Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Methods Cell Biol ; 188: 237-254, 2024.
Article in English | MEDLINE | ID: mdl-38880526

ABSTRACT

The prevalence of central nervous system (CNS) dysfunction as a result of disease or trauma remains a clinically unsolved problem which is raising increased awareness in our aging society. Human Dental Pulp Stem Cells (hDPSCs) are excellent candidates to be used in tissue engineering and regenerative therapies of the CNS due to their neural differentiation ability and lack of tumorigenicity. Accordingly, they have been successfully used in animal models of spinal cord injury, stroke and peripheral neuropathies. The ideal therapy in brain injury should combine strategies aiming to protect the damaged lesion and, at the same time, accelerate brain tissue regeneration, thus promoting fast recovery while minimizing side or long-term effects. The use of bioresorbable nanopatterned poly(lactide-co-ɛ-caprolactone) (PLCL) polymeric scaffolds as hDPCSs carriers can represent an advantage for tissue regeneration. In this chapter, we describe the surgical procedures to implant functionalized bioresorbable scaffolds loaded with hDPSCs to improve the brain lesion microenvironment in an intracranial stab wound injury model severing the rostral migratory stream (RMS) that connects the brain subventricular zone (SVZ) and the olfactory bulb in nude mice. Additionally, we also describe the technical steps after animal sacrifice for histological tissue observation and characterization.


Subject(s)
Dental Pulp , Disease Models, Animal , Mice, Nude , Stem Cells , Tissue Scaffolds , Dental Pulp/cytology , Animals , Humans , Tissue Scaffolds/chemistry , Mice , Stem Cells/cytology , Stem Cell Transplantation/methods , Wounds, Stab/therapy , Absorbable Implants , Brain Injuries/therapy , Brain Injuries/pathology , Tissue Engineering/methods
2.
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
3.
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
4.
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
5.
Int J Mol Sci ; 22(9)2021 May 01.
Article in English | MEDLINE | ID: mdl-34062897

ABSTRACT

Cancer is a phenomenon broadly related to ageing in various ways such as cell cycle deregulation, metabolic defects or telomerases dysfunction as principal processes. Although the tumor cell is the main actor in cancer progression, it is not the only element of the disease. Cells and the matrix surrounding the tumor, called the tumor microenvironment (TME), play key roles in cancer progression. Phenotypic changes of the TME are indispensable for disease progression and a few of these transformations are produced by epigenetic changes including miRNA dysregulation. In this study, we found that a specific group of miRNAs in the liver TME produced by colon cancer called geromiRs, which are miRNAs related to the ageing process, are significantly downregulated. The three principal cell types involved in the liver TME, namely, liver sinusoidal endothelial cells, hepatic stellate (Ito) cells and Kupffer cells, were isolated from a murine hepatic metastasis model, and the miRNA and gene expression profiles were studied. From the 115 geromiRs and their associated hallmarks of aging, which we compiled from the literature, 75 were represented in the used microarrays, 26 out of them were downregulated in the TME cells during colon cancer colonization of the liver, and none of them were upregulated. The histone modification hallmark of the downregulated geromiRs is significantly enriched with the geromiRs miR-15a, miR-16, miR-26a, miR-29a, miR-29b and miR-29c. We built a network of all of the geromiRs downregulated in the TME cells and their gene targets from the MirTarBase database, and we analyzed the expression of these geromiR gene targets in the TME. We found that Cercam and Spsb4, identified as prognostic markers in a few cancer types, are associated with downregulated geromiRs and are upregulated in the TME cells.


Subject(s)
Colonic Neoplasms/genetics , Hepatic Stellate Cells/metabolism , Liver/metabolism , MicroRNAs/genetics , Animals , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic/genetics , Hepatic Stellate Cells/pathology , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Kupffer Cells/metabolism , Kupffer Cells/pathology , Liver/pathology , Mice , MicroRNAs/classification
6.
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
7.
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
8.
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
9.
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.

10.
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
11.
Cancers (Basel) ; 12(9)2020 Aug 22.
Article in English | MEDLINE | ID: mdl-32842712

ABSTRACT

(1) Background & Aims: The roles of different cells in the tumor microenvironment (TME) are critical to the metastatic process. The phenotypic transformation of the liver cells is one of the most important stages of the hepatic metastasis progression of colorectal cancer (CRC). Our aim was to identify the major molecules (i.e., genes, miRNAs and proteins) involved in this process. (2) Methods: We isolated and performed whole-genome analysis of gene, miRNA, and protein expression in three types of liver cells (Ito cells, Kupffer cells, and liver sinusoidal endothelial cells) from the TME of a murine model of CRC liver metastasis. We selected the statistically significant differentially expressed molecules using the Student's t-test with Benjamini-Hochberg correction and performed functional statistically-significant enrichment analysis of differentially expressed molecules with hypergeometric distribution using the curated collection of molecular signatures, MSigDB. To build a gene-miRNA-protein network centered in Brca1, we developed a software package (miRDiana) that collects miRNA targets from the union of the TargetScan, MicroCosm, mirTarBase, and miRWalk databases. This was used to search for miRNAs targeting Brca1. We validated the most relevant miRNAs with real-time quantitative PCR. To investigate BRCA1 protein expression, we built tissue microarrays (TMAs) from hepatic metastases of 34 CRC patients. (3) Results: Using integrated omics analyses, we observed that the Brca1 gene is among the twenty transcripts simultaneously up-regulated in all three types of TME liver cells during metastasis. Further analysis revealed that Brca1 is the last BRCA1-associated genome surveillance complex (BASC) gene activated in the TME. We confirmed this finding in human reanalyzing transcriptomics datasets from 184 patients from non-tumor colorectal tissue, primary colorectal tumor and colorectal liver metastasis of the GEO database. We found that the most probable sequence of cell activation during metastasis is Endothelial→Ito→Kupffer. Immunohistochemical analysis of human liver metastases showed the BRCA1 protein was co-localized in Ito, Kupffer, and endothelial cells in 81.8% of early or synchronous metastases. However, in the greater part of the metachronous liver metastases, this protein was not expressed in any of these TME cells. (4) Conclusions: These results suggest a possible role of the co-expression of BRCA1 in Ito, Kupffer, and sinusoidal endothelial cells in the early occurrence of CRC liver metastases, and point to BRCA1 as a potential TME biomarker.

12.
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
13.
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
14.
Oncol Rep ; 41(2): 742-752, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30483808

ABSTRACT

Colon cancer is one of the most frequently occurring types of cancers in the world. Primary tumours are treated very efficiently, but the metastatic cases are known to have severe outcomes. Therefore, the aim of the present study was to obtain a greater understanding of the transformation of primary colon cancer cells into metastatic phenotypes. Small changes in protein expression provoke the metastatic phenotype transformation. More sensitive methods to detect small variations are required. A murine colon cancer cell line with metastatic characteristics in a very early phase was created in order to investigate the first steps of transformation using a murine liver metastasis model. The protein expression patterns of metastatic and non­metastatic cells were compared using the stable isotope labelling by amino acids in cell culture method in combination with mass spectrometry. Quantitative proteomics data indicated that nicotinamide adenine dinucleotide hydride (NADH) dehydrogenase complex I was overexpressed in metastatic cells with respect to non­metastatic cells. Since the NADH dehydrogenase complex catalyses the oxidation of NADH to NAD+, the functionality of the complex was studied by measuring the amount of NADH. The results revealed that metastatic cells accumulate more NADH and reactive oxygen species. In addition, the mitochondrial membrane potential of metastatic cells was lower than that of non­metastatic cells, indicating that the activity of NADH dehydrogenase and the mitochondrial oxidative chain were decreased in metastatic cells. During the incipient transformation of primary cancer cells, NADH dehydrogenase complex I was overexpressed but then became inactive due to the Warburg effect, which inhibits mitochondrial activity. In the first step of transformation, the high energy demand required in an adverse environment is fulfilled by overexpressing components of the respiratory chain, a fact that should be considered for future anti­metastatic therapies.


Subject(s)
Colonic Neoplasms/pathology , Electron Transport Complex I/metabolism , Liver Neoplasms/pathology , Mitochondria/pathology , NADH Dehydrogenase/metabolism , Animals , Cell Line, Tumor , Liver Neoplasms/secondary , Male , Mice , Mice, Inbred BALB C , Mitochondria/metabolism , NAD/analysis , NAD/metabolism , Reactive Oxygen Species
15.
Int J Cancer ; 143(3): 709-719, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29492958

ABSTRACT

Phenotypic transformation of liver sinusoidal endothelial cells is one of the most important stages of liver metastasis progression. The miRNA effects on liver sinusoidal endothelial cells during liver metastasis have not yet been studied. Herein, whole genome analysis of miRNA expression in these cells during colorectal liver metastasis revealed repressed expression of microRNA-20a. Importantly, downregulation of miR-20a occurs in parallel with upregulation of its known protein targets. To restore normal miR-20a levels in liver sinusoidal endothelial cells, we developed chondroitin sulfate-sorbitan ester nanoparticles conjugated with miR-20a in a delivery system that specifically targets liver sinusoidal endothelial cells. The restoration of normal mir-20a levels in these cells induced downregulation of the expression of its protein targets, and this also resulted in a reduction of in vitro LSEC migration and a reduction of in vivo activation and tumor-infiltrating capacity and ability of the tumor decreased by ∼80% in a murine liver metastasis model.


Subject(s)
Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Endothelial Cells/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/secondary , MicroRNAs/genetics , Nanoparticles , Animals , Biomarkers , Cell Line, Tumor , Cells, Cultured , Colonic Neoplasms/metabolism , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Heterografts , Liver Neoplasms/metabolism , Liver Neoplasms/therapy , Male , Mice , MicroRNAs/chemistry , Nanoparticles/chemistry , Signal Transduction
16.
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
17.
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.

18.
Oncol Rep ; 35(3): 1265-72, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26676882

ABSTRACT

Liver metastatic disease is the main cause of death in colorectal cancer (CRC) patients. During metastatic spread of the disease an imbalance in the oxidative stress and inflammation plays a crucial role in tumor progression. In order to improve the efficacy of current therapies, new complementary therapeutic approaches are being analyzed including biologically active compounds with low side effects. The anti-inflammatory and anti-oxidant properties of Ocoxin® oral solution (OOS) prompt us to analyze its effect on the metastatic development of CRC to the liver. First, in vitro effect of OOS in tumor cell viability and migration was analyzed. Second, in vivo effect of different dosage patterns and concentrations in the development of hepatic metastasis was analyzed by intra-splenic inoculation of C26 colon carcinoma cells in Balb/c mice. Third, the expression of alpha smooth muscle actin, caspase-3 and Ki-67 expression was quantified by immunohistochemistry, then gene expression levels of inflammatory factors were measured by quantitative RT-PCR. According to our results, OOS reduced tumor cell viability and migration in vitro. Moreover, in vivo daily administration of OOS from the 7th day after tumor cell inoculation decreased the total area and size of metastatic foci in the liver. Furthermore, cell proliferation and fibroblast recruitment was decreased in tumor foci while a higher number of apoptotic cells were observed. Finally, RNA levels for the inflammatory mediators COX-2, IFNγ, IL1ß, IL6 and TNFα were reduced in total liver. In conclusion, OOS reduced the metastatic development of colorectal cancer to the liver by increasing apoptosis, and decreasing tumor cell proliferation and fibroblast recruitment in the tumor foci, as well as the expression of inflammatory mediators in total liver. These results point out OOS as a potential supplement to be applied as complementary therapy for the treatment of liver metastasis from colorectal cancer.


Subject(s)
Ascorbic Acid/administration & dosage , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Glycyrrhizic Acid/administration & dosage , Liver Neoplasms/drug therapy , Plant Extracts/administration & dosage , Vitamin B 12/administration & dosage , Vitamin B 6/administration & dosage , Zinc/administration & dosage , Animals , Apoptosis/drug effects , Caspase 3/biosynthesis , Colorectal Neoplasms/pathology , Folic Acid , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ki-67 Antigen/biosynthesis , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Mice , Mice, Inbred BALB C , Neoplasm Proteins/biosynthesis , Pantothenic Acid , Vitamin B 12 Deficiency , Zinc Sulfate
19.
Histol Histopathol ; 29(11): 1355-64, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24781958

ABSTRACT

Until recently, collagen interactions with cells had been ascribed to integrins. The identification of the Discoidin Domain Receptor (DDR) family as collagen receptors represents a new paradigm in the regulation of collagen-cell interactions. How DDR signaling is biochemically linked to specific cell regulatory functions remains largely unknown. Moreover, the characteristic slow and substained phosphorylation of DDRs and the elevated expression of DDR2 in the myofibroblasts of healing wounds suggest a role for DDR2 in physiological and pathological wound healing. In fact, DDR2 signaling regulates cell proliferation and extracellular matrix synthesis, which are key aspects of fibroblast contribution to tissue healing. In this review we summarize evidence in favor of this concept.


Subject(s)
Fibroblasts/metabolism , Gene Expression Regulation , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Mitogen/metabolism , Skin/metabolism , Wound Healing/physiology , Animals , Cell Proliferation , Collagen/chemistry , Discoidin Domain Receptors , Extracellular Matrix/metabolism , Humans , Integrins/metabolism , Mice , Signal Transduction
20.
Int J Cancer ; 133(9): 2113-22, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23629598

ABSTRACT

Random homozygous gene perturbation (RHGP), in combination with liver sinusoidal endothelial cell (LSEC) adhesion screening of clonal colon cancer cells with perturbed genes, was used to identify genes contributing to the hepatic microvascular adhesion of colon cancer cells. Plasmid vector encoding transactivator and gene search vector were transfected into HT-29 human colorectal cancer cells to create a HT-29 RHGP cell library; the adhesion of these library cells to primary cultured mouse LSEC significantly decreased in the presence of RSL1 ligand (inducer), indicating that most of the genes contributing to HT-29 adhesion to LSEC were altered. Next, HT-29 RHGP cell library fractions with upregulated or silenced LSEC adhesion-related genes were isolated. Around 160 clones having altered expression in LSEC adhesion-related genes were obtained, and nine relevant protein-coding genes were identified. Some were proadhesive genes detected because of their overexpression in adherent HT-29 cells (DGCR8 and EFEMP1 genes) and their silenced status in nonadherent HT-29 cells (DGKE, DPY19L1, KIAA0753, PVR and USP11 genes). Others were antiadhesive genes detected because of their overexpression in nonadherent HT-29 cells (ITPKC gene) and their silenced status in adherent HT-29 cells (PPP6R2 gene). Silencing of PVR, DGCR8 and EFEMP1 genes decreased adhesion to LSEC and hepatic microvascular retention of HT-29 cells. The results conclude that RHGP was a valuable strategy for the discovery of mechanisms regulating microvascular adhesion of circulating colon cancer cells before hepatic metastasis formation. Identified genes may contribute to understand the metastatic process of colon cancer and to discovering molecular targets for hepatic metastasis therapeutics.


Subject(s)
Biomarkers, Tumor/genetics , Cell Adhesion/genetics , Colonic Neoplasms/genetics , Endothelial Cells/metabolism , Liver Neoplasms/genetics , Liver/blood supply , Animals , Apoptosis/drug effects , Biomarkers, Tumor/metabolism , Blotting, Western , Cells, Cultured , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Endothelial Cells/pathology , Flow Cytometry , Gene Expression Profiling , HT29 Cells , Humans , Liver/metabolism , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL