1
|
Jiménez C, Garrote-de-Barros A, López-Portugués C, Hernández-Sánchez M, Díez P. Characterization of Human B Cell Hematological Malignancies Using Protein-Based Approaches. Int J Mol Sci 2024; 25:4644. [PMID: 38731863 PMCID: PMC11083628 DOI: 10.3390/ijms25094644] [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: 03/19/2024] [Revised: 04/19/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
The maturation of B cells is a complex, multi-step process. During B cell differentiation, errors can occur, leading to the emergence of aberrant versions of B cells that, finally, constitute a malignant tumor. These B cell malignancies are classified into three main groups: leukemias, myelomas, and lymphomas, the latter being the most heterogeneous type. Since their discovery, multiple biological studies have been performed to characterize these diseases, aiming to define their specific features and determine potential biomarkers for diagnosis, stratification, and prognosis. The rise of advanced -omics approaches has significantly contributed to this end. Notably, proteomics strategies appear as promising tools to comprehensively profile the final molecular effector of these cells. In this narrative review, we first introduce the main B cell malignancies together with the most relevant proteomics approaches. Then, we describe the core studies conducted in the field and their main findings and, finally, we evaluate the advantages and drawbacks of flow cytometry, mass cytometry, and mass spectrometry for the profiling of human B cell disorders.
Collapse
Affiliation(s)
- Cristina Jiménez
- Hematology Department, University Hospital of Salamanca (HUS/IBSAL), CIBERONC and Cancer Research Institute of Salamanca-IBMCC (USAL-CSIC), 37007 Salamanca, Spain;
| | - Alba Garrote-de-Barros
- Department of Biochemistry and Molecular Biology, Pharmacy School, Universidad Complutense de Madrid, 28040 Madrid, Spain; (A.G.-d.-B.); (M.H.-S.)
- Department of Translational Hematology, Instituto de Investigación Hospital 12 de Octubre (imas12), Hematological Malignancies Clinical Research Unit H12O-CNIO, 28029 Madrid, Spain
| | - Carlos López-Portugués
- Department of Physical and Analytical Chemistry Chemistry, Faculty of Chemistry, University of Oviedo, 33006 Oviedo, Spain;
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
| | - María Hernández-Sánchez
- Department of Biochemistry and Molecular Biology, Pharmacy School, Universidad Complutense de Madrid, 28040 Madrid, Spain; (A.G.-d.-B.); (M.H.-S.)
- Department of Translational Hematology, Instituto de Investigación Hospital 12 de Octubre (imas12), Hematological Malignancies Clinical Research Unit H12O-CNIO, 28029 Madrid, Spain
| | - Paula Díez
- Department of Physical and Analytical Chemistry Chemistry, Faculty of Chemistry, University of Oviedo, 33006 Oviedo, Spain;
- Health Research Institute of the Principality of Asturias (ISPA), 33011 Oviedo, Spain
- Department of Functional Biology, Faculty of Medicine and Health Science, University of Oviedo, 33006 Oviedo, Spain
| |
Collapse
|
2
|
Psatha K, Kollipara L, Drakos E, Deligianni E, Brintakis K, Patsouris E, Sickmann A, Rassidakis GZ, Aivaliotis M. Interruption of p53-MDM2 Interaction by Nutlin-3a in Human Lymphoma Cell Models Initiates a Cell-Dependent Global Effect on Transcriptome and Proteome Level. Cancers (Basel) 2023; 15:3903. [PMID: 37568720 PMCID: PMC10417430 DOI: 10.3390/cancers15153903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 08/13/2023] Open
Abstract
In most lymphomas, p53 signaling pathway is inactivated by various mechanisms independent to p53 gene mutations or deletions. In many cases, p53 function is largely regulated by alterations in the protein abundance levels by the action of E3 ubiquitin-protein ligase MDM2, targeting p53 to proteasome-mediated degradation. In the present study, an integrating transcriptomics and proteomics analysis was employed to investigate the effect of p53 activation by a small-molecule MDM2-antagonist, nutlin-3a, on three lymphoma cell models following p53 activation. Our analysis revealed a system-wide nutlin-3a-associated effect in all examined lymphoma types, identifying in total of 4037 differentially affected proteins involved in a plethora of pathways, with significant heterogeneity among lymphomas. Our findings include known p53-targets and novel p53 activation effects, involving transcription, translation, or degradation of protein components of pathways, such as a decrease in key members of PI3K/mTOR pathway, heat-shock response, and glycolysis, and an increase in key members of oxidative phoshosphorylation, autophagy and mitochondrial translation. Combined inhibition of HSP90 or PI3K/mTOR pathway with nutlin-3a-mediated p53-activation enhanced the apoptotic effects suggesting a promising strategy against human lymphomas. Integrated omic profiling after p53 activation offered novel insights on the regulatory role specific proteins and pathways may have in lymphomagenesis.
Collapse
Affiliation(s)
- Konstantina Psatha
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology, 70013 Heraklion, Greece; (K.P.); (E.D.)
- Department of Pathology, Medical School, University of Crete, 70013 Heraklion, Greece;
- First Department of Pathology, National and Kapodistrian University of Athens, 15772 Athens, Greece;
- Functional Proteomics and Systems Biology (FunPATh), Center for Interdisciplinary Research and Innovation (CIRI-AUTH), 54124 Thessaloniki, Greece
| | - Laxmikanth Kollipara
- Leibniz-Institut für Analytische Wissenschaften–ISAS–e.V., 44139 Dortmund, Germany; (L.K.); (A.S.)
| | - Elias Drakos
- Department of Pathology, Medical School, University of Crete, 70013 Heraklion, Greece;
| | - Elena Deligianni
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology, 70013 Heraklion, Greece; (K.P.); (E.D.)
| | - Konstantinos Brintakis
- Institute of Electronic Structure and Laser, Foundation for Research and Technology—Hellas, 71110 Heraklion, Greece;
| | - Eustratios Patsouris
- First Department of Pathology, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften–ISAS–e.V., 44139 Dortmund, Germany; (L.K.); (A.S.)
- Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen AB24 3FX, UK
- Medizinische Fakultät, Medizinische Proteom-Center (MPC), Ruhr-Universität Bochum, 44801 Bochum, Germany
| | - George Z. Rassidakis
- Department of Oncology-Pathology, Karolinska Institute, 17164 Stockholm, Sweden;
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Solna, 17176 Stockholm, Sweden
| | - Michalis Aivaliotis
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology, 70013 Heraklion, Greece; (K.P.); (E.D.)
- Functional Proteomics and Systems Biology (FunPATh), Center for Interdisciplinary Research and Innovation (CIRI-AUTH), 54124 Thessaloniki, Greece
- Basic and Translational Research Unit, Special Unit for Biomedical Research and Education, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
- Laboratory of Biological Chemistry, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
3
|
Zhu L, Zhang L, Tang Y, Zhang F, Wan C, Xu L, Guo P. MicroRNA-363-3p inhibits tumor cell proliferation and invasion in oral squamous cell carcinoma cell lines by targeting SSFA2. Exp Ther Med 2021; 21:549. [PMID: 33850521 DOI: 10.3892/etm.2021.9981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Accepted: 06/10/2020] [Indexed: 01/28/2023] Open
Abstract
The aim of the present study was to evaluate the expression levels of microRNA (miR)-363-3p and its underlying physiological function in oral squamous cell carcinoma (OSCC). miR-363-3p expression levels were measured in OSCC cell lines using reverse transcription-quantitative PCR. The role of miR-363-3p in OSCC cells was examined using gain-of-function assays in vitro. Cell proliferation was assessed using Cell Counting Kit-8, 5-ethynyl-2'-deoxyuridine assays and flow cytometry. Cell migration and invasion were evaluated in wound-healing and Transwell Matrigel assays. In addition, bioinformatics analysis predicted binding sites of miR-363-3p on sperm-specific antigen 2 (SSFA2). Luciferase reporter and RNA pull-down assays were conducted to test whether miR-363-3p interacted with SSFA2. miR-363-3p expression was downregulated in OSCC cell lines compared with that in the normal epithelial cell line (NHOK). Additionally, miR-363-3p overexpression suppressed OSCC cell proliferation, migration and invasion in vitro. SSFA2 was verified as a direct target of miR-363-3p, and SSFA2 overexpression partially counteracted the inhibitory effects of miR-363-3p on cell proliferation, migration and invasion in OSCC cell lines. Thus, miR-363-3p may serve as a tumor suppressor via targeting SSFA2 and may represent a potential therapeutic target for OSCC.
Collapse
Affiliation(s)
- Liangming Zhu
- Department of Stomatology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| | - Lei Zhang
- Jiangcheng Dental Clinic, Wuhu, Anhui 241000, P.R. China
| | - Ying Tang
- Department of Endocrinology, Wuhu Hospital of Traditional Chinese Medicine, Wuhu, Anhui 241000, P.R. China
| | - Fang Zhang
- Department of Stomatology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| | - Chao Wan
- Department of Stomatology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| | - Liang Xu
- Department of Stomatology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| | - Ping Guo
- Department of Stomatology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241000, P.R. China
| |
Collapse
|
4
|
Large-Scale Proteomic Analysis of Follicular Lymphoma Reveals Extensive Remodeling of Cell Adhesion Pathway and Identifies Hub Proteins Related to the Lymphomagenesis. Cancers (Basel) 2021; 13:cancers13040630. [PMID: 33562532 PMCID: PMC7915278 DOI: 10.3390/cancers13040630] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/27/2021] [Accepted: 02/01/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Follicular lymphoma represents the major subtype of indolent B-cell non-Hodgkin lymphomas, ranging from about 20 to 30% of all B-NHLs cases in western countries. Yet, the global proteome profile of follicular lymphoma remains largely undocumented; thus, we aimed to employ for the first time a comprehensive proteomic analysis to outline its molecular landscape. A total of 15 lymphoma fine-needle aspiration biopsy samples and 14 controls were evaluated by label-free quantitative proteomics. Among the 7673 proteins identified in our dataset, 1186 proteins were differentially expressed between lymphoma and control samples. Importantly, dysregulated proteins were enriched in biological processes such as B-cell receptor signaling pathway, cellular adhesion molecules pathway, or membrane trafficking. Additionally, we identified several novel hub proteins related to lymphomagenesis. To summarize, we have determined the molecular characteristics of follicular lymphoma and discovered proteins which may hold potential for biomarkers or therapeutic targets. Abstract Follicular lymphoma (FL) represents the major subtype of indolent B-cell non-Hodgkin lymphomas (B-NHLs) and results from the malignant transformation of mature B-cells in lymphoid organs. Although gene expression and genomic studies have identified multiple disease driving gene aberrations, only a few proteomic studies focused on the protein level. The present work aimed to examine the proteomic profiles of follicular lymphoma vs. normal B-cells obtained by fine-needle aspiration biopsy (FNAB) to gain deep insight into the most perturbed pathway of FL. The cells of interest were purified by magnetic-activated cell sorting (MACS). High-throughput proteomic profiling was performed using liquid chromatography-tandem mass spectrometry (LC-MS/MS) and allowed to identify of 6724 proteins in at least 75% of each group of samples. The ‘Total Protein Approach’ (TPA) was applied to the absolute quantification of proteins in this study. We identified 1186 differentially abundant proteins (DAPs) between FL and control samples, causing an extensive remodeling of several molecular pathways, including the B-cell receptor signaling pathway, cellular adhesion molecules, and PPAR pathway. Additionally, the construction of protein–protein interactions networks (PPINs) and identification of hub proteins allowed us to indicate the key player proteins for FL pathology. Finally, ICAM1, CD9, and CD79B protein expression was validated in an independent cohort by flow cytometry (FCM), and the results were consistent with the mass spectrometry (MS) data.
