1
|
de la Fuente‐Vivas D, Cappitelli V, García‐Gómez R, Valero‐Díaz S, Amato C, Rodriguéz J, Duro‐Sánchez S, von Kriegsheim A, Grusch M, Lozano J, Arribas J, Casar B, Crespo P. ERK1/2 mitogen-activated protein kinase dimerization is essential for the regulation of cell motility. Mol Oncol 2025; 19:452-473. [PMID: 39263917 PMCID: PMC11792999 DOI: 10.1002/1878-0261.13732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 07/12/2024] [Accepted: 08/29/2024] [Indexed: 09/13/2024] Open
Abstract
ERK1/2 mitogen-activated protein kinases (ERK) are key regulators of basic cellular processes, including proliferation, survival, and migration. Upon phosphorylation, ERK becomes activated and a portion of it dimerizes. The importance of ERK activation in specific cellular events is generally well documented, but the role played by dimerization is largely unknown. Here, we demonstrate that impeding ERK dimerization precludes cellular movement by interfering with the molecular machinery that executes the rearrangements of the actin cytoskeleton. We also show that a constitutively dimeric ERK mutant can drive cell motility per se, demonstrating that ERK dimerization is both necessary and sufficient for inducing cellular migration. Importantly, we unveil that the scaffold protein kinase suppressor of Ras 1 (KSR1) is a critical element for endowing external agonists, acting through tyrosine kinase receptors, with the capacity to induce ERK dimerization and, subsequently, to unleash cellular motion. In agreement, clinical data disclose that high KSR1 expression levels correlate with greater metastatic potential and adverse evolution of mammary tumors. Overall, our results portray both ERK dimerization and KSR1 as essential factors for the regulation of cell motility and mammary tumor dissemination.
Collapse
Affiliation(s)
- Dalia de la Fuente‐Vivas
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC)Consejo Superior de Investigaciones Científicas (CSIC) – Universidad de CantabriaSantanderSpain
- Present address:
Universidad de BurgosBurgosSpain
| | - Vincenzo Cappitelli
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC)Consejo Superior de Investigaciones Científicas (CSIC) – Universidad de CantabriaSantanderSpain
| | - Rocío García‐Gómez
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC)Consejo Superior de Investigaciones Científicas (CSIC) – Universidad de CantabriaSantanderSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)Instituto de Salud Carlos IIIMadridSpain
| | - Sara Valero‐Díaz
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC)Consejo Superior de Investigaciones Científicas (CSIC) – Universidad de CantabriaSantanderSpain
| | - Camilla Amato
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC)Consejo Superior de Investigaciones Científicas (CSIC) – Universidad de CantabriaSantanderSpain
| | - Javier Rodriguéz
- Cancer Research UK Scotland Centre, Institute of Genetics and CancerUniversity of EdinburghUK
| | - Santiago Duro‐Sánchez
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)Instituto de Salud Carlos IIIMadridSpain
- Cancer Research ProgramHospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Department of Biochemistry and Molecular BiologyUniversitat Autónoma de BarcelonaSpain
- Preclinical and Translational Research ProgramVall d'Hebron Institute of Oncology (VHIO)BarcelonaSpain
| | | | - Michael Grusch
- Center for Cancer ResearchMedical University of ViennaAustria
| | - José Lozano
- Universidad de Málaga and Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina – IBIMA, Plataforma BionandSpain
| | - Joaquín Arribas
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)Instituto de Salud Carlos IIIMadridSpain
- Cancer Research ProgramHospital del Mar Medical Research Institute (IMIM)BarcelonaSpain
- Department of Biochemistry and Molecular BiologyUniversitat Autónoma de BarcelonaSpain
- Preclinical and Translational Research ProgramVall d'Hebron Institute of Oncology (VHIO)BarcelonaSpain
| | - Berta Casar
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC)Consejo Superior de Investigaciones Científicas (CSIC) – Universidad de CantabriaSantanderSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)Instituto de Salud Carlos IIIMadridSpain
| | - Piero Crespo
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC)Consejo Superior de Investigaciones Científicas (CSIC) – Universidad de CantabriaSantanderSpain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC)Instituto de Salud Carlos IIIMadridSpain
| |
Collapse
|
2
|
Li T, Gui X, Li B, Hu X, Wang Y. LSP1 promotes the progression of acute myelogenous leukemia by regulating KSR/ERK signaling pathway and cell migration. Hematology 2024; 29:2330285. [PMID: 38511641 DOI: 10.1080/16078454.2024.2330285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 03/10/2024] [Indexed: 03/22/2024] Open
Abstract
We aimed to investigate the role and mechanism of LSP1 in the progression of acute myelogenous leukemia. In this study, we established shLSP1 cell line to analyze the function of LSP1 in AML. We observed high expression of LSP1 in AML patients, whereas it showed no expression in normal adults. Furthermore, we found that LSP1 expression was associated with disease prognosis. Our results indicate that LSP1 plays a crucial role in mediating proliferation and survival of leukemia cells through the KSR/ERK signaling pathway. Additionally, LSP1 promotes cell chemotaxis and homing by enhancing cell adhesion and migration. We also discovered that LSP1 confers chemotactic ability to leukemia cells in vivo. Finally, our study identified 12 genes related to LSP1 in AML, which indicated poor survival outcome in AML patients and were enriched in Ras and cell adhesion signaling pathways. Our results revealed that the overexpression of LSP1 is related to the activation of the KSR/ERK signaling pathway, as well as cell adhesion and migration in AML patients. Reducing LSP1 expression impair AML progression, suggesting that LSP1 may serve as a potential drug therapy target for more effective treatment of AML.
Collapse
Affiliation(s)
- Tan Li
- Department of Hematology, Hefei City First People's Hospital, Hefei, People's Republic of China
| | - Xiaochen Gui
- College & Hospital of Stomatology, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, Hefei, People's Republic of China
| | - Bin Li
- Department of Hematology, Hefei City First People's Hospital, Hefei, People's Republic of China
| | - Xueying Hu
- Department of Hematology, Hefei City First People's Hospital, Hefei, People's Republic of China
| | - Yuanyin Wang
- College & Hospital of Stomatology, Anhui Medical University, Key Lab of Oral Diseases Research of Anhui Province, Hefei, People's Republic of China
| |
Collapse
|
3
|
Hilchey SP, Palshikar MG, Mendelson ES, Shen S, Rasam S, Emo JA, Qu J, Thakar J, Zand MS. Cyclosporine A Modulates LSP1 Protein Levels in Human B Cells to Attenuate B Cell Migration at Low O 2 Levels. LIFE (BASEL, SWITZERLAND) 2022; 12:life12081284. [PMID: 36013463 PMCID: PMC9410508 DOI: 10.3390/life12081284] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 12/20/2022]
Abstract
Coordinated migration of B cells within and between secondary lymphoid tissues is required for robust antibody responses to infection or vaccination. Secondary lymphoid tissues normally expose B cells to a low O2 (hypoxic) environment. Recently, we have shown that human B cell migration is modulated by an O2-dependent molecular switch, centrally controlled by the hypoxia-induced (transcription) factor-1α (HIF1A), which can be disrupted by the immunosuppressive calcineurin inhibitor, cyclosporine A (CyA). However, the mechanisms by which low O2 environments attenuate B cell migration remain poorly defined. Proteomics analysis has linked CXCR4 chemokine receptor signaling to cytoskeletal rearrangement. We now hypothesize that the pathways linking the O2 sensing molecular switch to chemokine receptor signaling and cytoskeletal rearrangement would likely contain phosphorylation events, which are typically missed in traditional transcriptomic and/or proteomic analyses. Hence, we have performed a comprehensive phosphoproteomics analysis of human B cells treated with CyA after engagement of the chemokine receptor CXCR4 with CXCL12. Statistical analysis of the separate and synergistic effects of CyA and CXCL12 revealed 116 proteins whose abundance is driven by a synergistic interaction between CyA and CXCL12. Further, we used our previously described algorithm BONITA to reveal a critical role for Lymphocyte Specific Protein 1 (LSP1) in cytoskeletal rearrangement. LSP1 is known to modulate neutrophil migration. Validating these modeling results, we show experimentally that LSP1 levels in B cells increase with low O2 exposure, and CyA treatment results in decreased LSP1 protein levels. This correlates with the increased chemotactic activity observed after CyA treatment. Lastly, we directly link LSP1 levels to chemotactic capacity, as shRNA knock-down of LSP1 results in significantly increased B cell chemotaxis at low O2 levels. These results directly link CyA to LSP1-dependent cytoskeletal regulation, demonstrating a previously unrecognized mechanism by which CyA modulates human B cell migration. Data are available via ProteomeXchange with identifier PXD036167.
