Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
J Oral Pathol Med ; 53(9): 567-576, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39160673

ABSTRACT

OBJECTIVE: Tumor hypoxia is associated with a poorer prognosis in cancer patients and can diminish the efficacy of radiation therapy (RT). This study investigates the potential of metformin to enhance radiosensitivity in hypoxic cancer cells. METHODS: Preliminary experiments were conducted to validate the impact of hypoxia on radiation response. Reactive oxygen species (ROS) levels, cell migration, and cell death were assessed in hypoxic, radiated cells treated with metformin. Proteomic and ontological analyses were employed to identify molecular targets associated with the radiosensitizing effect of metformin. Proteomic and ontological findings were validated through patient samples and in vitro studies. RESULTS: Metformin amplified cell death, induced DNA fragmentation, decreased cell migration, and elevated ROS levels in hypoxic, radiated cells. Proteomic analyses revealed that GAPDH and TAGLN2 were identified as pivotal targets linked to the radiosensitizing effect of metformin. Oral cancer patients exhibited elevated levels of TAGLN2 and reduced levels of GAPDH. Metformin downregulated TAGLN2 and upregulated GAPDH in hypoxic, radiated cells. Additionally, metformin reduced levels of mutated p53. CONCLUSIONS: This study suggests that metformin can enhance radiosensitivity in hypoxic cells, operating through modulation of GAPDH and TAGLN2. Furthermore, metformin effectively reduces mutated p53 levels in radiated cells under hypoxic conditions.


Subject(s)
Carcinoma, Squamous Cell , Metformin , Mouth Neoplasms , Radiation-Sensitizing Agents , Humans , Metformin/pharmacology , Metformin/therapeutic use , Mouth Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Radiation Tolerance/drug effects , Reactive Oxygen Species/metabolism , Proteomics , Glyceraldehyde-3-Phosphate Dehydrogenases , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating) , Cell Hypoxia/drug effects , Tumor Hypoxia/drug effects
2.
Mol Biol Rep ; 49(5): 3885-3891, 2022 May.
Article in English | MEDLINE | ID: mdl-35277789

ABSTRACT

BACKGROUND: Hypoxia pathways are deregulated in clear renal cell carcinoma (ccRCC) because of the loss of the von Hippel-Lindau tumor suppressor function. Quantitative PCR is a powerful tool for quantifying differential expression between normal and cancer cells. Reliable gene expression analysis requires the use of genes encoding housekeeping genes. Therefore, in this study, eight reference candidate genes were evaluated to determine their stability in 786-0 cells under normoxic and hypoxic conditions. METHODS AND RESULTS: Four different tools were used to rank the most stable genes-geNorm, NormFinder, BestKeeper, and Comparative Ct (ΔCt), and a general ranking was performed using RankAggreg. According to the four algorithms, the TFRC reference gene was identified as the most stable. There was no agreement among the results from the algorithms for the 2nd and 3rd positions. A general classification was then established using the RankAggreg tool. Finally, the three most suitable reference genes for use in 786-0 cells under normoxic and hypoxic conditions were TFRC, RPLP0, and SDHA. CONCLUSIONS: To the best of our knowledge, this is the first study to identify reliable genes that can be used for gene expression analysis in ccRCC in a hypoxic environment.


Subject(s)
Carcinoma, Renal Cell , Genes, Essential , Kidney Neoplasms , Tumor Hypoxia , Carcinoma, Renal Cell/genetics , Gene Expression , Gene Expression Profiling/methods , Humans , Kidney Neoplasms/genetics , Real-Time Polymerase Chain Reaction , Reference Standards
3.
Int J Mol Sci ; 23(3)2022 Feb 03.
Article in English | MEDLINE | ID: mdl-35163668

ABSTRACT

Breast cancer is characterized by a hypoxic microenvironment inside the tumor mass, contributing to cell metastatic behavior. Hypoxia induces the expression of hypoxia-inducible factor (HIF-1α), a transcription factor for genes involved in angiogenesis and metastatic behavior, including the vascular endothelial growth factor (VEGF), matrix metalloproteinases (MMPs), and integrins. Integrin receptors play a key role in cell adhesion and migration, being considered targets for metastasis prevention. We investigated the migratory behavior of hypoxia-cultured triple-negative breast cancer cells (TNBC) and endothelial cells (HUVEC) upon αvß3 integrin blocking with DisBa-01, an RGD disintegrin with high affinity to this integrin. Boyden chamber, HUVEC transmigration, and wound healing assays in the presence of DisBa-01 were performed in hypoxic conditions. DisBa-01 produced similar effects in the two oxygen conditions in the Boyden chamber and transmigration assays. In the wound healing assay, hypoxia abolished DisBa-01's inhibitory effect on cell motility and decreased the MMP-9 activity of conditioned media. These results indicate that αvß3 integrin function in cell motility depends on the assay and oxygen levels, and higher inhibitor concentrations may be necessary to achieve the same inhibitory effect as in normoxia. These versatile responses add more complexity to the role of the αvß3 integrin during tumor progression.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Endothelial Cells/metabolism , Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/metabolism , Tumor Hypoxia , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Shape/drug effects , Crotalid Venoms/pharmacology , Culture Media, Conditioned/pharmacology , Disintegrins/pharmacology , Endothelial Cells/pathology , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Matrix Metalloproteinases/metabolism , Neovascularization, Physiologic/drug effects , Oxygen , Protein Subunits/metabolism , Tumor Hypoxia/drug effects
4.
BMC Cancer ; 21(1): 1248, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34798868

