1
|
Alvarez MRS, Moreno PG, Grijaldo-Alvarez SJB, Yadlapati A, Zhou Q, Narciso MP, Completo GC, Nacario RC, Rabajante JF, Heralde FM, Lebrilla CB. The effects of immortalization on the N-glycome and proteome of CDK4-transformed lung cancer cells. Glycobiology 2024; 34:cwae030. [PMID: 38579012 PMCID: PMC11041852 DOI: 10.1093/glycob/cwae030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/26/2024] [Accepted: 04/02/2024] [Indexed: 04/07/2024] Open
Abstract
Biological experiments are often conducted in vitro using immortalized cells due to their accessibility and ease of propagation compared to primary cells and live animals. However, immortalized cells may present different proteomic and glycoproteomic characteristics from the primary cell source due to the introduction of genes that enhance proliferation (e.g. CDK4) or enable telomere lengthening. To demonstrate the changes in phenotype upon CDK4-transformation, we performed LC-MS/MS glycomic and proteomic characterizations of a human lung cancer primary cell line (DTW75) and a CDK4-transformed cell line (GL01) derived from DTW75. We observed that the primary and CDK4-transformed cells expressed significantly different levels of sialylated, fucosylated, and sialofucosylated N-glycans. Specifically, the primary cells expressed higher levels of hybrid- and complex-type sialylated N-glycans, while CDK4-transformed cells expressed higher levels of complex-type fucosylated and sialofucosylated N-glycans. Further, we compared the proteomic differences between the cell lines and found that CDK4-transformed cells expressed higher levels of RNA-binding and adhesion proteins. Further, we observed that the CDK4-transformed cells changed N-glycosylation after 31 days in cell culture, with a decrease in high-mannose and increase in fucosylated, sialylated, and sialofucosylated N-glycans. Identifying these changes between primary and CDK4-transformed cells will provide useful insight when adapting cell lines that more closely resemble in vivo physiological conditions.
Collapse
Affiliation(s)
- Michael Russelle S Alvarez
- Department of Chemistry, University of California, Davis, 1 Shields Avenue, Davis, California, 95616, USA
| | - Patrick Gabriel Moreno
- Molecular Diagnostics and Cellular Therapeutics Laboratory, Lung Center of the Philippines, Quezon City, 1100, Philippines
| | - Sheryl Joyce B Grijaldo-Alvarez
- Department of Chemistry, University of California, Davis, 1 Shields Avenue, Davis, California, 95616, USA
- Institute of Chemistry, College of Arts and Sciences, University of the Philippines Los Baños, 4031, Philippines
| | - Anirudh Yadlapati
- Department of Chemistry, University of California, Davis, 1 Shields Avenue, Davis, California, 95616, USA
| | - Qingwen Zhou
- Department of Chemistry, University of California, Davis, 1 Shields Avenue, Davis, California, 95616, USA
| | - Michelle P Narciso
- Institute of Mathematical Sciences and Physics, College of Arts and Sciences, University of the Philippines Los Baños, 4031, Philippines
| | - Gladys Cherisse Completo
- Institute of Chemistry, College of Arts and Sciences, University of the Philippines Los Baños, 4031, Philippines
| | - Ruel C Nacario
- Institute of Chemistry, College of Arts and Sciences, University of the Philippines Los Baños, 4031, Philippines
| | - Jomar F Rabajante
- Institute of Mathematical Sciences and Physics, College of Arts and Sciences, University of the Philippines Los Baños, 4031, Philippines
| | - Francisco M Heralde
- Molecular Diagnostics and Cellular Therapeutics Laboratory, Lung Center of the Philippines, Quezon City, 1100, Philippines
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, 1000, Philippines
| | - Carlito B Lebrilla
- Department of Chemistry, University of California, Davis, 1 Shields Avenue, Davis, California, 95616, USA
- Department of Chemistry, Biochemistry, Molecular, Cellular and Developmental Biology Group, University of California, Davis, 1 Shields Avenue, Davis, California, 95616, USA
| |
Collapse
|
2
|
Zhang Y, Lu W, Chen Y, Lin Y, Yang X, Wang H, Liu Z. The miR-19b-3p-MAP2K3-STAT3 feedback loop regulates cell proliferation and invasion in esophageal squamous cell carcinoma. Mol Oncol 2021; 15:1566-1583. [PMID: 33660414 PMCID: PMC8096789 DOI: 10.1002/1878-0261.12934] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 01/22/2021] [Accepted: 02/26/2021] [Indexed: 02/05/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is one of the most refractory malignancies worldwide. Mitogen-activated protein kinase 3 (MAP2K3) has a contradictory role in tumor progression, and the function and expression patterns of MAP2K3 in ESCC remain to be determined. We found that MAP2K3 expression to be downregulated in ESCC, and MAP2K3 downregulation correlated with clinically poor survival. MAP2K3 inhibited ESCC cell proliferation and invasion in vitro and in vivo. MAP2K3 suppressed STAT3 expression and activation. Mechanistically, MAPSK3 interacted with MDM2 to promote STAT3 degradation via the ubiquitin-proteasome pathway. Furthermore, exosomal miR-19b-3p derived from the plasma of patients with ESCC could suppress MAP2K3 expression to promote ESCC tumorigenesis. STAT3 was found to bind to the MIR19B promoter and increased the expression of miR-19b-3p in ESCC cells. In summary, our results demonstrated that the miR-19b-3p-MAP2K3-STAT3 feedback loop regulates ESCC tumorigenesis and elucidates the potential of therapeutically targeting this pathway in ESCC.