Collapse
|
5
|
Nisar S, Hashem S, Macha MA, Yadav SK, Muralitharan S, Therachiyil L, Sageena G, Al-Naemi H, Haris M, Bhat AA. Exploring Dysregulated Signaling Pathways in Cancer. Curr Pharm Des 2020; 26:429-445. [PMID: 31939726 DOI: 10.2174/1381612826666200115095937] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 11/27/2019] [Indexed: 02/08/2023]
Abstract
Cancer cell biology takes advantage of identifying diverse cellular signaling pathways that are disrupted in cancer. Signaling pathways are an important means of communication from the exterior of cell to intracellular mediators, as well as intracellular interactions that govern diverse cellular processes. Oncogenic mutations or abnormal expression of signaling components disrupt the regulatory networks that govern cell function, thus enabling tumor cells to undergo dysregulated mitogenesis, to resist apoptosis, and to promote invasion to neighboring tissues. Unraveling of dysregulated signaling pathways may advance the understanding of tumor pathophysiology and lead to the improvement of targeted tumor therapy. In this review article, different signaling pathways and how their dysregulation contributes to the development of tumors have been discussed.
Collapse
Affiliation(s)
- Sabah Nisar
- Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| | - Sheema Hashem
- Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| | - Muzafar A Macha
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, United States.,Department of Biotechnology, Central University of Kashmir, Ganderbal, Jammu and Kashmir, India
| | - Santosh K Yadav
- Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| | | | - Lubna Therachiyil
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | | | - Hamda Al-Naemi
- Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Mohammad Haris
- Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar.,Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Ajaz A Bhat
- Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| |
Collapse
|
6
|
Ding QY, Zhang Y, Ma L, Chen YG, Wu JH, Zhang HF, Wang X. Inhibiting MAPK14 showed anti-prolactinoma effect. BMC Endocr Disord 2020; 20:138. [PMID: 32894113 PMCID: PMC7487756 DOI: 10.1186/s12902-020-00619-z] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 08/31/2020] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The specific underlying pathogenesis of prolactinoma has not been clarified yet, to the best of our knowledge. p38 mitogen-activated protein kinase (MAPK) signaling including p38α MAPK (MAPK14), p38β (MAPK11), p38γ (MAPK12) and p38δ (MAPK13) is associated with the development and progression of several types of cancer. METHODS Immunofluorescence analysis was performed on the prolactin (PRL) and MAPK14 expressions of pituitary gland in C57BL/6 mice and human prolactinoma specimen. In the present study, the role of MAPK14 in prolactinoma was determined using estradiol-induced mice and dopamine D2 receptor knockout (DRD2-/-) mice models in C57BL/6 wild-type (WT), MAPK14-/- and DRD2-/-MAPK14+/- mice. GH3 cells were transfected with different sets of MAPK14 small interfering RNA, which to study MAPK14 and PRL expression in GH3 cells. RESULTS Immunofluorescence analysis showed that PRL and MAPK14 expression were colocalized and increased in the pituitary gland of mice and human prolactinoma specimen compared with the control specimen. It was shown that PRL and MAPK14 expression was colocalized and increased significantly in the pituitary gland of estradiol-injected prolactinoma mice compared with the control mice. Knockout of MAPK14 significantly inhibited tumor overgrowth, and PRL expression was decreased in estradiol-induced mice. Furthermore, MAPK14 knockout of DRD2-/-MAPK14+/- mice significantly reduced the overgrowth of pituitary gland and PRL production and secretion compared with DRD2-/- mice. MAPK14 knockout using siRNA inhibited PRL production in GH3 cells. CONCLUSION These results suggest that MAPK14 serves a promoting role in the formation of prolactinoma, and highlights the potential of MAPK14 as a potential therapeutic target in the treatment of prolactinoma.
Collapse
Affiliation(s)
- Qiao-Yan Ding
- Central lab, Tongren Hospital Affiliated to Wuhan University, The Third Hospital of Wuhan, 241 Pengliuyang Road, Wuchang District, Wuhan, 430060, Hubei, China
- Department of Pharmacy, Tongren Hospital Affiliated to Wuhan University, The Third Hospital of Wuhan, Wuhan, 430060, Hubei, China
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, Hubei, China
| | - Yu Zhang
- College of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, Hubei, China
| | - Li Ma
- Central lab, Tongren Hospital Affiliated to Wuhan University, The Third Hospital of Wuhan, 241 Pengliuyang Road, Wuchang District, Wuhan, 430060, Hubei, China
- Department of Pharmacy, Tongren Hospital Affiliated to Wuhan University, The Third Hospital of Wuhan, Wuhan, 430060, Hubei, China
| | - Yong-Gang Chen
- Department of Pharmacy, Tongren Hospital Affiliated to Wuhan University, The Third Hospital of Wuhan, Wuhan, 430060, Hubei, China
| | - Jin-Hu Wu
- Central lab, Tongren Hospital Affiliated to Wuhan University, The Third Hospital of Wuhan, 241 Pengliuyang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Hong-Feng Zhang
- Department of Pathology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430014, P. R. China
| | - Xiong Wang
- Central lab, Tongren Hospital Affiliated to Wuhan University, The Third Hospital of Wuhan, 241 Pengliuyang Road, Wuchang District, Wuhan, 430060, Hubei, China.
- Department of Pharmacy, Tongren Hospital Affiliated to Wuhan University, The Third Hospital of Wuhan, Wuhan, 430060, Hubei, China.
| |
Collapse
|
7
|
Antiprolactinoma Effect of Hordenine by Inhibiting MAPK Signaling Pathway Activation in Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:3107290. [PMID: 32382283 PMCID: PMC7195642 DOI: 10.1155/2020/3107290] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 02/20/2020] [Accepted: 03/09/2020] [Indexed: 11/17/2022]
Abstract
Prolactinomas are harmful to human health, and the clinical first-line treatment drug is bromocriptine. However, 20% prolactinomas patients did not respond to bromocriptine. Hordenine is an alkaloid separated from Fructus Hordei Germinatus, which showed significant antihyperprolactinemia activity in rats. The aim of this study was to explore the effect and mechanism of hordenine on prolactinomas in rats. The study used estradiol to induce prolactinomas, which caused the activation of the pituitary mitogen-activated protein kinase (MAPK) pathway in rats significantly. The treatment of hordenine restored estradiol, induced the overgrowth of pituitary gland, and reduced the prolactin (PRL) accumulation in the serum and pituitary gland of rats by blocking the MAPK (p38, ERK1/2, and JNK) activation and production of inflammatory cytokines, tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6). The antiprolactinoma effect of hordenine was mediated by inhibiting the MAPK signaling pathway activation in rats.
Collapse
|
8
|
Han H, Zhan Z, Xu J, Song Z. TMEFF2 inhibits pancreatic cancer cells proliferation, migration, and invasion by suppressing phosphorylation of the MAPK signaling pathway. Onco Targets Ther 2019; 12:11371-11382. [PMID: 31920328 PMCID: PMC6939404 DOI: 10.2147/ott.s210619] [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: 03/30/2019] [Accepted: 05/30/2019] [Indexed: 12/27/2022] Open
Abstract
PURPOSE This paper studied the effect of TMEFF2 expression on pancreatic cancer and its mechanism. METHODS A total of 72 pancreatic cancer patients were enrolled. AsPC1 and Panc1 cells were transfected. SB203580 was used to treat AsPC1 cells. CCK8 assay, colony formation analysis, Transwell experiment and Tunel test were performed. In vivo studies in nude mice were conducted. Immunohistochemistry, qRT-PCR and Western blot were used to detect genes expression. RESULTS TMEFF2 was downregulated in pancreatic cancer tissues and cells (P<0.001). Low TMEFF2 expression was associated with larger tumor size and advanced stage and poor differentiation (P<0.01). Compared with the NC group, AsPC1 and Panc1 cells of the TMEFF2 group exhibited much lower OD450 values, colony number, tumor volume and weight, migration and invasion cell numbers, obviously higher E-cadherin protein expression, lower Snail, Vimentin, MMP-2 and MMP-9 proteins expression, lower phosphorylation level of MAPK signaling pathway, and more apoptotic cells. AsPC1 cells of the SB203580 group showed much lower OD450 value when compared with the siTMEFF2 group. Significantly decreased colony number, migration and invasion number, higher E-cadherin protein expression and lower Snail, Vimentin, MMP-2 and MMP-9 proteins expression were found in AsPC1 cells of the siTMEFF2+ SB203580 group when compared with the siTMEFF2+ DMSO group. CONCLUSION TMEFF2 inhibits pancreatic cancer cells proliferation, migration, and invasion by suppressing the phosphorylation of the MAPK signaling pathway.