ABSTRACT

BACKGROUND: Glioblastoma is a fatal brain tumour with a poor patient survival outcome. Hypoxia has been shown to reprogram cells towards a stem cell phenotype associated with self-renewal and drug resistance properties. Activation of hypoxia-inducible factors (HIFs) helps in cellular adaptation mechanisms under hypoxia. Similarly, miRNAs are known to be dysregulated in GBM have been shown to act as critical mediators of the hypoxic response and to regulate key processes involved in tumorigenesis. METHODS: Glioblastoma (GBM) cells were exposed to oxygen deprivation to mimic a tumour microenvironment and different cell aspects were analysed such as morphological changes and gene expression of miRNAs and survival genes known to be associated with tumorigenesis. RESULTS: It was observed that miR-128a-3p, miR-34-5p, miR-181a/b/c, were down-regulated in 6 GBM cell lines while miR-17-5p and miR-221-3p were upregulated when compared to a non-GBM control. When the same GBM cell lines were cultured under hypoxic microenvironment, a further 4-10-fold downregulation was observed for miR-34-5p, miR-128a-3p and 181a/b/c while a 3-6-fold upregulation was observed for miR-221-3p and 17-5p for most of the cells. Furthermore, there was an increased expression of SOX2 and Oct4, GLUT-1, VEGF, Bcl-2 and survivin, which are associated with a stem-like state, increased metabolism, altered angiogenesis and apoptotic escape, respectively. CONCLUSION: This study shows that by mimicking a tumour microenvironment, miRNAs are dysregulated, stemness factors are induced and alteration of the survival genes necessary for the cells to adapt to the micro-environmental factors occurs. Collectively, these results might contribute to GBM aggressiveness.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , MicroRNAs/metabolism , Tumor Hypoxia/genetics , Tumor Microenvironment/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Down-Regulation , Genotype , Glioblastoma/metabolism , Glioblastoma/pathology , Glucose Transporter Type 1/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Octamer Transcription Factor-3/metabolism , Phenotype , Proto-Oncogene Proteins c-bcl-2/metabolism , SOXB1 Transcription Factors/metabolism , Survivin/metabolism , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
5.
Clin Transl Oncol ; 23(9): 1874-1884, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33811629

ABSTRACT

PURPOSE: Molecular mechanisms of uveal melanoma development in association with high pigmentation are unclear. Tyrosinase Related Protein (TYRP1) is not only one of the important melanogenesis marker that contributes to melanin synthesis, but can also prevents the melanocyte death. The induction of melanogenesis leads to induction of HIF-1α which can affect the behavior of melanoma cells and its surrounding environment. The aim of our study was to determine the expression of TYRP1 and HIF-1α at the protein and RNA level and determine its prognostic significance. METHODS: In the present study, the expression of TYRP1 and HIF-1α was investigated on 61 formalin-fixed paraffin-embedded choroidal melanoma samples by immunohistochemistry. Fresh 50 samples were validated by real-time PCR. Results were correlated with clinicopathological parameters and Kaplan-Meier was performed to determine the prognostic significance. RESULTS: High immunoexpression of TYRP1 and HIF-1α was present in 61 and 54% of patients, respectively. Both TYRP1 and HIF-1α correlated well with high pigmentation and BAP1 (BRCA1 Associated Protein-1) loss (p < 0.05) at IHC level as well as transcriptional level. There was reduced metastatic free survival in patients with necrosis and this was statistically significant (p = 0.010). CONCLUSION: Our findings indicate that TYRP1 can be used as a potential biomarker in the development of targeted therapy in UM. Further studies on melanogenesis markers associated with TYRP1 could provide us a better understanding in this field.


Subject(s)
Biomarkers, Tumor/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Melanoma/metabolism , Membrane Glycoproteins/metabolism , Oxidoreductases/metabolism , Tumor Hypoxia , Uveal Neoplasms/metabolism , Adult , Choroid , Female , Humans , Kaplan-Meier Estimate , Male , Melanins/biosynthesis , Melanoma/mortality , Melanoma/pathology , Pigmentation , Risk Factors , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism , Uveal Neoplasms/mortality , Uveal Neoplasms/pathology
6.
Clin Transl Oncol ; 23(8): 1678-1687, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33625672

