Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Oncotarget ; 15: 302-311, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38742684

ABSTRACT

The main goal of the present study was to analyze the expression profile of cyclin D1 in patients with PC, and to determine possible correlations with clinical and histopathological features. A survey was conducted with 100 patients diagnosed with PC, who were treated at two reference hospitals in São Luís, Maranhão, Brazil, between 2013 and 2017. A review of clinical, epidemiological, and histopathological data was performed, Human Papillomavírus (HPV) DNA was detected using polymerase chain reaction (PCR) and cyclin D1 expression analysis was performed using immunohistochemical techniques. The data revealed that the absence of cyclin D1 expression was significantly associated with HPV-positive histological subtypes (p = 0.001), while its expression was associated with high-grade tumors (p = 0.014), histological subtype (p = 0.001), presence of sarcomatoid transformation (p = 0.04), and perineural invasion (p = 0.023). Patients with cyclin D1 expression exhibited lower disease-free survival compared to the cyclin D1-negative group, although the difference was not statistically significant. The results suggest that cyclin D1 may be a potential biomarker for PC, especially for poorer prognosis.


Subject(s)
Biomarkers, Tumor , Cyclin D1 , Penile Neoplasms , Adult , Aged , Aged, 80 and over , Humans , Male , Middle Aged , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Brazil/epidemiology , Cyclin D1/metabolism , Cyclin D1/genetics , Disease-Free Survival , Immunohistochemistry , Papillomavirus Infections/virology , Papillomavirus Infections/metabolism , Penile Neoplasms/genetics , Penile Neoplasms/pathology , Penile Neoplasms/virology , Prognosis
2.
Sci Rep ; 11(1): 21159, 2021 10 27.
Article in English | MEDLINE | ID: mdl-34707101

ABSTRACT

Using chip array assays, we identified differentially expressed genes via a comparison between luminal A breast cancer subtype and normal mammary ductal cells from healthy donors. In silico analysis confirmed by western blot and immunohistochemistry revealed that C-JUN and C-FOS transcription factors are activated in luminal A patients as potential upstream regulators of these differentially expressed genes. Using a chip-on-chip assay, we identified potential C-JUN and C-FOS targets. Among these genes, the NRIP1 gene was revealed to be targeted by C-JUN and C-FOS. This was confirmed after identification and validation with transfection assays specific binding of C-JUN and C-FOS at consensus binding sites. NRIP1 is not only upregulated in luminal A patients and cell lines but also regulates breast cancer-related genes, including PR, ESR1 and CCND1. These results were confirmed by NRIP1 siRNA knockdown and chip array assays, thus highlighting the putative role of NRIP1 in PGR, ESR1 and CCND1 transcriptional regulation and suggesting that NRIP1 could play an important role in breast cancer ductal cell initiation.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Gene Expression Regulation, Neoplastic , Nuclear Receptor Interacting Protein 1/metabolism , Adult , Aged , Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Cyclin D1/genetics , Cyclin D1/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , MCF-7 Cells , Middle Aged , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Receptor Interacting Protein 1/genetics , Proto-Oncogene Proteins c-fos/metabolism , Transcriptome
3.
Clin Transl Oncol ; 23(9): 1866-1873, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33772723

ABSTRACT

PURPOSE: The aim of the present study was to elucidate the functional role of hsa-miR-328-3p/STAT3 pathway in the effects of propofol on gastric cancer proliferation. METHODS: Bioinformatics was used to analyze the molecular expression differences of hsa-miR-328-3p/STAT3 axis in stomach adenocarcinoma (n = 435) and normal samples (n = 41) from TCGA database. The expression of the above molecules in gastric cancer cells SGC-7901 and normal gastric mucosal cells GES-1 was verified via qPCR. The dual-luciferase assay was carried out to confirm the interaction between hsa-miR-328-3p and STAT3. Subsequently, the cell proliferation and the expression of the above molecules in SGC-7901 and GES-1 cells were evaluated after 10 µM propofol treatment. Finally, we analyzed whether propofol still inhibited the proliferation of gastric cancer by suppressing STAT3 pathway after hsa-miR-328-3p down-regulation. RESULTS: Compared with normal samples, the expression of hsa-miR-328-3p was significantly down-regulated in stomach adenocarcinoma samples, while the expression of STAT3 and downstream target genes (MMP2, CCND1 and COX2) was up-regulated. The results were consistent with those in GES-1 and SGC-7901 cell lines. Meanwhile, we found that hsa-miR-328-3p can bind to the 3'-UTR of the potential target gene STAT3. Furthermore, propofol significantly inhibited the proliferation of gastric cancer cell line SGC-7901, where hsa-miR-328-3p was up-regulated and the expression of STAT3 and downstream proliferation-related target genes were down-regulated. However, the growth inhibition of propofol on SGC-7901 cell was significantly reversed after the inhibition of hsa-miR-328-3p. CONCLUSIONS: To sum up, propofol suppressed the STAT3 pathway via up-regulating hsa-miR-328-3p to inhibit gastric cancer proliferation.


