1
|
Pérez CN, Falcón CR, Mons JD, Orlandi FC, Sangiacomo M, Fernandez-Muñoz JM, Guerrero M, Benito PG, Colombo MI, Zoppino FCM, Alvarez SE. Melanoma cells with acquired resistance to vemurafenib have decreased autophagic flux and display enhanced ability to transfer resistance. Biochim Biophys Acta Mol Basis Dis 2023:166801. [PMID: 37419396 DOI: 10.1016/j.bbadis.2023.166801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 05/31/2023] [Accepted: 06/28/2023] [Indexed: 07/09/2023]
Abstract
Over the last years, the incidence of melanoma, the deadliest form of skin cancer, has risen significantly. Nearly half of the melanoma patients exhibit the BRAFV600E mutation. Although the use of BRAF and MEK inhibitors (BRAFi and MEKi) showed an impressive success rate in melanoma patients, durability of response remains an issue because tumor quickly becomes resistant. Here, we generated and characterized Lu1205 and A375 melanoma cells resistant to vemurafenib (BRAFi). Resistant cells (Lu1205R and A375R) exhibit higher IC50 (5-6 fold increase) and phospho-ERK levels and 2-3 times reduced apoptosis than their sensitive parents (Lu1205S and A375S). Moreover, resistant cells are 2-3 times bigger, display a more elongated morphology and have a modulation the migration capacity. Interestingly, pharmacological inhibition of sphingosine kinases, that prevents sphingosine-1-phosphate production, reduces migration of Lu1205R cells by 50 %. In addition, although Lu1205R cells showed increased basal levels of the autophagy markers LC3II and p62, they have decreased autophagosome degradation and autophagy flux. Remarkably, expression of Rab27A and Rab27B, which are involved in the release of extracellular vesicles are dramatically augmented in resistant cells (i.e. 5-7 fold increase). Indeed, conditioned media obtained from Lu1205R cells increased the resistance to vemurafenib of sensitive cells. Hence, these results support that resistance to vemurafenib modulates migration and the autophagic flux and may be transferred to nearby sensitive melanoma cells by factors that are released to the extracellular milieu by resistant cells.
Collapse
Affiliation(s)
- Celia N Pérez
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis (UNSL), Ejército de los Andes 950, 5700 San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), CONICET, Argentina
| | - Cristian R Falcón
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis (UNSL), Ejército de los Andes 950, 5700 San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), CONICET, Argentina
| | - Johinna Delgado Mons
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis (UNSL), Ejército de los Andes 950, 5700 San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), CONICET, Argentina
| | - Federico Cuello Orlandi
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis (UNSL), Ejército de los Andes 950, 5700 San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), CONICET, Argentina
| | - Mercedes Sangiacomo
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis (UNSL), Ejército de los Andes 950, 5700 San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), CONICET, Argentina
| | | | - Martín Guerrero
- Instituto de Biología y Medicina Experimental de Cuyo (IMBECU), CONICET, Argentina
| | - Paula G Benito
- Instituto de Histología y Embriología de Mendoza (IHEM), Universidad Nacional de Cuyo-CONICET, Argentina
| | - María I Colombo
- Instituto de Histología y Embriología de Mendoza (IHEM), Universidad Nacional de Cuyo-CONICET, Argentina
| | - Felipe C M Zoppino
- Instituto de Biología y Medicina Experimental de Cuyo (IMBECU), CONICET, Argentina
| | - Sergio E Alvarez
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis (UNSL), Ejército de los Andes 950, 5700 San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), CONICET, Argentina.
| |
Collapse
|
2
|
Abutorabi ES, Poursheikhani A, Kashani B, Shamsaiegahkani S, Haghpanah V, Bashash D, Mousavi SA, Momeny M, Ghaffari SH. The effects of Abemaciclib on cell cycle and apoptosis regulation in anaplastic thyroid cancer cells. Mol Biol Rep 2023; 50:4073-4082. [PMID: 36877344 DOI: 10.1007/s11033-023-08255-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 01/05/2023] [Indexed: 03/07/2023]
Abstract
BACKGROUND Anaplastic thyroid cancer (ATC) is an aggressive subtype of thyroid cancer, accounting for 1 to 2% of all cases. Deregulations of cell cycle regulatory genes including cyclins, cyclin-dependent kinases (CDKs), and endogenous inhibitors of CDKs (CKIs) are hallmarks of cancer cells and hence, studies indicate the inhibition of CDK4/6 kinases and cell cycle progression as potent therapeutic strategies. In this study, we investigated the anti-tumor activity of Abemaciclib, a CDK4 and CDK6 inhibitor, in ATC cell lines. METHODS AND RESULTS The ATC cell lines C643 and SW1736 were selected to study the antiproliferative effects of Abemaciclib using a cell proliferation assay and crystal violet staining assay. Annexin V/PI staining and cell cycle analysis by flow cytometry were also performed to examine the effects on apoptosis induction and cell cycle arrest. Wound healing assay and zymography analysis examined the effects of the drug on invasive abilities of ATC cells and Western blot analyses were applied to further study the anti-tumor mechanism of Abemaciclib, in addition to combination treatment with alpelisib. Our data demonstrated that Abemaciclib significantly inhibited cell proliferation and increased cellular apoptosis and cell cycle arrest in ATC cell lines, while considerably reducing cell migration and colony formation. The mechanism seemed to involve the PI3K pathway. CONCLUSION Our preclinical data highlight CDK4/6 as interesting therapeutic targets in ATC and suggest CDK4/6-blockade therapies as promising strategies in this malignancy.
Collapse
Affiliation(s)
- Elaheh S Abutorabi
- Hematology/Oncology and Stem Cell Transplantation Research Center, School of Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Arash Poursheikhani
- Medical Genetics Research Center, Department of Medical Genetics and Molecular Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bahareh Kashani
- Hematology/Oncology and Stem Cell Transplantation Research Center, School of Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Sahar Shamsaiegahkani
- Hematology/Oncology and Stem Cell Transplantation Research Center, School of Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Haghpanah
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seied A Mousavi
- Hematology/Oncology and Stem Cell Transplantation Research Center, School of Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Momeny
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Seyed H Ghaffari
- Hematology/Oncology and Stem Cell Transplantation Research Center, School of Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Poursheikhani A, Abbaszadegan MR, Kerachian MA. Long non-coding RNA AC087388.1 as a novel biomarker in colorectal cancer. BMC Cancer 2022; 22:196. [PMID: 35193569 PMCID: PMC8862536 DOI: 10.1186/s12885-022-09282-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 02/09/2022] [Indexed: 12/24/2022] Open
Abstract
Background Several investigations have reported diverse roles of long non-coding RNA (lncRNA) in biological processes, tumor development, and progression of colorectal cancer (CRC). In this study, we investigated the lncRNA AC087388.1 tumorigenic role in CRC cells. Methods The CRC tissues were collected at the Reza Radiotherapy and Oncology Center, Mashhad, Iran. The human SW-48 and HT-29 CRC cell lines were obtained from the national cell bank of Iran. The cells were cultured according to ATCC (the American Type Culture Collection) recommendations. Quantitative real-time PCR was applied to assess the RNA expression. ShRNA transfection was done to downregulate the target gene. MTT and apoptosis assays were conducted to evaluate cell proliferation and viability, respectively. Colony formation assay, wound healing assay, and invasion assay were applied to determine growth, motility, and invasion of the cells, respectively. ENCORI online tool was used as downstream enrichment analysis. Results Forty CRC patients were encompassed in this study. The results demonstrated that the lncRNA SLC16A1-AS1, AC087388.1, and ELFN1-AS1 were significantly overexpressed in the CRC tissues in comparison to their normal counterpart margins. All the lncRNAs have shown significant Area Under Curve (AUC) values in the patients. Downregulation of lncRNA AC087388.1 remarkably decreased the cell proliferation and viability of the CRC cells. In addition, the data demonstrated that the downregulation of lncRNA AC087388.1 significantly suppressed cell growth and colony formation capability in the cells. Also, downregulation of lncRNA AC087388.1 attenuated motility and invasion of CRC cells, and significantly decreased the expression of invasion genes. In-silico functional enrichment analysis indicated that the lncRNA AC087388.1 has contributed to crucial signaling pathways in tumorigenesis such as the p53 and Wnt signaling pathways, apoptosis, and cell cycle. Conclusions Altogether, we showed that lncRNA AC087388.1 has an oncogenic role in tumorigenesis of CRC, and it can be considered as a novel diagnostic and prognostic biomarker in CRC. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09282-0.
Collapse
Affiliation(s)
- Arash Poursheikhani
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Abbaszadegan
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Amin Kerachian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran. .,Department of Medical Genetics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran. .,Cancer Genetics Research Unit, Reza Radiotherapy and Oncology Center, Mashhad, Iran.
| |
Collapse
|
4
|
Rodriguez YI, Campos LE, Castro MG, Bannoud N, Blidner AG, Filippa VP, Croci DO, Rabinovich GA, Alvarez SE. Tumor Necrosis Factor Receptor-1 (p55) Deficiency Attenuates Tumor Growth and Intratumoral Angiogenesis and Stimulates CD8 + T Cell Function in Melanoma. Cells 2020; 9:cells9112469. [PMID: 33202705 PMCID: PMC7696624 DOI: 10.3390/cells9112469] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 10/24/2020] [Accepted: 10/30/2020] [Indexed: 12/26/2022] Open
Abstract
The role of tumor necrosis factor-α (TNF-α) in shaping the tumor microenvironment is ambiguous. Consistent with its uncertain role in melanoma, TNF-α plays a dual role, either acting as a cytotoxic cytokine or favoring a tumorigenic inflammatory microenvironment. TNF-α signals via two cognate receptors, namely TNFR1 (p55) and TNFR2 (p75), which mediate divergent biological activities. Here, we analyzed the impact of TNFR1 deficiency in tumor progression in the B16.F1 melanoma model. Tumors developed in mice lacking TNFR1 (TNFR1 knock-out; KO) were smaller and displayed lower proliferation compared to their wild type (WT) counterpart. Moreover, TNFR1 KO mice showed reduced tumor angiogenesis. Although no evidence of spontaneous metastases was observed, conditioned media obtained from TNFR1 KO tumors increased tumor cell migration. Whereas the analysis of tumor-associated immune cell infiltrates showed similar frequency of total and M2-polarized tumor-associated macrophages (TAMs), the percentage of CD8+ T cells was augmented in TNFR1 KO tumors. Indeed, functional ex vivo assays demonstrated that CD8+ T cells obtained from TNFR1KO mice displayed an increased cytotoxic function. Thus, lack of TNFR1 attenuates melanoma growth by modulating tumor cell proliferation, migration, angiogenesis and CD8+ T cell accumulation and activation, suggesting that interruption of TNF-TNFR1 signaling may contribute to control tumor burden.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/immunology
- Cell Proliferation
- Lymphocyte Activation/immunology
- Melanins/metabolism
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasm Invasiveness
- Neovascularization, Pathologic/immunology
- Receptors, Tumor Necrosis Factor, Type I/deficiency
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Signal Transduction
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Yamila I. Rodriguez
- Instituto Multidisciplinario de Investigaciones Biológicas San Luis (IMIBIO-SL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) and Universidad Nacional de San Luis (UNSL), San Luis D5700, Argentina; (Y.I.R.); (L.E.C.); (M.G.C.); (V.P.F.)
| | - Ludmila E. Campos
- Instituto Multidisciplinario de Investigaciones Biológicas San Luis (IMIBIO-SL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) and Universidad Nacional de San Luis (UNSL), San Luis D5700, Argentina; (Y.I.R.); (L.E.C.); (M.G.C.); (V.P.F.)
| | - Melina G. Castro
- Instituto Multidisciplinario de Investigaciones Biológicas San Luis (IMIBIO-SL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) and Universidad Nacional de San Luis (UNSL), San Luis D5700, Argentina; (Y.I.R.); (L.E.C.); (M.G.C.); (V.P.F.)
| | - Nadia Bannoud
- Laboratorio de Inmunopatología, Facultad de Ciencias Exactas y Naturales, Instituto de Histología y Embriología de Mendoza (IHEM), Universidad Nacional de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza C5500, Argentina; (N.B.); (D.O.C.)
| | - Ada G. Blidner
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428, Argentina; (A.G.B.); (G.A.R.)
| | - Verónica P. Filippa
- Instituto Multidisciplinario de Investigaciones Biológicas San Luis (IMIBIO-SL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) and Universidad Nacional de San Luis (UNSL), San Luis D5700, Argentina; (Y.I.R.); (L.E.C.); (M.G.C.); (V.P.F.)
| | - Diego O. Croci
- Laboratorio de Inmunopatología, Facultad de Ciencias Exactas y Naturales, Instituto de Histología y Embriología de Mendoza (IHEM), Universidad Nacional de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza C5500, Argentina; (N.B.); (D.O.C.)
| | - Gabriel A. Rabinovich
- Laboratorio de Inmunopatología, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires C1428, Argentina; (A.G.B.); (G.A.R.)
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428, Argentina
| | - Sergio E. Alvarez
- Instituto Multidisciplinario de Investigaciones Biológicas San Luis (IMIBIO-SL), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) and Universidad Nacional de San Luis (UNSL), San Luis D5700, Argentina; (Y.I.R.); (L.E.C.); (M.G.C.); (V.P.F.)
- Correspondence:
| |
Collapse
|
5
|
Teng Z, Xu S, Lei Q. Tanshinone IIA enhances the inhibitory effect of imatinib on proliferation and motility of acute leukemia cell line TIB‑152 in vivo and in vitro by inhibiting the PI3K/AKT/mTOR signaling pathway. Oncol Rep 2020; 43:503-515. [PMID: 31894340 PMCID: PMC6967082 DOI: 10.3892/or.2019.7453] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a malignant hematological disease. Tanshinone IIA (Tan IIA) has antitumor activity in vitro and in vivo. The aim of the present study was to investigate the effects of Tan IIA in combination with imatinib (IM) on the proliferation, apoptosis, migration and invasion of acute T lymphocytic leukemia TIB‑152 cells in vivo and in vitro, and analyze the potential underlying mechanism. Tan IIA and IM, alone and in combination, significantly inhibited proliferation, migration and invasion of TIB‑152 cells, and promoted apoptosis; the effect of co‑treatment with Tan IIA plus IM was enhanced. IGF‑1 promoted the proliferation, migration and invasion of TIB‑152 cells and inhibited apoptosis, while Tan IIA treatment significantly reversed these effects. In vivo experiments demonstrated that treatment with Tan IIA and IM, alone or in combination, significantly inhibited tumor growth in TIB‑152 xenograft mice; the growth inhibition of Tan IIA plus IM was the strongest observed. Western blot analysis revealed that the combination of Tan IIA and IM resulted in significantly lower levels of p‑PI3K, p‑AKT and p‑mTOR in cells and tissues compared with the IM and Tan alone treatment groups. In addition, the combination of Tan IIA and IM significantly decreased the levels of Ki67, cleaved caspase‑3, VEGF and MMP‑9 in cells and tissues, and the level of caspase‑3 was significantly increased. Taken together, the results revealed that Tan IIA enhanced the inhibitory effect of imatinib on TIB‑152 cell proliferation, migration and invasion, and induced apoptosis, which may be associated with inhibition of the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Zhi Teng
- Department of Hematology, 215 Hospital of Shanxi Nuclear Industry, Xianyang, Shanxi 712000, P.R. China
| | - Shijuan Xu
- Department of Hematology, 215 Hospital of Shanxi Nuclear Industry, Xianyang, Shanxi 712000, P.R. China
| | - Qin Lei
- Department of Hematology, 215 Hospital of Shanxi Nuclear Industry, Xianyang, Shanxi 712000, P.R. China
| |
Collapse
|
6
|
Campos LE, Garibotto FM, Angelina E, Kos J, Tomašič T, Zidar N, Kikelj D, Gonec T, Marvanova P, Mokry P, Jampilek J, Alvarez SE, Enriz RD. Searching new structural scaffolds for BRAF inhibitors. An integrative study using theoretical and experimental techniques. Bioorg Chem 2019; 91:103125. [PMID: 31401373 DOI: 10.1016/j.bioorg.2019.103125] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/04/2019] [Accepted: 07/11/2019] [Indexed: 01/12/2023]
Abstract
The identification of the V600E activating mutation in the protein kinase BRAF in around 50% of melanoma patients has driven the development of highly potent small inhibitors (BRAFi) of the mutated protein. To date, Dabrafenib and Vemurafenib, two specific BRAFi, have been clinically approved for the treatment of metastatic melanoma. Unfortunately, after the initial response, tumors become resistant and patients develop a progressive and lethal disease, making imperative the development of new therapeutic options. The main objective of this work was to find new BRAF inhibitors with different structural scaffolds than those of the known inhibitors. Our study was carried out in different stages; in the first step we performed a virtual screening that allowed us to identify potential new inhibitors. In the second step, we synthesized and tested the inhibitory activity of the novel compounds founded. Finally, we conducted a molecular modelling study that allowed us to understand interactions at the molecular level that stabilize the formation of the different molecular complexes. Our theoretical and experimental study allowed the identification of four new structural scaffolds, which could be used as starting structures for the design and development of new inhibitors of BRAF. Our experimental data indicate that the most active compounds reduced significantly ERK½ phosphorylation, a measure of BRAF inhibition, and cell viability. Thus, from our theoretical and experimental results, we propose new substituted hydroxynaphthalenecarboxamides, N-(hetero)aryl-piperazinylhydroxyalkylphenylcarbamates, substituted piperazinylethanols and substituted piperazinylpropandiols as initial structures for the development of new inhibitors for BRAF. Moreover, by performing QTAIM analysis, we are able to describe in detail the molecular interactions that stabilize the different Ligand-Receptor complexes. Such analysis indicates which portion of the different molecules must be changed in order to obtain an increase in the binding affinity of these new ligands.
Collapse
Affiliation(s)
- Ludmila E Campos
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de los Andes 950, 5700 San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700 San Luis, Argentina
| | - Francisco M Garibotto
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de los Andes 950, 5700 San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700 San Luis, Argentina
| | - Emilio Angelina
- Laboratorio de Estructura Molecular y Propiedades, Área de Química Física, Departamento de Química, Facultad de Ciencias Exactas y Naturales y Agrimensura, Universidad Nacional del Nordeste, Avda. Libertad 5460, 3400 Corrientes, Argentina
| | - Jiri Kos
- Division of Biologically Active Complexes and Molecular Magnets, Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacky University Olomouc, Slechtitelu 27, 78371 Olomouc, Czech Republic
| | - Tihomir Tomašič
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Nace Zidar
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Danijel Kikelj
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, 1000 Ljubljana, Slovenia
| | - Tomas Gonec
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho 1, 61242 Brno, Czech Republic
| | - Pavlina Marvanova
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho 1, 61242 Brno, Czech Republic
| | - Petr Mokry
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences Brno, Palackeho 1, 61242 Brno, Czech Republic
| | - Josef Jampilek
- Division of Biologically Active Complexes and Molecular Magnets, Regional Centre of Advanced Technologies and Materials, Faculty of Science, Palacky University Olomouc, Slechtitelu 27, 78371 Olomouc, Czech Republic; Department of Analytical Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 84215 Bratislava, Slovakia
| | - Sergio E Alvarez
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de los Andes 950, 5700 San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700 San Luis, Argentina.
| | - Ricardo D Enriz
- Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejército de los Andes 950, 5700 San Luis, Argentina; Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO-SL), Ejército de los Andes 950, 5700 San Luis, Argentina.
| |
Collapse
|
7
|
Liu X, Su C, Xu J, Zhou D, Yan H, Li W, Chen G, Zhang N, Xu D, Hu H. Immunohistochemical analysis of matrix metalloproteinase-9 predicts papillary thyroid carcinoma prognosis. Oncol Lett 2019; 17:2308-2316. [PMID: 30675296 PMCID: PMC6341782 DOI: 10.3892/ol.2018.9850] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 11/14/2018] [Indexed: 02/06/2023] Open
Abstract
The present study aimed to explore the association between immunohistochemical matrix metalloproteinase-9 (MMP-9) expression and the clinicopathological characteristics of patients with papillary thyroid carcinoma (PTC), and to determine whether it may be used as a diagnostic or prognostic tool for PTC. Immunohistochemical staining of MMP-9 was performed in thyroid tissues obtained from 112 patients with PTC and 42 subjects with benign thyroid nodules (BTNs). The receiver operating characteristic curve was used to evaluate the legitimacy of MMP-9 as a diagnostic tool for PTC, and a predictor for structurally persistent/recurrent disease (SPRD) and disease status. Cox regression was applied to identify the risk factors of disease status and SPRD. The present study revealed that MMP-9 was overexpressed in PTC tissues, compared with in BTN tissues. Furthermore, MMP-9 scores yielded an area under the curve (AUC) of 0.842 (95% CI, 0.776-0.908) for differentially diagnosing PTC from BTN. In addition, the MMP-9 score was greater if patients previously had central lymph node metastasis, lateral lymph node metastasis or an advanced tumor-node-metastasis stage (III+IV). When MMP-9 was employed to predict disease status and SPRD, an AUC of 0.811 (95% CI, 0.706-0.917) and 0.806 (95% CI, 0.620-0.992) was obtained, respectively. A tumor size of >2 cm and an MMP-9 staining score of ≥6 were independent risk factors for predicting disease status, whereas vascular invasion and an MMP-9 staining score of ≥8 were risk factors for predicting SPRD. Furthermore, an MMP-9 staining score of ≥6 and ≥8 indicated shortened disease-free survival and survival without SPRD, respectively. In conclusion, the assessment of MMP-9 expression in thyroid carcinoma samples may represent a potential and supplementary tool for the diagnosis and prognostic prediction of PTC.
Collapse
Affiliation(s)
- Xingkai Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Chang Su
- Department of Thyroid Surgery, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jing Xu
- Cardiovascular Disease Center, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Dan Zhou
- Department of Pediatrics, The Second Hospital of Jilin University, Changchun, Jilin 130041, P.R. China
| | - He Yan
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Wei Li
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Guihui Chen
- Department of Pathology, Jilin City People's Hospital, Jilin, Jilin 132000, P.R. China
| | - Nan Zhang
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Dahai Xu
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Haixia Hu
- Department of Emergency, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
8
|
Tan Z, Wang C, Li X, Guan F. Bisecting N-Acetylglucosamine Structures Inhibit Hypoxia-Induced Epithelial-Mesenchymal Transition in Breast Cancer Cells. Front Physiol 2018; 9:210. [PMID: 29593568 PMCID: PMC5854678 DOI: 10.3389/fphys.2018.00210] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 02/23/2018] [Indexed: 01/04/2023] Open
Abstract
The epithelial-mesenchymal transition (EMT) process plays a key role in many biological processes, including tissue fibrosis, metastatic diseases, and cancer progression. EMT can be induced by certain factors, notably hypoxia, in the tumor microenvironment. Aberrant levels of certain N-glycans is associated with cancer progression. We used an integrated strategy (mass spectrometry in combination with lectin microarray analysis) to elucidate aberrant glycosylation in a hypoxia-induced EMT model using breast cancer cell lines MCF7 and MDA-MB-231. The model showed reduced levels of bisecting GlcNAc structures, and downregulated expression of the corresponding glycosyltransferase MGAT3. MGAT3 overexpression in MCF7 suppressed cell migration, proliferation, colony formation, expression of EMT markers, and AKT signaling pathway, whereas MGAT3 knockdown (shRNA silencing) had opposite effects. Our findings clearly demonstrate the functional role (and effects of dysregulation) of bisecting GlcNAc structures in hypoxia-induced EMT, and provide a useful basis for further detailed studies of physiological functions of these structures in breast cancer.
Collapse
Affiliation(s)
- Zengqi Tan
- College of Life Science, Northwest University, Xi'an, China
| | - Chenxing Wang
- School of Biotechnology, Jiangnan University, Wuxi, China
| | - Xiang Li
- College of Life Science, Northwest University, Xi'an, China.,Wuxi Medical School, Jiangnan University, Wuxi, China
| | - Feng Guan
- College of Life Science, Northwest University, Xi'an, China
| |
Collapse
|