Collapse
Affiliation(s)
- Shannon P. Hilchey
- Department of Medicine, Division of Nephrology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Mukta G. Palshikar
- Biophysics, Structural, and Computational Biology Program, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Eric S. Mendelson
- Department of Medicine, Division of Nephrology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Shichen Shen
- Department of Pharmaceutical Sciences, State University of New York (SUNY) at Buffalo, Buffalo, NY 14203, USA
- New York State Center of Excellence in Bioinformatics & Life Sciences, State University of New York (SUNY) at Buffalo, Buffalo, NY 14203, USA
| | - Sailee Rasam
- Department of Pharmaceutical Sciences, State University of New York (SUNY) at Buffalo, Buffalo, NY 14203, USA
- New York State Center of Excellence in Bioinformatics & Life Sciences, State University of New York (SUNY) at Buffalo, Buffalo, NY 14203, USA
| | - Jason A. Emo
- Department of Medicine, Division of Nephrology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jun Qu
- Department of Pharmaceutical Sciences, State University of New York (SUNY) at Buffalo, Buffalo, NY 14203, USA
- New York State Center of Excellence in Bioinformatics & Life Sciences, State University of New York (SUNY) at Buffalo, Buffalo, NY 14203, USA
| | - Juilee Thakar
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Martin S. Zand
- Department of Medicine, Division of Nephrology, University of Rochester Medical Center, Rochester, NY 14642, USA
- Clinical and Translational Science Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
- Correspondence:
| |
Collapse
|
4
|
Thanh Thi Ngoc Nguyen, Nguyen THN, Phan HN, Nguyen HT. Seven-Single Nucleotide Polymorphism Polygenic Risk Score for Breast Cancer Risk Prediction in a Vietnamese Population. CYTOL GENET+ 2022. [DOI: 10.3103/s0095452722040065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
5
|
Koral K, Bhushan B, Orr A, Stoops J, Bowen WC, Copeland MA, Locker J, Mars WM, Michalopoulos GK. Lymphocyte-Specific Protein-1 Suppresses Xenobiotic-Induced Constitutive Androstane Receptor and Subsequent Yes-Associated Protein-Activated Hepatocyte Proliferation. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:887-903. [PMID: 35390317 PMCID: PMC9194659 DOI: 10.1016/j.ajpath.2022.03.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 02/23/2022] [Accepted: 03/10/2022] [Indexed: 06/03/2023]
Abstract
Activation of constitutive androstane receptor (CAR) transcription factor by xenobiotics promotes hepatocellular proliferation, promotes hypertrophy without liver injury, and induces drug metabolism genes. Previous work demonstrated that lymphocyte-specific protein-1 (LSP1), an F-actin binding protein and gene involved in human hepatocellular carcinoma, suppresses hepatocellular proliferation after partial hepatectomy. The current study investigated the role of LSP1 in liver enlargement induced by chemical mitogens, a regenerative process independent of tissue loss. 1,4-Bis [2-(3,5-dichloropyridyloxy)] benzene (TCPOBOP), a direct CAR ligand and strong chemical mitogen, was administered to global Lsp1 knockout and hepatocyte-specific Lsp1 transgenic (TG) mice and measured cell proliferation, hypertrophy, and expression of CAR-dependent drug metabolism genes. TG livers displayed a significant decrease in Ki-67 labeling and liver/body weight ratios compared with wild type on day 2. Surprisingly, this was reversed by day 5, due to hepatocyte hypertrophy. There was no difference in CAR-regulated drug metabolism genes between wild type and TG. TG livers displayed increased Yes-associated protein (YAP) phosphorylation, decreased nuclear YAP, and direct interaction between LSP1 and YAP, suggesting LSP1 suppresses TCPOBOP-driven hepatocellular proliferation, but not hepatocyte volume, through YAP. Conversely, loss of LSP1 led to increased hepatocellular proliferation on days 2, 5, and 7. LSP1 selectively suppresses CAR-induced hepatocellular proliferation, but not drug metabolism, through the interaction of LSP1 with YAP, supporting the role of LSP1 as a selective growth suppressor.
Collapse
Affiliation(s)
- Kelly Koral
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Bharat Bhushan
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Anne Orr
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - John Stoops
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - William C Bowen
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Matthew A Copeland
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Joseph Locker
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wendy M Mars
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - George K Michalopoulos
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.
| |
Collapse
|
6
|
Schäringer K, Maxeiner S, Schalla C, Rütten S, Zenke M, Sechi A. LSP1-myosin1e bimolecular complex regulates focal adhesion dynamics and cell migration. FASEB J 2021; 35:e21268. [PMID: 33470457 DOI: 10.1096/fj.202000740rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 11/24/2020] [Accepted: 11/30/2020] [Indexed: 01/22/2023]
Abstract
Several cytoskeleton-associated proteins and signaling pathways work in concert to regulate actin cytoskeleton remodeling, cell adhesion, and migration. Although the leukocyte-specific protein 1 (LSP1) has been shown to interact with the actin cytoskeleton, its function in the regulation of actin cytoskeleton dynamics is, as yet, not fully understood. We have recently demonstrated that the bimolecular complex between LSP1 and myosin1e controls actin cytoskeleton remodeling during phagocytosis. In this study, we show that LSP1 downregulation severely impairs cell migration, lamellipodia formation, and focal adhesion dynamics in macrophages. Inhibition of the interaction between LSP1 and myosin1e also impairs these processes resulting in poorly motile cells, which are characterized by few and small lamellipodia. Furthermore, cells in which LSP1-myosin1e interaction is inhibited are typically associated with inefficient focal adhesion turnover. Collectively, our findings show that the LSP1-myosin1e bimolecular complex plays a pivotal role in the regulation of actin cytoskeleton remodeling and focal adhesion dynamics required for cell migration.
Collapse
Affiliation(s)
- Katja Schäringer
- Department of Cell Biology, Institute of Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Sebastian Maxeiner
- Department of Cell Biology, Institute of Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Carmen Schalla
- Department of Cell Biology, Institute of Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Stephan Rütten
- Electron Microscopy Facility, Institute of Pathology, RWTH Aachen University, Aachen, Germany
| | - Martin Zenke
- Department of Cell Biology, Institute of Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| | - Antonio Sechi
- Department of Cell Biology, Institute of Biomedical Engineering, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
7
|
Afzaljavan F, Moezzi A, Vahednia E, Khorshid Shamshiri A, Vakili F, Homaei Shandiz F, Pasdar A. Predictive and prognostic value of LSP1 rs3817198 in sporadic breast cancer in northeastern population of Iran. Exp Mol Pathol 2020; 116:104514. [PMID: 32738313 DOI: 10.1016/j.yexmp.2020.104514] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/25/2020] [Accepted: 07/25/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Fahimeh Afzaljavan
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Atefeh Moezzi
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elham Vahednia
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran
| | - Asma Khorshid Shamshiri
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran; Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Vakili
- Midwifery department, Faculty of Nursing and Midwifery, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Alireza Pasdar
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Science, Mashhad, Iran; Division of Applied Medicine, Medical School, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, UK; Bioinformatics Research Group, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
8
|
Capasso M, Lasorsa VA, Cimmino F, Avitabile M, Cantalupo S, Montella A, De Angelis B, Morini M, de Torres C, Castellano A, Locatelli F, Iolascon A. Transcription Factors Involved in Tumorigenesis Are Over-Represented in Mutated Active DNA-Binding Sites in Neuroblastoma. Cancer Res 2019; 80:382-393. [PMID: 31784426 DOI: 10.1158/0008-5472.can-19-2883] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 10/24/2019] [Accepted: 11/22/2019] [Indexed: 11/16/2022]
Abstract
The contribution of coding mutations to oncogenesis has been largely clarified, whereas little is known about somatic mutations in noncoding DNA and their role in driving tumors remains controversial. Here, we used an alternative approach to interpret the functional significance of noncoding somatic mutations in promoting tumorigenesis. Noncoding somatic mutations of 151 neuroblastomas were integrated with ENCODE data to locate somatic mutations in regulatory elements specifically active in neuroblastoma cells, nonspecifically active in neuroblastoma cells, and nonactive. Within these types of elements, transcription factors (TF) were identified whose binding sites were enriched or depleted in mutations. For these TFs, a gene expression signature was built to assess their implication in neuroblastoma. DNA- and RNA-sequencing data were integrated to assess the effects of those mutations on mRNA levels. The pathogenicity of mutations was significantly higher in transcription factor binding site (TFBS) of regulatory elements specifically active in neuroblastoma cells, as compared with the others. Within these elements, there were 18 over-represented TFs involved mainly in cell-cycle phase transitions and 15 under-represented TFs primarily regulating cell differentiation. A gene expression signature based on over-represented TFs correlated with poor survival and unfavorable prognostic markers. Moreover, recurrent mutations in TFBS of over-represented TFs such as EZH2 affected MCF2L and ADP-ribosylhydrolase like 1 expression, among the others. We propose a novel approach to study the involvement of regulatory variants in neuroblastoma that could be extended to other cancers and provide further evidence that alterations of gene expression may have relevant effects in neuroblastoma development. SIGNIFICANCE: These findings propose a novel approach to study regulatory variants in neuroblastoma and suggest that noncoding somatic mutations have relevant implications in neuroblastoma development.
Collapse
Affiliation(s)
- Mario Capasso
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli Federico II, Napoli, Italy. .,CEINGE Biotecnologie Avanzate, Napoli, Italy.,IRCCS SDN, Napoli, Italy
| | - Vito Alessandro Lasorsa
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli Federico II, Napoli, Italy.,CEINGE Biotecnologie Avanzate, Napoli, Italy
| | - Flora Cimmino
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli Federico II, Napoli, Italy.,CEINGE Biotecnologie Avanzate, Napoli, Italy
| | - Marianna Avitabile
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli Federico II, Napoli, Italy.,CEINGE Biotecnologie Avanzate, Napoli, Italy
| | | | - Annalaura Montella
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli Federico II, Napoli, Italy.,CEINGE Biotecnologie Avanzate, Napoli, Italy
| | - Biagio De Angelis
- Department of Pediatric Haematology and Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Roma, Italy
| | - Martina Morini
- Laboratory of Molecular Biology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Carmen de Torres
- Developmental Tumor Biology Laboratory, Department of Oncology, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Aurora Castellano
- Department of Pediatric Haematology and Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Roma, Italy
| | - Franco Locatelli
- Department of Pediatric Haematology and Oncology, IRCCS Ospedale Pediatrico Bambino Gesù, Roma, Italy.,Department of Paediatrics, Sapienza University of Rome, Roma, Italy
| | - Achille Iolascon
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli Federico II, Napoli, Italy. .,CEINGE Biotecnologie Avanzate, Napoli, Italy
| |
Collapse
|
9
|
Koral K, Haynes M, Bowen WC, Orr A, Mars W, Michalopoulos GK. Lymphocyte-Specific Protein-1 Controls Sorafenib Sensitivity and Hepatocellular Proliferation through Extracellular Signal-Regulated Kinase 1/2 Activation. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2074-2086. [PMID: 30126548 PMCID: PMC6854472 DOI: 10.1016/j.ajpath.2018.06.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 05/21/2018] [Accepted: 06/11/2018] [Indexed: 02/07/2023]
Abstract
The gene leukocyte-specific protein-1 (LSP1), encodes an F-actin binding protein that directly interacts with the mitogen-activated protein kinase pathway. LSP1 has copy number variations in 52% of human hepatocellular carcinoma (HCC). LSP1 suppresses proliferation and migration in hepatocytes. LSP1 binds to the rapidly accelerated fibrosarcoma (RAF)/mitogen-activated protein/extracellular signal-regulated kinase (ERK)/ERK signaling cassette, the target for sorafenib, a crucial chemotherapeutic agent for HCC. This study addresses the role of LSP1 in liver regeneration and sensitivity to sorafenib in normal and neoplastic hepatocytes. Two mouse models, an Lsp1 global knockout (LSP1KO) and a hepatocyte-specific Lsp1 transgenic (LSP1TG) mouse, were used. After two-thirds hepatectomy (PHx), LSP1KO mice displayed increased proliferation and ERK activation, whereas LSP1TG mice displayed suppressed proliferation and decreased ERK activation. LSP1KO hepatocytes cultured without growth factors exhibited increased proliferation, whereas LSP1TG hepatocytes showed decreased proliferation. Rat and human hepatoma cells expressing Lsp1 shRNA displayed increased sensitivity to sorafenib, as evidenced by decreased cell numbers and phosphorylated ERK expression compared with control. LSP1 KO mice treated with sorafenib before PHx displayed decreased hepatocyte proliferation. Our data show that loss of LSP1 function, observed in HCC, leads to increased sensitivity to sorafenib treatment and enhanced hepatocellular proliferation after PHx in vivo and in cultured cells.
Collapse
Affiliation(s)
- Kelly Koral
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Meagan Haynes
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - William C Bowen
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Anne Orr
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wendy Mars
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | |
Collapse
|
10
|
Zeng SX, Zhu Y, Ma AH, Yu W, Zhang H, Lin TY, Shi W, Tepper CG, Henderson PT, Airhart S, Guo JM, Xu CL, deVere White RW, Pan CX. The Phosphatidylinositol 3-Kinase Pathway as a Potential Therapeutic Target in Bladder Cancer. Clin Cancer Res 2017; 23:6580-6591. [PMID: 28808038 DOI: 10.1158/1078-0432.ccr-17-0033] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 07/07/2017] [Accepted: 08/08/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Activation of the PI3K pathway occurs in over 40% of bladder urothelial cancers. The aim of this study is to determine the therapeutic potential, the underlying action, and the resistance mechanisms of drugs targeting the PI3K pathway.Experimental Design: Urothelial cancer cell lines and patient-derived xenografts (PDXs) were analyzed for alterations of the PI3K pathway and for their sensitivity to the small-molecule inhibitor pictilisib alone and in combination with cisplatin and/or gemcitabine. Potential predictive biomarkers for pictilisib were evaluated, and RNA sequencing was performed to explore drug resistance mechanisms.Results: The bladder cancer cell line TCCSUP, which harbors a PIK3CA E545K mutation, was sensitive to pictilisib compared to cell lines with wild-type PIK3CA Pictilisib exhibited stronger antitumor activity in bladder cancer PDX models with PI3KCA H1047R mutation or amplification than the control PDX model. Pictilisib synergized with cisplatin and/or gemcitabine in vitro, significantly delayed tumor growth, and prolonged survival compared with single-drug treatment in the PDX models. The phosphorylation of ribosomal protein S6 correlated with response to pictilisib both in vitro and in vivo, and could potentially serve as a biomarker to predict response to pictilisib. Pictilisib activated the compensatory MEK/ERK pathway that likely contributed to pictilisib resistance, which was reversed by cotreatment with the RAF inhibitor sorafenib. RNA sequencing of tumors resistant to treatment suggested that LSP1 downregulation correlated with drug resistance.Conclusions: These preclinical results provide new insights into the therapeutic potential of targeting the PI3K pathway for the treatment of bladder cancer. Clin Cancer Res; 23(21); 6580-91. ©2017 AACR.
Collapse
Affiliation(s)
- Shu-Xiong Zeng
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, California.,Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yanjun Zhu
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, California.,Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ai-Hong Ma
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, California
| | - Weimin Yu
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, California.,Department of Urology, Renmin Hospital, Wuhan University, Wuhan, Hubei Province, China
| | - Hongyong Zhang
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, California
| | - Tzu-Yin Lin
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, California
| | - Wei Shi
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, California.,Department of Urology, Xijing Hospital, The Fourth Military Medical University, Shanxi Province, China
| | - Clifford G Tepper
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California Davis, Sacramento, California
| | - Paul T Henderson
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, California
| | | | - Jian-Ming Guo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chuan-Liang Xu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Ralph W deVere White
- Department of Urology, University of California Davis Comprehensive Cancer Center, Sacramento, California
| | - Chong-Xian Pan
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, California. .,Department of Urology, University of California Davis Comprehensive Cancer Center, Sacramento, California.,VA Northern California Health Care System, Mather, California
| |
Collapse
|
11
|
Zhang H, Wang Y, Liu Z, Yao B, Dou C, Xu M, Li Q, Jia Y, Wu S, Tu K, Liu Q. Lymphocyte-specific protein 1 inhibits the growth of hepatocellular carcinoma by suppressing ERK1/2 phosphorylation. FEBS Open Bio 2016; 6:1227-1237. [PMID: 28255535 PMCID: PMC5324767 DOI: 10.1002/2211-5463.12139] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 08/29/2016] [Accepted: 09/27/2016] [Indexed: 12/13/2022] Open
Abstract
Lymphocyte‐specific protein 1 (LSP1) has been reported to regulate cell biology in several human cancers including lymphoma and breast cancer. However, the functions of LSP1 in human hepatocellular carcinoma (HCC) are still unknown. In this study, we found that LSP1 expression was downregulated in HCC tissues and cell lines, and lower LSP1 expression was correlated with poor clinicopathological features including large tumor size, high Edmondson–Steiner grading and advanced tumor–node–metastasis (TNM) stage. Additionally, we demonstrated that patients with high LSP1 expression had significantly better overall survival and disease‐free survival. Moreover, LSP1 was found to be an independent factor for predicting the prognosis of HCC patients. In vitro and in vivo assays showed that overexpressing LSP1 inhibited HCC growth by inducing both apoptosis and growth arrest. Mechanistically, we found that expression of phosphorylated extracellular regulated protein kinases 1 and 2 (ERK1/2) was downregulated after LSP1 overexpression, indicating LSP1 could suppress HCC growth by inhibiting the ERK pathway in HCC cells. Taken together, these results indicate that LSP1 may serve as a prognostic marker and a potential therapeutic target in human HCC.
Collapse
Affiliation(s)
- Hongyong Zhang
- Department of Hepatobiliary Surgery The First Affiliated Hospital of Xi'an Jiaotong University China
| | - Yufeng Wang
- Department of Hepatobiliary Surgery The First Affiliated Hospital of Xi'an Jiaotong University China
| | - Zhikui Liu
- Department of Hepatobiliary Surgery The First Affiliated Hospital of Xi'an Jiaotong University China
| | - Bowen Yao
- Department of Hepatobiliary Surgery The First Affiliated Hospital of Xi'an Jiaotong University China
| | - Changwei Dou
- Department of Hepatobiliary Surgery The First Affiliated Hospital of Xi'an Jiaotong University China
| | - Meng Xu
- Department of Hepatobiliary Surgery The First Affiliated Hospital of Xi'an Jiaotong University China
| | - Qing Li
- Department of Hepatobiliary Surgery The First Affiliated Hospital of Xi'an Jiaotong University China
| | - Yuli Jia
- Department of Hepatobiliary Surgery The First Affiliated Hospital of Xi'an Jiaotong University China
| | - Shengli Wu
- Department of Hepatobiliary Surgery The First Affiliated Hospital of Xi'an Jiaotong University China
| | - Kangsheng Tu
- Department of Hepatobiliary Surgery The First Affiliated Hospital of Xi'an Jiaotong University China
| | - Qingguang Liu
- Department of Hepatobiliary Surgery The First Affiliated Hospital of Xi'an Jiaotong University China
| |
Collapse
|
12
|
Wu JL, Wu HY, Tsai DY, Chiang MF, Chen YJ, Gao S, Lin CC, Lin CH, Khoo KH, Chen YJ, Lin KI. Temporal regulation of Lsp1 O-GlcNAcylation and phosphorylation during apoptosis of activated B cells. Nat Commun 2016; 7:12526. [PMID: 27555448 PMCID: PMC4999498 DOI: 10.1038/ncomms12526] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 07/11/2016] [Indexed: 01/08/2023] Open
Abstract
Crosslinking of B-cell receptor (BCR) sets off an apoptosis programme, but the underlying pathways remain obscure. Here we decipher the molecular mechanisms bridging B-cell activation and apoptosis mediated by post-translational modification (PTM). We find that O-GlcNAcase inhibition enhances B-cell activation and apoptosis induced by BCR crosslinking. This proteome-scale analysis of the functional interplay between protein O-GlcNAcylation and phosphorylation in stimulated mouse primary B cells identifies 313 O-GlcNAcylation-dependent phosphosites on 224 phosphoproteins. Among these phosphoproteins, temporal regulation of the O-GlcNAcylation and phosphorylation of lymphocyte-specific protein-1 (Lsp1) is a key switch that triggers apoptosis in activated B cells. O-GlcNAcylation at S209 of Lsp1 is a prerequisite for the recruitment of its kinase, PKC-β1, to induce S243 phosphorylation, leading to ERK activation and downregulation of BCL-2 and BCL-xL. Thus, we demonstrate the critical PTM interplay of Lsp1 that transmits signals for initiating apoptosis after BCR ligation. B cell receptor (BCR) activation can trigger signalling causing apoptosis in order to eliminate auto-reactive B cells. Here the authors show that the O-GlcNAcylation and phosphorylation of lymphocyte-specific protein-1 are involved in a switch that regulates the initiation of apoptosis induced by BCR cross-linking.
Collapse
Affiliation(s)
- Jung-Lin Wu
- Institute of Microbiology and Immunology, National Yang-Ming University, Taipei 112, Taiwan.,Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Hsin-Yi Wu
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Dong-Yan Tsai
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan.,Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei 112, Taiwan
| | | | - Yi-Ju Chen
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Shijay Gao
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Chun-Cheng Lin
- Department of Chemistry, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Chun-Hung Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Kay-Hooi Khoo
- Institute of Biological Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Yu-Ju Chen
- Institute of Chemistry, Academia Sinica, Taipei 115, Taiwan
| | - Kuo-I Lin
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
13
|
Schevzov G, Kee AJ, Wang B, Sequeira VB, Hook J, Coombes JD, Lucas CA, Stehn JR, Musgrove EA, Cretu A, Assoian R, Fath T, Hanoch T, Seger R, Pleines I, Kile BT, Hardeman EC, Gunning PW. Regulation of cell proliferation by ERK and signal-dependent nuclear translocation of ERK is dependent on Tm5NM1-containing actin filaments. Mol Biol Cell 2015; 26:2475-90. [PMID: 25971798 PMCID: PMC4571302 DOI: 10.1091/mbc.e14-10-1453] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 05/07/2015] [Indexed: 12/27/2022] Open
Abstract
Tropomyosin Tm5NM1 regulates cell proliferation and organ size. It mediates this effect by regulating the interaction of pERK and Imp7, leading to the regulation of pERK nuclear translocation. This demonstrates a role for a specific population of actin filaments in regulating a critical step in the MAPK/ERK signaling pathway. ERK-regulated cell proliferation requires multiple phosphorylation events catalyzed first by MEK and then by casein kinase 2 (CK2), followed by interaction with importin7 and subsequent nuclear translocation of pERK. We report that genetic manipulation of a core component of the actin filaments of cancer cells, the tropomyosin Tm5NM1, regulates the proliferation of normal cells both in vitro and in vivo. Mouse embryo fibroblasts (MEFs) lacking Tm5NM1, which have reduced proliferative capacity, are insensitive to inhibition of ERK by peptide and small-molecule inhibitors, indicating that ERK is unable to regulate proliferation of these knockout (KO) cells. Treatment of wild-type MEFs with a CK2 inhibitor to block phosphorylation of the nuclear translocation signal in pERK resulted in greatly decreased cell proliferation and a significant reduction in the nuclear translocation of pERK. In contrast, Tm5NM1 KO MEFs, which show reduced nuclear translocation of pERK, were unaffected by inhibition of CK2. This suggested that it is nuclear translocation of CK2-phosphorylated pERK that regulates cell proliferation and this capacity is absent in Tm5NM1 KO cells. Proximity ligation assays confirmed a growth factor–stimulated interaction of pERK with Tm5NM1 and that the interaction of pERK with importin7 is greatly reduced in the Tm5NM1 KO cells.
Collapse
Affiliation(s)
- Galina Schevzov
- Oncology Research Unit, School of Medical Sciences, University of New South Wales, Australia, Sydney, NSW 2052, Australia
| | - Anthony J Kee
- Cellular and Genetic Medicine Unit, University of New South Wales, Australia, Sydney, NSW 2052, Australia
| | - Bin Wang
- Oncology Research Unit, School of Medical Sciences, University of New South Wales, Australia, Sydney, NSW 2052, Australia
| | - Vanessa B Sequeira
- Oncology Research Unit, School of Medical Sciences, University of New South Wales, Australia, Sydney, NSW 2052, Australia
| | - Jeff Hook
- Oncology Research Unit, School of Medical Sciences, University of New South Wales, Australia, Sydney, NSW 2052, Australia
| | - Jason D Coombes
- Oncology Research Unit, School of Medical Sciences, University of New South Wales, Australia, Sydney, NSW 2052, Australia Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
| | - Christine A Lucas
- Cellular and Genetic Medicine Unit, University of New South Wales, Australia, Sydney, NSW 2052, Australia
| | - Justine R Stehn
- Oncology Research Unit, School of Medical Sciences, University of New South Wales, Australia, Sydney, NSW 2052, Australia
| | - Elizabeth A Musgrove
- Kinghorn Cancer Centre, Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW 2010, Australia
| | - Alexandra Cretu
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6160
| | - Richard Assoian
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6160
| | - Thomas Fath
- Neurodegeneration and Repair Laboratory, School of Medical Sciences, University of New South Wales, Australia, Sydney, NSW 2052, Australia
| | - Tamar Hanoch
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Rony Seger
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Irina Pleines
- Cancer and Hematology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Benjamin T Kile
- Cancer and Hematology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Edna C Hardeman
- Cellular and Genetic Medicine Unit, University of New South Wales, Australia, Sydney, NSW 2052, Australia
| | - Peter W Gunning
- Oncology Research Unit, School of Medical Sciences, University of New South Wales, Australia, Sydney, NSW 2052, Australia
| |
Collapse
|
14
|
Maxeiner S, Shi N, Schalla C, Aydin G, Hoss M, Vogel S, Zenke M, Sechi AS. Crucial role for the LSP1-myosin1e bimolecular complex in the regulation of Fcγ receptor-driven phagocytosis. Mol Biol Cell 2015; 26:1652-64. [PMID: 25717183 PMCID: PMC4436777 DOI: 10.1091/mbc.e14-05-1005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 02/19/2015] [Indexed: 01/24/2023] Open
Abstract
The actin cytoskeleton is fundamental for the innate immune process of phagocytosis. This study shows that LSP1 plays a pivotal role in the regulation of actin cytoskeleton remodeling during Fcγ receptor–mediated phagocytosis and that its interactions with myosin1e and actin are crucial for the efficiency of this actin-driven process. Actin cytoskeleton remodeling is fundamental for Fcγ receptor–driven phagocytosis. In this study, we find that the leukocyte-specific protein 1 (LSP1) localizes to nascent phagocytic cups during Fcγ receptor–mediated phagocytosis, where it displays the same spatial and temporal distribution as the actin cytoskeleton. Down-regulation of LSP1 severely reduces the phagocytic activity of macrophages, clearly demonstrating a crucial role for this protein in Fcγ receptor–mediated phagocytosis. We also find that LSP1 binds to the class I molecular motor myosin1e. LSP1 interacts with the SH3 domain of myosin1e, and the localization and dynamics of both proteins in nascent phagocytic cups mirror those of actin. Furthermore, inhibition of LSP1–myosin1e and LSP1–actin interactions profoundly impairs pseudopodial formation around opsonized targets and their subsequent internalization. Thus the LSP1–myosin1e bimolecular complex plays a pivotal role in the regulation of actin cytoskeleton remodeling during Fcγ receptor–driven phagocytosis.
Collapse
Affiliation(s)
- Sebastian Maxeiner
- Institute of Biomedical Engineering, Department of Cell Biology, Applied Ecology, D-52074 Aachen, Germany
| | - Nian Shi
- Institute of Biomedical Engineering, Department of Cell Biology, Applied Ecology, D-52074 Aachen, Germany
| | - Carmen Schalla
- Institute of Biomedical Engineering, Department of Cell Biology, Applied Ecology, D-52074 Aachen, Germany
| | - Guelcan Aydin
- Institute of Biomedical Engineering, Department of Cell Biology, Applied Ecology, D-52074 Aachen, Germany
| | - Mareike Hoss
- Electron Microscopy Facility, Uniklinik RWTH Aachen, Applied Ecology, D-52074 Aachen, Germany
| | - Simon Vogel
- Fraunhofer Institute for Molecular Biology and Applied Ecology, D-52074 Aachen, Germany
| | - Martin Zenke
- Institute of Biomedical Engineering, Department of Cell Biology, Applied Ecology, D-52074 Aachen, Germany
| | - Antonio S Sechi
- Institute of Biomedical Engineering, Department of Cell Biology, Applied Ecology, D-52074 Aachen, Germany
| |
Collapse
|
15
|
Koral K, Paranjpe S, Bowen WC, Mars W, Luo J, Michalopoulos GK. Leukocyte-specific protein 1: a novel regulator of hepatocellular proliferation and migration deleted in human hepatocellular carcinoma. Hepatology 2015; 61:537-547. [PMID: 25234543 PMCID: PMC4303494 DOI: 10.1002/hep.27444] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 09/14/2014] [Indexed: 01/18/2023]
Abstract
UNLABELLED Hepatocellular carcinoma (HCC) is the most commonly diagnosed form of liver cancer with high morbidity and mortality. Copy number variation (CNV) analysis of human HCC revealed that leukocyte-specific protein 1 (LSP1) had the highest number of cases with CNV. LSP1, a F-actin-binding protein, is expressed in hematopoietic cells and interacts with kinase suppressor of Ras (KSR), a scaffold for the extracellular signal-related kinase/mitogen-activated protein kinase pathway. Expression of LSP1 in liver, and its role in normal hepatocellular function and carcinogenesis, remains unknown. Therefore, LSP1 messenger RNA and protein levels were analyzed in normal hepatocytes in culture, rat liver following partial hepatectomy (PHx), and hepatoma cell lines. In culture and after PHx, LSP1 increased after the termination of hepatocyte proliferation. To investigate LSP1 function in HCC, short hairpin RNA was utilized to stably knock down LSP1 expression in the JM1 rat hepatoma cell line. Loss of LSP1 in JM1 cells resulted in dramatic up-regulation of cyclin D1 and phosphorylated ERK2, increased cell proliferation, and migration. Coimmunoprecipitation and immunofluorescence analysis displayed an interaction and colocalization between LSP1, KSR, and F-actin in JM1 cells and liver during regeneration. Conversely, expression of LSP1 in the JM2 rat hepatoma cell line led to decreased proliferation. Enhanced expression of LSP1 in mouse hepatocytes during liver regeneration after injection of an LSP1 expression plasmid also led to decreased hepatocyte proliferation. CONCLUSION LSP1 is expressed in normal hepatocytes and liver after PHx after termination of proliferation. In rat hepatoma cell lines and mouse liver in vivo, LSP1 functions as a negative regulator of proliferation and migration. Given the high frequency of LSP1 CNV in human HCC, LSP1 may be a novel target for diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Kelly Koral
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA
| | | | | | | | | | | |
Collapse
|
16
|
Visentini M, Marrapodi R, Conti V, Mitrevski M, Camponeschi A, Lazzeri C, Carbonari M, Catizone A, Quinti I, Fiorilli M. Dysregulated extracellular signal-regulated kinase signaling associated with impaired B-cell receptor endocytosis in patients with common variable immunodeficiency. J Allergy Clin Immunol 2014; 134:401-10. [PMID: 24792875 DOI: 10.1016/j.jaci.2014.03.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 03/17/2014] [Accepted: 03/21/2014] [Indexed: 10/25/2022]
Abstract
BACKGROUND Common variable immunodeficiency (CVID) is a heterogeneous disorder characterized by B-cell dysfunction and, in a subgroup, by expansion of CD21(low) B cells. The CD21(low) B cells display defects in early B-cell receptor (BCR) signaling resembling those of anergic B cells. OBJECTIVE We sought to investigate whether B cells from patients with CVID, like anergic B cells, have defects in extracellular signal-regulated kinase (ERK) phosphorylation and in endocytic trafficking of the BCR. METHODS Using flow cytometry, we evaluated phosphorylated ERK (pERK) expression and internalization of cross-linked BCR in B-cell subsets. The localization of internalized BCR to lysosome-associated membrane protein 1-positive late endosomes was evaluated with confocal microscopy. RESULTS Constitutive pERK levels were increased in naive and IgM(+) memory B cells of patients with CVID compared with those of healthy donors, whereas the pERK increment induced by BCR cross-linking was relatively reduced. Intravenous immunoglobulin administration enhanced these anomalies, but they appeared to be intrinsic to B cells from patients with CVID. Cross-linking-induced BCR endocytosis was decreased in the IgM(+) memory B cells, especially in those with a CD21(low) phenotype, but not in the naive B cells of patients with CVID with CD21(low) expansion. Internalized BCR localized normally to late endosomes. Pharmacologic inhibition of ERK phosphorylation suppressed BCR endocytosis in B cells of healthy patients and those with CVID. CONCLUSIONS The B cells of patients with CVID with CD21(low) B-cell expansion resemble anergic B cells based on high constitutive pERK expression. The IgM(+) memory B cells of these patients, especially those that are CD21(low), have a defect in BCR endocytosis seemingly caused by dysregulated ERK signaling.
Collapse
Affiliation(s)
- Marcella Visentini
- Department of Clinical Immunology, Sapienza University of Rome, Rome, Italy
| | - Ramona Marrapodi
- Department of Clinical Immunology, Sapienza University of Rome, Rome, Italy
| | - Valentina Conti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Milica Mitrevski
- Department of Clinical Immunology, Sapienza University of Rome, Rome, Italy
| | | | - Cristina Lazzeri
- Department of Clinical Immunology, Sapienza University of Rome, Rome, Italy
| | - Maurizio Carbonari
- Department of Clinical Immunology, Sapienza University of Rome, Rome, Italy
| | - Angela Catizone
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
| | - Isabella Quinti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Massimo Fiorilli
- Department of Clinical Immunology, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
17
|
Jia Y, Zhou J, Liu H, Huang K. Effect of methionine sulfoxide reductase B1 (SelR) gene silencing on peroxynitrite-induced F-actin disruption in human lens epithelial cells. Biochem Biophys Res Commun 2013; 443:876-81. [PMID: 24342607 DOI: 10.1016/j.bbrc.2013.12.055] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 12/10/2013] [Indexed: 11/15/2022]
Abstract
F-actin plays a crucial role in fundamental cellular processes, and is extremely susceptible to peroxynitrite attack due to the high abundance of tyrosine in the peptide. Methionine sulfoxide reductase (Msr) B1 is a selenium-dependent enzyme (selenoprotein R) that may act as a reactive oxygen species (ROS) scavenger. However, its function in coping with reactive nitrogen species (RNS)-mediated stress and the physiological significance remain unclear. Thus, the present study was conducted to elucidate the role and mechanism of MsrB1 in protecting human lens epithelial (hLE) cells against peroxynitrite-induced F-actin disruption. While exposure to high concentrations of peroxynitrite and gene silencing of MsrB1 by siRNA alone caused disassembly of F-actin via inactivation of extracellular signal-regulated kinase (ERK) in hLE cells, the latter substantially aggravated the disassembly of F-actin triggered by the former. This aggravation concurred with elevated nitration of F-actin and inactivation of ERK compared with that induced by the peroxynitrite treatment alone. In conclusion, MsrB1 protected hLE cells against the peroxynitrite-induced F-actin disruption, and the protection was mediated by inhibiting the resultant nitration of F-actin and inactivation of ERKs.
Collapse
Affiliation(s)
- Yi Jia
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, 1037 Luoyu Road, Hongshan, Wuhan, Hubei 430074, People's Republic of China.
| | - Jun Zhou
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, 1037 Luoyu Road, Hongshan, Wuhan, Hubei 430074, People's Republic of China
| | - Hongmei Liu
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, 1037 Luoyu Road, Hongshan, Wuhan, Hubei 430074, People's Republic of China
| | - Kaixun Huang
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, 1037 Luoyu Road, Hongshan, Wuhan, Hubei 430074, People's Republic of China.
| |
Collapse
|
18
|
Sabbagh L, Andreeva D, Laramée GD, Oussa NAE, Lew D, Bisson N, Soumounou Y, Pawson T, Watts TH. Leukocyte-specific protein 1 links TNF receptor-associated factor 1 to survival signaling downstream of 4-1BB in T cells. J Leukoc Biol 2013; 93:713-21. [PMID: 23446150 DOI: 10.1189/jlb.1112579] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
4-1BB is a member of the TNFR superfamily, which contributes to the activation of signaling pathways required for the survival of activated and memory T cells. We have shown previously that TRAF1, an adaptor protein recruited to 4-1BB, is required for 4-1BB-mediated CD8 T cell survival in vivo. With the use of a proteomics approach in primary T cells, we have identified LSP1 as a novel protein recruited to the 4-1BB signaling complex in a TRAF1-dependent manner. Further characterization of the interaction between TRAF1 and LSP1 revealed that LSP1 requires the TRAF-N domain of TRAF1 for direct association. Similarly to TRAF1(-/-) T cells, LSP1(-/-) T cells exhibit impaired ERK activation following stimulation through 4-1BB and consequently, are unable to down-modulate expression of the proapoptotic Bcl-2 family member Bim. Moreover, we demonstrate that the absence of LSP1 expression leads to defective expansion and survival of T cells in response to 4-1BB stimulation. Thus, we have identified LSP1 as a new mediator involved in 4-1BB signaling and T cell survival. Collectively, our work shows that TRAF1 and LSP1 cooperate downstream of 4-1BB to activate ERK signaling and down-modulate the levels of Bim leading to enhanced T cell survival.
Collapse
Affiliation(s)
- Laurent Sabbagh
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, Quebec, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Nalesnik MA, Tseng G, Ding Y, Xiang GS, Zheng ZL, Yu Y, Marsh JW, Michalopoulos GK, Luo JH. Gene deletions and amplifications in human hepatocellular carcinomas: correlation with hepatocyte growth regulation. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 180:1495-1508. [PMID: 22326833 PMCID: PMC3657620 DOI: 10.1016/j.ajpath.2011.12.021] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 12/09/2011] [Accepted: 12/22/2011] [Indexed: 12/29/2022]
Abstract
Tissues from 98 human hepatocellular carcinomas (HCCs) obtained from hepatic resections were subjected to somatic copy number variation (CNV) analysis. Most of these HCCs were discovered in livers resected for orthotopic transplantation, although in a few cases, the tumors themselves were the reason for the hepatectomies. Genomic analysis revealed deletions and amplifications in several genes, and clustering analysis based on CNV revealed five clusters. The LSP1 gene had the most cases with CNV (46 deletions and 5 amplifications). High frequencies of CNV were also seen in PTPRD (21/98), GNB1L (18/98), KIAA1217 (18/98), RP1-1777G6.2 (17/98), ETS1 (11/98), RSU1 (10/98), TBC1D22A (10/98), BAHCC1 (9/98), MAML2 (9/98), RAB1B (9/98), and YIF1A (9/98). The existing literature regarding hepatocytes or other cell types has connected many of these genes to regulation of cytoskeletal architecture, signaling cascades related to growth regulation, and transcription factors directly interacting with nuclear signaling complexes. Correlations with existing literature indicate that genomic lesions associated with HCC at the level of resolution of CNV occur on many genes associated directly or indirectly with signaling pathways operating in liver regeneration and hepatocyte growth regulation.
Collapse
Affiliation(s)
- Michael A. Nalesnik
- Department of Pathology, the Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - George Tseng
- Departments of Biostatistics, the Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ying Ding
- Department of Surgery, the Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
- Departments of Biostatistics, the Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Guo-Sheng Xiang
- Department of Pathology, the Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Zhong-liang Zheng
- Department of Pathology, the Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - YanPing Yu
- Department of Pathology, the Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - James W. Marsh
- Joint CMU-Pitt Ph.D. Program in Computational Biology, the Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - George K. Michalopoulos
- Department of Pathology, the Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jian-Hua Luo
- Department of Pathology, the Graduate School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
20
|
Wortzel I, Seger R. The ERK Cascade: Distinct Functions within Various Subcellular Organelles. Genes Cancer 2011; 2:195-209. [PMID: 21779493 DOI: 10.1177/1947601911407328] [Citation(s) in RCA: 406] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The extracellular signal-regulated kinase 1/2 (ERK1/2) cascade is a central signaling pathway that regulates a wide variety of stimulated cellular processes, including mainly proliferation, differentiation, and survival, but apoptosis and stress response as well. The ability of this linear cascade to induce so many distinct and even opposing effects after various stimulations raises the question as to how the signaling specificity of the cascade is regulated. Over the past years, several specificity-mediating mechanisms have been elucidated, including temporal regulation, scaffolding interactions, crosstalks with other signaling components, substrate competition, and multiple components in each tier of the cascade. In addition, spatial regulation of various components of the cascade is probably one of the main ways by which signals can be directed to some downstream targets and not to others. In this review, we describe first the components of the ERK1/2 cascade and their mode of regulation by kinases, phosphatases, and scaffold proteins. In the second part, we focus on the role of MEK1/2 and ERK1/2 compartmentalization in the nucleus, mitochondria, endosomes, plasma membrane, cytoskeleton, and Golgi apparatus. We explain that this spatial distribution may direct ERK1/2 signals to regulate the organelles' activities. However, it can also direct the activity of the cascade's components to the outer surface of the organelles in order to bring them to close proximity to specific cytoplasmic targets. We conclude that the dynamic localization of the ERK1/2 cascade components is an important regulatory mechanism in determining the signaling specificity of the cascade, and its understanding should shed a new light on the understanding of many stimulus-dependent processes.
Collapse
Affiliation(s)
- Inbal Wortzel
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
21
|
Stadler ZK, Vijai J, Thom P, Kirchhoff T, Hansen NA, Kauff ND, Robson M, Offit K. Genome-wide Association Studies of Cancer Predisposition. Hematol Oncol Clin North Am 2010; 24:973-96. [DOI: 10.1016/j.hoc.2010.06.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
22
|
Chang M, Kanwar N, Feng E, Siu A, Liu X, Ma D, Jongstra J. PIM kinase inhibitors downregulate STAT3(Tyr705) phosphorylation. Mol Cancer Ther 2010; 9:2478-87. [PMID: 20667852 DOI: 10.1158/1535-7163.mct-10-0321] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Using a cell-based high-throughput screen designed to detect small chemical compounds that inhibit cell growth and survival, we identified three structurally related compounds, 21A8, 21H7, and 65D4, with differential activity on cancer versus normal cells. Introduction of structural modifications yielded compound M-110, which inhibits the proliferation of prostate cancer cell lines with IC(50)s of 0.6 to 0.9 μmol/L, with no activity on normal human peripheral blood mononuclear cells up to 40 μmol/L. Screening of 261 recombinant kinases and subsequent analysis revealed that M-110 is a selective inhibitor of the PIM kinase family, with preference for PIM-3. The prostate cancer cell line DU-145 and the pancreatic cancer cell line MiaPaCa2 constitutively express activated STAT3 (pSTAT3(Tyr705)). Treatment of DU-145 cells with M-110 or with a structurally unrelated PIM inhibitor, SGI-1776, significantly reduces pSTAT3(Tyr705) expression without affecting the expression of STAT3. Furthermore, treatment of DU-145 cells with M-110 attenuates the interleukin-6-induced increase in pSTAT3(Tyr705). To determine which of the three PIM kinases is most likely to inhibit expression of pSTAT3(Tyr705), we used PIM-1-, PIM-2-, or PIM-3-specific siRNA and showed that knockdown of PIM-3, but not of PIM-1 or PIM-2, in DU-145 cells results in a significant downregulation of pSTAT3(Tyr705). The phosphorylation of STAT5 on Tyr694 in 22Rv1 cells is not affected by M-110 or SGI-1776, suggesting specificity for pSTAT3(Tyr705). These results identify a novel role for PIM-3 kinase as a positive regulator of STAT3 signaling and suggest that PIM-3 inhibitors cause growth inhibition of cancer cells by downregulating the expression of pSTAT3(Tyr705).
Collapse
Affiliation(s)
- Marisa Chang
- Genetics and Development Division, Toronto Western Research Institute, University Health Network, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
23
|
Schibler L, Gibbs L, Benoist-Lasselin C, Decraene C, Martinovic J, Loget P, Delezoide AL, Gonzales M, Munnich A, Jais JP, Legeai-Mallet L. New insight on FGFR3-related chondrodysplasias molecular physiopathology revealed by human chondrocyte gene expression profiling. PLoS One 2009; 4:e7633. [PMID: 19898608 PMCID: PMC2764091 DOI: 10.1371/journal.pone.0007633] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Accepted: 10/03/2009] [Indexed: 11/18/2022] Open
Abstract
Endochondral ossification is the process by which the appendicular skeleton, facial bones, vertebrae and medial clavicles are formed and relies on the tight control of chondrocyte maturation. Fibroblast growth factor receptor (FGFR)3 plays a role in bone development and maintenance and belongs to a family of proteins which differ in their ligand affinities and tissue distribution. Activating mutations of the FGFR3 gene lead to craniosynostosis and multiple types of skeletal dysplasia with varying degrees of severity: thanatophoric dysplasia (TD), achondroplasia and hypochondroplasia. Despite progress in the characterization of FGFR3-mediated regulation of cartilage development, many aspects remain unclear. The aim and the novelty of our study was to examine whole gene expression differences occurring in primary human chondrocytes isolated from normal cartilage or pathological cartilage from TD-affected fetuses, using Affymetrix technology. The phenotype of the primary cells was confirmed by the high expression of chondrocytic markers. Altered expression of genes associated with many cellular processes was observed, including cell growth and proliferation, cell cycle, cell adhesion, cell motility, metabolic pathways, signal transduction, cell cycle process and cell signaling. Most of the cell cycle process genes were down-regulated and consisted of genes involved in cell cycle progression, DNA biosynthesis, spindle dynamics and cytokinesis. About eight percent of all modulated genes were found to impact extracellular matrix (ECM) structure and turnover, especially glycosaminoglycan (GAG) and proteoglycan biosynthesis and sulfation. Altogether, the gene expression analyses provide new insight into the consequences of FGFR3 mutations in cell cycle regulation, onset of pre-hypertrophic differentiation and concomitant metabolism changes. Moreover, impaired motility and ECM properties may also provide clues about growth plate disorganization. These results also suggest that many signaling pathways may be directly or indirectly altered by FGFR3 and confirm the crucial role of FGFR3 in the control of growth plate development.
Collapse
Affiliation(s)
- Laurent Schibler
- Unité U781, Institut National de la Santé et de la Recherche Médicale, Université Paris Descartes-Hôpital Necker, Paris, France
- Unité Mixte de Recherche 1313, Institut National de la Recherche Agronomique, Jouy-en-Josas, France
| | - Linda Gibbs
- Unité U781, Institut National de la Santé et de la Recherche Médicale, Université Paris Descartes-Hôpital Necker, Paris, France
- 4Clinics, Waterloo, Belgique
| | - Catherine Benoist-Lasselin
- Unité U781, Institut National de la Santé et de la Recherche Médicale, Université Paris Descartes-Hôpital Necker, Paris, France
| | | | - Jelena Martinovic
- Service de Fœtopathologie, Hôpital Necker, Université Paris Descartes, Paris, France
| | - Philippe Loget
- Centre Pluridisciplinaire de Diagnostic Prénatal de Rennes, Hôpital de Rennes, Rennes, France
| | - Anne-Lise Delezoide
- Service de Biologie du développement, Hôpital Robert Debré, Université Paris Diderot, Paris, France
| | - Marie Gonzales
- Service de Génétique et d'Embryologie Médicales, Hôpital Armand Trousseau, Université Pierre et Marie Curie, Paris, France
| | - Arnold Munnich
- Unité U781, Institut National de la Santé et de la Recherche Médicale, Université Paris Descartes-Hôpital Necker, Paris, France
| | - Jean-Philippe Jais
- Service de Biostatistique et Informatique Médicale, Hôpital Necker, Université Paris Descartes, Paris, France
| | - Laurence Legeai-Mallet
- Unité U781, Institut National de la Santé et de la Recherche Médicale, Université Paris Descartes-Hôpital Necker, Paris, France
- * E-mail:
| |
Collapse
|
24
|
Abstract
The extracellular signal-regulated kinase cascade is a central signaling pathway that is stimulated by various extracellular stimuli. The signals of these stimuli are then transferred by the cascade's components to a large number of targets at distinct subcellular compartments, which in turn induce and regulate a large number of cellular processes. To achieve these functions, the cascade exhibits versatile and dynamic subcellular distribution that allows proper temporal and spatial modulation of the appropriate processes. In this review, we discuss the intracellular localizations of different components of the ERK cascade, and the impact of these localizations on their activation and specificity.
Collapse
Affiliation(s)
- Zhong Yao
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
25
|
Gringhuis SI, den Dunnen J, Litjens M, van der Vlist M, Geijtenbeek TBH. Carbohydrate-specific signaling through the DC-SIGN signalosome tailors immunity to Mycobacterium tuberculosis, HIV-1 and Helicobacter pylori. Nat Immunol 2009; 10:1081-8. [PMID: 19718030 DOI: 10.1038/ni.1778] [Citation(s) in RCA: 344] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Accepted: 07/07/2009] [Indexed: 12/16/2022]
Abstract
Cooperation between different innate signaling pathways induced by pattern-recognition receptors (PRRs) on dendritic cells (DCs) is crucial for tailoring adaptive immunity to pathogens. Here we show that carbohydrate-specific signaling through the C-type lectin DC-SIGN tailored cytokine production in response to distinct pathogens. DC-SIGN was constitutively associated with a signalosome complex consisting of the scaffold proteins LSP1, KSR1 and CNK and the kinase Raf-1. Mannose-expressing Mycobacterium tuberculosis and human immunodeficiency virus type 1 (HIV-1) induced the recruitment of effector proteins to the DC-SIGN signalosome to activate Raf-1, whereas fucose-expressing pathogens such as Helicobacter pylori actively dissociated the KSR1-CNK-Raf-1 complex from the DC-SIGN signalosome. This dynamic regulation of the signalosome by mannose- and fucose-expressing pathogens led to the enhancement or suppression of proinflammatory responses, respectively. Our study reveals another level of plasticity in tailoring adaptive immunity to pathogens.
Collapse
Affiliation(s)
- Sonja I Gringhuis
- Center of Infection and Immunity Amsterdam, University of Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | |
Collapse
|
26
|
Yin X, Zafrullah M, Lee H, Haimovitz-Friedman A, Fuks Z, Kolesnick R. A ceramide-binding C1 domain mediates kinase suppressor of ras membrane translocation. Cell Physiol Biochem 2009; 24:219-30. [PMID: 19710537 DOI: 10.1159/000233248] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2009] [Indexed: 11/19/2022] Open
Abstract
Genetic and biochemical data support Kinase Suppressor of Ras 1 (KSR1) as a positive regulator of the Ras-Raf-MAPK pathway, functioning as a kinase and/or scaffold to regulate c-Raf-1 activation. Membrane translocation mediated by the KSR1 CA3 domain, which is homologous to the atypical PKC C1 lipid-binding domain, is a critical step of KSR1-mediated c-Raf-1 activation. In this study, we used an ELISA to characterize the KSR1 CA3 domain as a lipid-binding moiety. Purified GST-KSR1-CA3 protein effectively binds ceramide but not other lipids including 1,2-diacylglyceol, dihydroceramide, ganglioside GM1, sphingomyelin and phosphatidylcholine. Upon epidermal growth factor stimulation of COS-7 cells, KSR1 translocates into and is activated within glycosphingolipid-enriched plasma membrane platforms. Pharmacologic inhibition of ceramide generation attenuates KSR1 translocation and KSR1 kinase activation in COS-7 cells. Disruption of two cysteines, which are indispensable for maintaining ternary structure of all C1 domains and their lipid binding capability, mitigates ceramide-binding capacity of purified GST-KSR1-CA3 protein, and inhibits full length KSR1 membrane translocation and kinase activation. These studies provide evidence for a mechanism by which the second messenger ceramide can target proteins to subcellular compartments in the process of transmembrane signal transduction.
Collapse
Affiliation(s)
- Xianglei Yin
- Laboratory of Signal Transduction, Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | | | | | | | | | | |
Collapse
|
27
|
Anglesio MS, Arnold JM, George J, Tinker AV, Tothill R, Waddell N, Simms L, Locandro B, Fereday S, Traficante N, Russell P, Sharma R, Birrer MJ, AOCS Study Group, deFazio A, Chenevix-Trench G, Bowtell DDL. Mutation of ERBB2 provides a novel alternative mechanism for the ubiquitous activation of RAS-MAPK in ovarian serous low malignant potential tumors. Mol Cancer Res 2008; 6:1678-90. [PMID: 19010816 PMCID: PMC6953412 DOI: 10.1158/1541-7786.mcr-08-0193] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Collaborators] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Approximately, 10% to 15% of serous ovarian tumors fall into the category designated as tumors of low malignant potential (LMP). Like their invasive counterparts, LMP tumors may be associated with extraovarian disease, for example, in the peritoneal cavity and regional lymph nodes. However, unlike typical invasive carcinomas, patients generally have a favorable prognosis. The mutational profile also differs markedly from that seen in most serous carcinomas. Typically, LMP tumors are associated with KRAS and BRAF mutations. Interrogation of expression profiles in serous LMP tumors suggested overall redundancy of RAS-MAPK pathway mutations and a distinct mechanism of oncogenesis compared with high-grade ovarian carcinomas. Our findings indicate that activating mutation of the RAS-MAPK pathway in serous LMP may be present in >70% of cases compared with approximately 12.5% in serous ovarian carcinomas. In addition to mutations of KRAS (18%) and BRAF (48%) mutations, ERBB2 mutations (6%), but not EGFR, are prevalent among serous LMP tumors. Based on the expression profile signature observed throughout our serous LMP cohort, we propose that RAS-MAPK pathway activation is a requirement of serous LMP tumor development and that other activators of this pathway are yet to be defined. Importantly, as few nonsurgical options exist for treatment of recurrent LMP tumors, therapeutic targeting of this pathway may prove beneficial, especially in younger patients where maintaining fertility is important.
Collapse
Collaborators
D Bowtell, G Chenevix-Trench, A deFazio, D Gertig, A Green, P Webb,
Collapse
|
28
|
Jackson WM, Jaasma MJ, Tang RY, Keaveny TM. Mechanical loading by fluid shear is sufficient to alter the cytoskeletal composition of osteoblastic cells. Am J Physiol Cell Physiol 2008; 295:C1007-15. [DOI: 10.1152/ajpcell.00509.2007] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Many structural modifications have been observed as a part of the cellular response to mechanical loading in a variety of cell types. Although changes in morphology and cytoskeletal rearrangement have been widely reported, few studies have investigated the change in cytoskeletal composition. Measuring how the amounts of specific structural proteins in the cytoskeleton change in response to mechanical loading will help to elucidate cellular mechanisms of functional adaptation to the applied forces. Therefore, the overall hypothesis of this study was that osteoblasts would respond to fluid shear stress by altering the amount of specific cross-linking proteins in the composition of the cytoskeleton. Mouse osteoblats cell line MC3T3-E1 and human fetal osteoblasts (hFOB) were exposed to 2 Pa of steady fluid shear for 2 h in a parallel plate flow chamber, and then the amount of actin, vimentin, α-actinin, filamin, and talin in the cytoskeleton was measured using Western blot analyses. After mechanical loading, there was no change in the amount of actin monomers in the cytoskeleton, but the cross-linking proteins α-actinin and filamin that cofractionated with the cytoskeleton increased by 29% ( P < 0.01) and 18% ( P < 0.02), respectively. Localization of the cross-linking proteins by fluorescent microscopy revealed that they were more widely distributed throughout the cell after exposure to fluid shear. The amount of vimentin in the cytoskeleton also increased by 15% ( P < 0.01). These results indicate that osteoblasts responded to mechanical loading by altering the cytoskeletal composition, which included an increase in specific proteins that would likely enhance the mechanical resistance of the cytoskeleton.
Collapse
|
29
|
Parsa KVL, Butchar JP, Rajaram MVS, Cremer TJ, Tridandapani S. The tyrosine kinase Syk promotes phagocytosis of Francisella through the activation of Erk. Mol Immunol 2008; 45:3012-21. [PMID: 18295889 PMCID: PMC2408723 DOI: 10.1016/j.molimm.2008.01.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2007] [Accepted: 01/15/2008] [Indexed: 11/29/2022]
Abstract
Francisella tularensis is a highly infectious, Gram-negative intra-cellular pathogen that can cause the zoonotic disease tularemia. Although the receptors critical for internalization of Francisella by macrophages are beginning to be defined, the identity of the downstream signaling pathways essential for the engulfment are not yet identified. In this study we have tested the role of Syk in the phagocytosis of Francisella. We report that Syk is activated during Francisella infection and is critical for the uptake of the organisms. Pharmacologic inhibition of Syk almost completely abrogated uptake, whereas the overexpression of Syk significantly enhanced uptake. However, Syk appears to be dispensable during initial host-pathogen contact. Further analyses of the molecular mechanism of Syk influence on Francisella uptake revealed that the MAPK Erk but not the phosphatidylinositol 3 kinase (PI3K)/Akt pathway is the downstream effector of Syk. Thus, the inhibition of Erk in Syk-overexpressing cells or the inhibition of Syk in Erk-overexpressing cells led to a significant attenuation of uptake. Collectively, these data identify Syk and Erk as key players in the phagocytosis of Francisella.
Collapse
Affiliation(s)
- Kishore V L Parsa
- Ohio State Biochemistry Program, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | |
Collapse
|
30
|
Pal NR, Aguan K, Sharma A, Amari SI. Discovering biomarkers from gene expression data for predicting cancer subgroups using neural networks and relational fuzzy clustering. BMC Bioinformatics 2007; 8:5. [PMID: 17207284 PMCID: PMC1770936 DOI: 10.1186/1471-2105-8-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2006] [Accepted: 01/06/2007] [Indexed: 11/10/2022] Open
Abstract
Background The four heterogeneous childhood cancers, neuroblastoma, non-Hodgkin lymphoma, rhabdomyosarcoma, and Ewing sarcoma present a similar histology of small round blue cell tumor (SRBCT) and thus often leads to misdiagnosis. Identification of biomarkers for distinguishing these cancers is a well studied problem. Existing methods typically evaluate each gene separately and do not take into account the nonlinear interaction between genes and the tools that are used to design the diagnostic prediction system. Consequently, more genes are usually identified as necessary for prediction. We propose a general scheme for finding a small set of biomarkers to design a diagnostic system for accurate classification of the cancer subgroups. We use multilayer networks with online gene selection ability and relational fuzzy clustering to identify a small set of biomarkers for accurate classification of the training and blind test cases of a well studied data set. Results Our method discerned just seven biomarkers that precisely categorized the four subgroups of cancer both in training and blind samples. For the same problem, others suggested 19–94 genes. These seven biomarkers include three novel genes (NAB2, LSP1 and EHD1 – not identified by others) with distinct class-specific signatures and important role in cancer biology, including cellular proliferation, transendothelial migration and trafficking of MHC class antigens. Interestingly, NAB2 is downregulated in other tumors including Non-Hodgkin lymphoma and Neuroblastoma but we observed moderate to high upregulation in a few cases of Ewing sarcoma and Rabhdomyosarcoma, suggesting that NAB2 might be mutated in these tumors. These genes can discover the subgroups correctly with unsupervised learning, can differentiate non-SRBCT samples and they perform equally well with other machine learning tools including support vector machines. These biomarkers lead to four simple human interpretable rules for the diagnostic task. Conclusion Although the proposed method is tested on a SRBCT data set, it is quite general and can be applied to other cancer data sets. Our scheme takes into account the interaction between genes as well as that between genes and the tool and thus is able find a very small set and can discover novel genes. Our findings suggest the possibility of developing specialized microarray chips or use of real-time qPCR assays or antibody based methods such as ELISA and western blot analysis for an easy and low cost diagnosis of the subgroups.
Collapse
Affiliation(s)
- Nikhil R Pal
- Electronics and Communication Sciences Unit, Indian Statistical Institute, 203, B. T. Road, Calcutta – 700108, India
| | - Kripamoy Aguan
- Neurogenetics Laboratory, RIKEN Brain Science Institute, Saitama, Japan
| | - Animesh Sharma
- Department of Computer Science and Electrical Engineering, University of Missouri, Kansas city, USA
| | - Shun-ichi Amari
- Lab for Mathematical Neuroscience, RIKEN Brain Science Institute, Saitama, Japan
| |
Collapse
|
31
|
Jongstra-Bilen J, Jongstra J. Leukocyte-specific protein 1 (LSP1): a regulator of leukocyte emigration in inflammation. Immunol Res 2006; 35:65-74. [PMID: 17003510 DOI: 10.1385/ir:35:1:65] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 01/22/2023]
Abstract
LSP1 is an F-actin bundling cytoskeletal protein expressed in hematopoietic lineage and endothelial cells. We investigated the function of this protein by generating and analyzing an LSP1-deficient mouse strain and in this review we describe our findings together with those of other investigators. The results show a complex function of LSP1 in regulating leukocyte recruitment to inflamed sites. Based on current evidence, we propose that the levels of LSP1 on the cytoskeleton and the type of integrin involved are some of the critical elements which affect LSP1 function in modulating the threshold for transmigration.
Collapse
Affiliation(s)
- Jenny Jongstra-Bilen
- Cell and Molecular Biology Division, Toronto General Research Institute, University Health Network, Toronto Western Research Institute, University Health Network, Ontario, Canada.
| | | |
Collapse
|
32
|
Abstract
Hairy-cell leukaemia (HCL) has long been recognized as distinct from other chronic B-cell malignancies, but several questions remain unanswered. What is the HCL cell of origin? Why does HCL lack the hallmarks of most mature B-cell tumours (for example, chromosomal translocations and consistent lymph node involvement) and show unique features like 'hairy' morphology and bone-marrow fibrosis? Gene-expression profiling and other studies have recently provided new insights into HCL biology and have the potential to affect clinical practice.
Collapse
Affiliation(s)
- Enrico Tiacci
- Institute of Haematology, University of Perugia, Italy.
| | | | | | | |
Collapse
|
33
|
Delogu A, Schebesta A, Sun Q, Aschenbrenner K, Perlot T, Busslinger M. Gene repression by Pax5 in B cells is essential for blood cell homeostasis and is reversed in plasma cells. Immunity 2006; 24:269-81. [PMID: 16546096 DOI: 10.1016/j.immuni.2006.01.012] [Citation(s) in RCA: 256] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2005] [Revised: 01/17/2006] [Accepted: 01/20/2006] [Indexed: 11/15/2022]
Abstract
The transcription factor Pax5 represses lineage-inappropriate genes and activates B cell-specific genes in B lymphocytes. By identifying 110 Pax5-repressed genes, we now demonstrate that Pax5 downregulates diverse biological activities including receptor signaling, cell adhesion, migration, transcriptional control, and cellular metabolism at B cell commitment. The T lymphoid or myeloid expression of these genes demonstrates that Pax5(-/-) pro-B cells and common lymphoid progenitors display lymphoid and myeloid promiscuity of gene expression. These lineage-inappropriate genes require continuous Pax5 activity for their repression, as they are reactivated in committed pro-B cells and mature B cells following conditional Pax5 deletion. Pax5-repressed genes are also reexpressed in plasma cells, which depend for normal function on Cd28 and Ccr2 reactivation. The loss of Pax5 during terminal differentiation thus contributes to the plasma cell transcription program. Finally, ectopic expression of the Pax5-repressed chemokine gene Ccl3 in B cells results in increased osteoclast formation and bone loss, demonstrating that Pax5-mediated gene repression is essential for normal homeostasis of hematopoietic development.
Collapse
Affiliation(s)
- Alessio Delogu
- Research Institute of Molecular Pathology, Vienna Biocenter, Dr. Bohr-Gasse 7, A-1030 Vienna, Austria
| | | | | | | | | | | |
Collapse
|
34
|
Ebisuya M, Kondoh K, Nishida E. The duration, magnitude and compartmentalization of ERK MAP kinase activity: mechanisms for providing signaling specificity. J Cell Sci 2006; 118:2997-3002. [PMID: 16014377 DOI: 10.1242/jcs.02505] [Citation(s) in RCA: 263] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
ERK MAP kinase signaling plays a pivotal role in diverse cellular functions, including cell proliferation, differentiation, migration and survival. One of the central questions concerning this signaling is how activation of the same protein kinase, ERK, elicits distinct cellular outcomes. Recent progress has demonstrated that differences in the duration, magnitude and subcellular compartmentalization of ERK activity generate variations in signaling output that regulate cell fate decisions. Furthermore, several molecules have been identified as spatial, temporal or strength-controlling regulators of ERK activity. Signaling by various extracellular stimuli thus could be modulated by these regulators to give qualitative and quantitative differences in ERK activity, which are then interpreted by the cells as determinants for appropriate responses.
Collapse
Affiliation(s)
- Miki Ebisuya
- Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan
| | | | | |
Collapse
|
35
|
Kolch W. Coordinating ERK/MAPK signalling through scaffolds and inhibitors. Nat Rev Mol Cell Biol 2005; 6:827-37. [PMID: 16227978 DOI: 10.1038/nrm1743] [Citation(s) in RCA: 826] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The pathway from Ras through Raf and MEK (MAPK and ERK kinase) to ERK/MAPK (extracellular signal-regulated kinase/mitogen-activated protein kinase) regulates many fundamental cellular processes. Recently, a number of scaffolding proteins and endogenous inhibitors have been identified, and their important roles in regulating signalling through this pathway are now emerging. Some scaffolds augment the signal flux, but also mediate crosstalk with other pathways; certain adaptors target MEK-ERK/MAPK complexes to subcellular localizations; others provide regulated inhibition. Computational modelling indicates that, together, these modulators can determine the dynamic biological behaviour of the pathway.
Collapse
Affiliation(s)
- Walter Kolch
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK.
| |
Collapse
|
36
|
Liu L, Cara DC, Kaur J, Raharjo E, Mullaly SC, Jongstra-Bilen J, Jongstra J, Kubes P. LSP1 is an endothelial gatekeeper of leukocyte transendothelial migration. ACTA ACUST UNITED AC 2005; 201:409-18. [PMID: 15684321 PMCID: PMC2213033 DOI: 10.1084/jem.20040830] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Leukocyte-specific protein 1 (LSP1), an F-actin binding protein and a major downstream substrate of p38 mitogen-activated protein kinase as well as protein kinase C, has been reported to be important in leukocyte chemotaxis. Although its distribution has been thought to be restricted to leukocytes, herein we report that LSP1 is expressed in endothelium and is essential to permit neutrophil emigration. Using intravital microscopy to directly visualize leukocyte rolling, adhesion, and emigration in postcapillary venules in LSP1-deficient (Lsp1−/−) mice, we found that LSP1 deficiency inhibits neutrophil extravasation in response to various cytokines (tumor necrosis factor-α and interleukin-1β) and to neutrophil chemokine keratinocyte-derived chemokine in vivo. LSP1 deficiency did not affect leukocyte rolling or adhesion. Generation of Lsp1−/− chimeric mice using bone marrow transplantation revealed that in mice with Lsp1−/− endothelial cells and wild-type leukocytes, neutrophil transendothelial migration out of postcapillary venules is markedly restricted. In contrast, Lsp1−/− neutrophils in wild-type mice were able to extravasate normally. Consistent with altered endothelial function was a reduction in vascular permeability to histamine in Lsp1−/− animals. Western blot analysis and immunofluorescence microscopy examination confirmed the presence of LSP1 in wild-type but not in Lsp1−/− mouse microvascular endothelial cells. Cultured human endothelial cells also stained positive for LSP1. Our results suggest that LSP1 expressed in endothelium regulates neutrophil transendothelial migration.
Collapse
Affiliation(s)
- Lixin Liu
- Immunology Research Group, Department of Physiology and Biophysics, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | | | | | | | | | | | | | | |
Collapse
|