Collapse
Affiliation(s)
- Ying Zhang
- Department of PathologySun Yat‐Sen University Cancer CenterGuangzhouChina
- Sun Yat‐Sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Weiqing Lu
- Department of OrthopaedicsFirst Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Yelong Chen
- Department of OrthopaedicsFirst Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Youbin Lin
- Department of OrthopaedicsFirst Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Xia Yang
- Sun Yat‐Sen University Cancer CenterState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Hu Wang
- Department of OrthopaedicsFirst Affiliated Hospital of Shantou University Medical CollegeShantouChina
| | - Zhaoyong Liu
- Department of OrthopaedicsFirst Affiliated Hospital of Shantou University Medical CollegeShantouChina
| |
Collapse
|
3
|
Fernández Muñoz B, Lopez-Navas L, Gonzalez Bermejo M, Lomas Romero IM, Montiel Aguilera MÁ, Campos Cuerva R, Arribas Arribas B, Nogueras S, Carmona Sánchez G, Santos González M. A PROPRIETARY GMP HUMAN PLATELET LYSATE FOR THE EXPANSION OF DERMAL FIBROBLASTS FOR CLINICAL APPLICATIONS. Platelets 2021; 33:98-109. [PMID: 33393414 DOI: 10.1080/09537104.2020.1856356] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Recent years have witnessed the introduction of ex vivo expanded dermal fibroblasts for several cell therapy and tissue-engineering applications, including the treatment of facial scars and burns, representing a promising cell type for regenerative medicine. We tested different in-house produced human platelet lysate (HPL) solutions against fetal bovine serum as supplements for in vitro fibroblast expansion by comparing cell yield, molecular marker expression, extracellular matrix (ECM) generation, genomic stability and global gene expression. Our in-house produced HPL supported fibroblast growth at levels similar to those for FBS and commercial HPL products and was superior to AB human serum. Cells grown in HPL maintained a fibroblast phenotype (VIM+, CD44+, CD13+, CD90+), ECM generation capacity (FN+, COL1+) and a normal karyotype, although gene expression profiling revealed changes related to cell metabolism, adhesion and cellular senescence. The HPL manufacturing process was validated within a GMP compliant system and the solution was stable at -80ºC and -20ºC for 2 years. Dermal fibroblasts expanded in vitro with HPL maintain a normal karyotype and expression of fibroblast markers, with only minor changes in their global gene expression profile. Our in-house produced GMP-HPL is an efficient, safe and economical cell culture supplement that can help increase the healthcare activity of blood transfusion centers through the re-use of transfusional plasma and platelets approaching their expiration date. Currently, our HPL solution is approved by the Spanish Agency of Medicines and Medical Devices and is being used in the manufacture of cell therapy products.
Collapse
Affiliation(s)
- Beatriz Fernández Muñoz
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC), Red Andaluza de Diseño y Traslación de Terapias Avanzadas (RADyTTA), Seville, Spain.,Departamento de Neurociencia Aplicada, Instituto de Investigaciones Biomédicas de Sevilla (IBIS), Seville, Spain
| | - Luis Lopez-Navas
- Unidad de Coordinación, Red Andaluza de Diseño y Traslación de Terapias Avanzadas (RADyTTA), Seville, Spain
| | - María Gonzalez Bermejo
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC), Red Andaluza de Diseño y Traslación de Terapias Avanzadas (RADyTTA), Seville, Spain.,Program in Biología Molecular, Biomedicina e Investigación Clínica, University of Seville, Seville, Spain
| | - Isabel María Lomas Romero
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC), Red Andaluza de Diseño y Traslación de Terapias Avanzadas (RADyTTA), Seville, Spain
| | - Miguel Ángel Montiel Aguilera
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC), Red Andaluza de Diseño y Traslación de Terapias Avanzadas (RADyTTA), Seville, Spain
| | - Rafael Campos Cuerva
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC), Red Andaluza de Diseño y Traslación de Terapias Avanzadas (RADyTTA), Seville, Spain.,Program in Biología Molecular, Biomedicina e Investigación Clínica, University of Seville, Seville, Spain.,Centro de Transfusiones, Tejidos y Células de Sevilla (CTTS), Fundación Pública Andaluza para la Gestión de la Investigación en Salud en Sevilla (FISEVI), Seville, Spain
| | - Blanca Arribas Arribas
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC), Red Andaluza de Diseño y Traslación de Terapias Avanzadas (RADyTTA), Seville, Spain.,Program in Pharmaceutical Technology and Medicine Sciences (Pharmacy), University of Seville, Seville, Spain
| | - Sonia Nogueras
- Departamento de Terapia Celular, Instituto Maimónides de Investigación Biomédica of Córdoba (IMIBIC), Córdoba, Spain.,Unidad de Terapia Celular, Hospital Universitario Reina Sofía, Cordoba, Spain
| | - Gloria Carmona Sánchez
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC), Red Andaluza de Diseño y Traslación de Terapias Avanzadas (RADyTTA), Seville, Spain.,Unidad de Coordinación, Red Andaluza de Diseño y Traslación de Terapias Avanzadas (RADyTTA), Seville, Spain.,Program in Biomedicine, University of Granada, Granada, Spain
| | - Mónica Santos González
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC), Red Andaluza de Diseño y Traslación de Terapias Avanzadas (RADyTTA), Seville, Spain.,Centro de Transfusiones, Tejidos y Células de Sevilla (CTTS), Fundación Pública Andaluza para la Gestión de la Investigación en Salud en Sevilla (FISEVI), Seville, Spain
| |
Collapse
|
4
|
Anjitha R, Antony A, Shilpa O, Anupama KP, Mallikarjunaiah S, Gurushankara HP. Malathion induced cancer-linked gene expression in human lymphocytes. ENVIRONMENTAL RESEARCH 2020; 182:109131. [PMID: 32069766 DOI: 10.1016/j.envres.2020.109131] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND Malathion is the most widely used organophosphate pesticide in agriculture. Increasing cancer incidence in agricultural workers and their children links to the exposure of malathion. Identification of genes involved in the process of carcinogenesis is essential for exploring the role of malathion. The alteration in gene expression by malathion in human lymphocytes has not been explored yet, although hematological malignancies are rampant in humans. OBJECTIVE This study investigates the malathion induced expression of cancer associated genes in human lymphocytes. METHODS Human lymphocyte viability and colony-forming ability were analyzed in malathion treated and control groups. Gene expression profile in control and malathion treated human lymphocytes were performed using a microarray platform. The genes which have significant functions and those involved in different pathways were analyzed using the DAVID database. Differential gene expression upon malathion exposure was validated by quantitative real-time (qRT)-PCR. RESULTS Malathion caused a concentration-dependent reduction in human lymphocyte viability. At low concentration (50 μg/mL) of malathion treatment, human lymphocytes were viable indicating that low concentration of malathion is not cytotoxic and induces the colony formation. Total of 659 genes (15%) were up regulated and 3729 genes (85%) were down regulated in malathion treated human lymphocytes. About 57 cancer associated genes related to the growth and differentiation of B and T cells, immunoglobulin production, haematopoiesis, tumor suppression, oncogenes and signal transduction pathways like MAPK and RAS were induced by malathion. CONCLUSION This study evidences the carcinogenic nature of malathion. Low concentration of this pesticide is not cytotoxic and induces differentially regulated genes in human lymphocytes, which are involved in the initiation, progression, and pathogenesis of cancer.