Subject(s)
Adenocarcinoma/pathology , Anesthetics, Intravenous/pharmacology , Cell Proliferation/drug effects , MicroRNAs/metabolism , Propofol/pharmacology , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/pathology , 3' Untranslated Regions , Adenocarcinoma/metabolism , Cell Line, Tumor , Computational Biology , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Down-Regulation , Gastric Mucosa/drug effects , Gastric Mucosa/metabolism , Humans , Luciferases/metabolism , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , MicroRNAs/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Stomach Neoplasms/metabolism , Up-Regulation
4.
BMC Cancer ; 21(1): 76, 2021 Jan 18.
Article in English | MEDLINE | ID: mdl-33461524

ABSTRACT

BACKGROUND: Breast cancer is the most frequently diagnosed malignancy among women. However, the role of microRNA (miRNA) expression in breast cancer progression is not fully understood. In this study we examined predictive interactions between differentially expressed miRNAs and mRNAs in breast cancer cell lines representative of the common molecular subtypes. Integrative bioinformatics analysis identified miR-193 and miR-210 as potential regulatory biomarkers of mRNA in breast cancer. Several recent studies have investigated these miRNAs in a broad range of tumors, but the mechanism of their involvement in cancer progression has not previously been investigated. METHODS: The miRNA-mRNA interactions in breast cancer cell lines were identified by parallel expression analysis and miRNA target prediction programs. The expression profiles of mRNA and miRNAs from luminal (MCF-7, MCF-7/AZ and T47D), HER2 (BT20 and SK-BR3) and triple negative subtypes (Hs578T e MDA-MB-231) could be clearly separated by unsupervised analysis using HB4A cell line as a control. Breast cancer miRNA data from TCGA patients were grouped according to molecular subtypes and then used to validate these findings. Expression of miR-193 and miR-210 was investigated by miRNA transient silencing assays using the MCF7, BT20 and MDA-MB-231 cell lines. Functional studies included, xCELLigence system, ApoTox-Glo triplex assay, flow cytometry and transwell inserts were performed to determine cell proliferation, cytotoxicity, apoptosis, migration and invasion, respectively. RESULTS: The most evident effects were associated with cell proliferation after miR-210 silencing in triple negative subtype cell line MDA-MB-231. Using in silico prediction algorithms, TNFRSF10 was identified as one of the potential regulated downstream targets for both miRNAs. The TNFRSF10C and TNFRSF10D mRNA expression inversely correlated with the expression levels of miR-193 and miR210 in breast cell lines and breast cancer patients, respectively. Other potential regulated genes whose expression also inversely correlated with both miRNAs were CCND1, a known mediator on invasion and metastasis, and the tumor suppressor gene RUNX3. CONCLUSIONS: In summary, our findings identify miR-193 and miR-210 as potential regulatory miRNA in different molecular subtypes of breast cancer and suggest that miR-210 may have a specific role in MDA-MB-231 proliferation. Our results highlight important new downstream regulated targets that may serve as promising therapeutic pathways for aggressive breast cancers.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/metabolism , Biomarkers, Tumor/analysis , Breast/pathology , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Computational Biology , Core Binding Factor Alpha 3 Subunit/genetics , Cyclin D1/genetics , Female , GPI-Linked Proteins/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , MicroRNAs/analysis , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Oligonucleotide Array Sequence Analysis , Receptors, Tumor Necrosis Factor, Member 10c/genetics , Tumor Necrosis Factor Decoy Receptors/genetics
5.
Clin Transl Oncol ; 23(4): 874-881, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32880048

ABSTRACT

PURPOSE: Endocrine therapy is a mainstay for the treatment of hormone receptor-positive breast cancer (BC); however, only a fraction of patients experience a pronounced response to antagonists of estrogen signaling. There is a need to identify predictors for efficacy of this treatment. METHODS: This study included 138 patients with newly diagnosed metastatic BC, who received upfront endocrine therapy. Archival biopsy specimens were tested for CCND1 and FGFR1 gene amplification and mRNA expression by PCR-based methods. RESULTS: CCND1 and FGFR1 amplification was detected in 24 (17.9%) and 28 (20.9%) of 134 evaluable cases, respectively; 9 carcinomas had concurrent alterations of these two genes. Presence of amplification in at least one locus was more common in tumors of higher grade (p = 0.018) and was associated with higher Ki-67 proliferation index (p = 0.036). CCND1 gene amplification was associated with shorter progression-free survival (PFS) in patients receiving aromatase inhibitors (AI) [16.0 months vs. 32.4 months, HR = 3.16 (95% CI 1.26-7.93), p = 0.014]. FGFR1 status did not significantly affect PFS of AI-treated women; however, objective response to AI was observed less frequently in FGFR1-amplified BC as compared to cases with normal FGFR1 copy number [2/15 (13.3%) vs. 22/46 (47.8%), p = 0.031]. Meanwhile, CCND1/FGFR1 gene status did not influence the outcome of tamoxifen-treated patients. CONCLUSION: Presence of CCND1 and/or FGFR1 amplification is associated with worse outcomes of AI therapy in patients with metastatic BC.


