by targeting tumor microenvironment (TME). However, due to complexity of tumor microenvironment, it remained difficult for accurate targeting. Dwarf-lillytruf tuber monomer-13 (DT-13) possess good potential against TME. OBJECTIVE: As TME is supportive for tumor metastasis, alternatively it is a challenging for therapeutic intervention. In our present study, we explored molecular mechanism through which TME induced cell migration and how DT-13 interferes in this mechanism. METHODS: We used a novel model of co-culture system which is eventually developed in our lab. Tumor cells were co-cultured with hypoxia induced cancer-associated fibroblasts (CAF) or with chemically induced cancer-associated adipocytes (CAA). The effect of hypoxia in conditioned medium for CAF was assessed through expression of α-SMA and HIF by western blotting while oil red staining was done to assess the successful chemical induction for adipocytes (CAA), the effect of TME through conditioned medium on cell migration was analyzed by trans-well cell migration, and cell motility (wound healing) analyses. The expression changes in cellular proteins were assessed through western blotting and immunofluorescent studies. RESULTS AND CONCLUSION: Our results showed that tumor microenvironment has a direct role in promoting breast cancer cell migration by stromal cells; moreover, we found that DT-13 restricts this TME regulated cell migration via targeting stromal cells in vitro. Additionally we also found that DT-13 targets NMII-A for its effect on breast cancer cell migration for the regulation of stromal cells in TME.


Subject(s)
Breast Neoplasms/drug therapy , Myosin Heavy Chains/metabolism , Saponins/pharmacology , Animals , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Cell Movement/immunology , Female , Humans , Liriope Plant/chemistry , Mice , Myosin Heavy Chains/genetics , Signal Transduction , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
13.
Cells ; 8(12)2019 12 04.
Article in English | MEDLINE | ID: mdl-31817155

ABSTRACT

Breast cancer stromal compartment, may influence responsiveness to chemotherapy. Our aim was to detect a stromal cell signature (using a direct approach of microdissected stromal cells) associated with response to neoadjuvant chemotherapy (neoCT) in locally advanced breast cancer (LABC). The tumor samples were collected from 44 patients with LABC (29 estrogen receptor (ER) positive and 15 ER negative) before the start of any treatment. Neoadjuvant chemotherapy consisted of doxorubicin and cyclophosphamide, followed by paclitaxel. Response was defined as downstaging to maximum ypT1a-b/ypN0. The stromal cells, mainly composed of fibroblast and immune cells, were microdissected from fresh frozen tumor samples and gene expression profile was determined using Agilent SurePrint G3 Human Gene Expression microarrays. Expression levels were compared using MeV (MultiExperiment Viewer) software, applying SAM (significance analysis of microarrays). To classify samples according to tumor response, the order of median based on confidence statements (MedOr) was used, and to identify gene sets correlated with the phenotype downstaging, gene set enrichment analysis (GSEA). Nine patients presented disease downstaging. Eleven sequences (FDR 17) were differentially expressed, all of which (except H2AFJ) more expressed in responsive tumors, including PTCHD1 and genes involved in abnormal cytotoxic T cell physiology, TOX, LY75, and SH2D1A. The following four pairs of markers could correctly classify all tumor samples according to response: PTCHD1/PDXDC2P, LOC100506731/NEURL4, SH2D1A/ENST00000478672, and TOX/H2AFJ. Gene sets correlated with tumor downstaging (FDR < 0.01) were mainly involved in immune response or lymphocyte activation, including CD47, LCK, NCK1, CD24, CD3E, ZAP70, FOXP3, and CD74, among others. In locally advanced breast cancer, stromal cells may present specific features of immune response that may be associated with chemotherapy response.


Subject(s)
Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Stromal Cells/metabolism , Transcriptome , Adult , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cancer-Associated Fibroblasts/metabolism , Cyclophosphamide/administration & dosage , Cyclophosphamide/therapeutic use , Doxorubicin/administration & dosage , Doxorubicin/therapeutic use , Drug Administration Schedule , Female , Humans , Middle Aged , Neoadjuvant Therapy , Paclitaxel/administration & dosage , Paclitaxel/therapeutic use
14.
Front Immunol ; 10: 1394, 2019.
Article in English | MEDLINE | ID: mdl-31281317