Collapse
Affiliation(s)
- Ramakrishnan Anjitha
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Periya, 671 320, Kasaragod, Kerala, India
| | - Anet Antony
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Periya, 671 320, Kasaragod, Kerala, India
| | - Olakkaran Shilpa
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Periya, 671 320, Kasaragod, Kerala, India
| | - Kizhakke P Anupama
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Periya, 671 320, Kasaragod, Kerala, India
| | - Shanthala Mallikarjunaiah
- Center for Applied Genetics, Department of Studies in Zoology, Bangalore University, Jnanabharathi, Bengaluru, 560 056, Karnataka, India
| | - Hunasanahally P Gurushankara
- Department of Zoology, School of Biological Sciences, Central University of Kerala, Periya, 671 320, Kasaragod, Kerala, India.
| |
Collapse
|
5
|
PTK7 promotes the malignant properties of cancer stem-like cells in esophageal squamous cell lines. Hum Cell 2020; 33:356-365. [PMID: 31894477 DOI: 10.1007/s13577-019-00309-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/02/2019] [Indexed: 01/08/2023]
Abstract
This study was performed to investigate the role of PTK7 in esophageal squamous cell carcinoma (ESCC) stem-like cells (CSCs). PTK7 expression in ESCCs identified by RT-qPCR, and CSC-like cells were isolated from populations of NEC and TE-1 cells. The CSC-like cells were verified by flow cytometric analyses performed using CD34 and CD133 antibodies, and RT-qPCR and western blot assays were used to examine the self-renewal capability of CSC-like cells. CSC-like cells treated with PTK7 siRNA or a P53-specific inhibitor (PFTα) were analyzed for their sphere formation capacity and their apoptosis and migration/invasion capabilities by sphere formation, flow cytometry, and transwell assay, respectively. Their levels of P53, MKK3, and cleaved caspase 3 expression were examined by western blot analysis. Our results revealed that a majority of the isolated CSC-like cells were CD34+/CD133+ double positive cells. Nango, Sox2, and OCT4 were dramatically increased in the separated CSC-like cells, which had the pluripotency and self-renewal properties of stem cells. Additional, PTK7 was dramatically upregulated in the ESCC tissues and CSC-like cells. An investigation of the function of CSC-like cells revealed that knockdown of PTK7 reduced their sphere formation, promoted apoptosis, and suppressed their migration and invasion abilities, all of which could be significantly reversed by PFTα. Mechanistic studies showed that PFTα could attenuate the upregulation of P53, MKK3, and cleaved caspase 3 expression that was induced by PTK7 knockdown in CSC-like cells. PTK7 increased the malignant behaviors of CSC-like cells derived from ESCC cells by regulating p53. Therefore, this study suggests PTK7 as an underlying target for therapy against ESCC.
Collapse
|
6
|
Wang J, Zhao Q. Linc02381 Exacerbates Rheumatoid Arthritis Through Adsorbing miR-590-5p and Activating the Mitogen-Activated Protein Kinase Signaling Pathway in Rheumatoid arthritis-fibroblast-like synoviocytes. Cell Transplant 2020; 29:963689720938023. [PMID: 32608996 PMCID: PMC7563894 DOI: 10.1177/0963689720938023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/17/2020] [Accepted: 06/01/2020] [Indexed: 12/29/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease. New evidence suggested that linc02381 suppressed colorectal cancer progression by regulating PI3 K signaling pathway, but the role of linc02381 in other diseases, such as RA, remains unclear. This study aimed to reveal the mechanism of linc02381 in RA progression. In vivo and in vitro, we found that linc02381 was upregulated in RA synovial tissues or RA fibroblast-like synoviocytes (RA-FLSs, P < 0.01), which were detected by quantitative real-time polymerase chain reaction. Cell Counting Kit-8, EDU, and Transwell assays revealed that linc02381 overexpression enhanced cell proliferation and invasion, and linc02381 knockdown inhibited cell proliferation and invasion in FLSs. Moreover, the results of bioinformatics analysis, luciferase reporter gene assay, and pull-down assay verified that linc02381 could directly bind with miR-590-5p. MiR-590-5p was downregulated in RA-FLSs, and overexpression of linc02381 suppressed expression of miR-590-5p that post-transcriptionally suppressed the expression of mitogen-activated protein kinase kinase 3 (MAP2K3), and overexpression of miR-590-5p reversed the effect of linc02381 overexpression on MAP2K3 expression. MiR-590-5p inhibitor reversed the inhibition effect of linc02381 knockdown on proliferation and invasion of FLSs, which enhanced expression of MAP2K3, and activation of p38 and AP-1 in the MAPK signaling pathway. In summary, linc02381 was upregulated in RA synovial tissues and RA-FLSs, and it exacerbated RA by adsorbing miR-590-5p to activate the MAPK signaling pathway.
Collapse
Affiliation(s)
- Jing Wang
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, People’s Republic of China
| | - Qing Zhao
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, People’s Republic of China
| |
Collapse
|
7
|
Blunder S, Messner B, Scharinger B, Doppler C, Zeller I, Zierer A, Laufer G, Bernhard D. Targeted gene expression analyses and immunohistology suggest a pro-proliferative state in tricuspid aortic valve-, and senescence and viral infections in bicuspid aortic valve-associated thoracic aortic aneurysms. Atherosclerosis 2018; 271:111-119. [DOI: 10.1016/j.atherosclerosis.2018.02.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 01/11/2018] [Accepted: 02/02/2018] [Indexed: 01/13/2023]
|
8
|
Abbadie C, Pluquet O, Pourtier A. Epithelial cell senescence: an adaptive response to pre-carcinogenic stresses? Cell Mol Life Sci 2017; 74:4471-4509. [PMID: 28707011 PMCID: PMC11107641 DOI: 10.1007/s00018-017-2587-9] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/27/2017] [Accepted: 07/06/2017] [Indexed: 01/01/2023]
Abstract
Senescence is a cell state occurring in vitro and in vivo after successive replication cycles and/or upon exposition to various stressors. It is characterized by a strong cell cycle arrest associated with several molecular, metabolic and morphologic changes. The accumulation of senescent cells in tissues and organs with time plays a role in organismal aging and in several age-associated disorders and pathologies. Moreover, several therapeutic interventions are able to prematurely induce senescence. It is, therefore, tremendously important to characterize in-depth, the mechanisms by which senescence is induced, as well as the precise properties of senescent cells. For historical reasons, senescence is often studied with fibroblast models. Other cell types, however, much more relevant regarding the structure and function of vital organs and/or regarding pathologies, are regrettably often neglected. In this article, we will clarify what is known on senescence of epithelial cells and highlight what distinguishes it from, and what makes it like, replicative senescence of fibroblasts taken as a standard.
Collapse
Affiliation(s)
- Corinne Abbadie
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161-M3T-Mechanisms of Tumorigenesis and Targeted Therapies, 59000, Lille, France.
| | - Olivier Pluquet
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161-M3T-Mechanisms of Tumorigenesis and Targeted Therapies, 59000, Lille, France
| | - Albin Pourtier
- Univ. Lille, CNRS, Institut Pasteur de Lille, UMR 8161-M3T-Mechanisms of Tumorigenesis and Targeted Therapies, 59000, Lille, France
| |
Collapse
|
9
|
Mozdoori N, Safarian S, Sheibani N. Augmentation of the cytotoxic effects of zinc oxide nanoparticles by MTCP conjugation: Non-canonical apoptosis and autophagy induction in human adenocarcinoma breast cancer cell lines. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 78:949-959. [PMID: 28576071 PMCID: PMC6018014 DOI: 10.1016/j.msec.2017.03.300] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 03/27/2017] [Accepted: 03/31/2017] [Indexed: 01/04/2023]
Abstract
Zinc oxide nanoparticles are very toxic, but their agglomeration reduces their lethal cytotoxic effects. Here we tested the hypothesis that conjugation of ZnO nanoparticles via Meso-Tetra (4-Carboxyphenyl) Porphyrin (MTCP) could provide electrostatic or steric stabilization of ZnO nanoparticles and increase their cytotoxic effects. The cytotoxicity and cell death induction were assessed using two human breast adenocarcinoma cell lines (MCF-7 and MDA-MB-468). The MTT results indicated that the toxicity of ZnO nanoparticles was significantly increased upon MTCP conjugation. Annexin/PI and real time RT-PCR results demonstrated that the ZnO-MTCP nanoparticles induced cell death via different non-canonical pathways that are under ca2+ control. Calcium signaling could regulate lysosomal dependent apoptosis and death autophagy, and killing of the two selected types of breast cancer cells.
Collapse
Affiliation(s)
- Najmeh Mozdoori
- Cell and Molecular Biology Department, School of Biology, College of Science, University of Tehran, Tehran 1417614411, Iran
| | - Shahrokh Safarian
- Cell and Molecular Biology Department, School of Biology, College of Science, University of Tehran, Tehran 1417614411, Iran.
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, Biomedical Engineering, University of Wisconsin, School of Medicine and Public Health, Madison, WI, USA; Department of Cell and Regenerative Biology, University of Wisconsin, School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
10
|
Ratushnyy A, Lobanova M, Buravkova LB. Expansion of adipose tissue-derived stromal cells at "physiologic" hypoxia attenuates replicative senescence. Cell Biochem Funct 2017; 35:232-243. [PMID: 28589682 DOI: 10.1002/cbf.3267] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 03/02/2017] [Accepted: 03/28/2017] [Indexed: 12/16/2022]
Abstract
Multipotent mesenchymal stromal cells are considered as a perspective tool in cell therapy and regenerative medicine. Unfortunately, autologous cell therapy does not always provide positive outcomes in elder donors, perhaps as a result of the alterations of stem cell compartments. The mechanisms of stem and progenitor cell senescence and the factors engaged are investigated intensively. In present paper, we elucidated the effects of tissue-related O2 on morphology, functions, and transcriptomic profile of adipose tissue-derived stromal cells (ASCs) in replicative senescence in vitro model. Replicatively senescent ASCs at ambient (20%) O2 (12-21 passages) demonstrated an increased average cell size, granularity, reactive oxygen species level, including anion superoxide, lysosomal compartment activity, and IL-6 production. Decreased ASC viability and proliferation, as well as the change of more than 10 senescence-associated gene expression were detected (IGF1, CDKN1C, ID1, CCND1, etc). Long-term ASC expansion at low O2 (5%) revoked in part the replicative senescence-associated alterations.
Collapse
Affiliation(s)
- Andrey Ratushnyy
- Lab. of Cell Physiology, Institute of Biomedical Problems of Russian Academy of Science, Moscow, Russia
| | - Margarita Lobanova
- Lab. of Cell Physiology, Institute of Biomedical Problems of Russian Academy of Science, Moscow, Russia
| | - Ludmila B Buravkova
- Lab. of Cell Physiology, Institute of Biomedical Problems of Russian Academy of Science, Moscow, Russia
| |
Collapse
|
11
|
Klahan S, Huang CC, Chien SC, Wu MS, Wong HSC, Huang CY, Chang WC, Wei PL. Bioinformatic analyses revealed underlying biological functions correlated with oxaliplatin responsiveness. Tumour Biol 2015; 37:583-90. [PMID: 26232912 DOI: 10.1007/s13277-015-3807-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/20/2015] [Indexed: 11/27/2022] Open
Abstract
Colorectal cancer is one of the most common cancers worldwide. Surgery is usually the primary treatment for colon cancers that have not spread to distant sites. However, chemotherapy may be considered after surgery to eliminate remaining cancer cells or in case the cancer has a high risk of recurrence. Oxaliplatin is often used in combination regimens such as FOLFOX, CapeOX, and FOLFOXIRI because of the cost-effectiveness of adjuvant treatment for patients and also the good tolerability profile. However, some patients show resistance to oxaliplatin which causes poor treatment outcomes. Most colon cancer studies focused on treatments and patient survival. Some studies focused on genetic associations of specific genes. However, pathway and network analyses of oxaliplatin resistance in colon cancer cells using gene expression patterns are still lacking. We performed a microarray analysis and found that endothelin-1 (EDN1), dishevelled segment polarity protein (DV1), toll-like receptor 5(TLR5), mitogen-activated protein kinase 3 (MAP2K3), phosphatidylinositol-4,5-bisphosphate 3-kinase, and catalytic subunit beta (PIK3CB) were closely related to responsiveness to oxaliplatin treatment. Furthermore, we found that the signal transduction, melanogenesis, and toll-like receptor signaling pathways might be involved in oxaliplatin-resistant colon cancer. These genes and pathways might be potential targets for improving oxaliplatin treatment in colon cancer patients.
Collapse
Affiliation(s)
- Sukhontip Klahan
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Chi-Cheng Huang
- Department of Surgery, Cathay General Hospital, SiJhih, New Taipei City, Taiwan
- School of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
- School of Medicine, Taipei Medical University, Taipei City, Taiwan
| | - Shu-Chen Chien
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Master Program for Clinical Pharmacogenomics and Pharmacoproteomics, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Mei-Shin Wu
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Henry Sung-Ching Wong
- Master Program for Clinical Pharmacogenomics and Pharmacoproteomics, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Chien-Yu Huang
- Division of General Surgery, Department of Surgery, Department of Neurosurgery Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - Wei-Chiao Chang
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan.
- Master Program for Clinical Pharmacogenomics and Pharmacoproteomics, School of Pharmacy, Taipei Medical University, Taipei, Taiwan.
| | - Po-Li Wei
- Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University, Taipei, Taiwan.
- Division of General Surgery, Department of Surgery, Department of Neurosurgery Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan.
- Department of Surgery, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan.
- Cancer Center, Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan.
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
12
|
Tebaldi T, Zaccara S, Alessandrini F, Bisio A, Ciribilli Y, Inga A. Whole-genome cartography of p53 response elements ranked on transactivation potential. BMC Genomics 2015; 16:464. [PMID: 26081755 PMCID: PMC4470028 DOI: 10.1186/s12864-015-1643-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 05/20/2015] [Indexed: 11/18/2022] Open
Abstract
Background Many recent studies using ChIP-seq approaches cross-referenced to trascriptome data and also to potentially unbiased in vitro DNA binding selection experiments are detailing with increasing precision the p53-directed gene regulatory network that, nevertheless, is still expanding. However, most experiments have been conducted in established cell lines subjected to specific p53-inducing stimuli, both factors potentially biasing the results. Results We developed p53retriever, a pattern search algorithm that maps p53 response elements (REs) and ranks them according to predicted transactivation potentials in five classes. Besides canonical, full site REs, we developed specific pattern searches for non-canonical half sites and 3/4 sites and show that they can mediate p53-dependent responsiveness of associated coding sequences. Using ENCODE data, we also mapped p53 REs in about 44,000 distant enhancers and identified a 16-fold enrichment for high activity REs within those sites in the comparison with genomic regions near transcriptional start sites (TSS). Predictions from our pattern search were cross-referenced to ChIP-seq, ChIP-exo, expression, and various literature data sources. Based on the mapping of predicted functional REs near TSS, we examined expression changes of thirteen genes as a function of different p53-inducing conditions, providing further evidence for PDE2A, GAS6, E2F7, APOBEC3H, KCTD1, TRIM32, DICER, HRAS, KITLG and TGFA p53-dependent regulation, while MAP2K3, DNAJA1 and potentially YAP1 were identified as new direct p53 target genes. Conclusions We provide a comprehensive annotation of canonical and non-canonical p53 REs in the human genome, ranked on predicted transactivation potential. We also establish or corroborate direct p53 transcriptional control of thirteen genes. The entire list of identified and functionally classified p53 REs near all UCSC-annotated genes and within ENCODE mapped enhancer elements is provided. Our approach is distinct from, and complementary to, existing methods designed to identify p53 response elements. p53retriever is available as an R package at: http://tomateba.github.io/p53retriever. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1643-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Toma Tebaldi
- Centre for Integrative Biology (CIBIO), University of Trento, via delle Regole 101, 38123, Mattarello, TN, Italy.
| | - Sara Zaccara
- Centre for Integrative Biology (CIBIO), University of Trento, via delle Regole 101, 38123, Mattarello, TN, Italy.
| | - Federica Alessandrini
- Centre for Integrative Biology (CIBIO), University of Trento, via delle Regole 101, 38123, Mattarello, TN, Italy.
| | - Alessandra Bisio
- Centre for Integrative Biology (CIBIO), University of Trento, via delle Regole 101, 38123, Mattarello, TN, Italy.
| | - Yari Ciribilli
- Centre for Integrative Biology (CIBIO), University of Trento, via delle Regole 101, 38123, Mattarello, TN, Italy.
| | - Alberto Inga
- Centre for Integrative Biology (CIBIO), University of Trento, via delle Regole 101, 38123, Mattarello, TN, Italy.