Subject(s)
Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cyclin D1/genetics , Gene Amplification , Receptor, Fibroblast Growth Factor, Type 1/genetics , Adult , Aged , Aged, 80 and over , Breast Neoplasms/chemistry , Breast Neoplasms/pathology , Cell Proliferation , Drug Resistance, Neoplasm , Female , Humans , Kaplan-Meier Estimate , Ki-67 Antigen/metabolism , Middle Aged , Progression-Free Survival , RNA, Messenger/metabolism , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Tamoxifen/therapeutic use , Treatment Outcome
6.
Toxicol In Vitro ; 68: 104927, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32634469

ABSTRACT

Breast cancer is a complex disease and encompassing different types of tumor. Although advances in understanding of the molecular bases of breast cancer biology, the therapeutic proposals available still are not effective. In this scenario, the present study aimed to evaluate the mechanisms associated to antitumor activity of 7-Epiclusianone (7-Epi), a tetraprenylated benzophenone, on luminal A (MCF-7) and claudin-low (Hs 578T) breast cancer cell lines. We found that 7-Epi efficiently inhibited cell proliferation and migration of these cells; however MCF-7 was slightly more responsive than Hs 578T. Cell cycle analysis showed accumulation of cells at G0/G1 phase with drastic reduction of S population in treated cultures. This effect was associated to downregulation of CDKN1A (p21) and cyclin E in both cell lines. In addition, 7-Epi reduced cyclin D1 and p-ERK expression levels in MCF-7 cell line. Cytotoxic effect of 7-Epi on breast cancer cell lines was associated to its ability to increase BAX/BCL-2 ratio. In conclusion, our findings showed that 7-Epi is a promising antitumor agent against breast cancer by modulating critical regulators of the cell cycle and apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Benzophenones/pharmacology , Benzoquinones/pharmacology , Breast Neoplasms/drug therapy , Apoptosis/drug effects , Breast Neoplasms/genetics , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cyclin D1/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Female , Humans , Proto-Oncogene Proteins c-bcl-2/genetics
7.
Ann Hepatol ; 19(1): 99-106, 2020.
Article in English | MEDLINE | ID: mdl-31558421

ABSTRACT

INTRODUCTION AND OBJECTIVES: Liver regeneration plays a valuable significance for hepatectomies, and is mainly attributed to hepatocyte proliferation. MicroRNA-125a-3p was reported to be highly associated with liver regeneration process. We studied the underlying mechanism of the functional role of miR-125a-3p in liver regeneration. MATERIALS AND METHODS: The miR-125a-3p mimics and inhibitor vector were constructed and transfected into primary human liver HL-7702 cells, the transfected cell viability was detected using cell counting kit-8 (CCK-8). Cell cycle distribution was analyzed by flow cytometry. With Targetscan and OUGene prediction, the potential targets of miR-125 were verified by real-time quantitative PCR (qPCR) and luciferase reporter assays in turn. The overexpression vector of proline-rich acidic protein 1 (PRAP1) was constructed and co-transfected with miR-125a-3p mimics into HL-7702 cells, detecting the changes of proliferative capacity and cell cycle distribution. Western blot and qPCR performed to analyze gene expressions. RESULTS: Overexpressed miR-125a-3p notably increased the hepatocyte viability at 48h, and decreased the number of G1 phase cells (p<0.05). However, miR-125a-3p inhibition suppressed the development of hepatocytes. PRAP1 was the target of miR-125a-3p. After co-transfection with PRAP1 vector, hepatocyte viability was decrease and the G1 phase cell number was increased (p<0.05). More importantly, overexpressed PRAP1 notably decreased the mRNA and protein levels of cyclin D1, cyclin-dependent kinase 2 (CDK2) and cell division cycle 25A (CDC25A). CONCLUSION: The elevated miR-125a-3p positively correlated with hepatocyte viability and cell cycle progression due to the modulation of PRAP1, and miR-125a-3p may contribute to improving liver regeneration.


Subject(s)
Cell Proliferation/genetics , Hepatocytes/metabolism , Liver Regeneration/genetics , Liver/physiology , MicroRNAs/genetics , Pregnancy Proteins/genetics , Blotting, Western , Cell Cycle/genetics , Cell Line , Cell Survival/genetics , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , G1 Phase , Humans , Polymerase Chain Reaction , Pregnancy Proteins/metabolism , RNA, Messenger/metabolism , cdc25 Phosphatases/genetics , cdc25 Phosphatases/metabolism
8.
Sci Rep ; 9(1): 14193, 2019 10 02.
Article in English | MEDLINE | ID: mdl-31578445

ABSTRACT

The placental stem cells have called the focus of attention for their therapeutic potential to treat different diseases, including cancer. There is plenty evidence about the antiproliferative, antiangiogenic and proapoptotic properties of the amniotic membrane. Liver cancer is the fifth cause of cancer in the world, with a poor prognosis and survival. Alternative treatments to radio- or chemotherapy have been searched. In this work we aimed to study the antiproliferative properties of the human amniotic membrane conditioned medium (AM-CM) in hepatocarcinoma cells. In addition, we have analyzed the regulation of pro and antiOncomiRs expression involved in hepatocarcinoma physiology. We have determined by 3H-thymidine incorporation assay that AM-CM inhibits DNA synthesis in HepG2 cells after 72 h of treatment. AM-CM pure or diluted at 50% and 25% also diminished HepG2 and HuH-7 cells viability and cell number. Furthermore, AM-CM induced cell cycle arrest in G2/M. When proliferation mechanisms were analyzed we found that AM-CM reduced the expression of both Cyclin D1 mRNA and protein. Nuclear expression of Ki-67 was also reduced. We observed that this CM was able to promote the expression of p53 and p21 mRNA and proteins, leading to cell growth arrest. Moreover, AM-CM induced an increase in nuclear p21 localization, observed by immunofluorescence. As p53 levels were increased, Mdm-2 expression was downregulated. Interestingly, HepG2 and HuH-7 cells treatment with AM-CM during 24 and 72 h produced an upregulation of antiOncomiRs 15a and 210, and a downregulation of proOncomiRs 206 and 145. We provide new evidence about the promising novel applications of human amniotic membrane in liver cancer.


Subject(s)
Amnion/metabolism , Carcinoma, Hepatocellular/drug therapy , Culture Media, Conditioned/pharmacology , Liver Neoplasms/drug therapy , Amnion/growth & development , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Culture Media, Conditioned/metabolism , Cyclin D1/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , MicroRNAs/genetics , Placenta/metabolism , Pregnancy , Proto-Oncogene Proteins c-mdm2/genetics , Stem Cells/metabolism , Tumor Suppressor Protein p53/genetics
9.
Asian Pac J Cancer Prev ; 20(8): 2345-2351, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31450905

ABSTRACT

Background: Glioma, most common primary malignant brain tumor in adults, is highly aggressive and associated with a poor prognosis. Evaluate the association of polymorphisms related of to the cell cycle, integrity and DNA repair with gliomas, as well as lifestyle habits, comorbidities, survival and response to treatment. Methods: Were studied 303 individuals distributed into: Study Group - 100 patients with gliomas, regardless of the degree of malignancy, and Control Group - 203 individuals without clinical signs of the disease. These polymorphisms were genotyped by TaqMan® SNP Genotyping Assay. Significance level was set at 5%. Results: Smoking, alcohol consumption, systemic arterial hypertension (SAH) and diabetes mellitus (DM) prevailed in patients, compared to controls (P=0.0088, P=0.0001, P=0.0001, P=0.0011, respectively). In the logistic regression analysis, alcohol consumption and SAH were identified as independent risk factors for gliomas (P=0.0001, P=0.0027, respectively). Patients with low-grade gliomas showed survival in one year (92.0±6.8%), compared to patients with high-grade gliomas (24.0±5.3; P=0.011). Conclusion: Polymorphisms involved in cell cycle, telomere protection and stability and DNA repair are not associated with gliomas. On the other hand, alcohol consumption and SAH stand out as independent risk factors for the disease. Low-grade gliomas, response to treatment and the combination of chemotherapy with Temozolomide and radiation therapy show increased survival of patients.


Subject(s)
Biomarkers, Tumor/genetics , Cyclin D1/genetics , DNA Helicases/genetics , Glioma/genetics , Glioma/pathology , Polymorphism, Genetic , X-ray Repair Cross Complementing Protein 1/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Case-Control Studies , Child , Child, Preschool , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Genotype , Humans , Infant , Male , Middle Aged , Neoplasm Grading , Survival Rate , Telomere/chemistry , Telomere/genetics , Young Adult
10.
Mol Biol Rep ; 46(5): 4827-4841, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31270760

ABSTRACT

Salvia lachnostachys is an herbaceous plant with anti-inflammatory, analgesic and cytotoxic properties. This study investigated the antitumor effect of an ethanolic extract of Salvia lachnostachys leaves (EES) in a solid Ehrlich carcinoma model. Ehrlich cells were inoculated subcutaneously in the right pelvic member (2 × 106 cells) in female Swiss mice. The animals were treated with vehicle (10 mL kg-1, p.o.), EES (30 and 100 mg kg-1, p.o.), or methotrexate (2.5 mg kg-1, i.p.) for 21 days (early treatment) or 14 days (late treatment) after tumor inoculation, or 10 days before tumor inoculation and continued for 21 days after tumor inoculation (chemopreventive treatment). The acute toxicity test was performed according OECD guidelines Late treatment with EES had no antitumor effect. Early treatment with 100 mg kg-1 EES prevented tumor development, increased tumor necrosis factor-α (TNF-α) levels and decreased tumor superoxide dismutase (SOD) activity, interleukin-10 (IL-10) levels and Cyclin D1 expression, and tumor cell necrosis was observed. Chemopreventive treatment with EES for 10 and 31 days prevented tumor development in the same manner. EES treatment for 31 days decreased hepatic and tumor SOD activity, tumor IL-10 levels and Cyclin D1 expression, and increased tumor reduced glutathione, N-acetylglucosaminidase, reactive oxygen species, lipid peroxidation, TNF-α levels and Nrf2 expression. No toxicity was observed in the acute toxicity assay. In conclusion, EES had an antitumor effect by inhibiting Cyclin D1 expression and increasing inflammation with early and chemopreventive treatment. Modulation of the antioxidant system also contribute for the antitumor effects of EES.


Subject(s)
Anticarcinogenic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Plant Extracts/pharmacology , Salvia/chemistry , Animals , Anticarcinogenic Agents/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Carcinoma, Ehrlich Tumor/genetics , Carcinoma, Ehrlich Tumor/metabolism , Chemoprevention , Chromatography, High Pressure Liquid , Cyclin D1/genetics , Cyclin D1/metabolism , Female , Gene Expression Regulation, Neoplastic , Mice , Molecular Structure , Oxidative Stress/drug effects , Plant Extracts/chemistry , Plant Leaves/chemistry , Reactive Oxygen Species/metabolism
11.
Cell Death Dis ; 10(6): 423, 2019 05 29.
Article in English | MEDLINE | ID: mdl-31142736

ABSTRACT

The family of long noncoding mitochondrial RNAs (ncmtRNAs), comprising sense (SncmtRNA), and antisense (ASncmtRNA-1 and ASncmtRNA-2) members, are differentially expressed according to cell proliferative status; SncmtRNA is expressed in all proliferating cells, while ASncmtRNAs are expressed in normal proliferating cells, but is downregulated in tumor cells. ASncmtRNA knockdown with an antisense oligonucleotide induces massive apoptosis in tumor cell lines, without affecting healthy cells. Apoptotic death is preceded by proliferation blockage, suggesting that these transcripts are involved in cell cycle regulation. Here, we show that ASncmtRNA knockdown induces cell death preceded by proliferative blockage in three different human breast cancer cell lines. This effect is mediated by downregulation of the key cell cycle progression factors cyclin B1, cyclin D1, CDK1, CDK4, and survivin, the latter also constituting an essential inhibitor of apoptosis, underlying additionally the onset of apoptosis. The treatment also induces an increase in the microRNA hsa-miR-4485-3p, whose sequence maps to ASncmtRNA-2 and transfection of MDA-MB-231 cells with a mimic of this miRNA induces cyclin B1 and D1 downregulation. Other miRNAs that are upregulated include nuclear-encoded hsa-miR-5096 and hsa-miR-3609, whose mimics downregulate CDK1. Our results suggest that ASncmtRNA targeting blocks tumor cell proliferation through reduction of essential cell cycle proteins, mediated by mitochondrial and nuclear miRNAs. This work adds to the elucidation of the molecular mechanisms behind cell cycle arrest preceding tumor cell apoptosis induced by ASncmtRNA knockdown. As proof-of-concept, we show that in vivo knockdown of ASncmtRNAs results in drastic inhibition of tumor growth in a xenograft model of MDA-MB-231 subcutaneous tumors, further supporting this approach for the development of new therapeutic strategies against breast cancer.


Subject(s)
Apoptosis , Cell Cycle Checkpoints , Mitochondria/genetics , RNA, Long Noncoding/metabolism , Animals , Antagomirs/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CDC2 Protein Kinase/chemistry , CDC2 Protein Kinase/genetics , CDC2 Protein Kinase/metabolism , Cell Line, Tumor , Cell Proliferation , Cyclin B1/genetics , Cyclin B1/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , Down-Regulation , Female , Humans , Mice , Mice, Inbred BALB C , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , MicroRNAs/metabolism , RNA Interference , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism
12.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1540-1554, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30890427

ABSTRACT

Growth differentiation factor 11 (GDF11) has been characterized as a key regulator of differentiation in cells that retain stemness features, despite some controversies in age-related studies. GDF11 has been poorly investigated in cancer, particularly in those with stemness capacity, such as hepatocellular carcinoma (HCC), one of the most aggressive cancers worldwide. Here, we focused on investigating the effects of GDF11 in liver cancer cells. GDF11 treatment significantly reduced proliferation, colony and spheroid formation in HCC cell lines. Consistently, down-regulation of CDK6, cyclin D1, cyclin A, and concomitant upregulation of p27 was observed after 24 h of treatment. Interestingly, cell viability was unchanged, but cell functionality was compromised. These effects were potentially induced by the expression of E-cadherin and occludin, as well as Snail and N-cadherin repression, in a time-dependent manner. Furthermore, GDF11 treatment for 72 h induced that cells were incapable of sustaining colony and sphere capacity in the absent of GDF11, up to 5 days, indicating that the effect of GDF11 on self-renewal capacity is not transient. Finally, in vivo invasion studies revealed a significant decrease in cell migration of hepatocellular carcinoma cells treated with GDF11 associated to a decreased proliferation judged by Ki67 staining. Data show that exogenous GDF11 displays tumor suppressor properties in HCC cells.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic , Growth Differentiation Factors/pharmacology , Neovascularization, Pathologic/prevention & control , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Chick Embryo , Chorioallantoic Membrane/blood supply , Chorioallantoic Membrane/drug effects , Cyclin A/genetics , Cyclin A/metabolism , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Growth Differentiation Factors/genetics , Growth Differentiation Factors/metabolism , Hep G2 Cells , Humans , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Occludin/genetics , Occludin/metabolism , Signal Transduction , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology
13.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1502-1515, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30853403

ABSTRACT

Abnormalities in cerebellar structure and function may cause ataxia, a neurological dysfunction of motor coordination. In the course of the present study, we characterized a mutant mouse lineage with an ataxia-like phenotype. We localized the mutation on chromosome 17 and mapped it to position 1534 of the Nox3 gene, resulting in p.Asn64Tyr change. The primary defect observed in Nox3eqlb mice was increased proliferation of cerebellar granule cell precursors (GCPs). cDNA microarray comparing Nox3eqlb and BALB/c neonatal cerebellum revealed changes in the expression of genes involved in the control of cell proliferation. Nox3eqlb GCPs and NSC produce higher amounts of reactive oxygen species (ROS) and upregulate the expression of SHH target genes, such as Gli1-3 and Ccnd1 (CyclinD1). We hypothesize that this new mutation is responsible for an increase in proliferation via stimulation of the SHH pathway. We suggest this mutant mouse lineage as a new model to investigate the role of ROS in neuronal precursor cell proliferation.


Subject(s)
Ataxia/genetics , Cerebellum/enzymology , Hedgehog Proteins/genetics , NADPH Oxidases/genetics , Neural Stem Cells/enzymology , Signal Transduction/genetics , Animals , Ataxia/enzymology , Ataxia/physiopathology , Cell Differentiation , Cell Proliferation , Cerebellum/growth & development , Cerebellum/pathology , Chromosome Mapping , Chromosomes, Mammalian , Cyclin D1/genetics , Cyclin D1/metabolism , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Developmental , Hedgehog Proteins/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , Motor Activity/genetics , Mutation , NADPH Oxidases/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/pathology , Primary Cell Culture , Reactive Oxygen Species/metabolism , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli2/metabolism , Zinc Finger Protein Gli3/genetics , Zinc Finger Protein Gli3/metabolism
14.
Appl Immunohistochem Mol Morphol ; 27(1): 8-14, 2019 01.
Article in English | MEDLINE | ID: mdl-28549034

ABSTRACT

Non-Hodgkin lymphoma may occasionally contain large transformed cells resembling Hodgkin and Reed-Sternberg cells (HRS cells). We report a 63-year-old man with HRS cells in a recurrent mantle cell lymphoma (MCL). The patient initially presented with orbital MCL and recurred after 8 years with widespread involvement. The HRS cells were present in the recurrent disease but not in the initial orbital lesions, suggesting a transformed event after a prolonged disease course. Morphologically, the HRS cells were single cells and small clusters among the MCL cells and were frequently accompanied by histiocytes but without eosinophils or other inflammatory cells. The HRS cells showed a phenotype of classic Hodgkin lymphoma (cHL). The HRS cells were clonally related to the MCL, which was demonstrated by the presence of identical t(11;14) that resulted in productive cyclin D1 expression in both cell types. Review of the literature identified 7 additional MCL cases that showed a spectrum of clinical and pathologic features ranging from scattered HRS cells to true composite MCL and cHL. The HRS cells were clonally related to MCL in 4 cases (including the current case) and unrelated in 2 cases. These findings suggest MCL with HRS cells is a heterogeneous group that may represent a spectrum of transformation at the various stages. Proof of clonal relationship between HRS cells and MCL is useful to distinguish these cases from true composite MCL and cHL.


Subject(s)
Eye Neoplasms/pathology , Hodgkin Disease/pathology , Lymphoma, Mantle-Cell/pathology , Reed-Sternberg Cells/pathology , Cell Transformation, Neoplastic , Clone Cells , Cyclin D1/genetics , Cyclin D1/metabolism , Eye Neoplasms/diagnosis , Gene Expression Regulation, Neoplastic , Hodgkin Disease/diagnosis , Humans , Lymphoma, Mantle-Cell/diagnosis , Male , Middle Aged , Tumor Cells, Cultured
15.
Biofactors ; 45(1): 24-34, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30521071

ABSTRACT

The effects of radiation are known to be potentiated by N-3 polyunsaturated fatty acids, which modulate several signaling pathways, but the molecular mechanisms through which these fatty acids enhance the anticancer effects of irradiation in colorectal cancer (CRC) treatment remain poorly elucidated. Here, we aimed to ascertain whether the fatty acid docosahexaenoic acid (DHA) exerts a modulating effect on the response elicited by radiation treatment (RT). Two CRC cell lines, Caco-2 and HT-29, were exposed to RT, DHA, or both (DHA + RT) for various times, and then cell viability, proliferation, and clonogenicity were assessed. Moreover, cell cycle, apoptosis, and necrosis were analyzed using flow cytometry, and the involvement of WNT/ß-catenin signaling was investigated by immunofluorescence to determine nuclear ß-catenin, GSK3ß phosphorylation status, and TCF/LEF-activity reporter. DHA and RT applied separately diminished the viability of both HT-29 and Caco-2 cells, and DHA + RT caused a further reduction in proliferation mainly in HT-29 cells, particularly in terms of colony formation. Concomitantly, our results verified cell cycle arrest in G0/G1 phase, a reduction of cyclin D1 expression, and a decrease in GSK3ß phosphorylation after the combined treatment. Furthermore, immunofluorescence quantification revealed that nuclear ß-catenin was increased in RT-exposed cells, but this effect was abrogated in cells exposed to DHA + RT, and the results of TCF/LEF-activity assays confirmed that DHA attenuated the increase in nuclear ß-catenin activity induced by irradiation. Our finding shows that DHA applied in combination with RT enhanced the antitumor effects of irradiation on CRC cells, and that the underlying mechanism involved the WNT/ß-catenin pathway. © 2018 BioFactors, 45(1):24-34, 2019.