ABSTRACT

BACKGROUND: The study examines the function of hypoxia-mediated down-regulation of microRNAs (miRNAs) (mir-30c, mir-135a, and mir-27a) in the process of bladder cancer immune escape. METHODS: Quantitative Real-time PCR (qRT-PCR) was carried out to determine gene expression levels of Drosha and Dicer under hypoxia treatment, while western blotting and flow cytometry were used to determine protein expression. Seven reported miRNAs were identified via qRT-PCR assay. Flow cytometry detection of CD3/CD4/CD8-positive expression and statistics. Enzyme-linked immunosorbent assay (ELISA) detected cellular immune factors content. Cell apoptosis was checked via flow cytometry assay. Luciferase report assay and western blot assays were both used to verify the relationship between miRNAs and Casitas B-lineage lymphoma proto-oncogene b (Cbl-b). The animal model was established and Hematoxylin-eosin (HE) staining, TdT-mediated dUTP Nick-End Labeling (TUNEL) staining, and immunohistochemistry (IHC) assays were separately used to verify the conclusions. RESULTS: The CD3 + /CD4 + expression was increased in the hypoxia group, while CD3 + /CD8 + expression, the cellular immune factors content Interleukin-2 (IL-2) and Tumor Necrosis Factor-α (TNFα) along with the cell apoptosis were suppressed. The protein expression of Cbl-b was found to be up-regulated in the hypoxia group. After constructing the overexpression/ knockdown of Cbl-b in peripheral blood mononuclear cell (PBMC), Cbl-b has been found to promote tumor immune escape in bladder cancer. Furthermore, Cbl-b had been identified as the co-targets of mir-30c, mir-135a, and mir-27a and down-regulation of miRNA biogenesis promotes Cbl-b expression and deactivating T cells in vitro/in vivo. CONCLUSION: Hypoxia-mediated down-regulation of miRNAs' biogenesis promotes tumor immune escape in bladder cancer, which could bring much more advance to the medical research on tumors.


Subject(s)
Down-Regulation/immunology , MicroRNAs/metabolism , Tumor Escape/immunology , Tumor Hypoxia/immunology , Urinary Bladder Neoplasms/immunology , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis , CD3 Complex/metabolism , CD4 Antigens/metabolism , CD8 Antigens/metabolism , DEAD-box RNA Helicases/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Interleukin-2/metabolism , Leukocytes, Mononuclear/metabolism , Male , Mice, Inbred C57BL , MicroRNAs/biosynthesis , Prospective Studies , Proto-Oncogene Mas , Proto-Oncogene Proteins c-cbl/genetics , Random Allocation , Ribonuclease III/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation , Urinary Bladder Neoplasms/metabolism
7.
Mol Biol Rep ; 48(2): 1161-1169, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33547534

ABSTRACT

Hypoxia is associated with tumor aggressiveness and poor prognosis, including breast cancer. Low oxygen levels induces global genomic hypomethylation and hypermethylation of specific loci in tumor cells. DNA methylation is a reversible epigenetic modification, usually associated with gene silencing, contributing to carcinogenesis and tumor progression. Since the effects of DNA methyltransferase inhibitor are context-dependent and as there is little data comparing their molecular effects in normoxic and hypoxic microenvironments in breast cancer, this study aimed to understand the gene expression profiles and molecular effects in response to treatment with DNA methyltransferase inhibitor in normoxia and hypoxia, using the breast cancer model. For this, a cDNA microarray was used to analyze the changes in the transcriptome upon treatment with DNA methyltransferase inhibitor (5-Aza-2'-deoxycytidine: 5-Aza-2'-dC), in normoxia and hypoxia. Furthermore, immunocytochemistry was performed to investigate the effect of 5-Aza-2'-dC on NF-κB/p65 inflammation regulator subcellular localization and expression, in normoxia and hypoxia conditions. We observed that proinflammatory pathways were upregulated by treatment with 5-Aza-2'-dC, in both conditions. However, treatment with 5-Aza-2'-dC in normoxia showed a greater amount of overexpressed proinflammatory pathways than 5-Aza-2'-dC in hypoxia. In this sense, we observed that the NF-κB expression increased only upon 5-Aza-2'-dC in normoxia. Moreover, nuclear staining for NF-κB and NF-κB target genes upregulation, IL1A and IL1B, were also observed after 5-Aza-2'-dC in normoxia. Our results suggest that 5-Aza-2'-dC induces a greater inflammatory change, at the molecular levels, in normoxic than hypoxic tumor microenvironment. These data may support further studies and expand the understanding of the DNA methyltransferase inhibitor effects in different tumor contexts.


Subject(s)
DNA Methylation/drug effects , DNA Modification Methylases/genetics , Decitabine/pharmacology , Inflammation/genetics , Acetylation/drug effects , Cell Line, Tumor , DNA Modification Methylases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/genetics , Humans , Inflammation/chemically induced , Inflammation/pathology , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/genetics , Transcription Factor RelA/genetics , Tumor Hypoxia/drug effects , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics
8.
ACS Appl Bio Mater ; 4(1): 195-228, 2021 01 18.
Article in English | MEDLINE | ID: mdl-35014281

ABSTRACT

Photodynamic therapy (PDT) is a minimally invasive clinical protocol that combines a nontoxic photosensitizer (PS), appropriate visible light, and molecular oxygen for cancer treatment. This triad generates reactive oxygen species (ROS) in situ, leading to different cell death pathways and limiting the arrival of nutrients by irreversible destruction of the tumor vascular system. Despite the number of formulations and applications available, the advancement of therapy is hindered by some characteristics such as the hypoxic condition of solid tumors and the limited energy density (light fluence) that reaches the target. As a result, the use of PDT as a definitive monotherapy for cancer is generally restricted to pretumor lesions or neoplastic tissue of approximately 1 cm in size. To expand this limitation, researchers have synthesized functional nanoparticles (NPs) capable of carrying classical photosensitizers with self-supplying oxygen as well as targeting specific organelles such as mitochondria and lysosomes. This has improved outcomes in vitro and in vivo. This review highlights the basis of PDT, many of the most commonly used strategies of functionalization of smart NPs, and their potential to break the current limits of the classical protocol of PDT against cancer. The application and future perspectives of the multifunctional nanoparticles in PDT are also discussed in some detail.


Subject(s)
Nanostructures/chemistry , Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/therapeutic use , Animals , Apoptosis/drug effects , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Nanostructures/therapeutic use , Nanostructures/toxicity , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology , Reactive Oxygen Species/metabolism , Tumor Hypoxia/drug effects
9.
Methods Mol Biol ; 2174: 3-12, 2021.
Article in English | MEDLINE | ID: mdl-32813240

ABSTRACT

The inherent or developed resistance of many cancer cells to chemotherapy and irradiation is actually the main challenge to overcome in cancer treatment. It is well known that cancer cells are characterized by several hallmarks, and it seems that the ability to evolve ways to evade stressful conditions and killing therapies must be consider another typical characteristic displayed by all malignant cells. This overview aims to provide a concise description of the main mechanisms involved in the promotion of resistance to anticancer therapy and to describe the most frequent challenges faced in the war against cancer therapy resistance.


Subject(s)
Drug Resistance, Neoplasm/physiology , Neoplasms/drug therapy , Neoplasms/radiotherapy , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/radiation effects , Humans , Immunotherapy/methods , Molecular Targeted Therapy , Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , Tumor Hypoxia
10.
Clin Transl Oncol ; 23(2): 246-256, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32519176

ABSTRACT

PURPOSE: Clinically, hypoxia is associated with increased distant metastasis and poor survival in gastric cancer (GC). In this study, we set out from the cellular interaction to further explain the molecular mechanism of invasion in GC cells under hypoxic conditions. METHODS: Gastric cancer cells were cultured under 1% O2 (hypoxia-cultured gastric cancer cells, HGC) and 20% O2 condition (normoxic-cultured gastric cancer cells, NGC). NGC was co-cultured with HGC-medium. Scrape and Transwell were used to evaluate invasion and migration. Exosomes from GC were extracted by ultracentrifugation. Electron microscopy images, western-blot used to analyze the size distributions and the number of exosomes. RESULTS: HGC-medium induced NGC dissociated. Long non-coding RNA (lncRNA) prostate cancer gene expression marker 1 (PCGEM1) was specifically expressed in HGC exosomes. HGC-derived PCGEM1-riched exosomes could promote the invasion and migration of NGC. On the mechanism, PCGEM1 maintained stability and reduced the degradation of SNAI1, which could induce the epithelial-mesenchymal transition of GC. CONCLUSION: LncRNA PCGEM1 was overexpressed in GC cells. And part of the PCGEM1 can be encapsulated into exosomes. These exosomes promoted invasion and migration of other GC cells. We considered PCGEM1 might act as a "scaffold" combined with SNAI1 and prompt the invasion and migration of GC.


Subject(s)
Exosomes/metabolism , RNA, Long Noncoding/metabolism , Snail Family Transcription Factors/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Tumor Hypoxia , Cell Communication , Cell Movement/genetics , Coculture Techniques/methods , Culture Media/pharmacology , Epithelial-Mesenchymal Transition/physiology , Exosomes/ultrastructure , Humans , Neoplasm Invasiveness , Neoplasm Metastasis , RNA, Messenger/metabolism , Snail Family Transcription Factors/genetics , Tumor Microenvironment , Up-Regulation
11.
Cell Biochem Funct ; 39(4): 478-487, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33377261