| |
Collapse
|
13
|
Raval AN, Schmuck EG, Tefera G, Leitzke C, Ark CV, Hei D, Centanni JM, de Silva R, Koch J, Chappell RG, Hematti P. Bilateral administration of autologous CD133+ cells in ambulatory patients with refractory critical limb ischemia: lessons learned from a pilot randomized, double-blind, placebo-controlled trial. Cytotherapy 2014; 16:1720-32. [PMID: 25239491 PMCID: PMC4253573 DOI: 10.1016/j.jcyt.2014.07.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Revised: 07/31/2014] [Accepted: 07/31/2014] [Indexed: 01/18/2023]
Abstract
BACKGROUND AIMS CD133+ cells confer angiogenic potential and may be beneficial for the treatment of critical limb ischemia (CLI). However, patient selection, blinding methods and end points for clinical trials are challenging. We hypothesized that bilateral intramuscular administration of cytokine-mobilized CD133+ cells in ambulatory patients with refractory CLI would be feasible and safe. METHODS In this double-blind, randomized sham-controlled trial, subjects received subcutaneous injections of granulocyte colony-stimulating factor (10 μg/kg per day) for 5 days, followed by leukapheresis, and intramuscular administration of 50-400 million sorted CD133+ cells delivered into both legs. Control subjects received normal saline injections, sham leukapheresis and intramuscular injection of placebo buffered solution. Subjects were followed for 1 year. An aliquot of CD133+ cells was collected from each subject to test for genes associated with cell senescence. RESULTS Seventy subjects were screened, of whom 10 were eligible. Subject enrollment was suspended because of a high rate of mobilization failure in subjects randomly assigned to treatment. Of 10 subjects enrolled (7 randomly assigned to treatment, 3 randomly assigned to control), there were no differences in serious adverse events at 12 months, and blinding was preserved. There were non-significant trends toward improved amputation-free survival, 6-minute walk distance, walking impairment questionnaire and quality of life in subjects randomly assigned to treatment. Successful CD133+ mobilizers expressed fewer senescence-associated genes compared with poor mobilizers. CONCLUSIONS Bilateral administration of autologous CD133+ cells in ambulatory CLI subjects was safe, and blinding was preserved. However, poor mobilization efficiency combined with high CD133+ senescence suggests futility in this approach.
Collapse
Affiliation(s)
- Amish N Raval
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA.
| | - Eric G Schmuck
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Girma Tefera
- Division of Vascular Surgery, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Cathlyn Leitzke
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Cassondra Vander Ark
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Derek Hei
- Waisman Biomanufacturing Facility, Madison, Wisconsin, USA
| | - John M Centanni
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Ranil de Silva
- National Heart and Lung Institute, Imperial College London and NIHR Cardiovascular Biomedical Research Unit, Royal Brompton and Harefield NHS Foundation Trust, London, United Kingdom
| | - Jill Koch
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Richard G Chappell
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Peiman Hematti
- Division of Hematology/Oncology, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| |
Collapse
|
14
|
Noderer WL, Flockhart RJ, Bhaduri A, Diaz de Arce AJ, Zhang J, Khavari PA, Wang CL. Quantitative analysis of mammalian translation initiation sites by FACS-seq. Mol Syst Biol 2014; 10:748. [PMID: 25170020 PMCID: PMC4299517 DOI: 10.15252/msb.20145136] [Citation(s) in RCA: 132] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
An approach combining fluorescence-activated cell sorting and high-throughput DNA sequencing
(FACS-seq) was employed to determine the efficiency of start codon recognition for all possible
translation initiation sites (TIS) utilizing AUG start codons. Using FACS-seq, we measured
translation from a genetic reporter library representing all 65,536 possible TIS sequences spanning
the −6 to +5 positions. We found that the motif RYMRMVAUGGC enhanced start codon
recognition and translation efficiency. However, dinucleotide interactions, which cannot be conveyed
by a single motif, were also important for modeling TIS efficiency. Our dataset combined with
modeling allowed us to predict genome-wide translation initiation efficiency for all mRNA
transcripts. Additionally, we screened somatic TIS mutations associated with tumorigenesis to
identify candidate driver mutations consistent with known tumor expression patterns. Finally, we
implemented a quantitative leaky scanning model to predict alternative initiation sites that produce
truncated protein isoforms and compared predictions with ribosome footprint profiling data. The
comprehensive analysis of the TIS sequence space enables quantitative predictions of translation
initiation based on genome sequence.
Collapse
Affiliation(s)
- William L Noderer
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Ross J Flockhart
- The Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Aparna Bhaduri
- The Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA The Program in Cancer Biology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Jiajing Zhang
- The Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Paul A Khavari
- The Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, USA Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA, USA
| | - Clifford L Wang
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| |
Collapse
|
15
|
MacNeil AJ, Jiao SC, McEachern LA, Yang YJ, Dennis A, Yu H, Xu Z, Marshall JS, Lin TJ. MAPK kinase 3 is a tumor suppressor with reduced copy number in breast cancer. Cancer Res 2013; 74:162-72. [PMID: 24233520 DOI: 10.1158/0008-5472.can-13-1310] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cancers are initiated as a result of changes that occur in the genome. Identification of gains and losses in the structure and expression of tumor-suppressor genes and oncogenes lies at the root of the understanding of cancer cell biology. Here, we show that the mitogen-activated protein kinase (MAPK) MKK3 suppresses the growth of breast cancer, in which it varies in copy number. A pervasive loss of MKK3 gene copy number in patients with breast cancer is associated with an impairment of MKK3 expression and protein level in malignant tissues. To assess the functional role of MKK3 in breast cancer, we showed in an animal model that MKK3 activity is required for suppression of tumor growth. Active MKK3 enhanced expression of the cyclin-dependent kinase inhibitors p21(Cip1/Waf1) and p27(Kip1), leading to increased cell-cycle arrest in G1 phase of the cell cycle. Our results reveal the functional significance of MKK3 as a tumor suppressor and improve understanding of the dynamic role of the MAPK pathway in tumor progression.