ABSTRACT

In colorectal cancer (CRC), cancer-associated fibroblasts (CAFs) are the most abundant component from the tumor microenvironment (TM). CAFs facilitate tumor progression by inducing angiogenesis, immune suppression and invasion, thus altering the organization/composition of the extracellular matrix (i.e., desmoplasia) and/or activating epithelial-mesenchymal transition (EMT). Soluble factors from the TM can also contribute to cell invasion through secretion of cytokines and recently, IL-33/ST2 pathway has gained huge interest as a protumor alarmin, promoting progression to metastasis by inducing changes in TM. Hence, we analyzed IL-33 and ST2 content in tumor and healthy tissue lysates and plasma from CRC patients. Tissue localization and distribution of these molecules was evaluated by immunohistochemistry (using localization reference markers α-smooth muscle actin or α-SMA and E-cadherin), and clinical/histopathological information was obtained from CRC patients. In vitro experiments were conducted in primary cultures of CAFs and normal fibroblasts (NFs) isolated from tumor and healthy tissue taken from CRC patients. Additionally, migration and proliferation analysis were performed in HT29 and HCT116 cell lines. It was found that IL-33 content increases in left-sided CRC patients with lymphatic metastasis, with localization in tumor epithelia associated with abundant desmoplasia. Although ST2 content showed similarities between tumor and healthy tissue, a decreased immunoreactivity was observed in left-sided tumor stroma, associated to metastasis related factors (advanced stages, abundant desmoplasia, and presence of tumor budding). A principal component analysis (including stromal and epithelial IL-33/ST2 and α-SMA immunoreactivity with extent of desmoplasia) allowed us to distinguish clusters of low, intermediate and abundant desmoplasia, with potential to develop a diagnostic signature with benefits for further therapeutic targets. IL-33 transcript levels from CAFs directly correlated with CRC cell line migration induced by CAFs conditioned media, with rhIL-33 inducing a mesenchymal phenotype in HT29 cells. These results indicate a role of IL-33/ST2 in tumor microenvironment, specifically in the interaction between CAFs and epithelial tumor cells, thus contributing to invasion and metastasis in left-sided CRC, most likely by activating desmoplasia.


Subject(s)
Colorectal Neoplasms/pathology , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Neoplasm Invasiveness/pathology , Tumor Microenvironment/physiology , Adult , Aged , Cancer-Associated Fibroblasts/metabolism , Cell Movement/physiology , Cell Proliferation/physiology , Colorectal Neoplasms/metabolism , Female , Humans , Male , Middle Aged
15.
Biochem Biophys Res Commun ; 516(1): 96-101, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31200958

ABSTRACT

Ovarian cancer (OC) is a highly prevalent gynecological malignancy worldwide. Throughout ovarian carcinogenesis, the crosstalk between cellular components of the microenvironment, including tumor cells and fibroblasts, is proposed to play critical roles in cancer progression. The dysregulation of microRNA expression is also a pronounced feature of the OC. The screening of microRNAs, mainly those involved in OC microenvironment, could have diagnostic and/or therapeutic potential for this malignancy. Thus, we assessed the influence of fibroblasts on microRNA expression and the motility of OC cells. To achieve this goal, SKOV-3 cancer cells were co-cultured with human normal fibroblasts derived from primary culture (FP-96). Cell viability, expression of tumor suppressor microRNAs and oncomiRs by RT-qPCR, cell migration by wound healing assay and analysis of MMP-2 activity by zymography were performed in SKOV-3 cells. Moreover, α-smooth muscle actin (α-SMA) expression was evaluated by Western blot in FP-96 fibroblasts. Notably, the co-culture downregulated the tumor suppressor miR-29b and increased migration of SKOV-3 cells. In addition, co-culture increased the activity of MMP-2, which is a miR-29 target, and accounted for extracellular matrix remodeling and augmented cellular motility. Concomitantly, the co-culture system induced α-SMA expression in FP-96 fibroblasts, the commonly expressed marker in cancer-associated fibroblasts (CAFs). Our findings suggest that the potential crosstalk between OC cells and fibroblasts in tumor microenvironment may play a key role in the progression of OC.


Subject(s)
Fibroblasts/pathology , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Ovarian Neoplasms/pathology , Tumor Microenvironment , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Cell Line , Cell Line, Tumor , Cell Movement , Coculture Techniques , Down-Regulation , Female , Fibroblasts/metabolism , Humans , Ovarian Neoplasms/genetics
16.
Sci Rep ; 9(1): 8028, 2019 05 29.
Article in English | MEDLINE | ID: mdl-31142771

ABSTRACT

Thyroid cancer is the most common endocrine malignancy. Anaplastic thyroid cancer is one of the most aggressive thyroid tumors. It is known that activation of oncogenes and/or inactivation of tumor suppressor genes in tumor cells promotes tumorigenesis. The microenvironment of the tumor also plays a key role on cancer development and progression in a variety of tumors. However, the mechanisms by which tumor-stroma crosstalk in thyroid cancer remains poorly characterized. In this study we aimed to understand how interactions between fibroblasts and anaplastic thyroid cancer cells contribute to thyroid carcinogenesis. We first characterized the phenotypic changes of human fibroblasts in vitro through co-cultures by using transwells as well as by using anaplastic thyroid cancer cells-derived conditioned media. We found that fibroblasts acquired an activated phenotype or also known as cancer-associated fibroblast phenotype after being in contact with soluble factors secreted from anaplastic thyroid cancer cells, compared to the fibroblasts in mono-cultures. All the changes were partly mediated through Src/Akt activation. Treatment with the antioxidant N-acetyl-cysteine reversed in part the metabolic phenotype of activated fibroblasts. Remarkably, conditioned media obtained from these activated fibroblasts promoted cell proliferation and invasion of follicular thyroid cancer cell line, FTC-133 cells. Thus, a reciprocal and dynamic interaction exists between tumor and stromal cells, which results in the promotion of thyroid tumorigenesis. The present studies have advanced the understanding of the molecular basis of tumor-stroma communications, enabling identification and targeting of tumor-supportive mechanisms for novel treatment modalities.


Subject(s)
Adenocarcinoma, Follicular/pathology , Cancer-Associated Fibroblasts/metabolism , Stromal Cells/pathology , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Neoplasms/pathology , Carcinogenesis/pathology , Cell Communication , Cell Culture Techniques , Cell Dedifferentiation , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Culture Media, Conditioned/metabolism , Disease Progression , Humans , Neoplasm Invasiveness/pathology , Paracrine Communication , Thyroid Gland/cytology , Thyroid Gland/pathology , Tumor Microenvironment
18.
J Proteomics ; 174: 1-8, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29275043

ABSTRACT

The imbalance of cellular homeostasis during oncogenesis together with the high heterogeneity of tumor-associated stromal cells have a marked effect on the repertoire of the proteins secreted by malignant cells (the secretome). Hence, the study of tumoral secretomes provides insights for understanding the cross-talk between cells within the tumor microenvironment as well as the key effectors for the establishment of the pre-metastatic niche in distant tumor sites. In this context, we performed a proteomic analysis of the secretomes derived from four cell lines: a paired set of fibroblasts - Hs 895. T, a cell line obtained from a lung node metastatic site from a patient who had melanoma and Hs 895.Sk, a skin fibroblast cell line (derived from the same patient); two malignant metastatic melanoma cell lines - A375, a malignant melanoma cell line from primary source and SH-4, a cell line derived from pleural effusion of a patient with metastatic melanoma. Clustering of expression profiles together with functional enrichment analysis resulted in patterns that mirrored each cell type. In addition, these patterns might be the result of cell-specific protein expression programs and reveal the emergence of trends in the co-expression of functionally related proteins in cellular melanoma models. SIGNIFICANCE: Melanoma is an aggressive skin cancer and a lethal melanocytic neoplasm with increasing annual number of cases, faster than any other solid tumor. In this context, the imbalance of cellular homeostasis during oncogenesis together with the high heterogeneity of tumor-associated stromal cells have a marked effect on the repertoire of the proteins secreted by malignant cells (the secretome). Therefore, the identification of protein expression patterns in malignant cells together with functional enrichment analysis provide insights into cell-specific protein expression programs and may reveal the emergence of trends in the co-expression of functionally related proteins regardless of cell type. Moreover, the identification of networks of protein interactions together with their expression profiles can be used for the targeted analysis of co-expressed proteins, allowing the identification of regulatory motifs in melanoma protein-protein interaction networks.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Melanoma/metabolism , Proteomics/methods , Cell Line, Tumor , Cluster Analysis , Gene Expression Profiling , Humans , Neoplasm Proteins/metabolism , Protein Interaction Mapping , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL