Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Microbes Infect ; 26(4): 105305, 2024.
Article in English | MEDLINE | ID: mdl-38296157

ABSTRACT

The liver, and more specifically, the liver sinusoidal endothelial cells, constitute the beginning of one of the most important responses for the elimination of hematogenously disseminated Candida albicans. Therefore, we aimed to study the mechanisms involved in the interaction between these cells and C. albicans. Transcriptomics-based analysis showed an increase in the expression of genes related to the immune response (including receptors, cytokines, and adhesion molecules), as well as to aerobic glycolysis. Further in vitro analyses showed that IL-6 production in response to C. albicans is controlled by MyD88- and SYK-pathways, suggesting an involvement of Toll-like and C-type lectin receptors and the subsequent activation of the MAP-kinases and c-Fos/AP-1 transcription factor. In addition, liver sinusoidal endothelial cells undergo metabolic reprogramming towards aerobic glycolysis induced by C. albicans, as confirmed by the increased Extracellular Acidification Rate and the overexpression of enolase (Eno2), hexonikase (Hk2) and glucose transporter 1 (Slc2a1). In conclusion, these results indicate that the hepatic endothelium responds to C. albicans by increasing aerobic glycolysis and promoting an inflammatory environment.


Subject(s)
Candida albicans , Endothelial Cells , Glycolysis , Liver , Candida albicans/immunology , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Animals , Liver/metabolism , Liver/microbiology , Syk Kinase/metabolism , Interleukin-6/metabolism , Interleukin-6/genetics , Mice , Myeloid Differentiation Factor 88/metabolism , Inflammation/metabolism , Gene Expression Profiling , Candidiasis/immunology , Candidiasis/microbiology , Candidiasis/metabolism
2.
Nutrients ; 15(11)2023 May 29.
Article in English | MEDLINE | ID: mdl-37299500

ABSTRACT

Prostate cancer is one of the most common cancers among men. Although many patients respond favorably to first-line treatments, castration-and chemotherapy-resistance arises after a few years, leading to metastasis. Thus, new approaches are being investigated using natural supplements to reinforce current therapies. Ocoxin is a plant-based mixture with antitumor properties that have been proved in several cancers. Here, we evaluated the cytotoxic capacity of this compound itself and combined with Docetaxel, Enzalutamide and Olaparib as an adjuvant agent. We observed that Ocoxin reduced tumor cell viability; slowed down cell cycles; altered the expression of genes involved in DNA replication, cell cycles and the p53 signaling pathway; and reduced migratory capacity after stimulation with cancer-associated fibroblasts (CAFs) and osteoblasts in vitro and reduced tumor volume in vivo. The combination of the nutritional supplement with chemotherapy showed a higher cytotoxic effect than chemotherapy alone and reverted chemoresistance conferred by CAFs and osteoblasts. Moreover, the adjuvant therapy also improved the outcome in vivo compared to the treatment with solo chemotherapy, where mice developed smaller tumors and less angiogenesis. Therefore, Ocoxin arises as a good candidate for further studies in combination with current treatments for prostate-cancer patients.


Subject(s)
Antineoplastic Agents , Prostatic Neoplasms , Humans , Male , Animals , Mice , Taxoids/pharmacology , Taxoids/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Docetaxel/pharmacology , Docetaxel/therapeutic use , Prostatic Neoplasms/drug therapy , Adjuvants, Immunologic , Cell Line, Tumor
3.
J Extracell Vesicles ; 12(5): e12326, 2023 05.
Article in English | MEDLINE | ID: mdl-37194998

ABSTRACT

The capture of tumour-derived extracellular vesicles (TEVs) by cells in the tumour microenvironment (TME) contributes to metastasis and notably to the formation of the pre-metastatic niche (PMN). However, due to the challenges associated with modelling release of small EVs in vivo, the kinetics of PMN formation in response to endogenously released TEVs have not been examined. Here, we have studied the endogenous release of TEVs in mice orthotopically implanted with metastatic human melanoma (MEL) and neuroblastoma (NB) cells releasing GFP-tagged EVs (GFTEVs) and their capture by host cells to demonstrate the active contribution of TEVs to metastasis. Human GFTEVs captured by mouse macrophages in vitro resulted in transfer of GFP vesicles and the human exosomal miR-1246. Mice orthotopically implanted with MEL or NB cells showed the presence of TEVs in the blood between 5 and 28 days after implantation. Moreover, kinetic analysis of TEV capture by resident cells relative to the arrival and outgrowth of TEV-producing tumour cells in metastatic organs demonstrated that the capture of TEVs by lung and liver cells precedes the homing of metastatic tumour cells, consistent with the critical roles of TEVs in PMN formation. Importantly, TEV capture at future sites of metastasis was associated with the transfer of miR-1246 to lung macrophages, liver macrophages, and stellate cells. This is the first demonstration that the capture of endogenously released TEVs is organotropic as demonstrated by the presence of TEV-capturing cells only in metastatic organs and their absence in non-metastatic organs. The capture of TEVs in the PMN induced dynamic changes in inflammatory gene expression which evolved to a pro-tumorigenic reaction as the niche progressed to the metastatic state. Thus, our work describes a novel approach to TEV tracking in vivo that provides additional insights into their role in the earliest stages of metastatic progression.


Subject(s)
Extracellular Vesicles , Melanoma , MicroRNAs , Humans , Animals , Mice , Extracellular Vesicles/metabolism , Kinetics , MicroRNAs/metabolism , Melanoma/metabolism , Inflammation/metabolism , Tumor Microenvironment
4.
Pharmaceuticals (Basel) ; 15(10)2022 Sep 20.
Article in English | MEDLINE | ID: mdl-36297277

ABSTRACT

The endocannabinoid system is widespread through the body and carries out a wide variety of functions. However, its involvement in other pathologies, such as cancer, still needs further attention. We aim to investigate the role of CB2 receptor during melanoma and colorectal cancer (CRC) aggressiveness and metastatic growth in the liver. We used the synthetic cannabinoid URB447, a known CB2 agonist and CB1 antagonist drug, and studied prometastatic ability of mouse B16 melanoma and MCA38 CRC cells, by means of proliferation, apoptosis, cell cycle, migration and matrix degradation in vitro upon URB447 treatment. We reported a dose-dependent viability decrease in both tumor types. This result is partly mediated by apoptotic cell death and cell cycle arrest in G1/G0 phase, as observed through flow cytometry. Melanoma and CRC cell migration was affected in a dose-dependent fashion as observed through scratch assay, whereas the secretion of matrix degrading proteins metalloprotease 2 (MMP2) and 9 (MMP9) in tumor cells did not significantly change. Moreover, daily treatment of tumor bearing mice with URB447 decreased the development of liver metastasis in a melanoma model in vivo. This proof of concept study points out to the synthetic cannabinoid URB447 as a potential candidate for deeper studies to confirm its potential as antitumor therapy and liver metastasis treatment for CRC and melanoma.

5.
Breast J ; 2022: 5169405, 2022.
Article in English | MEDLINE | ID: mdl-35711892

ABSTRACT

Discoidin domain receptor 2 (DDR2) is arising as a promising therapeutic target in breast carcinoma (BC). The ability of DDR2 to bind to collagen promotes protumoral responses in cancer cells that influence the tumor microenvironment (TME). Nonetheless, the interrelation between DDR2 expression and TME modulation during BC progression remains poorly known. For this reason, we aim to evaluate the correlation between intratumoral expression of DDR2 and the infiltration of the main TME cell populations, cancer-associated fibroblasts (CAFs), and tumor-associated macrophages (TAMs). First, collagen and DDR2 expression levels were analyzed in human invasive BC samples. Then, DDR2 status correlation with tumor aggressiveness and patient survival were retrieved from different databases. Subsequently, the main pathways, cell types, and tissues correlated with DDR2 expression in BC were obtained through bioinformatics approach. Finally, we studied the association of DDR2 expression with the recruitment of CAFs and TAMs. Our findings showed that, together with the expected overexpression of TME markers, DDR2 was upregulated in tumor samples. Besides, we uncovered that altered TME markers were linked to DDR2 expression in invasive BC patients. Consequently, DDR2 modulates the stromal reaction through CAFs and TAMs infiltration and could be used as a potential worse prognostic factor in the treatment response of invasive BC.


Subject(s)
Breast Neoplasms , Cancer-Associated Fibroblasts , Discoidin Domain Receptor 2 , Tumor-Associated Macrophages , Breast Neoplasms/pathology , Collagen/metabolism , Discoidin Domain Receptor 2/genetics , Discoidin Domain Receptor 2/metabolism , Female , Humans , Prognosis , Tumor Microenvironment
6.
Biomol Ther (Seoul) ; 30(2): 162-169, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34873071

ABSTRACT

We utilized Fas21, a resveratrol analog, to modulate the function of hepatic stellate cells (HSCs) and liver sinusoidal endothelial cells (LSECs) during the angiogenic phase of murine liver metastasis by B16 melanoma and 51b colorectal carcinoma. Preangiogenic micrometastases were treated with Fas21 (1 mg/kg/day) or vehicle during the development of intra-angiogenic tracts. Mice treated with Fas21 showed reduced liver tumor foci in both liver metastasis models. Micrometastases were classified immunohistochemically, as well as according to their position coordinates and connection to local microvasculature. The volume of liver occupied by sinusoidal-type foci, containing infiltrating angiogenic capillaries, decreased by ~50% in Fas21-treated mice compared to vehicle-treated ones in both tumor metastasis models. The volume of portal foci, containing peripheral neoangiogenesis within a discontinuous layer of myofibroblasts, was similar in all experimental groups in both tumor metastasis models, but displayed enhanced necrotic central areas devoid of angiogenesis following Fas21 treatment. As a result, sinusoidal tumors from mice treated with Fas21 showed a 50% reduction in desmin(+)/asma(+) HSCs and CD31(+) vessel density, and a 45% reduction in intrametastatic VEGF mRNA compared with sinusoidal tumors from vehicle-treated mice. Necrotic portal metastases increased 2-4-fold in treated mice. In vitro, Fas21 reduced VEGF secretion by HSCs and 51b cells dose-dependently. Additionally, HSCs migration in response to tumor soluble factors was dose-dependently diminished by Fas21, as was LSEC migration in response to HSCs and tumor soluble factors. Resveratrol analog Fas21 inhibits the proangiogenic response of HSCs and LSECs during the development of murine liver metastasis.

7.
World J Gastroenterol ; 27(24): 3516-3529, 2021 Jun 28.
Article in English | MEDLINE | ID: mdl-34239266

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has a tremendous impact on the health of millions of people worldwide. Unfortunately, those suffering from previous pathological conditions are more vulnerable and tend to develop more severe disease upon infection with the new SARS-CoV-2. This coronavirus interacts with the angiotensin-converting enzyme 2 receptor to invade the cells. Recently, another receptor, neuropilin-1 (NRP-1), has been reported to amplify the viral infection. Interestingly, NRP-1 is expressed in nonparenchymal liver cells and is related to and upregulated in a wide variety of liver-related pathologies. It has been observed that SARS-CoV-2 infection promotes liver injury through several pathways that may be influenced by the previous pathological status of the patient and liver expression of NRP-1. Moreover, coronavirus disease 2019 causes an inflammatory cascade called cytokine storm in patients with severe disease. This cytokine storm may influence liver sinusoidal-cell phenotype, facilitating viral invasion. In this review, the shreds of evidence linking NRP-1 with liver pathologies such as hepatocellular carcinoma, liver fibrosis, nonalcoholic fatty liver disease and inflammatory disorders are discussed in the context of SARS-CoV-2 infection. In addition, the involvement of the infection-related cytokine storm in NRP-1 overexpression and the subsequent increased risk of SARS-CoV-2 infection are also analyzed. This review aims to shed some light on the involvement of liver NRP-1 during SARS-CoV-2 infection and emphasizes the possible involvement this receptor with the observed liver damage.


Subject(s)
COVID-19 , Cytokine Release Syndrome , Humans , Liver , Neuropilin-1 , SARS-CoV-2
8.
Am J Physiol Gastrointest Liver Physiol ; 320(6): G1002-G1013, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33851541

ABSTRACT

Tumor DDR1 acts as a key factor during the desmoplastic response surrounding hepatic colorectal metastasis. Hepatic sinusoidal cell-derived soluble factors stimulate tumor DDR1 activation. DDR1 modulates matrix remodeling to promote metastasis in the liver through the interaction with hepatic stromal cells, specifically liver sinusoidal endothelial cells and hepatic stellate cells.


Subject(s)
Carcinoma/genetics , Colonic Neoplasms/genetics , Discoidin Domain Receptor 1/genetics , Liver Neoplasms/genetics , Liver/pathology , Animals , Carcinoma/metabolism , Carcinoma/secondary , Cell Line, Tumor , Cell Proliferation , Colon/metabolism , Colon/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Discoidin Domain Receptor 1/metabolism , Hepatic Stellate Cells/pathology , Humans , Liver/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Male , Mice , Phosphorylation , Prognosis , Stromal Cells/metabolism , Stromal Cells/pathology
9.
Nutrients ; 13(2)2021 Feb 21.
Article in English | MEDLINE | ID: mdl-33669949

ABSTRACT

Whereas the prevalence of several cancer types is decreasing, skin malignancies are growing more common every year. Malignant melanoma is the most aggressive form of skin cancer with high metastatic capacity. In most cases, malignant melanoma shows acquired therapy resistance. We evaluated the ability of Ocoxin, a natural compound-based antioxidant and anti-inflammatory nutritional complement, to exert an antitumor effect in melanoma. To do so, the cytotoxicity of Ocoxin in a panel of BRAF-mutated murine and human melanoma cell lines was tested alone and in combination with BRAF inhibitor Vemurafenib. Our results revealed a potent cytotoxic effect of Ocoxin against melanoma cells and a synergic effect when combined with Vemurafenib, reducing viability and increasing apoptosis. Besides, Ocoxin interferes with the cell cycle, impairs the inherent and fibroblast-mediated melanoma cell migration, and reduces resistance to BRAF inhibition. Proteomic analysis revealed reduced tumor secretion of inflammatory factors Galectin-1, Osteopontin, CCL5, and CCL9 upon treatment with Ocoxin. Moreover, RNASeq showed that Ocoxin downregulated the cell cycle and proliferation-related genes. In vivo, Ocoxin reduced the number of lung metastasis of YUMM-1.7 melanoma cells. Therefore, Ocoxin arises as a good candidate for clinical trials analyzing the beneficial effects in patients suffering from this cutaneous malignancy.


Subject(s)
Antineoplastic Agents/pharmacology , Ascorbic Acid/pharmacology , Folic Acid/pharmacology , Melanoma/drug therapy , Pantothenic Acid/pharmacology , Plant Extracts/pharmacology , Protein Kinase Inhibitors/pharmacology , Skin Neoplasms/drug therapy , Vitamin B 12/pharmacology , Vitamin B 6/pharmacology , Zinc Sulfate/pharmacology , Animals , Cancer-Associated Fibroblasts/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Humans , Melanoma/genetics , Mice , Proteomics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/genetics , Vemurafenib/pharmacology , Melanoma, Cutaneous Malignant
10.
Sci Rep ; 11(1): 3583, 2021 02 11.
Article in English | MEDLINE | ID: mdl-33574425

ABSTRACT

Human sperm protein associated with the nucleus on the X chromosome (SPANX) genes encode a protein family (SPANX-A, -B, -C and -D), whose expression is limited to the testis and spermatozoa in normal tissues and various tumour cells. SPANX-A/D proteins have been detected in metastatic melanoma cells, but their contribution to cancer development and the underlying molecular mechanisms of skin tumourigenesis remain unknown. Combining functional and proteomic approaches, the present work describes the presence of SPANX-A/D in primary and metastatic human melanoma cells and how it promotes pro-tumoural processes such as cell proliferation, motility and migration. We provide insights into the molecular features of skin tumourigenesis, describing for the first time a multifunctional role of the SPANX-A/D protein family in nuclear function, energy metabolism and cell survival, considered key hallmarks of cancer. A better comprehension of the SPANX-A/D protein subfamily and its molecular mechanisms will help to describe new aspects of tumour cell biology and develop new therapeutic targets and tumour-directed pharmacological drugs for skin tumours.


Subject(s)
Carcinogenesis/genetics , Melanoma/genetics , Nuclear Proteins/genetics , Proteomics , Amino Acid Sequence/genetics , Cell Nucleus/genetics , Cell Nucleus/pathology , Chromosomes, Human, X/genetics , Humans , Male , Melanoma/pathology , Nuclear Proteins/classification , Sequence Homology, Amino Acid , Spermatozoa/metabolism , Spermatozoa/pathology , Testis/growth & development , Testis/pathology
11.
Biomol Ther (Seoul) ; 29(3): 342-351, 2021 May 01.
Article in English | MEDLINE | ID: mdl-33455946

ABSTRACT

Liver colonization is initiated through the interplay between tumor cells and adhesion molecules present in liver sinusoidal endothelial cells (LSECs). This crosstalk stimulates tumor COX-2 upregulation and PGE2 secretion. To elucidate the role of the LSEC intercellular adhesion molecule-1 (ICAM-1) in the prometastatic response exerted by tumor and stromal COX-2, we utilized celecoxib (CLX) as a COX-2 inhibitory agent. We analyzed the in vitro proliferative and secretory responses of murine C26 colorectal cancer (CRC) cells to soluble ICAM-1 (sICAM-1), cultured alone or with LSECs, and their effect on LSEC and hepatic stellate cell (HSC) migration and in vivo liver metastasis. CLX reduced sICAM-1-stimulated COX-2 activation and PGE2 secretion in C26 cells cultured alone or cocultured with LSECs. Moreover, CLX abrogated sICAM-1-induced C26 cell proliferation and C26 secretion of promigratory factors for LSECs and HSCs. Interestingly, CLX reduced the protumoral response of HSC, reducing their migratory potential when stimulated with C26 secretomes and impairing their secretion of chemotactic factors for LSECs and C26 cells and proliferative factors for C26 cells. In vivo, CLX abrogated the prometastatic ability of sICAM-1-activated C26 cells while reducing liver metastasis. COX-2 inhibition blocked the creation of a favorable tumor microenvironment (TME) by hindering the intratumoral recruitment of activated HSCs and macrophages in addition to the accumulation of fibrillar collagen. These results point to COX-2 being a key modulator of processes initiated by host ICAM-1 during tumor cell/LSEC/HSC crosstalk, leading to the creation of a prometastatic TME in the liver.

12.
Int J Med Sci ; 18(3): 835-845, 2021.
Article in English | MEDLINE | ID: mdl-33437220

ABSTRACT

Chemotherapy and radiotherapy are the most frequent treatment for patients suffering from malignant progression of cancer. Even though new treatments are now being implemented, administration of these chemotherapeutic agents remains as the first line option in many tumor types. However, the secondary effects of these compounds represent one of the main reasons cancer patients lose life quality during disease progression. Recent data suggests that Ocoxin, a plant extract and natural compound based nutritional complement rich in antioxidants and anti-inflammatory mediators exerts a positive effect in patients receiving chemotherapy and radiotherapy. This mixture attenuates the chemotherapy and radiotherapy-related side effects such as radiation-induced skin burns and mucositis, chemotherapy-related diarrhea, hepatic toxicity and blood-infection. Moreover, it has been proven to be effective as anticancer agent in different tumor models both in vitro and in vivo, potentiating the cytotoxic effect of several chemotherapy compounds such as Lapatinib, Gemcitabine, Paclitaxel, Sorafenib and Irinotecan. The aim of this review is to put some light on the potential of this nutritional mixture as an anticancer agent and complement for the standard chemotherapy routine.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Ascorbic Acid/administration & dosage , Drug-Related Side Effects and Adverse Reactions/prevention & control , Folic Acid/administration & dosage , Neoplasms/therapy , Pantothenic Acid/administration & dosage , Plant Extracts/administration & dosage , Radiation Injuries/prevention & control , Vitamin B 12/administration & dosage , Vitamin B 6/administration & dosage , Zinc Sulfate/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Ascorbic Acid/pharmacokinetics , Chemoradiotherapy/adverse effects , Chemoradiotherapy/methods , Clinical Trials as Topic , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Drug-Related Side Effects and Adverse Reactions/etiology , Folic Acid/pharmacokinetics , Humans , Pantothenic Acid/pharmacokinetics , Plant Extracts/pharmacokinetics , Radiation Injuries/etiology , Radiation Tolerance/drug effects , Treatment Outcome , Vitamin B 12/pharmacokinetics , Vitamin B 6/pharmacokinetics , Zinc Sulfate/pharmacokinetics
13.
Cancers (Basel) ; 12(11)2020 Oct 29.
Article in English | MEDLINE | ID: mdl-33138017

ABSTRACT

The role of innate lymphoid cells (ILCs) in cancer progression has been uncovered in recent years. ILCs are classified as Type 1, Type 2, and Type 3 ILCs, which are characterized by the transcription factors necessary for their development and the cytokines and chemokines they produce. ILCs are a highly heterogeneous cell population, showing both anti- and protumoral properties and capable of adapting their phenotypes and functions depending on the signals they receive from their surrounding environment. ILCs are considered the innate counterparts of the adaptive immune cells during physiological and pathological processes, including cancer, and as such, ILC subsets reflect different types of T cells. In cancer, each ILC subset plays a crucial role, not only in innate immunity but also as regulators of the tumor microenvironment. ILCs' interplay with other immune and stromal cells in the metastatic microenvironment further dictates and influences this dichotomy, further strengthening the seed-and-soil theory and supporting the formation of more suitable and organ-specific metastatic environments. Here, we review the present knowledge on the different ILC subsets, focusing on their interplay with components of the tumor environment during the development of primary melanoma as well as on metastatic progression to organs, such as the liver or lung.

14.
Sci Rep ; 10(1): 18398, 2020 10 27.
Article in English | MEDLINE | ID: mdl-33110221

ABSTRACT

Liver metastasis depends on the collagenous microenvironment generated by hepatic sinusoidal cells (SCs). DDR1 is an atypical collagen receptor linked to tumor progression, but whether SCs express DDR1 and its implication in liver metastasis remain unknown. Freshly isolated hepatic stellate cells (HSCs), Kupffer cells (KCs), and liver sinusoidal endothelial cells (LSECs), that conform the SCs, expressed functional DDR1. HSCs expressed the largest amounts. C26 colon carcinoma secretomes increased DDR1 phosphorylation in HSCs and KCs by collagen I. Inhibition of kinase activity by DDR1-IN-1 or mRNA silencing of DDR1 reduced HSCs secretion of MMP2/9 and chemoattractant and proliferative factors for LSECs and C26 cells. DDR1-IN-1 did not modify MMP2/9 in KCs or LSECs secretomes, but decreased the enhancement of C26 migration and proliferation induced by their secretomes. Gene array showed that DDR1 silencing downregulated HSCs genes for collagens, MMPs, interleukins and chemokines. Silencing of DDR1 before tumor inoculation reduced hepatic C26 metastasis in mice. Silenced livers bore less tumor foci than controls. Metastatic foci in DDR1 silenced mice were smaller and contained an altered stroma with fewer SCs, proliferating cells, collagen and MMPs than foci in control mice. In conclusion, hepatic DDR1 promotes C26 liver metastasis and favors the pro-metastatic response of SCs to the tumor.


Subject(s)
Discoidin Domain Receptor 1/genetics , Liver Neoplasms, Experimental/prevention & control , Neoplasm Metastasis/genetics , Animals , Down-Regulation , Gene Silencing , Hepatic Stellate Cells/pathology , Liver Neoplasms, Experimental/genetics , Mice , Tumor Microenvironment
15.
Sci Rep ; 9(1): 13111, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31511625

ABSTRACT

The prometastatic stroma generated through tumor cells/host cells interaction is critical for metastatic growth. To elucidate the role of ICAM-1 on the crosstalk between tumor and primary liver sinusoidal endothelial cells (LSECs) and hepatic stellate cells (HSCs), implicated in tumor adhesion and angiogenesis, we performed in vitro cocultures and an in vivo model of liver metastasis of colorectal cancer (CRC). ICAM-1 blockade in the LSECs decreased the adhesion and transmigration of tumor cells through an LSEC in vitro and vivo. Cocultures of C26 cells and LSECs contained higher amounts of IL-1ß, IL-6, PGE-2, TNF-α and ICAM-1 than monocultures. C26 cells incubated with sICAM-1 secreted higher amounts of PGE-2, IL-6, VEGF, and MMPs, while enhanced the migration of LSECs and HSCs. HSCs cultures activated by media from C26 cells pretreated with sICAM-1 contained the largest amounts of VEGF and MMPs. C26 cell activation with sICAM-1 enhanced their metastasizing potential in vivo, while tumor LFA-1 blockade reduced tumor burden and LSECs and HSC-derived myofibroblasts recruitment. In vivo ICAM-1 silencing produced similar results. These findings uncover LSEC ICAM-1 as a mediator of the CRC metastatic cascade in the liver and identifies it as target for the inhibition of liver colonization and metastatic progression.


Subject(s)
Capillaries/pathology , Colonic Neoplasms/pathology , Endothelial Cells/pathology , Inflammation/complications , Intercellular Adhesion Molecule-1/metabolism , Liver Neoplasms/secondary , Neovascularization, Pathologic/complications , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Capillaries/immunology , Capillaries/metabolism , Cell Adhesion , Cell Communication , Cell Movement , Cell Proliferation , Colonic Neoplasms/etiology , Colonic Neoplasms/metabolism , Endothelial Cells/immunology , Endothelial Cells/metabolism , Gene Expression Regulation, Neoplastic , Humans , Inflammation/immunology , Intercellular Adhesion Molecule-1/genetics , Liver Neoplasms/etiology , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Neovascularization, Pathologic/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Pancreas ; 48(4): 555-567, 2019 04.
Article in English | MEDLINE | ID: mdl-30946238

ABSTRACT

OBJECTIVES: Pancreatic carcinoma is one of the most aggressive cancers overcoming chemoresistance. Thus, novel compounds to complement the current antitumor agents are in need. Ocoxin oral solution (OOS) has proven antioxidant, anti-inflammatory, and antistromagenic properties. The aim of this study was to analyze the effect of OOS in an experimental pancreatic cancer model and its implication in stroma-related chemoresistance to paclitaxel and gemcitabine. METHODS: Murine pancreatic carcinoma 266-6 cells were treated with OOS to analyze cell cycle and to perform a mRNA comparative microarray study. Then the viability was assessed in combination with paclitaxel and/or gemcitabine. Chemoresistance induced by the medium taken from fibroblast cultures was also investigated on 6 human pancreatic carcinoma cell lines. Furthermore, an experimental model of pancreatic cancer was carried out to study the effect of OOS in vivo. RESULTS: Ocoxin oral solution enhances the cytotoxic effect of paclitaxel and gemcitabine, while it ameliorates the chemoresistance induced by fibroblast-derived soluble factors in human pancreatic cancer cells. The OOS also promotes the regulation of the expression of genes that are altered in pancreatic carcinoma and slows down 266-6 cell pancreatic tumor development in vivo. CONCLUSIONS: Ocoxin oral solution could be a potential complement to the chemotherapeutic drugs for pancreatic adenocarcinoma.


Subject(s)
Adenocarcinoma/drug therapy , Ascorbic Acid/pharmacology , Drug Resistance, Neoplasm/drug effects , Neoplasms, Experimental/drug therapy , Pancreatic Neoplasms/drug therapy , Plant Extracts/pharmacology , Vitamin B 12/pharmacology , Vitamin B 6/pharmacology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Ascorbic Acid/administration & dosage , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Drug Resistance, Neoplasm/genetics , Folic Acid , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice, Inbred C57BL , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Paclitaxel/administration & dosage , Paclitaxel/pharmacology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pantothenic Acid , Plant Extracts/administration & dosage , Solutions , Vitamin B 12/administration & dosage , Vitamin B 6/administration & dosage , Zinc Sulfate , Gemcitabine
17.
Oncol Rep ; 39(4): 2022-2030, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29436696

ABSTRACT

The liver is a common site for the metastatic spread of primary malignancies including colorectal cancer, and liver metastasis is a main cause of death in cancer patients. This is due to the complexity of the interactions taking place in the liver between tumor and stromal cells. In fact, cancer­associated fibroblasts (CAFs) have been shown to support tumor growth through the CXCL12/CXCR4 axis. However, along with cancer cells, myeloid­derived suppressor cells (MDSCs), immature dendritic cells with immunosuppressive potential, also express CXCR4. It has recently been demonstrated that reducing CXCL12 availability in the tumor microenvironment decreases liver metastasis. Therefore, blocking CXCL12 chemokine receptor CXCR4 may be a successful approach to diminish the metastatic spread of colorectal cancer to the liver. However, the subjacent mechanisms by which this chemokine influences the tumor are not fully understood. Thus, in order to uncover the role of CXCR4 during tumor cell/liver fibroblast crosstalk driving liver metastasis, the CXCR4 antagonist AMD3100 was used for in vitro studies and in an in vivo approach using an orthotopic model of liver metastasis in immune competent mice through intrasplenic injection of grafted C26 cells. In vitro blockage of CXCR4 led to an impaired migratory potential of tumor and hepatic stellate cells (HSCs) and a reduced tumor response to CXCL12. In vivo administration of AMD3100 to tumor­bearing mice resulted in attenuated metastatic development in the liver, which was accompanied by an impaired infiltration of αSMA­expressing cells within the tumors. In addition, a reduced CD11+Ly6G+ cell count in the liver was directly correlated with a reduction in MDSC numbers in the blood of AMD3100­treated mice compared to the vehicle­treated mice. Therefore, disruption of the CXCR4/CXCL12 axis by CXCR4 antagonist AMD3100 blocked the contribution of both cancer and stromal cells to the metastatic cascade in the liver.


Subject(s)
Chemokine CXCL12/genetics , Colorectal Neoplasms/drug therapy , Liver Neoplasms/drug therapy , Receptors, CXCR4/genetics , Animals , Benzylamines , Cancer-Associated Fibroblasts/pathology , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Chemokine CXCL12/antagonists & inhibitors , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Cyclams , Gene Expression Regulation, Neoplastic/drug effects , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/pathology , Heterocyclic Compounds/administration & dosage , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Mice , Receptors, CXCR4/antagonists & inhibitors , Signal Transduction , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Xenograft Model Antitumor Assays
18.
BMC Cancer ; 17(1): 827, 2017 Dec 06.
Article in English | MEDLINE | ID: mdl-29207960

ABSTRACT

BACKGROUND: Lymphocyte Function-Associated Antigen-1 (LFA-1; CD18/CD11a) is one of the main adhesion molecules used by immune cells to infiltrate the liver under inflammatory conditions. Recently, the expression of this integrin has also been reported on several solid tumors, including colorectal cancer. However, its functional role in the metastatic progression to the liver remains unknown. Using in vitro assays and an experimental orthotopic in vivo model of liver metastasis, we aimed to elucidate the role of tumor LFA-1 in the metastatic progression by means of the partial depletion of the ß2 subunit of LFA-1, required for integrin activation, firm adhesion and signaling. METHODS: To do so, we evaluated the effects of ß2 reduction on the murine colon carcinoma C26 cell line on their pro-metastatic features in vitro and their metastatic potential in vivo in a mouse model of colon carcinoma metastasis to the liver. RESULTS: The reduction in ß2 integrin expression correlated with a slower proliferation, and a reduced adhesion and migration of C26 cells in an in vitro setting. Additionally, tumor cells with a reduced in ß2 integrin expression were unable to activate the liver sinusoidal endothelial cells (LSECs). This resulted in a recovery of the cytotoxic potential of liver lymphocytes which is compromised by LSECs activated by C26 cells. This was related to the abrogation of RNA expression of inflammatory and angiogenic cytokines by C26 cells after their activation with sICAM-1, the main ligand of ß2αL. Furthermore, in vivo tumor cell retention and metastasis were profoundly reduced, along with a decrease in the recruitment and infiltration of myeloid derived suppressor cells (MDSCs) and lymphocytes to the liver. CONCLUSION: Taken together, our findings uncovered the modulatory role for the tumor ß2 subunit of the LFA-1 integrin in the metastatic progression of colorectal cancer to the liver by impairing activation of liver endothelium and thus, the local immune response in the liver. Besides, this integrin also showed to be critical in vivo for tumor cell retention, cytokine release, leukocyte recruitment and metastasis development. These data support a therapeutical potential of the integrin LFA-1 as a target for the treatment of colorectal liver metastasis.


Subject(s)
CD18 Antigens/metabolism , Colorectal Neoplasms/metabolism , Liver Neoplasms/metabolism , Animals , BALB 3T3 Cells , Cell Adhesion , Cell Line, Tumor , Cell Movement , Cell Proliferation , Coculture Techniques , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Endothelium/immunology , Endothelium/pathology , Liver/pathology , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Lymphocyte Function-Associated Antigen-1/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Neoplasm Transplantation , T-Lymphocytes, Cytotoxic/immunology
19.
Oncol Lett ; 14(4): 3883-3892, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28943897

ABSTRACT

Intercellular adhesion molecule (ICAM)-1, is a transmembrane glycoprotein of the immunoglobulin (Ig)-like superfamily, consisting of five extracellular Ig-like domains, a transmembrane domain and a short cytoplasmic tail. ICAM-1 is expressed in various cell types, including endothelial cells and leukocytes, and is involved in several physiological processes. Furthermore, it has additionally been reported to be expressed in various cancer cells, including melanoma, colorectal cancer and lymphoma. The majority of studies to date have focused on the expression of the ICAM-1 on the surface of tumor cells, without research into ICAM-1 expression at sites of metastasis. Cancer cells frequently metastasize to the liver, due to its unique physiology and specialized liver sinusoid capillary network. Liver sinusoidal endothelial cells constitutively express ICAM-1, which is upregulated under inflammatory conditions. Furthermore, liver ICAM-1 may be important during the development of liver metastasis. Therefore, it is necessary to improve the understanding of the mechanisms mediated by this adhesion molecule in order to develop host-directed anticancer therapies.

20.
Oncol Lett ; 13(6): 4002-4012, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28599406

ABSTRACT

Colorectal cancer (CRC) is an aggressive disease in which patients usually die due to its metastatic progression to the liver. Up to date, irinotecan is one of the most used chemotherapeutic agents to treat CRC metastasis with demonstrated efficacy. However, the severity of the side effects constitute the main limitation to its use in the treatment. Consequently, new complementary therapies are being developed to avoid these adverse effects while maintaining the efficacy of the antitumoral drugs. Ocoxin oral solution (OOS®) is a nutritional mixture containing biologically active compounds with demonstrated antitumoral and immunomodulatory effects. Thus, we aimed to analyze the effect of OOS® as a suitable complement to irinotecan therapy in the treatment of CRC metastasis to the liver. First, the effect of OOS®, irinotecan and the combination of both on the viability of C26 cells was tested in vitro and in vivo. Second, the expression of caspase-3, Ki67 and the macrophage infiltration by F4/80 marker was quantified in liver tissue sections by immunohistochemistry. Finally, mRNA microarray study was carried out on tumor cells isolated from tumor-bearing livers collected from mice subjected to the above treatments. Our results show that OOS® administered as a complementary therapy to irinotecan reduced tumor cell viability in vitro. Moreover, irinotecan administered either alone or in combination with 100 µl OOS® from the 7th day after tumor cell inoculation decreased the metastatic growth in the liver. Besides, several genes with binding and catalytic activities showed to be deregulated by RNA microarray analysis. In conclusion, OOS®, when administered as a complement to irinotecan, reduced the metastatic development of colorectal cancer to the liver. Additionally, the overall health state of the animals improved. These results point out OOS® as a potential supplement to the anti-tumoral treatments used in clinical settings in patients suffering from disseminated colorectal cancer.

SELECTION OF CITATIONS
SEARCH DETAIL
...