Collapse
Affiliation(s)
- Hongchao Han
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Shanghai No. 10 People’s Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
- Department of General Surgery, Yancheng Third People’s Hospital, Yancheng, People’s Republic of China
| | - Zhilin Zhan
- Department of Hepatobiliary Surgery, Chizhou People’s Hospital, Chizhou, People’s Republic of China
| | - Jie Xu
- Department of General Surgery, Yancheng Third People’s Hospital, Yancheng, People’s Republic of China
| | - Zhenshun Song
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Shanghai No. 10 People’s Hospital, Nanjing Medical University, Nanjing, People’s Republic of China
| |
Collapse
|
9
|
Zhu A, Li X, Wu H, Miao Z, Yuan F, Zhang F, Wang B, Zhou Y. Molecular mechanism of SSFA2 deletion inhibiting cell proliferation and promoting cell apoptosis in glioma. Pathol Res Pract 2018; 215:600-606. [PMID: 30712887 DOI: 10.1016/j.prp.2018.12.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 12/09/2018] [Accepted: 12/30/2018] [Indexed: 10/27/2022]
Abstract
Gliomas are the most common primary brain malignant tumors in humans. Glioblastoma multiforme(GBM) is the most malignant intracranial tumor with a relatively poor prognosis. There promote us to find effective anti-cancer therapies to reduce cancer mortality. By using bioinformatic analysis, we found SSFA2 as a gene with elevated expression in the glioma tissues. We detected the expression of SSFA2 in glioma tissues and in the glioma cell lines, as well as in normal brain tissues. SSFA2 expression was higher in glioma tissues, especially in glioblastoma multiforme than normal brain tissues. Subsequently, we found that down-regulate SSFA2 in glioma cell lines can regulate the cell cycle to reduce the proliferation ability and induce the early apoptosis rate in shSSFA2 cells relative to control cells. Moreover, we found that down-regulate SSFA2 in glioma cell line U87(shSSFA2-U87) inhibited the growth effectiveness compared to the control cell line U87. These result reveals us that SSFA2 may act as oncogene to promote the progression of glioma. For further research specific mechanisms of SSFA2 in gliomas, we used the gene chip to detect the downstream gene in U87. We found that 30 genes also may be as target gene of SSFA2, and we testify the protein expression by western-blot. The result reveal that IL1A, IL1B and CDK6 as target gene of SSFA2 to regulate the progression of glioma. These finding suggest that SSFA2 could be a new therapeutic target for gliomas.
Collapse
Affiliation(s)
- Aihua Zhu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 899 Pinghai Street, Suzhou, 215006, Jiangsu, China; Department of Neurosurgery of Wuxi Third People's Hospital Research, The Third Affiliated Hospital of Nantong University, 585 Xingyuan North Road, Wuxi, 214041, Jiangsu, China
| | - Xuetao Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 899 Pinghai Street, Suzhou, 215006, Jiangsu, China
| | - Haibin Wu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 899 Pinghai Street, Suzhou, 215006, Jiangsu, China
| | - Zongning Miao
- The Research Institute of Wuxi Third People's Hospital, The Third Affiliated Hospital of Nantong University, 585 Xingyuan North Road, Wuxi, 214041, Jiangsu, China
| | - Fenglai Yuan
- The Research Institute of Wuxi Third People's Hospital, The Third Affiliated Hospital of Nantong University, 585 Xingyuan North Road, Wuxi, 214041, Jiangsu, China
| | - Feng Zhang
- The Research Institute of Wuxi Third People's Hospital, The Third Affiliated Hospital of Nantong University, 585 Xingyuan North Road, Wuxi, 214041, Jiangsu, China
| | - Bei Wang
- The Research Institute of Wuxi Third People's Hospital, The Third Affiliated Hospital of Nantong University, 585 Xingyuan North Road, Wuxi, 214041, Jiangsu, China
| | - Youxin Zhou
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, 899 Pinghai Street, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
10
|
Psatha K, Kollipara L, Voutyraki C, Divanach P, Sickmann A, Rassidakis GZ, Drakos E, Aivaliotis M. Deciphering lymphoma pathogenesis via state-of-the-art mass spectrometry-based quantitative proteomics. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1047:2-14. [PMID: 27979587 DOI: 10.1016/j.jchromb.2016.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/18/2016] [Accepted: 11/04/2016] [Indexed: 12/13/2022]
Abstract
Mass spectrometry-based quantitative proteomics specifically applied to comprehend the pathogenesis of lymphoma has incremental value in deciphering the heterogeneity in complex deregulated molecular mechanisms/pathways of the lymphoma entities, implementing the current diagnostic and therapeutic strategies. Essential global, targeted and functional differential proteomics analyses although still evolving, have been successfully implemented to shed light on lymphoma pathogenesis to discover and explore the role of potential lymphoma biomarkers and drug targets. This review aims to outline and appraise the present status of MS-based quantitative proteomic approaches in lymphoma research, introducing the current state-of-the-art MS-based proteomic technologies, the opportunities they offer in biological discovery in human lymphomas and the related limitation issues arising from sample preparation to data evaluation. It is a synopsis containing information obtained from recent research articles, reviews and public proteomics repositories (PRIDE). We hope that this review article will aid, assimilate and assess all the information aiming to accelerate the development and validation of diagnostic, prognostic or therapeutic targets for an improved and empowered clinical proteomics application in lymphomas in the nearby future.
Collapse
Affiliation(s)
- Konstantina Psatha
- Institute of Molecular Biology and Biotechnology, FORTH, Heraklion, Greece; School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Department of Pathology, School of Medicine, University of Crete, Heraklion, Greece
| | - Laxmikanth Kollipara
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany
| | | | - Peter Divanach
- Institute of Molecular Biology and Biotechnology, FORTH, Heraklion, Greece
| | - Albert Sickmann
- Leibniz-Institut für Analytische Wissenschaften - ISAS - e.V., Dortmund, Germany; Department of Chemistry, College of Physical Sciences, University of Aberdeen, Aberdeen, Scotland, United Kingdom; Medizinische Fakultät, Medizinische Proteom-Center (MPC), Ruhr-Universität Bochum, Bochum, Germany
| | - George Z Rassidakis
- School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Department of Pathology and Cytology, Karolinska University Hospital and Karolinska Institute, Radiumhemmet, Stockholm, SE-17176, Sweden
| | - Elias Drakos
- Department of Pathology, School of Medicine, University of Crete, Heraklion, Greece
| | | |
Collapse
|
11
|
3D-QSAR studies on disubstituted dibenzosuberone derivatives as p38α MAP kinase inhibitors using CoMFA and COMSIA. Med Chem Res 2016. [DOI: 10.1007/s00044-016-1642-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
12
|
Chen CY, Chung IH, Tsai MM, Tseng YH, Chi HC, Tsai CY, Lin YH, Wang YC, Chen CP, Wu TI, Yeh CT, Tai DI, Lin KH. Thyroid hormone enhanced human hepatoma cell motility involves brain-specific serine protease 4 activation via ERK signaling. Mol Cancer 2014; 13:162. [PMID: 24980078 PMCID: PMC4087245 DOI: 10.1186/1476-4598-13-162] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 06/19/2014] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The thyroid hormone, 3, 3', 5-triiodo-L-thyronine (T3), has been shown to modulate cellular processes via interactions with thyroid hormone receptors (TRs), but the secretory proteins that are regulated to exert these effects remain to be characterized. Brain-specific serine protease 4 (BSSP4), a member of the human serine protease family, participates in extracellular matrix remodeling. However, the physiological role and underlying mechanism of T3-mediated regulation of BSSP4 in hepatocellular carcinogenesis are yet to be established. METHODS The thyroid hormone response element was identified by reporter and chromatin immunoprecipitation assays. The cell motility was analyzed via transwell and SCID mice. The BSSP4 expression in clinical specimens was examined by Western blot and quantitative reverse transcription polymerase chain reaction. RESULTS Upregulation of BSSP4 at mRNA and protein levels after T3 stimulation is a time- and dose-dependent manner in hepatoma cell lines. Additionally, the regulatory region of the BSSP4 promoter stimulated by T3 was identified at positions -609/-594. BSSP4 overexpression enhanced tumor cell migration and invasion, both in vitro and in vivo. Subsequently, BSSP4-induced migration occurs through the ERK 1/2-C/EBPβ-VEGF cascade, similar to that observed in HepG2-TRα1 and J7-TRα1 cells. BSSP4 was overexpressed in clinical hepatocellular carcinoma (HCC) patients, compared with normal subjects, and positively associated with TRα1 and VEGF to a significant extent. Importantly, a mild association between BSSP4 expression and distant metastasis was observed. CONCLUSIONS Our findings collectively support a potential role of T3 in cancer cell progression through regulation of the BSSP4 protease via the ERK 1/2-C/EBPβ-VEGF cascade. BSSP4 may thus be effectively utilized as a novel marker and anti-cancer therapeutic target in HCC.
Collapse
Affiliation(s)
- Cheng-Yi Chen
- Department of Biochemistry, School of Medicine, Chang-Gung University, 259 Wen-hwa 1 Road, Taoyuan, Taiwan
- Department of Medical Research, Mackay Memorial Hospital, 251 Taipei, Taiwan
| | - I-Hsiao Chung
- Department of Biochemistry, School of Medicine, Chang-Gung University, 259 Wen-hwa 1 Road, Taoyuan, Taiwan
| | - Ming-Ming Tsai
- Department of Nursing, Chang-Gung University of Science and Technology, 333 Taoyuan, Taiwan
| | - Yi-Hsin Tseng
- Department of Biochemistry, School of Medicine, Chang-Gung University, 259 Wen-hwa 1 Road, Taoyuan, Taiwan
| | - Hsiang-Cheng Chi
- Department of Biochemistry, School of Medicine, Chang-Gung University, 259 Wen-hwa 1 Road, Taoyuan, Taiwan
| | - Chung-Ying Tsai
- Department of Biochemistry, School of Medicine, Chang-Gung University, 259 Wen-hwa 1 Road, Taoyuan, Taiwan
| | - Yang-Hsiang Lin
- Department of Biochemistry, School of Medicine, Chang-Gung University, 259 Wen-hwa 1 Road, Taoyuan, Taiwan
| | - You-Ching Wang
- Department of Biochemistry, School of Medicine, Chang-Gung University, 259 Wen-hwa 1 Road, Taoyuan, Taiwan
| | - Chie-Pein Chen
- Department of Medical Research, Mackay Memorial Hospital, 251 Taipei, Taiwan
- Division of High Risk Pregnancy, Mackay Memorial Hospital, 104 Taipei, Taiwan
| | - Tzu-I Wu
- Department of Biochemistry, School of Medicine, Chang-Gung University, 259 Wen-hwa 1 Road, Taoyuan, Taiwan
| | - Chau-Ting Yeh
- Medical Research Central, Chang Gung Memorial Hospital, 333 Taoyuan, Taiwan
| | - Dar-In Tai
- Medical Research Central, Chang Gung Memorial Hospital, 333 Taoyuan, Taiwan
| | - Kwang-Huei Lin
- Department of Biochemistry, School of Medicine, Chang-Gung University, 259 Wen-hwa 1 Road, Taoyuan, Taiwan
| |
Collapse
|
13
|
Abstract
Mitogen-activated protein kinases (MAPKs) mediate a wide variety of cellular behaviors in response to extracellular stimuli. One of the main subgroups, the p38 MAP kinases, has been implicated in a wide range of complex biologic processes, such as cell proliferation, cell differentiation, cell death, cell migration, and invasion. Dysregulation of p38 MAPK levels in patients are associated with advanced stages and short survival in cancer patients (e.g., prostate, breast, bladder, liver, and lung cancer). p38 MAPK plays a dual role as a regulator of cell death, and it can either mediate cell survival or cell death depending not only on the type of stimulus but also in a cell type specific manner. In addition to modulating cell survival, an essential role of p38 MAPK in modulation of cell migration and invasion offers a distinct opportunity to target this pathway with respect to tumor metastasis. The specific function of p38 MAPK appears to depend not only on the cell type but also on the stimuli and/or the isoform that is activated. p38 MAPK signaling pathway is activated in response to diverse stimuli and mediates its function by components downstream of p38. Extrapolation of the knowledge gained from laboratory findings is essential to address the clinical significance of p38 MAPK signaling pathways. The goal of this review is to provide an overview on recent progress made in defining the functions of p38 MAPK pathways with respect to solid tumor biology and generate testable hypothesis with respect to the role of p38 MAPK as an attractive target for intervention of solid tumors.
Collapse
Affiliation(s)
- Hari K Koul
- Department of Biochemistry & Molecular Biology, LSU Health Sciences Center, Shreveport, LA, USA ; Feist-Weiller Cancer Center, Shreveport, LA, USA ; Veterans Administration Medical Center, Shreveport, LA, USA
| | - Mantu Pal
- Department of Biochemistry & Molecular Biology, LSU Health Sciences Center, Shreveport, LA, USA ; Veterans Administration Medical Center, Shreveport, LA, USA
| | - Sweaty Koul
- Feist-Weiller Cancer Center, Shreveport, LA, USA ; Department of Urology, LSU Health Sciences Center, Shreveport, LA, USA
| |
Collapse
|
14
|
Abstract
Basic science research in hematology has been determining the functions of gene products using classical approaches that typically involve studying one or a few genes at a time. Proteomics, defined as the study of protein properties on a large scale, provides tools to globally analyze malignant hematologic cells. A major challenge in cancer therapy is the identification of drugs that kill tumor cells while preserving normal cells. Differential display via proteomics enables analysis of direct as well as side-effects of drugs at a molecular level. Proteomics also allows a better understanding of cell signaling pathways involved during apoptosis in hematologic cells. Storing the information in a 2D electrophoresis database enhances the efficiency of proteome research on malignant cells. Finally, the work needed to be carried out on proteomic analysis prior to routine clinical adoption is discussed, and the necessity for multi-institutional collaborations is emphasized.
Collapse
Affiliation(s)
- Michel Caron
- Protein Biochemistry and Proteomics Laboratory, Université Paris 13, UFR SMBH, 74, Rue Marcel Cachin, 93017 Bobigny Cedex, France.
| | | |
Collapse
|
15
|
He Y, Zhang M, Ju Y, Yu Z, Lv D, Sun H, Yuan W, He F, Zhang J, Li H, Li J, Wang-Sattler R, Li Y, Zhang G, Xie L. dbDEPC 2.0: updated database of differentially expressed proteins in human cancers. Nucleic Acids Res 2012; 40:D964-71. [PMID: 22096234 PMCID: PMC3245147 DOI: 10.1093/nar/gkr936] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 10/10/2011] [Accepted: 10/11/2011] [Indexed: 01/07/2023] Open
Abstract
A large amount of differentially expressed proteins (DEPs) have been identified in various cancer proteomics experiments, curation and annotation of these proteins are important in deciphering their roles in oncogenesis and tumor progression, and may further help to discover potential protein biomarkers for clinical applications. In 2009, we published the first database of DEPs in human cancers (dbDEPCs). In this updated version of 2011, dbDEPC 2.0 has more than doubly expanded to over 4000 protein entries, curated from 331 experiments across 20 types of human cancers. This resource allows researchers to search whether their interested proteins have been reported changing in certain cancers, to compare their own proteomic discovery with previous studies, to picture selected protein expression heatmap across multiple cancers and to relate protein expression changes with aberrance in other genetic level. New important developments include addition of experiment design information, advanced filter tools for customer-specified analysis and a network analysis tool. We expect dbDEPC 2.0 to be a much more powerful tool than it was in its first release and can serve as reference to both proteomics and cancer researchers. dbDEPC 2.0 is available at http://lifecenter.sgst.cn/dbdepc/index.do.
Collapse
Affiliation(s)
- Ying He
- Key Laboratory of Systems Biology, Chinese Academy of Sciences, Shanghai 200031, Shanghai Center for Bioinformation Technology, Shanghai 200235, Department of Bioinformatics and Biostatistics, Shanghai Jiaotong University, Shanghai 200240, P. R. China, Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg 85764, Germany and Biomedical Engineering for School of Life Sciences and Technology, Tongji University, Shanghai 200092, P. R. of China
| | - Menghuan Zhang
- Key Laboratory of Systems Biology, Chinese Academy of Sciences, Shanghai 200031, Shanghai Center for Bioinformation Technology, Shanghai 200235, Department of Bioinformatics and Biostatistics, Shanghai Jiaotong University, Shanghai 200240, P. R. China, Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg 85764, Germany and Biomedical Engineering for School of Life Sciences and Technology, Tongji University, Shanghai 200092, P. R. of China
| | - Yuanhu Ju
- Key Laboratory of Systems Biology, Chinese Academy of Sciences, Shanghai 200031, Shanghai Center for Bioinformation Technology, Shanghai 200235, Department of Bioinformatics and Biostatistics, Shanghai Jiaotong University, Shanghai 200240, P. R. China, Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg 85764, Germany and Biomedical Engineering for School of Life Sciences and Technology, Tongji University, Shanghai 200092, P. R. of China
| | - Zhonghao Yu
- Key Laboratory of Systems Biology, Chinese Academy of Sciences, Shanghai 200031, Shanghai Center for Bioinformation Technology, Shanghai 200235, Department of Bioinformatics and Biostatistics, Shanghai Jiaotong University, Shanghai 200240, P. R. China, Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg 85764, Germany and Biomedical Engineering for School of Life Sciences and Technology, Tongji University, Shanghai 200092, P. R. of China
| | - Daqing Lv
- Key Laboratory of Systems Biology, Chinese Academy of Sciences, Shanghai 200031, Shanghai Center for Bioinformation Technology, Shanghai 200235, Department of Bioinformatics and Biostatistics, Shanghai Jiaotong University, Shanghai 200240, P. R. China, Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg 85764, Germany and Biomedical Engineering for School of Life Sciences and Technology, Tongji University, Shanghai 200092, P. R. of China
| | - Han Sun
- Key Laboratory of Systems Biology, Chinese Academy of Sciences, Shanghai 200031, Shanghai Center for Bioinformation Technology, Shanghai 200235, Department of Bioinformatics and Biostatistics, Shanghai Jiaotong University, Shanghai 200240, P. R. China, Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg 85764, Germany and Biomedical Engineering for School of Life Sciences and Technology, Tongji University, Shanghai 200092, P. R. of China
| | - Weilan Yuan
- Key Laboratory of Systems Biology, Chinese Academy of Sciences, Shanghai 200031, Shanghai Center for Bioinformation Technology, Shanghai 200235, Department of Bioinformatics and Biostatistics, Shanghai Jiaotong University, Shanghai 200240, P. R. China, Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg 85764, Germany and Biomedical Engineering for School of Life Sciences and Technology, Tongji University, Shanghai 200092, P. R. of China
| | - Fei He
- Key Laboratory of Systems Biology, Chinese Academy of Sciences, Shanghai 200031, Shanghai Center for Bioinformation Technology, Shanghai 200235, Department of Bioinformatics and Biostatistics, Shanghai Jiaotong University, Shanghai 200240, P. R. China, Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg 85764, Germany and Biomedical Engineering for School of Life Sciences and Technology, Tongji University, Shanghai 200092, P. R. of China
| | - Jianshe Zhang
- Key Laboratory of Systems Biology, Chinese Academy of Sciences, Shanghai 200031, Shanghai Center for Bioinformation Technology, Shanghai 200235, Department of Bioinformatics and Biostatistics, Shanghai Jiaotong University, Shanghai 200240, P. R. China, Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg 85764, Germany and Biomedical Engineering for School of Life Sciences and Technology, Tongji University, Shanghai 200092, P. R. of China
| | - Hong Li
- Key Laboratory of Systems Biology, Chinese Academy of Sciences, Shanghai 200031, Shanghai Center for Bioinformation Technology, Shanghai 200235, Department of Bioinformatics and Biostatistics, Shanghai Jiaotong University, Shanghai 200240, P. R. China, Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg 85764, Germany and Biomedical Engineering for School of Life Sciences and Technology, Tongji University, Shanghai 200092, P. R. of China
| | - Jing Li
- Key Laboratory of Systems Biology, Chinese Academy of Sciences, Shanghai 200031, Shanghai Center for Bioinformation Technology, Shanghai 200235, Department of Bioinformatics and Biostatistics, Shanghai Jiaotong University, Shanghai 200240, P. R. China, Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg 85764, Germany and Biomedical Engineering for School of Life Sciences and Technology, Tongji University, Shanghai 200092, P. R. of China
| | - Rui Wang-Sattler
- Key Laboratory of Systems Biology, Chinese Academy of Sciences, Shanghai 200031, Shanghai Center for Bioinformation Technology, Shanghai 200235, Department of Bioinformatics and Biostatistics, Shanghai Jiaotong University, Shanghai 200240, P. R. China, Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg 85764, Germany and Biomedical Engineering for School of Life Sciences and Technology, Tongji University, Shanghai 200092, P. R. of China
| | - Yixue Li
- Key Laboratory of Systems Biology, Chinese Academy of Sciences, Shanghai 200031, Shanghai Center for Bioinformation Technology, Shanghai 200235, Department of Bioinformatics and Biostatistics, Shanghai Jiaotong University, Shanghai 200240, P. R. China, Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg 85764, Germany and Biomedical Engineering for School of Life Sciences and Technology, Tongji University, Shanghai 200092, P. R. of China
| | - Guoqing Zhang
- Key Laboratory of Systems Biology, Chinese Academy of Sciences, Shanghai 200031, Shanghai Center for Bioinformation Technology, Shanghai 200235, Department of Bioinformatics and Biostatistics, Shanghai Jiaotong University, Shanghai 200240, P. R. China, Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg 85764, Germany and Biomedical Engineering for School of Life Sciences and Technology, Tongji University, Shanghai 200092, P. R. of China
| | - Lu Xie
- Key Laboratory of Systems Biology, Chinese Academy of Sciences, Shanghai 200031, Shanghai Center for Bioinformation Technology, Shanghai 200235, Department of Bioinformatics and Biostatistics, Shanghai Jiaotong University, Shanghai 200240, P. R. China, Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, Neuherberg 85764, Germany and Biomedical Engineering for School of Life Sciences and Technology, Tongji University, Shanghai 200092, P. R. of China
| |
Collapse
|
16
|
Chapuy B, Schuelper N, Panse M, Dohm A, Hand E, Schroers R, Truemper L, Wulf GG. Multikinase inhibitor sorafenib exerts cytocidal efficacy against Non-Hodgkin lymphomas associated with inhibition of MAPK14 and AKT phosphorylation. Br J Haematol 2011; 152:401-12. [PMID: 21689083 DOI: 10.1111/j.1365-2141.2010.08526.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Intracellular signal transduction by kinase-mediated phosphorylation is essential for the survival and growth of lymphoma cells. This study analysed the multikinase inhibitor sorafenib for its cytotoxic activity against lymphoma cells. We found that sorafenib reduced cell viability at low micromolar concentrations in a time-dependent manner in cell lines and primary cell suspensions representing major types of aggressive B- and T-cell lymphomas. In cells surviving short term exposure, proliferative arrest occurred leading to complete loss of in vitro clonogenicity. Previously described sorafenib targets within the RAF kinase family were found to be expressed and phosphorylated in all cell lines, and sorafenib perturbed the activation of classical RAF/MEK/ERK pathway targets. However, using a global phoshoprotein array, the most consistent downstream effect of sorafenib in NHL cells was the inhibition of mitogen-activated protein kinase 14 (MAPK14) and panAKT phosphorylation. In conclusion, sorafenib has significant in vitro efficacy against aggressive B- and T-cell lymphoma cells, associated with inhibition of MAPK14 and panAKT.
Collapse
Affiliation(s)
- Bjoern Chapuy
- Department of Haematology and Oncology, Georg-August-University Goettingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Determination of the critical region of KRAS-induced actin-interacting protein for the interaction with inositol 1,4,5-trisphosphate receptor. Biochem Biophys Res Commun 2011; 408:282-6. [PMID: 21501587 DOI: 10.1016/j.bbrc.2011.04.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 04/03/2011] [Indexed: 11/23/2022]
Abstract
KRAS-induced actin-interacting protein (KRAP) was originally characterized as a filamentous-actin-interacting protein. We have recently found that KRAP is an associated molecule with inositol 1,4,5-trisphosphate receptor (IP(3)R) and is critical for the proper subcellular localization and function of IP(3)R. However, the molecular mechanisms underlying the regulation of IP(3)R by KRAP remain elusive. In this report, to determine the critical region of KRAP protein for the regulation of IP(3)R, we generate several mutants of KRAP and examine the association with IP(3)R using coimmunoprecipitation and confocal imaging assays. Coimmunoprecipitations using the deletion mutants reveal that amino-acid residues 1-218 but not 1-199 of KRAP interact with IP(3)R, indicating that the 19-length amino-acid residues (200-218) are essential for the association with IP(3)R. This critical region is highly conserved between human and mouse KRAP. Within the critical region, substitutions of two phenylalanine residues (Phe202/Phe203) in mouse KRAP to alanines result in failure of the association with IP(3)R, suggesting that the two consecutive phenylalanine residues are indispensable for the association. Moreover, the KRAP-knockdown stable HeLa cells exhibit the inappropriate subcellular localization of IP(3)R, in which exogenous expression of full-length of KRAP properly restores the subcellular localization of IP(3)R, but not the 1-218 or 1-236 mutant, indicating that the residual carboxyl-terminal region is also required for the proper subcellular localization of KRAP-IP(3)R complex. All these results provide insight into the understandings for the molecular mechanisms underlying the regulation of IP(3)R, and would reveal a potent strategy for the drug development targeting on IP(3)R.
Collapse
|
18
|
Abstract
Fire and Mello initiated the current explosion of interest in RNA interference (RNAi) biology with their seminal work in Caenorhabditis elegans. These observations were closely followed by the demonstration of RNAi in Drosophila melanogaster. However, the full potential of these new discoveries only became clear when Tuschl and colleagues showed that 21-22 bp RNA duplexes with 3" overhangs, termed small interfering (si)RNAs, could reliably execute RNAi in a range of mammalian cells. Soon afterwards, it became clear that many different human cell types had endogenous machinery, the RNA-induced silencing complex (RISC), which could be harnessed to silence any gene in the genome. Beyond the availability of a novel way to dissect biology, an important target validation tool was now available. More importantly, two key properties of the RNAi pathway - sequence-mediated specificity and potency - suggested that RNAi might be the most important pharmacological advance since the advent of protein therapeutics. The implications were profound. One could now envisage selecting disease-associated targets at will and expect to suppress proteins that had remained intractable to inhibition by conventional methods, such as small molecules. This review attempts to summarize the current understanding on siRNA lead discovery, the delivery of RNAi therapeutics, typical in vivo pharmacological profiles, preclinical safety evaluation and an overview of the 14 programs that have already entered clinical practice.
Collapse
|
19
|
Proteomic analysis of lymphoid and haematopoietic neoplasms: There's more than biomarker discovery. J Proteomics 2010; 73:508-20. [DOI: 10.1016/j.jprot.2009.08.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Revised: 08/26/2009] [Accepted: 08/27/2009] [Indexed: 12/29/2022]
|
20
|
Butler GS, Overall CM. Proteomic identification of multitasking proteins in unexpected locations complicates drug targeting. Nat Rev Drug Discov 2009; 8:935-48. [PMID: 19949400 DOI: 10.1038/nrd2945] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Proteomics has revealed that many proteins are present in unexpected cellular locations. Moreover, it is increasingly recognized that proteins can translocate between intracellular and extracellular compartments in non-conventional ways. This increases gene pleiotrophy as the diverse functions of the protein that the gene encodes are dependent on the cellular location. Given that trafficking drug targets may exist in various forms--often with completely different functions--in multiple cellular compartments, careful interpretation of proteomics data is needed for an accurate understanding of gene function. This Perspective is intended to inspire the investigation of unusual protein localizations, rather than assuming that they are due to mislocalization or artefacts. Given a fair chance, proteomics could reveal novel and unforeseen biology with important ramifications for target validation in drug discovery.
Collapse
Affiliation(s)
- Georgina S Butler
- Centre for Blood Research, Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, British Columbia, VT6 1Z3, Canada.
| | | |
Collapse
|
21
|
Ding H, Gabali AM, Jenson SD, Lim MS, Elenitoba-Johnson KSJ. P38 mitogen activated protein kinase expression and regulation by interleukin-4 in human B cell non-Hodgkin lymphomas. J Hematop 2009; 2:195-204. [PMID: 20309428 PMCID: PMC2798936 DOI: 10.1007/s12308-009-0049-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2009] [Accepted: 09/30/2009] [Indexed: 01/09/2023] Open
Abstract
The prevalence and regulation of p38 mitogen activated protein kinase (MAPK) expression in human lymphomas have not been extensively studied. In order to elucidate the role of p38 MAPK in lymphomagenesis, we examined the expression of native and phosphorylated p38 (p-p38) MAPK in cell lines derived from human hematopoietic neoplasms including B cell lymphoma-derived cell lines using Western blot analysis. The p-p38 MAPK protein was also analyzed in 30 B cell non-Hodgkin lymphoma (NHL) tissue biopsies by immunohistochemistry. Our results show that the expression of p38 MAPK was up-regulated in most of the cell lines as compared with peripheral blood lymphocytes, while the expression of p-p38 MAPK was more variable. A subset of B cell NHL biopsies showed increased expression of p-p38 MAPK relative to reactive germinal center cells. Interleukin-4 (IL-4) induced a dose-dependent increase in the expression of p-p38 MAPK (1.6- to 2.8-fold) in cell lines derived from activated B cell-like diffuse large B cell lymphoma (DLBCL) but not those from germinal center-like DLBCL. No change was seen in native p38 MAPK. The in vitro kinase activity of p38 MAPK, however, was induced (1.6- to 3.2-fold) in all five cell lines by IL-4. Quantitative fluorescent RT-PCR demonstrated that all four isoforms of p38 MAPK gene were expressed in the lymphoma cell lines, with p38γ and p38β isoforms being predominant. IL-4 stimulation increased the expression of β, γ, and δ isoforms but not α isoform in two cell lines. In conclusion, there is constitutive expression and activation of p38 MAPK in a large number of B-lymphoma-derived cell lines and primary lymphoma tissues, supportive of its role in lymphomagenesis. The differential IL-4 regulation of p38 MAPK expression in cell lines derived from DLBCL may relate to the cellular origin of these neoplasms.
Collapse
|
22
|
The proteomic signature of NPM/ALK reveals deregulation of multiple cellular pathways. Blood 2009; 114:1585-95. [PMID: 19531656 DOI: 10.1182/blood-2009-02-204735] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Constitutive expression of the chimeric NPM/ALK fusion protein encoded by the t(2;5)(p32;q35) is a key oncogenic event in the pathogenesis of most anaplastic large cell lymphomas (ALCLs). The proteomic network alterations produced by this aberration remain largely uncharacterized. Using a mass spectrometry (MS)-driven approach to identify changes in protein expression caused by the NPM/ALK fusion, we identified diverse NPM/ALK-induced changes affecting cell proliferation, ribosome synthesis, survival, apoptosis evasion, angiogenesis, and cytoarchitectural organization. MS-based findings were confirmed using Western blotting and/or immunostaining of NPM/ALK-transfected cells and ALK-deregulated lymphomas. A subset of the proteins distinguished NPM/ALK-positive ALCLs from NPM/ALK-negative ALCLs and Hodgkin lymphoma. The multiple NPM/ALK-deregulated pathways identified by MS analysis also predicted novel biologic effects of NPM/ALK expression. In this regard, we showed loss of cell adhesion as a consequence of NPM/ALK expression in a kinase-dependent manner, and sensitivity of NPM/ALK-positive ALCLs to inhibition of the RAS, p42/44ERK, and FRAP/mTOR signaling pathways. These findings reveal that the NPM/ALK alteration affects diverse cellular pathways, and provide novel insights into NPM/ALK-positive ALCL pathobiology. Our studies carry important implications for the use of MS-driven approaches for the elucidation of neoplastic pathobiology, the identification of novel diagnostic biomarkers, and pathogenetically relevant therapeutic targets.
Collapse
|
23
|
Epithelial cell survival by activating transcription factor 3 (ATF3) in response to chemical ribosome-inactivating stress. Biochem Pharmacol 2009; 77:1105-15. [DOI: 10.1016/j.bcp.2008.11.028] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2008] [Revised: 11/25/2008] [Accepted: 11/26/2008] [Indexed: 12/14/2022]
|
24
|
Liu T, Martin AM, Sinai AP, Lynn BC. Three-layer sandwich gel electrophoresis: a method of salt removal and protein concentration in proteome analysis. J Proteome Res 2008; 7:4256-65. [PMID: 18795766 DOI: 10.1021/pr800182b] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Sample preparation plays a critical role in successful proteomic applications. Features of electrospray mass spectrometry impose limits on the types of buffers, detergents and other reagents that can be used in sample preparation. Unfortunately, many of these mass spectrometry incompatible reagents significantly enhance protein recoveries from complex matrices. This problem prompted our search for a better cleanup protocol. Our data suggest that the Three-layer Sandwich Gel Electrophoresis (TSGE) protocol can solve this problem and provide near quantitative recovery of extremely low concentration proteins from harsh solutions, a feature not available from other cleanup protocols. The hallmark of the TSGE protocol is the combination of the properties of agarose gels (that serve as the matrix to immobilize the proteins of interest) with low- and high-percentage polyacrylamide gels (that serve as the concentration and sealing layers, respectively). By electrophoretically driving the proteins of interest from the agarose matrix into the concentration layer, the TSGE protocol simultaneously concentrates the sample in the concentration layer and provides an environment amenable to downstream buffer exchange and proteolytic digestion. In combination with 2D-LC-MS/MS, the TSGE protocol was evaluated in the analysis of a whole cell extract from the protozoan parasite Toxoplasma gondii. Comparison of our experimental proteomic results with in silico predictions from gene data indicated that TSGE did not bias the protein identification.
Collapse
Affiliation(s)
- Ting Liu
- Department of Chemistry, University of Kentucky, Lexington, Kentucky 40506-0055, USA
| | | | | | | |
Collapse
|
25
|
p38 MAPK as a signal transduction component of heavy metals stress in Euglena gracilis. Arch Microbiol 2008; 191:47-54. [DOI: 10.1007/s00203-008-0427-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2008] [Revised: 08/05/2008] [Accepted: 08/18/2008] [Indexed: 10/21/2022]
|
26
|
Chiacchiera F, Simone C. Signal-dependent regulation of gene expression as a target for cancer treatment: inhibiting p38alpha in colorectal tumors. Cancer Lett 2008; 265:16-26. [PMID: 18395970 DOI: 10.1016/j.canlet.2008.02.061] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2007] [Revised: 02/15/2008] [Accepted: 02/19/2008] [Indexed: 12/27/2022]
Abstract
In the last year, several evidences indicated that pharmacological manipulation of relevant signaling pathways could selectively affect gene expression to influence cell fate. These findings render of extreme importance the elucidation of how external stimuli are transduced to mediate chromatin modifications, resulting in a permissive or repressive environment for gene expression. These signaling cascades activate or repress the function of chromatin binding proteins that represent attractive pharmacological targets for human diseases. Actually, the closer the target is to chromatin, the more the transcriptional effect will be selective. Recent studies suggest that pharmacological manipulation of signaling pathways to modulate cell fate is indeed possible and that chromatin-associated kinases could represent an optimal target. The p38 MAPK is the prototype of this class of enzymes and its central role in the transcription process is evolutionary conserved. In this review we will focus on the possibility to inhibit p38alpha in colorectal cancer to arrest tumor progression and induce autophagic cell death.
Collapse
Affiliation(s)
- Fulvio Chiacchiera
- Laboratory of Signal-dependent Transcription, Department of Translational Pharmacology (DTP), Consorzio Mario Negri Sud, Via Nazionale 8/A, 66030 Santa Maria Imbaro (Chieti), Italy
| | | |
Collapse
|
27
|
Vivekanandan P, Singh OV. High-dimensional biology to comprehend hepatocellular carcinoma. Expert Rev Proteomics 2008; 5:45-60. [PMID: 18282123 DOI: 10.1586/14789450.5.1.45] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common cancer worldwide and is the third leading cause of death from cancer. The diverse etiology, high morbidity/mortality, lack of diagnostic markers for early diagnosis and the highly variable clinical course of HCC have hindered advances in diagnosis and treatment. Microsatellite instability, chromosomal aberrations, mutations in key cell cycle genes and epigenetic changes have been reported in HCC. Availability of modern technologies advance 'high-dimensional biology' (HDB), a term that refers to the simultaneous study of the genetic variants (genome), transcription (mRNA; transcriptome), peptides and proteins (proteomics), and metabolites (metabolomics) for the intermediate products of metabolism of an organ, tissue or organism. The growing interest in omics-based research has enabled the simultaneous examination of thousands of genes, transcripts and proteins of interest, with high-throughput techniques and advanced analytical tools for data analysis. The use of each approach towards functional omics has lead to the classification of HCC into molecular subgroups. Here we review the use of HDB as a tool for the identification of markers for screening, diagnosis, molecular classification and the discovery of new therapeutic drug targets of HCC. With the extensive use of HDB, it may be possible in the near future, to have custom-made therapeutic regimens for HCC based on the molecular subtype, ultimately leading to an improved survival of HCC patients.
Collapse
|
28
|
Molecular effectors and modulators of hypericin-mediated cell death in bladder cancer cells. Oncogene 2007; 27:1916-29. [PMID: 17952126 DOI: 10.1038/sj.onc.1210825] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Photodynamic therapy (PDT) is an anticancer approach utilizing a light-absorbing molecule and visible light irradiation to generate, in the presence of O(2), cytotoxic reactive oxygen species, which cause tumor ablation. Given that the photosensitizer hypericin is under consideration for PDT treatment of bladder cancer we used oligonucleotide microarrays in the T24 bladder cancer cell line to identify differentially expressed genes with therapeutic potential. This study reveals that the expression of several genes involved in various metabolic processes, stress-induced cell death, autophagy, proliferation, inflammation and carcinogenesis is strongly affected by PDT and pinpoints the coordinated induction of a cluster of genes involved in the unfolded protein response pathway after endoplasmic reticulum stress and in antioxidant response. Analysis of PDT-treated cells after p38(MAPK) inhibition or silencing unraveled that the induction of an important subset of differentially expressed genes regulating growth and invasion, as well as adaptive mechanisms against oxidative stress, is governed by this stress-activated kinase. Moreover, p38(MAPK) inhibition blocked autonomous regrowth and migration of cancer cells escaping PDT-induced cell death. This analysis identifies new molecular effectors of the cancer cell response to PDT opening attractive avenues to improve the therapeutic efficacy of hypericin-based PDT of bladder cancer.
Collapse
|
29
|
Schumacher JA, Crockett DK, Elenitoba-Johnson KSJ, Lim MS. Proteome-wide changes induced by the Hsp90 inhibitor, geldanamycin in anaplastic large cell lymphoma cells. Proteomics 2007; 7:2603-16. [PMID: 17610208 DOI: 10.1002/pmic.200700108] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The molecular chaperone heat shock protein 90 (Hsp90) affects the function of many oncogenic signaling proteins including nucleophosmin-anaplastic lymphoma kinase (NPM-ALK) expressed in anaplastic large cell lymphoma (ALCL). While ALK-positive ALCL cells are sensitive to the Hsp90 inhibitor and the geldanamycin (GA) analog, 17-allylamino-17-demethoxygeldanamycin (17-AAG), the proteomic effects of these drugs on ALK-positive ALCL cells are unpublished. In this study, we investigated the cellular, biologic, and proteomic changes occurring in ALK-positive ALCL cells in response to GA treatment. GA induced G2/M cell cycle arrest and caspase-3-mediated apoptosis. Furthermore, quantitative proteomic changes analyzed by cleavable isotope-coded affinity tag-LC-MS/MS (cICAT-LC-MS/MS) identified 176 differentially expressed proteins. Out of these, 49 were upregulated 1.5-fold or greater and 70 were downregulated 1.5-fold or greater in GA-treated cells. Analysis of biological functions of differentially expressed proteins revealed diverse changes, including induction of proteins involved in the 26S proteasome as well as downregulation of proteins involved in signal transduction and protein and nucleic acid metabolism. Pathway analysis revealed changes in MAPK, WNT, NF-kappaB, TGFbeta, PPAR, and integrin signaling components. Our studies reveal some of the molecular and proteomic consequences of Hsp90 inhibition in ALK-positive ALCL cells and provide novel insights into the mechanisms of its diverse cellular effects.
Collapse
MESH Headings
- Antibiotics, Antineoplastic/pharmacology
- Apoptosis/drug effects
- Apoptosis/physiology
- Benzoquinones/pharmacology
- Caspase 3/metabolism
- Cell Cycle/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- DNA, Neoplasm/analysis
- Enzyme Inhibitors/pharmacology
- HSP90 Heat-Shock Proteins/antagonists & inhibitors
- HSP90 Heat-Shock Proteins/physiology
- Humans
- Lactams, Macrocyclic/pharmacology
- Lymphoma, Large B-Cell, Diffuse/enzymology
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/pathology
- Models, Biological
- Proteome/analysis
- Reproducibility of Results
Collapse
Affiliation(s)
- Jonathan A Schumacher
- Associated and Regional University Pathologists (ARUP), Institute for Clinical and Experimental Pathology, Salt Lake City, UT, USA
| | | | | | | |
Collapse
|
30
|
Abstract
Proteomic studies have generated numerous datasets of potential diagnostic, prognostic, and therapeutic significance in human cancer. Two key technologies underpinning these studies in cancer tissue are two-dimensional polyacrylamide gel electrophoresis (2D-PAGE) and mass spectrometry (MS). Although surface-enhanced laser desorption/ionization time-of-flight (SELDI-TOF)-MS is the mainstay for serum or plasma analysis, other methods including isotope-coded affinity tag technology, reverse-phase protein arrays, and antibody microarrays are emerging as alternative proteomic technologies. Because there is little overlap between studies conducted with these approaches, confirmation of these advanced technologies remains an elusive goal. This problem is further exacerbated by lack of uniform patient inclusion and exclusion criteria, low patient numbers, poor supporting clinical data, absence of standardized sample preparation, and limited analytical reproducibility (in particular of 2D-PAGE). Despite these problems, there is little doubt that the proteomic approach has the potential to identify novel diagnostic biomarkers in cancer. In therapeutic proteomics, the challenge is significant due to the complexity systems under investigation (i.e., cells generate over 10(5) different polypeptides). However, the most significant contribution of therapeutic proteomics research is expected to derive not from single experiments, but from the synthesis and comparison of large datasets obtained under different conditions (e.g., normal, inflammation, cancer) and in different tissues and organs. Thus, standardized processes for storing and retrieving data obtained with different technologies by different research groups will have to be developed. Shifting the emphasis of cancer proteomics from technology development and data generation to careful study design, data organization, formatting, and mining is crucial to answer clinical questions in cancer research.
Collapse
Affiliation(s)
- M A Reymond
- Department of Surgery, University of Magdeburg, Germany
| | | |
Collapse
|
31
|
Zer C, Sachs G, Shin JM. Identification of genomic targets downstream of p38 mitogen-activated protein kinase pathway mediating tumor necrosis factor-alpha signaling. Physiol Genomics 2007; 31:343-51. [PMID: 17652167 PMCID: PMC2880477 DOI: 10.1152/physiolgenomics.00080.2007] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Inhibition of p38 MAPK suppresses the expression of proinflammatory cytokines such as TNF-alpha and IL-1 beta in macrophages and fibroblast-like synoviocytes (FLS). However, there have been no genomewide studies on the gene targets of p38 MAPK signaling in synoviocytes. Microarray technology was applied to generate a comprehensive analysis of all genes regulated by the p38 MAPK signaling pathway in FLS. Gene expression levels were measured with Agilent oligonucleotide microarrays. Four independent sets of mRNA modulated by TNF-alpha and vehicle were used to measure the change of gene expression due to TNF-alpha, and three experiments were done to ascertain the effect of SB-203580, a p38 MAPK inhibitor, on TNF-alpha-induced genes. Microarray data were validated by RT-quantitative polymerase chain reaction. One hundred forty-one significantly expressed genes were more than twofold upregulated by TNF-alpha. Thirty percent of these genes were downregulated by the p38 inhibitor SB-203580, whereas 67% of these genes were not significantly changed. The SB-203580-inhibited genes include proinflammatory cytokines such as interleukins and chemokines, proteases including matrix metallopeptidases, metabolism-related genes such as cyclooxygenases and phosphodiesterase, genes involved in signal transduction, and genes encoding for transcription factors, receptors, and transporters. Approximately one-third of the TNF-alpha-induced genes in FLS are regulated by the p38 MAPK signal pathway, showing that p38 MAPK is a possible target for suppressing proinflammatory gene expressions in rheumatoid arthritis.
Collapse
Affiliation(s)
- Cindy Zer
- Department of Physiology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | | | | |
Collapse
|
32
|
Okoli AS, Fox EM, Raftery MJ, Mendz GL. Effects of Helicobacter hepaticus on the proteome of HEp-2 cells. Antonie van Leeuwenhoek 2007; 92:289-300. [PMID: 17357813 DOI: 10.1007/s10482-007-9155-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2006] [Revised: 10/20/2006] [Accepted: 12/13/2006] [Indexed: 10/23/2022]
Abstract
Helicobacter hepaticus infects the bowel and biliary tree of several animals, producing inflammation. Colonisation of mouse livers can induce hepatocellular carcinomas. The effects of H. hepaticus on the proliferation and global protein expression of human HEp-2 cells were studied by examining the changes in the protein profiles of cells exposed to the bacterium. HEp-2 cells were grown for four days under a microaerobic atmosphere or under the same conditions in co-cultures with H. hepaticus at various inoculum densities. Enlargement, distension and elongation of HEp-2 cells were observed in co-cultures with H. hepaticus. The number of live cells declined by only an order of magnitude at bacterial inocula of approximately 10(9)cfu/ml, but were reduced to less than 10(3)cells/ml at approximately 10(10)cfu/ml bacteria inocula. Protein expression by HEp-2 cells was investigated employing two-dimensional gel electrophoresis. In cells grown with or without bacteria, 17 differentially expressed proteins were identified by tandem mass spectrometry. These proteins participated in several biological functions including amino acid metabolism, cell growth and proliferation, stress response, protein translation and modification, etc. The onset of a catastrophic killing of HEp-2 cells at a bacterial density of approximately 10(9)cfu/ml suggested a multimodal action for H. hepaticus infection, and the modulation of the expression of proteins involved in different biological functions showed that the presence of H. hepaticus has broad effects on the physiology of HEp-2 cells.
Collapse
Affiliation(s)
- Arinze S Okoli
- School of Medical Sciences, The University of New South Wales, Sydney, NSW 2052, Australia
| | | | | | | |
Collapse
|
33
|
Kuramitsu Y, Nakamura K. Proteomic analysis of cancer tissues: shedding light on carcinogenesis and possible biomarkers. Proteomics 2007; 6:5650-61. [PMID: 16972299 DOI: 10.1002/pmic.200600218] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Lung, gastric, colorectal, pancreatic, and esophageal cancers, as well as hepatocellular carcinoma (HCC), were the six most common and highly fatal cancers for Japanese men in Japan in 2003, while for women uterine cervical cancer could also be added to this list. To identify diagnostic or therapeutic biomarkers for these cancers, investigators are nowadays performing proteomic analyses of cancer tissues and cells, and revealing a large number of molecules which are diagnostic, prognostic and informative of carcinogenesis. From reports of proteomic analyses of cancerous tissues and noncancerous tissues sampled from HCC, and pancreatic, esophageal, gastric, colorectal, lung and uterine cervical cancers, we classified the proteins into digestive enzymes, growth factors, cell adhesion molecules, calcium-binding proteins, proteases, protease inhibitors, transporter proteins, structural molecules, apoptosis inhibitor, molecular chaperone, as well as proteins related to cell growth, cell differentiation, cell transformation, tumor invasion, carcinogen metabolism, and others. The aim of this study was to understand carcinogenesis of major cancers from a proteomics perspective using samples from cancer patients, and to elucidate their tumor biomarkers.
Collapse
Affiliation(s)
- Yasuhiro Kuramitsu
- Department of Biochemistry and Functional Proteomics, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | | |
Collapse
|
34
|
Kuramitsu Y, Nakamura K. Current progress in proteomic study of hepatitis C virus-related human hepatocellular carcinoma. Expert Rev Proteomics 2007; 2:589-601. [PMID: 16097891 DOI: 10.1586/14789450.2.4.589] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Chronic infection with hepatitis C virus (HCV) is known to be a risk factor for not only cirrhosis and steatosis but also hepatocellular carcinoma (HCC). A number of diagnostic and prognostic molecular markers are being identified by transcriptomic and proteomic analysis of HCC today. However, the analyses are performed on HCC in general, and the studied tissues are HCV infected, HBV infected, infected with both or neither, or the infection status may be unknown. The authors performed proteomic analysis of cancerous and noncancerous tissues from HCC patients with HCV infection, and determined that, in the cancerous tissues, HSP70 family proteins such as GRP78, HSC70, GRP75 and HSP70.1, glutaine synthetase isoforms, HSP60, alpha-enolase, phosphoglycerate mutase 1, ATP synthetase beta chain and triosephosphate isomerase were increased whereas albumin, ferritin light chain, smoothelin, tropomyosin beta chain, arginase 1, aldolase B and kietohexokinase were decreased. The aim of this study is to understand the pathogenesis of HCV-HCC using proteomic analysis of samples from HCV-HCC patients on which transcriptomics has already been performed.
Collapse
Affiliation(s)
- Yasuhiro Kuramitsu
- Department of Biochemistry and Biomolecular Recognition, Yamaguchi University School of Medicine, Minami-Kogushi, Ube, Yamaguchi 755-8505, Japan.
| | | |
Collapse
|
35
|
Tenhunen O, Rysä J, Ilves M, Soini Y, Ruskoaho H, Leskinen H. Identification of cell cycle regulatory and inflammatory genes as predominant targets of p38 mitogen-activated protein kinase in the heart. Circ Res 2006; 99:485-93. [PMID: 16873723 DOI: 10.1161/01.res.0000238387.85144.92] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mitogen-activated protein kinases (MAPKs) regulate cardiomyocyte growth and apoptosis in response to extracellular stimulation, but the downstream effectors that mediate their pathophysiological effects remain poorly understood. We determined the targets and role of p38 MAPK in the heart in vivo by using local adenovirus-mediated gene transfer of constitutively active upstream kinase mitogen-activated protein kinase kinase 3b (MKK3bE) and wild-type p38alpha in rats. DNA microarray analysis of animals with cardiac-specific overexpression of p38 MAPK revealed that 264 genes were upregulated more than 2-fold including multiple genes controlling cell division, cell signaling, inflammation, adhesion, and transcription. A large number of previously unknown p38 target genes were found. Using gel mobility-shift assays we identified several cardiac transcription factors that were directly activated by p38 MAPK. Finally, we determined the functional significance of the altered cardiac gene-expression profile by histological analysis and echocardiographic measurements, which indicated that p38 MAPK overexpression-induced gene expression results in myocardial cell proliferation, inflammation, and fibrosis. In conclusion, we defined the novel target genes and transcription factors as well as the functional effects of p38 MAPK in the heart. Expression profiling of p38 MAPK overexpression identified cell cycle regulatory and inflammatory genes critical for pathological processes in the adult heart.
Collapse
Affiliation(s)
- Olli Tenhunen
- Department of Pharmacology and Toxicology, Biocenter Oulu, Finland
| | | | | | | | | | | |
Collapse
|
36
|
Ambrosino C, Iwata T, Scafoglio C, Mallardo M, Klein R, Nebreda A. TEF-1 and C/EBPbeta are major p38alpha MAPK-regulated transcription factors in proliferating cardiomyocytes. Biochem J 2006; 396:163-72. [PMID: 16492136 PMCID: PMC1449985 DOI: 10.1042/bj20051502] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
p38 MAPKs (mitogen-activated protein kinases) play important roles in the regulation of cellular responses to environmental stress. Recently, this signalling pathway has also been implicated in the regulation of processes unrelated to stress, for example, in T lymphocytes and cardiomyocytes. In order to identify molecular targets responsible for the housekeeping functions of p38 MAPKs, we have analysed the differences in the transcriptomes of normally proliferating wild-type and p38alpha knockout immortalized embryonic cardiomyocytes. Interestingly, many potential components of the myocardium extracellular matrix were found to be upregulated in the absence of p38alpha. Further analysis of the microarray data identified TEF-1 (transcriptional enhancer factor-1), a known regulator of heart-specific gene expression, and C/EBPbeta (CCAAT/enhancer-binding protein beta), as the two transcription factors the binding sites of which were most enriched in the promoters of p38alpha-regulated genes. We have focused on the study of the extracellular matrix component COL1A1 (alpha1 chain of type I collagen) and found evidence for the involvement of both TEF-1 and C/EBPbeta in the p38alpha-dependent inhibition of COL1A1 transcription. Our data therefore show that p38 MAPKs regulate TEF-1 and C/EBPbeta transcriptional activity in the absence of environmental stress and suggests a role for p38alpha in the expression of extracellular matrix components that maintain organ architecture.
Collapse
Affiliation(s)
- Concetta Ambrosino
- *European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- †Dipartimento di Patologia Generale, Seconda Università degli Studi di Napoli, 80138 Napoli, Italy
| | - Tomoko Iwata
- ‡Department of Molecular Neurobiology, Max-Planck Institute of Neurobiology, 82152 Martinsried, Germany
| | - Claudio Scafoglio
- †Dipartimento di Patologia Generale, Seconda Università degli Studi di Napoli, 80138 Napoli, Italy
| | - Massimo Mallardo
- §Dipartimento di Biochimica e Biotecnologie Mediche, Università di Napoli ‘Federico II’, Italy
| | - Rüdiger Klein
- ‡Department of Molecular Neurobiology, Max-Planck Institute of Neurobiology, 82152 Martinsried, Germany
| | - Angel R. Nebreda
- *European Molecular Biology Laboratory, 69117 Heidelberg, Germany
- ∥CNIO (Spanish National Cancer Center), Melchor Fernández Almagro 3, E-28029 Madrid, Spain
- To whom correspondence should be addressed (email )
| |
Collapse
|
37
|
Thadikkaran L, Siegenthaler MA, Crettaz D, Queloz PA, Schneider P, Tissot JD. Recent advances in blood-related proteomics. Proteomics 2005; 5:3019-34. [PMID: 16041673 DOI: 10.1002/pmic.200402053] [Citation(s) in RCA: 142] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Blood is divided in two compartments, namely, plasma and cells. The latter contain red blood cells, leukocytes, and platelets. From a descriptive medical discipline, hematology has evolved towards a pioneering discipline where molecular biology has permitted the development of prognostic and diagnostic indicators for disease. The recent advance in MS and protein separation now allows similar progress in the analysis of proteins. Proteomics offers great promise for the study of proteins in plasma/serum, indeed a number of proteomics databases for plasma/serum have been established. This is a very complex body fluid containing lipids, carbohydrates, amino acids, vitamins, nucleic acids, hormones, and proteins. About 1500 different proteins have recently been identified, and a number of potential new markers of diseases have been characterized. Here, examples of the enormous promise of plasma/serum proteomic analysis for diagnostic/prognostic markers and information on disease mechanism are given. Within the blood are also a large number of different blood cell types that potentially hold similar information. Proteomics of red blood cells, until now, has not improved our knowledge of these cells, in contrast to the major progresses achieved while studying platelets and leukocytes. In the future, proteomics will change several aspects of hematology.
Collapse
Affiliation(s)
- Lynne Thadikkaran
- Service régional vaudois de transfusion sanguine, Rue du Bugnon 27, CH-1005 Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
38
|
Affiliation(s)
- Ali Hachem
- University of Maryland Greenebaum Cancer Center, 9-011 BRB, 655 West Baltimore St, Baltimore MD 21201, USA
| | | |
Collapse
|
39
|
Abstract
Shotgun proteomics has emerged as a powerful approach for the analysis of complex protein mixtures, including biofluids, tissues, cells, organelles or protein complexes. Having evolved from the integration of chromatography and mass spectrometry, innovations in sample preparation, multidimensional chromatography, mass spectrometry and proteomic informatics continually facilitate, enable and challenge shotgun proteomics. As a result, shotgun proteomics continues to evolve and enable new areas of biological research, and is beginning to impact human disease diagnosis and therapeutic intervention.
Collapse
Affiliation(s)
- Selene K Swanson
- Stowers Institute for Medical Research, 1000 E. 50th St., Kansas City, MO 64110, USA
| | | |
Collapse
|
40
|
Abstract
Abstract
The evolution of indolent lymphomas to aggressive histologies, known as histologic transformation (HT), is a frequent occurrence for all subtypes of low grade B cell lymphoproliferative disorders. The risk of developing HT is approximately 3% per year for patients with indolent lymphoma. Clinically these present with a rapid change in the behavior of the disease, with evidence of a highly proliferative malignancy with a propensity to involve extranodal sites. The prognosis of patients following transformation is generally poor, with median survival of about 12 months. Recent studies suggest that the development of HT is very complex with the acquisition of multiple cytogenetic abnormalities in the low-grade lymphoma cells prior to HT. To date, there are no biologic or genetic parameters predictive of the development of HT. A myriad of genetic lesions have been identified in HT, and provide insight into its pathogenesis. These include genes regulating proliferation (C-MYC and C-MYC-regulated genes); control of the cell cycle (CDKN2a and CDKN2B); and programmed cell death (TP53, C-MYC, and BCL2). Gene expression profiling has been applied to the study of HT and has increased our understanding of the transformation process. There has been limited progress in the treatment of patients with HT. Conventional chemotherapy is generally of limited benefit, although a subset of patients are long-term survivors following high-dose therapy and autologous stem cell transplantation. The use of radioimmunotherapy and new agents targeting specific lesions or aberrant pathways may impact on the management of these aggressive diseases.
Collapse
MESH Headings
- Cell Cycle/genetics
- Cell Death/genetics
- Cell Division/genetics
- Cell Transformation, Neoplastic
- Chromosome Aberrations
- Chromosomes, Human, Pair 14
- Chromosomes, Human, Pair 18
- Gene Expression Profiling
- Humans
- Lymphoma, B-Cell/genetics
- Lymphoma, B-Cell/mortality
- Lymphoma, B-Cell/pathology
- Lymphoma, Non-Hodgkin/genetics
- Lymphoma, Non-Hodgkin/mortality
- Lymphoma, Non-Hodgkin/pathology
- Mutation
- Prognosis
- Survival Analysis
- Time Factors
- Translocation, Genetic
Collapse
Affiliation(s)
- Arnold S Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.
| |
Collapse
|
41
|
Abstract
Proteomics is a multifaceted approach to study various aspects of protein expression, post-translational modification, interactions, organization and function at a global level. While DNA constitutes the 'information archive of the genome', it is the proteins that actually serve as the functional effectors of cellular processes. Thus, analysis of protein derangements on a proteome-wide scale will reveal insights into deregulated pathways and networks involved in the pathogenesis of disease. Although the field of proteomics has advanced tremendously in recent years, there are significant technical challenges that pose limitations to the routine application of mass spectrometry to clinical research. Despite these challenges, proteomic studies have yielded unparalleled information and understanding of the cellular biology of diseased states. The application of mass spectrometry to the study of diseases will ultimately lead to identification of biomarkers that are critical for the detection, diagnosis, prognosis and treatment of specific disease entities.
Collapse
Affiliation(s)
- Megan S Lim
- Department of Pathology, University of Utah Health Sciences Center, Salt Lake City, UT 84132, USA.
| | | |
Collapse
|