Collapse
Affiliation(s)
- Adam J MacNeil
- Authors' Affiliations: Departments of Microbiology and Immunology, Pediatrics, Physiology and Biophysics, and Pathology, Dalhousie University; Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada; Department of Medical Oncology, General Hospital of the People's Liberation Army, Beijing; and Institute of Zoonosis, College of Animal Sciences and Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Xu G, Zhang Y, Wei J, Jia W, Ge Z, Zhang Z, Liu X. MicroRNA-21 promotes hepatocellular carcinoma HepG2 cell proliferation through repression of mitogen-activated protein kinase-kinase 3. BMC Cancer 2013; 13:469. [PMID: 24112539 PMCID: PMC3852118 DOI: 10.1186/1471-2407-13-469] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 09/17/2013] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND microRNA 21 (miR-21) has been demonstrated to be significantly elevated in many types of cancers, including the hepatocellular carcinoma (HCC). In the present study, we investigated the role of miR-21 in HCC by identifying its novel targets, as well as its underlying molecular mechanism. METHODS The expression of mitogen-activated protein kinase-kinase 3 (MAP2K3) in human HCC tumor tissues and adjacent non-tumor tissues was determined by immunohistochemistry staining (IHC) analysis. The 3'-untranslated region (3'-UTR) of MAP2K3 combined with miR-21 was experimentally verified by a miRNA luciferase reporter approach. Moreover, the role of miR-21 in regulating HCC cell proliferation was analyzed by an MTT assay infected with miR-21mimics/sponge inhibitor Adenoviral viral vectors. RESULTS By immunohistochemistry staining analysis, we found that mitogen-activated protein kinase-kinase 3 (MAP2K3) was strikingly repressed in the human HCC tumor tissues, in comparison with the adjacent non-tumor tissues in clinical settings. More importantly, the repression of MAP2K3 was inversely correlated with the expression of miR-21 in HCC. Further study demonstrated that the MAP2K3 was a novel direct target of miR-21, which was experimentally validated by a miRNA luciferase reporter approach. In HepG2 cells, inhibition of miR-21 expression with an adenoviral miR-21 sponge vector profoundly suppressed cell proliferation by up-regulating MAP2K3 expression at both mRNA and protein levels. CONCLUSIONS These results provide a clinical evidence that MAP2K3 may be a tumor repressor gene, and it is a direct target of miR-21 in HCC, indicating an underlying mechanism by which miR-21 is able to directly target MAP2K3 and inhibit its expression during the carcinogenesis of HCC, at both transcriptional and post-translational levels. This study also suggests that targeting miR-21-MAP2K3 pathway may be a promising strategy in the prevention and treatment of HCC.
Collapse
Affiliation(s)
- Guangxian Xu
- General Hospital of Ningxia Medical University, Yinchuan 750004, China.
| | | | | | | | | | | | | |
Collapse
|
17
|
Gatta V, D'Aurora M, Lanuti P, Pierdomenico L, Sperduti S, Palka G, Gesi M, Marchisio M, Miscia S, Stuppia L. Gene expression modifications in Wharton's Jelly mesenchymal stem cells promoted by prolonged in vitro culturing. BMC Genomics 2013; 14:635. [PMID: 24053474 PMCID: PMC3849041 DOI: 10.1186/1471-2164-14-635] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Accepted: 09/17/2013] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND It has been demonstrated that the umbilical cord matrix, represented by the Wharton's Jelly (WJ), contains a great number of mesenchymal stem cells (MSCs), characterized by the expression of specific MSCs markers, shared by both human and animal models. The easy access to massive WJ amount makes it an attractive source of MSCs for cell-based therapies. However, as in other stem cell models, a deeper investigation of WJ-derived MSCs (WJ-MSCs) biological properties, probably modulated by their prolonged expansion and fast growth abilities, is required before their use in clinical settings. In this context, in order to analyze specific gene expression modifications occurring in WJ-MSCs, along with their culture prolongation, we investigated the transcriptomic profiles of WJ-MSCs after 4 and 12 passages of in vitro expansion by microarray analysis. RESULTS Hierarchical clustering analysis of the data set originated from a total of 6 experiments revealed that in vitro expansion of WJ-MSCs up to 12 passages promote selective over-expression of 157 genes and down-regulation of 440 genes compared to the 4th passage. IPA software analysis of the biological functions related to the identified sets of genes disclosed several transcripts related to inflammatory and cell stress response, cell proliferation and maturation, and apoptosis. CONCLUSIONS Taken together, these modifications may lead to an impairment of both cell expansion ability and resistance to apoptosis, two hallmarks of aging cells. In conclusion, results provided by the present study suggest the need to develop novel culture protocols able to preserve stem cell plasticity.
Collapse
Affiliation(s)
- Valentina Gatta
- Department of Medicine and Aging Science, School of Medicine and Health Sciences, University "G, d'Annunzio" Chieti-Pescara, via dei Vestini 31, 66013, Chieti, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Cifola I, Pietrelli A, Consolandi C, Severgnini M, Mangano E, Russo V, De Bellis G, Battaglia C. Comprehensive genomic characterization of cutaneous malignant melanoma cell lines derived from metastatic lesions by whole-exome sequencing and SNP array profiling. PLoS One 2013; 8:e63597. [PMID: 23704925 PMCID: PMC3660556 DOI: 10.1371/journal.pone.0063597] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Accepted: 04/04/2013] [Indexed: 02/08/2023] Open
Abstract
Cutaneous malignant melanoma is the most fatal skin cancer and although improved comprehension of its pathogenic pathways allowed to realize some effective molecular targeted therapies, novel targets and drugs are still needed. Aiming to add genetic information potentially useful for novel targets discovery, we performed an extensive genomic characterization by whole-exome sequencing and SNP array profiling of six cutaneous melanoma cell lines derived from metastatic patients. We obtained a total of 3,325 novel coding single nucleotide variants, including 2,172 non-synonymous variants. We catalogued the coding mutations according to Sanger COSMIC database and to a manually curated list including genes involved in melanoma pathways identified by mining recent literature. Besides confirming the presence of known melanoma driver mutations (BRAF(V600E), NRAS(Q61R) ), we identified novel mutated genes involved in signalling pathways crucial for melanoma pathogenesis and already addressed by current targeted therapies (such as MAPK and glutamate pathways). We also identified mutations in four genes (MUC19, PAICS, RBMXL1, KIF23) never reported in melanoma, which might deserve further investigations. All data are available to the entire research community in our Melanoma Exome Database (at https://155.253.6.64/MExDB/). In summary, these cell lines are valuable biological tools to improve the genetic comprehension of this complex cancer disease and to study functional relevance of individual mutational events, and these findings could provide insights potentially useful for identification of novel therapeutic targets for cutaneous malignant melanoma.
Collapse
Affiliation(s)
- Ingrid Cifola
- Institute for Biomedical Technologies, National Research Council, Milan, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Lahtela J, Corson LB, Hemmes A, Brauer MJ, Koopal S, Lee J, Hunsaker TL, Jackson PK, Verschuren EW. A high-content cellular senescence screen identifies candidate tumor suppressors, including EPHA3. Cell Cycle 2013; 12:625-34. [PMID: 23324396 PMCID: PMC3594263 DOI: 10.4161/cc.23515] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Activation of a cellular senescence program is a common response to prolonged oncogene activation or tumor suppressor loss, providing a physiological mechanism for tumor suppression in premalignant cells. The link between senescence and tumor suppression supports the hypothesis that a loss-of-function screen measuring bona fide senescence marker activation should identify candidate tumor suppressors. Using a high-content siRNA screening assay for cell morphology and proliferation measures, we identify 12 senescence-regulating kinases and determine their senescence marker signatures, including elevation of senescence-associated β-galactosidase, DNA damage and p53 or p16INK4a expression. Consistent with our hypothesis, SNP array CGH data supports loss of gene copy number of five senescence-suppressing genes across multiple tumor samples. One such candidate is the EPHA3 receptor tyrosine kinase, a gene commonly mutated in human cancer. We demonstrate that selected intracellular EPHA3 tumor-associated point mutations decrease receptor expression level and/or receptor tyrosine kinase (RTK) activity. Our study therefore describes a new strategy to mine for novel candidate tumor suppressors and provides compelling evidence that EPHA3 mutations may promote tumorigenesis only when key senescence-inducing pathways have been inactivated.
Collapse
Affiliation(s)
- Jenni Lahtela
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Prognostic Impact of del(17p) and del(22q) as assessed by interphase FISH in sporadic colorectal carcinomas. PLoS One 2012; 7:e42683. [PMID: 22912721 PMCID: PMC3422354 DOI: 10.1371/journal.pone.0042683] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 07/11/2012] [Indexed: 12/11/2022] Open
Abstract
Background Most sporadic colorectal cancer (sCRC) deaths are caused by metastatic dissemination of the primary tumor. New advances in genetic profiling of sCRC suggest that the primary tumor may contain a cell population with metastatic potential. Here we compare the cytogenetic profile of primary tumors from liver metastatic versus non-metastatic sCRC. Methodology/Principal Findings We prospectively analyzed the frequency of numerical/structural abnormalities of chromosomes 1, 7, 8, 13, 14, 17, 18, 20, and 22 by iFISH in 58 sCRC patients: thirty-one non-metastatic (54%) vs. 27 metastatic (46%) disease. From a total of 18 probes, significant differences emerged only for the 17p11.2 and 22q11.2 chromosomal regions. Patients with liver metastatic sCRC showed an increased frequency of del(17p11.2) (10% vs. 67%;p<.001) and del(22q11.2) (0% vs. 22%;p = .02) versusnon-metastatic cases. Multivariate analysis of prognostic factors for overall survival (OS) showed that the only clinical and cytogenetic parameters that had an independent adverse impact on patient outcome were the presence of del(17p) with a 17p11.2 breakpoint and del(22q11.2). Based on these two cytogenetic variables, patients were classified into three groups: low- (no adverse features), intermediate- (one adverse feature) and high-risk (two adverse features)- with significantly different OS rates at 5-years (p<.001): 92%, 53% and 0%, respectively. Conclusions/Significance Our results unravel the potential implication of del(17p11.2) in sCRC patients with liver metastasis as this cytogenetic alteration appears to be intrinsically related to an increased metastatic potential and a poor outcome, providing additional prognostic information to that associated with other cytogenetic alterations such as del(22q11.2). Additional prospective studies in larger series of patients would be required to confirm the clinical utility of the new prognostic markers identified.
Collapse
|
21
|
Jia M, Souchelnytskyi S. Proteome profiling of heat shock of human primary breast epithelial cells, a dataset report. Cell Stress Chaperones 2011; 16:459-67. [PMID: 21221872 PMCID: PMC3118829 DOI: 10.1007/s12192-010-0253-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Revised: 12/13/2010] [Accepted: 12/17/2010] [Indexed: 01/24/2023] Open
Abstract
Exposure to elevated temperatures has a strong effect on cell functions, and is used in clinical practice. Hyperthermia may affect multiple regulatory mechanisms in cells. To understand better the response to hyperthermia of immortalized primary human breast epithelial cells, we performed a proteomics study of these cells cultured at 34°C or 39°C. Twenty-four proteins were shown to be differentially expressed due to hyperthermia. Analysis of these proteins showed the potential involvement of various biological processes in response to hyperthermia, e.g., cell adhesion, cell communication, and cell cycle. Transforming growth factor-β2 (TGF-β2) and heat shock protein 27 (HSP27) were found to be upregulated at 39°C. TGF-β2 was found to affect expression of HSP27, and to have a protective role in hyperthermia-induced cell death. Thus, the dataset described here of hyperthermia-related proteins in human primary breast epithelial cells predicts a number of cellular activities affected by exposure to high temperatures and provides a set of proteins for further studies.
Collapse
Affiliation(s)
- Min Jia
- Department of Oncology-Pathology, Karolinska Biomics Center, Karolinska Institutet, Stockholm, Sweden.
| | | |
Collapse
|
22
|
Zakharchenko O, Greenwood C, Alldridge L, Souchelnytskyi S. Optimized Protocol for Protein Extraction from the Breast Tissue that is Compatible with Two-Dimensional Gel Electrophoresis. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2011; 5:37-42. [PMID: 21494400 PMCID: PMC3076014 DOI: 10.4137/bcbcr.s6263] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Proteomics is a highly informative approach to analyze cancer-associated transformation in tissues. The main challenge to use a tissue for proteomics studies is the small sample size and difficulties to extract and preserve proteins. The choice of a buffer compatible with proteomics applications is also a challenge. Here we describe a protocol optimized for the most efficient extraction of proteins from the human breast tissue in a buffer compatible with two-dimensional gel electrophoresis (2D-GE). This protocol is based on mechanically assisted disintegration of tissues directly in the 2D-GE buffer. Our method is simple, robust and easy to apply in clinical practice. We demonstrate high quality of separation of proteins prepared according to the reported here protocol.
Collapse
Affiliation(s)
- Olena Zakharchenko
- Karolinska Institutet, Dept. Oncology-Pathology, Karolinska Biomics Center, Cancer Translational Research Unit, Stockholm, Sweden
| | | | | | | |
Collapse
|