ABSTRACT

Low levels of oxygen (hypoxia) have been reported in solid tumours. This hypoxic microenvironment modulates the expression of genes linked to a more aggressive disease. However, it is unclear if the expression of drug-metabolizing enzymes as cytochromes P450 (CYPs) is affected by hypoxia in cancer. We aimed to define which cytochromes are affected by hypoxia using a liver cancer model in vitro. For this purpose, we assessed whole-genome expression microarrays of HepG2 liver cancer cell line from free repository databases, looking for gene expression hypoxia-associated profiles and selected those cytochromes with significant differences. Then, we corroborated their mRNA expression and protein levels by RT-qPCR and western blot, respectively, as well as immunofluorescence. Based on microarray analysis, we found that the expression of CYP2S1 and CYP24A1 were up-regulated with at least twice fold change compared with normoxia. The levels of mRNA and protein of CYP2S1 and CYP24A1 were increased significantly in hypoxic conditions (P < .05), and this tendency was also observed by immunofluorescence assays. Our data show that the expression of cytochromes CYP2S1 and CYP24A1 are induced in hypoxia, being the first time that CYP24A1 expression is associated with tumour hypoxia; which might have consequences in cancer progression and drug resistance. SIGNIFICANCE OF THE STUDY: Hypoxia is among the most important factors for cellular adaptation to stress. Especially in cancer, a major public health issue, hypoxia plays a substantial role in angiogenesis, metastasis and resistance to therapy. Tumoral hypoxia has been described at least in the brain, breast, cervical, liver, renal, lung, pancreatic and renal cancer. However, the understanding of how hypoxia drives cancer progression is still a major challenge. One emerging question is the role of hypoxia over the expression of drug-metabolizing enzymes, with a significant impact on drug treatment. In this context, our paper focus on the effect of hypoxia on CYPs, which is an essential group of drug-metabolizing enzymes. We show that hypoxia induces the expression of two members of the CYPs family: CYP2S1 and CYP24A1. Importantly, CYP2S1 is a major metabolizer of carcinogenic substances being relevant that hypoxia could promote this function. Interestingly, CYP24A1 limits the action of the active form of vitamin D, which is an anti-proliferative factor in cancer. Our evidence shows for the first time that hypoxia can induce CYP24A1 expression, with a potential effect on cancer progression. Our contribution clarifies a particular effect of tumoral hypoxia and the implications will be useful in the understanding of the progression of cancer, the resistance to treatment and the development of alternative therapies.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Hypoxia/metabolism , Liver Neoplasms/metabolism , Tumor Hypoxia , Vitamin D3 24-Hydroxylase/metabolism , Computational Biology , Cytochrome P-450 Enzyme System/genetics , Humans , Liver Neoplasms/pathology , Tumor Cells, Cultured , Vitamin D3 24-Hydroxylase/genetics
12.
Biochim Biophys Acta Rev Cancer ; 1875(1): 188455, 2021 01.
Article in English | MEDLINE | ID: mdl-33148499

ABSTRACT

Dysregulation of mRNA translation is involved in the onset and progression of different types of cancer. To gain insight into novel genetic strategies to avoid this malady, we reviewed the available genomic, transcriptomic, and proteomic data about the translational machinery from the naked-mole rat (NMR) Heterocephalus glaber, a new model of study that exhibits high resistance to cancer. The principal features that might confer cancer resistance are 28S rRNA fragmentation, RPL26 and eIF4G overexpression, global downregulation of mTOR pathway, specific amino acid residues in RAPTOR (P908) and RICTOR (V1695), and the absence of 4E-BP3. These features are not only associated with cancer but also might couple longevity and adaptation to hypoxia. We propose that the regulation of translation is among the strategies endowing NMR cancer resistance.


Subject(s)
Disease Resistance/genetics , Mole Rats/genetics , Neoplasms/genetics , Transcriptome/genetics , Animals , Eukaryotic Initiation Factor-4G/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Longevity/genetics , Neoplasms/pathology , RNA, Ribosomal, 28S/genetics , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Regulatory-Associated Protein of mTOR/genetics , Tumor Hypoxia/genetics
13.
Sci Rep ; 10(1): 22302, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33339852

ABSTRACT

Tumor hypoxia and the hypoxia inducible factor-1, HIF-1, play critical roles in cancer progression and metastasis. We previously showed that hypoxia activates the endosomal GTPase Rab5, leading to tumor cell migration and invasion, and that these events do not involve changes in Rab protein expression, suggesting the participation of intermediate activators. Here, we identified ALS2, a guanine nucleotide exchange factor that is upregulated in cancer, as responsible for increased Rab5-GTP loading, cell migration and metastasis in hypoxia. Specifically, hypoxia augmented ALS2 mRNA and protein levels, and these events involved HIF-1α-dependent transcription, as shown by RNAi, pharmacological inhibition, chromatin immunoprecipitation and bioinformatics analyses, which identified a functional HIF-1α-binding site in the proximal promoter region of ALS2. Moreover, ALS2 and Rab5 activity were elevated both in a model of endogenous HIF-1α stabilization (renal cell carcinoma) and by following expression of stable non-hydroxylatable HIF-1α. Strikingly, ALS2 upregulation in hypoxia was required for Rab5 activation, tumor cell migration and invasion, as well as experimental metastasis in C57BL/6 mice. Finally, immunohistochemical analyses in patient biopsies with renal cell carcinoma showed that elevated HIF-1α correlates with increased ALS2 expression. Hence, this study identifies ALS2 as a novel hypoxia-inducible gene associated with tumor progression and metastasis.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Carcinoma, Renal Cell/genetics , Guanine Nucleotide Exchange Factors/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Transcriptional Activation/genetics , Tumor Hypoxia , rab5 GTP-Binding Proteins/genetics
14.
Oxid Med Cell Longev ; 2020: 3176375, 2020.
Article in English | MEDLINE | ID: mdl-33149807

ABSTRACT

A hypoxic microenvironment is a hallmark in different types of tumors; this phenomenon participates in a metabolic alteration that confers resistance to treatments. Because of this, it was proposed that a combination of 2-methoxyestradiol (2-ME) and sodium dichloroacetate (DCA) could reduce this alteration, preventing proliferation through the reactivation of aerobic metabolism in lung adenocarcinoma cell line (A549). A549 cells were cultured in a hypoxic chamber at 1% O2 for 72 hours to determine the effect of this combination on growth, migration, and expression of hypoxia-inducible factors (HIFs) by immunofluorescence. The effect in the metabolism was evaluated by the determination of glucose/glutamine consumption and the lactate/glutamate production. The treatment of 2-ME (10 µM) in combination with DCA (40 mM) under hypoxic conditions showed an inhibitory effect on growth and migration. Notably, this reduction could be attributed to 2-ME, while DCA had a predominant effect on metabolic activity. Moreover, this combination decreases the signaling of HIF-3α and partially HIF-1α but not HIF-2α. The results of this study highlight the antitumor activity of the combination of 2-ME 10 µl/DCA 40 mM, even in hypoxic conditions.


Subject(s)
2-Methoxyestradiol/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Dichloroacetic Acid/therapeutic use , Lung Neoplasms/drug therapy , Tumor Hypoxia , Tumor Microenvironment , 2-Methoxyestradiol/pharmacology , A549 Cells , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis Regulatory Proteins/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Dichloroacetic Acid/pharmacology , Glucose/metabolism , Glutamic Acid/metabolism , Glutamine/metabolism , Glycolysis/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lactic Acid/metabolism , Lung Neoplasms/pathology , Repressor Proteins/metabolism , Signal Transduction/drug effects , Tumor Hypoxia/drug effects , Tumor Microenvironment/drug effects , Wound Healing/drug effects
15.
Int J Mol Sci ; 21(20)2020 Oct 13.
Article in English | MEDLINE | ID: mdl-33066331

ABSTRACT

Alterations in microRNA (miRNA) profiles, induced by tumor microenvironment stressors, like hypoxia, allow cancer cells to acquire immune-resistance phenotypes. Indeed, hypoxia-induced miRNAs have been implicated in cancer progression through numerous cancer cell non-autonomous mechanisms, including the direct transfer of hypoxia-responsive miRNA from cancer to immune cells via extracellular vesicles. Connexin-43 (Cx43)-constituted gap junctions (GJs) have also been involved in miRNA intercellular mobilization, in other biological processes. In this report, we aimed to evaluate the involvement of Cx43-GJs in the shift of miRNAs induced by hypoxia, from hypoxic melanoma cells to dendritic cells and melanoma-specific cytotoxic T lymphocytes (CTLs). Using qRT-PCR arrays, we identified that miR-192-5p was strongly induced in hypoxic melanoma cells. Immune cells acquired this miRNA after co-culture with hypoxic melanoma cells. The transfer of miR-192-5p was inhibited when hypoxic melanoma cells expressed a dominant negative Cx43 mutant or when Cx43 expression was silenced using specific short-hairpin RNAs. Interestingly, miR-192-5p levels on CTLs after co-culture with hypoxic melanoma cells were inversely correlated with the cytotoxic activity of T cells and with ZEB2 mRNA expression, a validated immune-related target of miR-192-5p, which is also observed in vivo. Altogether, our data suggest that hypoxic melanoma cells may suppress CTLs cytotoxic activity by transferring hypoxia-induced miR-192-5p through a Cx43-GJs driven mechanism, constituting a resistance strategy for immunological tumor escape.


Subject(s)
Connexin 43/metabolism , Dendritic Cells/metabolism , Melanoma/metabolism , MicroRNAs/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Tumor Hypoxia , Animals , Cell Line, Tumor , Connexin 43/genetics , Gap Junctions/metabolism , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/genetics
16.
Bol. méd. Hosp. Infant. Méx ; 77(4): 186-194, Jul.-Aug. 2020. graf
Article in English | LILACS | ID: biblio-1131975

ABSTRACT

Abstract Background: Acute lymphoblastic leukemia (ALL) is an aggressive malignant disease with high prevalence in pediatric patients. It has been shown that the downregulation of Fas expression is correlated with an inadequate response in ALL, although these mechanisms are still not well understood. Several reports demonstrated that hypoxia is involved in dysfunctional apoptosis. Yin-Yang-1 (YY1) transcription factor is involved in resistance to apoptosis, tumor progression, and it is increased in different types of cancer, including leukemia. The regulatory mechanism underlying YY1 expression in leukemia is still not understood, but it is known that YY1 negatively regulates Fas expression. The study aimed to evaluate the effect of YY1 on Fas expression under hypoxic conditions in ALL. Methods: Leukemia cell line RS4; 11 was cultured under normoxic and hypoxic conditions. YY1, Fas receptor, and hypoxia-inducible factor (HIF-1α) expression were analyzed. After treatment with a Fas agonist (DX2), apoptosis was analyzed through the detection of active caspase 3. Data were analyzed using Pearson’s correlation. Results: Leukemia cells co-expressed both HIF-1α and YY1 under hypoxia, which correlated with a downregulation of Fas expression. During hypoxia, the levels of apoptosis diminished after DX2 treatment. The analysis revealed that patients with high levels of HIF-1α also express high levels of YY1 and low levels of Fas. Conclusions: These results suggest that YY1 negatively regulates the expression of the Fas receptor, which could be involved in the escape of leukemic cells from the immune response contributing to the ALL pathogenesis.


Resumen Introducción: La leucemia linfoblástica aguda (LLA) es una enfermedad con alta prevalencia en la población pediátrica. El mecanismo por el cual el receptor de Fas participa en la regulación inmunitaria en los tumores es desconocido, pero se sabe que está subexpresado en LLA. El factor de transcripción Ying-Yang-1 (YY1) está involucrado en la resistencia a la apoptosis y la progresión tumoral; se encuentra aumentado en diferentes tumores, incluida la LLA. Aunque los mecanismos que regulan la expresión de YY1 en LLA son desconocidos, se sabe que YY1 regula la expresión del receptor de Fas. El objetivo de este trabajo fue evaluar el efecto de YY1 en la expresión de Fas en condiciones de hipoxia en la LLA. Métodos: Se cultivaron células RS4;11 en condiciones de hipoxia y se analizó la expresión de YY1, receptor de Fas y HIF-1α. La apoptosis fue inducida usando un agonista de Fas (DX2) y se analizó con la detección de caspasa 3 activa. Los datos se analizaron mediante correlación de Pearson. Resultados: Las células RS4;11 coexpresaron HIF-1αy YY1 en hipoxia, lo cual correlaciona con una baja expresión de Fas. La apoptosis se encontró disminuida durante condiciones de hipoxia, después del tratamiento con DX2. El análisis bioinformático mostró que los pacientes con altos niveles de HIF-1αpresentan YY1 elevado y bajos niveles del receptor de Fas. Conclusiones: Estos resultados sugieren que YY1 regula negativamente la expresión del receptor de Fas, lo cual podría estar involucrado en el escape de las células leucémicas a la respuesta inmunitaria, contribuyendo a la patogénesis de la LLA.


Subject(s)
Child , Humans , Cell Hypoxia/physiology , Apoptosis/physiology , fas Receptor/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , YY1 Transcription Factor/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Down-Regulation , Gene Expression , fas Receptor , Cell Line, Tumor , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , YY1 Transcription Factor/genetics , Caspase 3/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Immune Evasion , Tumor Hypoxia/physiology , Immunologic Surveillance
17.
Bol Med Hosp Infant Mex ; 77(4): 186-194, 2020.
Article in English | MEDLINE | ID: mdl-32713953

ABSTRACT

Background: Acute lymphoblastic leukemia (ALL) is an aggressive malignant disease with high prevalence in pediatric patients. It has been shown that the downregulation of Fas expression is correlated with an inadequate response in ALL, although these mechanisms are still not well understood. Several reports demonstrated that hypoxia is involved in dysfunctional apoptosis. Yin-Yang-1 (YY1) transcription factor is involved in resistance to apoptosis, tumor progression, and it is increased in different types of cancer, including leukemia. The regulatory mechanism underlying YY1 expression in leukemia is still not understood, but it is known that YY1 negatively regulates Fas expression. The study aimed to evaluate the effect of YY1 on Fas expression under hypoxic conditions in ALL. Methods: Leukemia cell line RS4; 11 was cultured under normoxic and hypoxic conditions. YY1, Fas receptor, and hypoxia-inducible factor (HIF-1α) expression were analyzed. After treatment with a Fas agonist (DX2), apoptosis was analyzed through the detection of active caspase 3. Data were analyzed using Pearson's correlation. Results: Leukemia cells co-expressed both HIF-1α and YY1 under hypoxia, which correlated with a downregulation of Fas expression. During hypoxia, the levels of apoptosis diminished after DX2 treatment. The analysis revealed that patients with high levels of HIF-1α also express high levels of YY1 and low levels of Fas. Conclusions: These results suggest that YY1 negatively regulates the expression of the Fas receptor, which could be involved in the escape of leukemic cells from the immune response contributing to the ALL pathogenesis.


Introducción: La leucemia linfoblástica aguda (LLA) es una enfermedad con alta prevalencia en la población pediátrica. El mecanismo por el cual el receptor de Fas participa en la regulación inmunitaria en los tumores es desconocido, pero se sabe que está subexpresado en LLA. El factor de transcripción Ying-Yang-1 (YY1) está involucrado en la resistencia a la apoptosis y la progresión tumoral; se encuentra aumentado en diferentes tumores, incluida la LLA. Aunque los mecanismos que regulan la expresión de YY1 en LLA son desconocidos, se sabe que YY1 regula la expresión del receptor de Fas. El objetivo de este trabajo fue evaluar el efecto de YY1 en la expresión de Fas en condiciones de hipoxia en la LLA. Métodos: Se cultivaron células RS4;11 en condiciones de hipoxia y se analizó la expresión de YY1, receptor de Fas y HIF-1α. La apoptosis fue inducida usando un agonista de Fas (DX2) y se analizó con la detección de caspasa 3 activa. Los datos se analizaron mediante correlación de Pearson. Resultados: Las células RS4;11 coexpresaron HIF-1αy YY1 en hipoxia, lo cual correlaciona con una baja expresión de Fas. La apoptosis se encontró disminuida durante condiciones de hipoxia, después del tratamiento con DX2. El análisis bioinformático mostró que los pacientes con altos niveles de HIF-1αpresentan YY1 elevado y bajos niveles del receptor de Fas. Conclusiones: Estos resultados sugieren que YY1 regula negativamente la expresión del receptor de Fas, lo cual podría estar involucrado en el escape de las células leucémicas a la respuesta inmunitaria, contribuyendo a la patogénesis de la LLA.


Subject(s)
Apoptosis/physiology , Cell Hypoxia/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , YY1 Transcription Factor/metabolism , fas Receptor/metabolism , Caspase 3/metabolism , Cell Line, Tumor , Child , Down-Regulation , Gene Expression , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immune Evasion , Immunologic Surveillance , Precursor Cell Lymphoblastic Leukemia-Lymphoma/immunology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tumor Hypoxia/physiology , YY1 Transcription Factor/genetics , fas Receptor/agonists
18.
Clin Transl Oncol ; 22(12): 2196-2205, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32445035

ABSTRACT

Radiotherapy is the treatment of choice for many cancer patients. Residual tumor leads to local recurrence after a period of an equilibrium created between proliferating, quiescent and dying cancer cells. The tumor microenvironment is a main obstacle for the efficacy of radiotherapy, as impaired blood flow leads to hypoxia, acidity and reduced accessibility of radiosensitizers. Eradication of remnant disease is an intractable clinical quest. After more than a century of research, anti-tumor immunity has gained a dominant position in oncology research and therapy. Immune cells play a significant role in the eradication of tumors during and after the completion of radiotherapy. The tumor equilibrium reached in the irradiated tumor may shift towards cancer cell eradication if the immune response is appropriately modulated. In the modern immunotherapy era, clinical trials are urged to standardize immunotherapy schemes that could be safely applied to improve clearance of the post-radiotherapy remnant disease.


Subject(s)
Immunity , Neoplasms/radiotherapy , Tumor Microenvironment , Humans , Immunotherapy , Neoplasm Recurrence, Local , Neoplasm, Residual/immunology , Neoplasm, Residual/pathology , Neoplasms/immunology , Neoplasms/pathology , Neovascularization, Pathologic , Radiation Tolerance , Radiation-Sensitizing Agents/therapeutic use , Tumor Hypoxia/immunology , Tumor Microenvironment/immunology
19.
Cell Death Dis ; 11(2): 105, 2020 02 06.
Article in English | MEDLINE | ID: mdl-32029741

ABSTRACT

Lipid droplets (also known as lipid bodies) are lipid-rich, cytoplasmic organelles that play important roles in cell signaling, lipid metabolism, membrane trafficking, and the production of inflammatory mediators. Lipid droplet biogenesis is a regulated process, and accumulation of these organelles within leukocytes, epithelial cells, hepatocytes, and other nonadipocyte cells is a frequently observed phenotype in several physiologic or pathogenic situations and is thoroughly described during inflammatory conditions. Moreover, in recent years, several studies have described an increase in intracellular lipid accumulation in different neoplastic processes, although it is not clear whether lipid droplet accumulation is directly involved in the establishment of these different types of malignancies. This review discusses current evidence related to the biogenesis, composition and functions of lipid droplets related to the hallmarks of cancer: inflammation, cell metabolism, increased proliferation, escape from cell death, and hypoxia. Moreover, the potential of lipid droplets as markers of disease and targets for novel anti-inflammatory and antineoplastic therapies will be discussed.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Lipid Droplets/metabolism , Neoplasms/metabolism , Animals , Cell Death , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Energy Metabolism , Humans , Inflammation Mediators/metabolism , Lipid Droplets/pathology , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction , Tumor Hypoxia , Tumor Microenvironment
20.
Exp Oncol ; 41(4): 318-322, 2019 12.
Article in English | MEDLINE | ID: mdl-31868335

ABSTRACT

This study aimed to investigate the effects of hypoxia and serum deprivation on regulation of fucosyltransferase-3 (FUT3) expression in breast cancer cells. MATERIALS AND METHODS: FUT3 expression was evaluated in T47D and MCF7 cells. Transcriptional and protein analysis was performed under hypoxia and serum deprivation conditions after 6 and 24 hours; and after 24 and 48 hours, respectively. RESULTS: In T47D cells, experimental conditions induced a significant decrease in FUT3 expression at both, transcriptional and protein levels, while in MCF7 cells the same conditions induced a significant increase of FUT3 expression. CONCLUSIONS: Regulation of FUT3 expression under hypoxic and serum deprivation conditions may be involved in the acquisition of advantages related to apoptosis resistance and metastasis promotion, according to the intrinsic differences presented by T47D and MCF7 cells.


Subject(s)
Breast Neoplasms/genetics , Fucosyltransferases/genetics , Gene Expression Regulation, Neoplastic , Apoptosis , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Humans , MCF-7 Cells , Serum/metabolism , Tumor Hypoxia
SELECTION OF CITATIONS
SEARCH DETAIL