Subject(s)
Cell Cycle Checkpoints/drug effects , Docosahexaenoic Acids/pharmacology , Gamma Rays , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3 beta/genetics , beta Catenin/genetics , Apoptosis/drug effects , Apoptosis/radiation effects , Caco-2 Cells , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Colony-Forming Units Assay , Cyclin D1/genetics , Cyclin D1/metabolism , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3 beta/metabolism , HT29 Cells , Humans , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Phosphorylation/drug effects , Phosphorylation/radiation effects , T Cell Transcription Factor 1/genetics , T Cell Transcription Factor 1/metabolism , Wnt Signaling Pathway , beta Catenin/metabolism
16.
J Cell Mol Med ; 22(10): 4899-4912, 2018 10.
Article in English | MEDLINE | ID: mdl-30079458

ABSTRACT

Tyrosine kinase inhibitors (TKI) have become a first-line treatment for chronic myeloid leuakemia (CML). TKIs efficiently target bulk CML cells; however, they are unable to eliminate the leukaemic stem cell (LSC) population that causes resistance and relapse in CML patients. In this study, we assessed the effects of parthenolide (PTL) and dimethyl amino parthenolide (DMAPT), two potent inhibitors of LSCs in acute myeloid leukaemia (AML), on CML bulk and CML primitive (CD34+ lin- ) cells. We found that both agents induced cell death in CML, while having little effect on the equivalent normal hematopoietic cells. PTL and DMAPT caused an increase in reactive oxygen species (ROS) levels and inhibited NF-κB activation. PTL and DMAPT inhibited cell proliferation and induced cell cycle arrest in G0 and G2 phases. Furthermore, we found cell cycle inhibition to correlate with down-regulation of cyclin D1 and cyclin A. In summary, our study shows that PTL and DMAPT have a strong inhibitory effect on CML cells. Given that cell cycle arrest was not dependent on ROS induction, we speculate that this effect could be a direct consequence of NF-κB inhibition and if this mechanism was to be evaded, PTL and DMAPT induced cell death would be potentiated.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Neoplasm Recurrence, Local/drug therapy , Sesquiterpenes/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin A/genetics , Cyclin D1/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , NF-kappa B/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
17.
Mol Oncol ; 12(7): 1026-1046, 2018 06.
Article in English | MEDLINE | ID: mdl-29689621

ABSTRACT

SALL2 is a poorly characterized transcription factor that belongs to the Spalt-like family involved in development. Mutations on SALL2 have been associated with ocular coloboma and cancer. In cancers, SALL2 is deregulated and is proposed as a tumor suppressor in ovarian cancer. SALL2 has been implicated in stemness, cell death, proliferation, and quiescence. However, mechanisms underlying roles of SALL2 related to cancer remain largely unknown. Here, we investigated the role of SALL2 in cell proliferation using mouse embryo fibroblasts (MEFs) derived from Sall2-/- mice. Compared to Sall2+/+ MEFs, Sall2-/- MEFs exhibit enhanced cell proliferation and faster postmitotic progression through G1 and S phases. Accordingly, Sall2-/- MEFs exhibit higher mRNA and protein levels of cyclins D1 and E1. Chromatin immunoprecipitation and promoter reporter assays showed that SALL2 binds and represses CCND1 and CCNE1 promoters, identifying a novel mechanism by which SALL2 may control cell cycle. In addition, the analysis of tissues from Sall2+/+ and Sall2-/- mice confirmed the inverse correlation between expression of SALL2 and G1-S cyclins. Consistent with an antiproliferative function of SALL2, immortalized Sall2-/- MEFs showed enhanced growth rate, foci formation, and anchorage-independent growth, confirming tumor suppressor properties for SALL2. Finally, cancer data analyses show negative correlations between SALL2 and G1-S cyclins' mRNA levels in several cancers. Altogether, our results demonstrated that SALL2 is a negative regulator of cell proliferation, an effect mediated in part by repression of G1-S cyclins' expression. Our results have implications for the understanding and significance of SALL2 role under physiological and pathological conditions.


Subject(s)
Cell Cycle/genetics , Cyclin D1/genetics , Cyclin E/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasms/genetics , Neoplasms/pathology , Repressor Proteins/metabolism , Animals , Cell Proliferation , Cell Transformation, Neoplastic/pathology , Cyclin D1/metabolism , Cyclin E/metabolism , DNA-Binding Proteins , Fibroblasts/metabolism , G1 Phase , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Mice, Knockout , Models, Biological , Phenotype , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , S Phase , Transcription Factors , Transcription, Genetic
18.
Cell Death Dis ; 9(3): 311, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29472557

ABSTRACT

The recurrent gain-of-function JAK2V617F mutation confers growth factor-independent proliferation for hematopoietic cells and is a major contributor to the pathogenesis of myeloproliferative neoplasms (MPN). The lack of complete response in most patients treated with the JAK1/2 inhibitor ruxolitinib indicates the need for identifying novel therapeutic strategies. Metformin is a biguanide that exerts selective antineoplastic activity in hematological malignancies. In the present study, we investigate and compare effects of metformin and ruxolitinib alone and in combination on cell signaling and cellular functions in JAK2V617F-positive cells. In JAK2V617F-expressing cell lines, metformin treatment significantly reduced cell viability, cell proliferation, clonogenicity, and cellular oxygen consumption and delayed cell cycle progression. Metformin reduced cyclin D1 expression and RB, STAT3, STAT5, ERK1/2 and p70S6K phosphorylation. Metformin plus ruxolitinib demonstrated more intense reduction of cell viability and induction of apoptosis compared to monotherapy. Notably, metformin reduced Ba/F3 JAK2V617F tumor burden and splenomegaly in Jak2V617F knock-in-induced MPN mice and spontaneous erythroid colony formation in primary cells from polycythemia vera patients. In conclusion, metformin exerts multitarget antileukemia activity in MPN: downregulation of JAK2/STAT signaling and mitochondrial activity. Our exploratory study establishes novel molecular mechanisms of metformin and ruxolitinib action and provides insights for development of alternative/complementary therapeutic strategies for MPN.


Subject(s)
Antineoplastic Agents/administration & dosage , Janus Kinase 2/metabolism , Metformin/administration & dosage , Myeloproliferative Disorders/drug therapy , Animals , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism , Female , Gene Knock-In Techniques , Humans , Janus Kinase 2/genetics , Mice , Mice, Inbred NOD , Mutation, Missense , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/physiopathology , Phosphorylation/drug effects , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/metabolism
19.
Rev. chil. dermatol ; 34(2): 60-67, 2018. ilus, tab
Article in Spanish | LILACS | ID: biblio-994875

ABSTRACT

En la última década se ha avanzado en la caracterización genética y mapeo molecular del melanoma cutáneo con el objetivo de identificar y comprender mejor los mecanismos patogénicos propios de cada subgrupo y así desarrollar tratamientos específicos. El melanoma lentiginoso acral (MLA) constituye un subtipo de melanoma con características clínicas, epidemiológicas, histopatológicas, pronósticas y terapéuticas distintivas y su perfil mutacional no es la excepción. A diferencia del melanoma ubicado en zonas fotoexpuestas, el MLA presenta una baja tasa de mutaciones BRAF (15%) y mayor frecuencia de amplificaciones y ganancias genéticas de KIT (15-30%), CCND1 (15-40%) y TERT (20%). En esta revisión se describen las características más relevantes del MLA con énfasis en el rol que cumplen los principales genes que participan en la patogenia del MLA.


Over the last decade, the genetic characterization and molecular mapping of cutaneous melanoma has been developed in order to identify and better understand the pathogenic mechanisms of each subgroup and to develop specific treatments. Acral lentiginous melanoma (ALM) is a melanoma subtype with distinctive clinical, epidemiological, histopathological, prognostic and therapeutic features and its mutational profile is not an exception. Unlike melanoma located in photoexposed areas, MLA has a low rate of BRAF mutations (15%) and a higher frequency of amplifications and genetic gains at KIT (15-30%), CCND1 (15-40%) and TERT (20%). In this review we will describe the most relevant characteristics of MLA with emphasis on the role of the main genes involved in its pathogenesis.


Subject(s)
Humans , Skin Neoplasms/genetics , Melanoma/genetics , Prognosis , Skin Neoplasms/pathology , Telomerase/genetics , Proto-Oncogene Proteins c-kit/genetics , Cyclin D1/genetics , Melanoma/pathology , Mutation
20.
J Mol Endocrinol ; 59(1): 13-27, 2017 07.
Article in English | MEDLINE | ID: mdl-28385776

ABSTRACT

Parathyroid hormone-related peptide (PTHrP) is associated with several human cancers such as colon carcinoma. This disease is a complex multistep process that involves enhanced cell cycle progression and migration. Recently we obtained evidence that in the human colorectal adenocarcinoma Caco2 cells, exogenous PTHrP increases the proliferation and positively modulates cell cycle progression via ERK1/2, p38 MAPK and PI3K. The purpose of this study was to explore if the serine/threonine kinase RSK, which is involved in the progress of many cancers and it is emerging as a potential therapeutic target, mediates PTHrP effects on cancer colon cells. Western blot analysis revealed that PTHrP increases RSK phosphorylation via ERK1/2 signaling pathway but not through p38 MAPK. By performing subcellular fractionation, we found that the peptide also induces the nuclear localization of activated RSK, where many of its substrates are located. RSK participates in cell proliferation, in the upregulation of cyclin D1 and CDK6 and in the downregulation of p53 induced by PTHrP. Wound healing and transwell filter assays revealed that cell migration increased after PTHrP treatment. In addition, the hormone increases the protein expression of the focal adhesion kinase FAK, a regulator of cell motility. We observed that PTHrP induces cell migration and modulates FAK protein expression through ERK/RSK signaling pathway but not via p38 MAPK pathway. Finally, in vivo studies revealed that the hormone activates RSK in xenografts tumor. Taken together, our findings provide new insights into the deregulated cell cycle and migration that is characteristic of tumor intestinal cells.


Subject(s)
Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , MAP Kinase Signaling System/genetics , Parathyroid Hormone-Related Protein/pharmacology , Protein Serine-Threonine Kinases/genetics , Animals , Caco-2 Cells , Cell Movement/drug effects , Cell Survival/drug effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , HCT116 Cells , Humans , Injections, Intralesional , Male , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Transplantation , Parathyroid Hormone-Related Protein/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL