1
|
Mekapogu AR, Xu Z, Pothula S, Perera C, Pang T, Hosen SMZ, Damalanka V, Janetka J, Goldstein D, Pirola R, Wilson J, Apte M. HGF/c-Met pathway inhibition combined with chemotherapy increases cytotoxic T-cell infiltration and inhibits pancreatic tumour growth and metastasis. Cancer Lett 2023:216286. [PMID: 37354984 DOI: 10.1016/j.canlet.2023.216286] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/17/2023] [Accepted: 06/20/2023] [Indexed: 06/26/2023]
Abstract
Pancreatic cancer (PC) is a deadly cancer with a high mortality rate. The unique characteristics of PC, including desmoplasia and immunosuppression, have made it difficult to develop effective treatment strategies. Pancreatic stellate cells (PSCs) play a crucial role in the progression of the disease by interacting with cancer cells. One of the key mediators of PSC - cancer cell interactions is the hepatocyte growth factor (HGF)/c-MET pathway. Using an immunocompetent in vivo model of PC as well as in vitro experiments, this study has shown that a combined approach using HGF/c-MET inhibitors to target stromal-tumour interactions and chemotherapy (gemcitabine) to target cancer cells effectively decreases tumour volume, EMT, and stemness, and importantly, eliminates metastasis. Notably, HGF/c-MET inhibition decreases TGF-β secretion by cancer cells, resulting in an increase in cytotoxic T-cell infiltration, thus contributing to cancer cell death in tumours. HGF/c-MET inhibition + chemotherapy was also found to normalise the gut microbiome and improve gut microbial diversity. These findings provide a strong platform for assessment of this triple therapy (HGF/c-MET inhibition + chemotherapy) approach in the clinical setting.
Collapse
Affiliation(s)
- Alpha Raj Mekapogu
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Zhihong Xu
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Srinivasa Pothula
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia; AbCellera, Beaconsfield, New South Wales, United Kingdom
| | - Chamini Perera
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Tony Pang
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia; Surgical Innovations Unit, Westmead Hospital, Sydney, Australia; Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - S M Zahid Hosen
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Vishnu Damalanka
- Department of Biochemistry and Molecular Biophysics, Washington University, St. Louis, USA
| | - James Janetka
- Department of Biochemistry and Molecular Biophysics, Washington University, St. Louis, USA
| | - David Goldstein
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia
| | - Romano Pirola
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia
| | - Jeremy Wilson
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia
| | - Minoti Apte
- Pancreatic Research Group, South West Sydney Clinical Campuses, School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Australia; Ingham Institute for Applied Medical Research, Sydney, Australia.
| |
Collapse
|
2
|
Cazes A, Childers BG, Esparza E, Lowy AM. The MST1R/RON Tyrosine Kinase in Cancer: Oncogenic Functions and Therapeutic Strategies. Cancers (Basel) 2022; 14:cancers14082037. [PMID: 35454943 PMCID: PMC9027306 DOI: 10.3390/cancers14082037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/10/2022] [Accepted: 04/13/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary MST1R/RON receptor tyrosine kinase is a highly conserved transmembrane protein present on epithelial cells, macrophages, and recently identified in a T-cell subset. RON activation attenuates inflammation in healthy tissue. Interestingly, it is overexpressed in several epithelial neoplasms with increasing levels of expression associated with worse outcomes. Though the mechanisms involved are still under investigation, RON is involved in carcinogenesis via immune modulation of the immune tumor microenvironment, activation of numerous oncogenic pathways, and is protective under cellular stress. Alternatively, inhibition of RON abrogates tumor progression in both animal and human tissue models. Given this, RON is a targetable protein of great interest for cancer treatment. Here, we review RON’s function in tissue inflammation and cancer progression, and review cancer clinical trials to date that have used agents targeting RON signaling. Abstract The MST1R/RON receptor tyrosine kinase is a homologue of the more well-known MET receptor. Like MET, RON orchestrates cell signaling pathways that promote oncogenesis and enable cancer cell survival; however, it has a more unique role in the regulation of inflammation. RON was originally described as a transmembrane receptor expressed on tissue resident macrophages and various epithelial cells. RON is overexpressed in a variety of cancers and its activation modifies multiple signaling pathways with resultant changes in epithelial and immune cells which together modulate oncogenic phenotypes. While several RON isoforms have been identified with differences in structure, activation, and pathway regulation, increased RON expression and/or activation is consistently associated with worse outcomes. Tyrosine kinase inhibitors targeting RON have been developed, making RON an actionable therapeutic target.
Collapse
|
3
|
Li C, Morvaridi S, Lam G, Chheda C, Kamata Y, Katsumata M, Edderkaoui M, Yuan X, Nissen N, Pandol SJ, Wang Q. MSP-RON Signaling Is Activated in the Transition From Pancreatic Intraepithelial Neoplasia (PanIN) to Pancreatic Ductal Adenocarcinoma (PDAC). Front Physiol 2019; 10:147. [PMID: 30863319 PMCID: PMC6399467 DOI: 10.3389/fphys.2019.00147] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 02/07/2019] [Indexed: 12/18/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the deadliest epithelial malignancies and remains difficult to treat. Pancreatic intraepithelial neoplasias (PanINs) represent the majority of the pre-cancer lesions in the pancreas. The PDAC microenvironment consists of activated pancreatic stellate cells (PSCs) and immune cells, which are thought to contribute to neoplastic transformation. However, the signaling events involved in driving the transition from the neoplastic precursor to the more advanced and aggressive forms in the pancreas are not well understood. Recepteur d’Origine Nantais (RON) is a c-MET family receptor tyrosine kinase that is implicated in playing a role in cell proliferation, migration and other aspects of tumorigenesis. Macrophage stimulating protein (MSP) is the ligand for RON and becomes activated upon proteolytic cleavage by matriptase (also known as ST14), a type II transmembrane serine protease. In the current study, by immunohistochemistry (IHC) analysis of human pancreatic tissues, we found that the expression levels MSP and matriptase are drastically increased during the transition from the preneoplastic PanIN stages to the more advanced and aggressive PDAC. Moreover, RON is highly expressed in both PDAC and in cancer-associated stellate cells. In contrast, MSP, RON, and matriptase are expressed at low levels, if any, in normal pancreas. Our study underscores an emerging role of MSP-RON autocrine and paracrine signaling events in driving malignant progression in the pancreas.
Collapse
Affiliation(s)
- Ce Li
- Department of Medical Oncology, The First Affiliated Hospital of China Medical University, Shenyang, China.,Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Susan Morvaridi
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Gloria Lam
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Chintan Chheda
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Yoshiko Kamata
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Makoto Katsumata
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Mouad Edderkaoui
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Xiaopu Yuan
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Nicholas Nissen
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Stephen J Pandol
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Qiang Wang
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
4
|
Park YL, Lee GH, Kim KY, Myung E, Kim JS, Myung DS, Park KJ, Cho SB, Lee WS, Jung YD, Kim HS, Joo YE. Expression of RON in colorectal cancer and its relationships with tumor cell behavior and prognosis. TUMORI JOURNAL 2018; 98:652-62. [DOI: 10.1177/030089161209800517] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Aims and background The aims of the current study were to evaluate whether recepteur d'origine nantais (RON) affects tumor cell behavior and oncogenic signaling pathways in colorectal cancer, and to examine the relationship of its expression with various clinicopathological parameters and patient survival. Methods Immunohistochemistry, Western blot and RT-PCR were used to detect the expression of the RON gene in human colorectal cancer tissue. To study the biological role of RON in tumor cell behavior and cellular signaling pathways, we used small interfering RNA (siRNA) to knock down RON gene expression in human colorectal cancer cell lines. Results Knockdown of RON inhibited the induction of the invasive growth phenotype and the activation of oncogenic signaling pathways including Akt, MAPK and β-catenin. RON overexpression was associated with tumor size, lymphovascular invasion, depth of invasion, lymph node metastasis, distant metastasis, tumor stage and poor survival. Conclusions These results suggest that RON overexpression may help in predicting poor clinical outcomes in colorectal cancer.
Collapse
Affiliation(s)
- Young-Lan Park
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Gi-Hoon Lee
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Kyu-Yeol Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Eun Myung
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Jong-Sun Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Dae-Seong Myung
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Kang-Jin Park
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Sung-Bum Cho
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Wan-Sik Lee
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Young-Do Jung
- Department of Biochemistry, Chonnam National University Medical School, Gwangju, Korea
| | - Hyun-Soo Kim
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | - Young-Eun Joo
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
5
|
LoRusso PM, Gounder M, Jalal SI, André V, Kambhampati SRP, Loizos N, Hall J, Holzer TR, Nasir A, Cosaert J, Kauh J, Chiorean EG. Phase 1 study of narnatumab, an anti-RON receptor monoclonal antibody, in patients with advanced solid tumors. Invest New Drugs 2017; 35:442-450. [PMID: 28161886 DOI: 10.1007/s10637-016-0413-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/22/2016] [Indexed: 01/21/2023]
Abstract
Purpose Macrophage-stimulating 1-receptor (RON) is expressed on macrophages, epithelial cells, and a variety of tumors. Narnatumab (IMC-RON8; LY3012219) is a neutralizing monoclonal antibody that blocks RON binding to its ligand, macrophage-stimulating protein (MSP). This study assessed safety, maximum tolerated dose (MTD), pharmacokinetics, pharmacodynamics, and efficacy of narnatumab in patients with advanced solid tumors. Methods Narnatumab was administered intravenously weekly at 5, 10, 15, or 20 mg/kg or every 2 weeks at 15, 20, 30, or 40 mg/kg in 4-week cycles. Results Thirty-nine patients were treated, and 1 dose-limiting toxicity (DLT) (grade 3 hyponatremia, 5 mg/kg) was reported. The most common narnatumab-related adverse events (AEs) were fatigue (20.5%) and decreased appetite, diarrhea, nausea, and vomiting (10.3% each). Except for 2 treatment-related grade 3 AEs (hyponatremia, hypokalemia), all treatment-related AEs were grade 1 or 2. Narnatumab had a short half-life (<7 days). After Cycle 2, no patients had concentrations above 140 μg/mL (concentration that demonstrated antitumor activity in animal models), except for 1 patient receiving 30 mg/kg biweekly. Eleven patients had a best response of stable disease, ranging from 6 weeks to 11 months. Despite only 1 DLT, due to suboptimal drug exposure, the dose was not escalated beyond 40 mg/kg biweekly. This decision was based on published data reporting that mRNA splice variants of RON are highly prevalent in tumors, accumulate in cytoplasm, and are not accessible by large-molecule monoclonal antibodies. Conclusions Narnatumab was well tolerated and showed limited antitumor activity with this dosing regimen.
Collapse
Affiliation(s)
- Patricia M LoRusso
- Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA. .,Yale Cancer Center, New Haven, CT, USA.
| | - Mrinal Gounder
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shadia I Jalal
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA
| | | | | | - Nick Loizos
- Eli Lilly and Company, Indianapolis, IN, USA
| | - Jennifer Hall
- Eli Lilly and Company, Indianapolis, IN, USA.,Boehringer Ingelheim, Ridgefield, CT, USA
| | | | - Aejaz Nasir
- Eli Lilly and Company, Indianapolis, IN, USA
| | - Jan Cosaert
- Eli Lilly and Company, Indianapolis, IN, USA.,Merck KGaA, Darmstadt, Germany
| | - John Kauh
- Eli Lilly and Company, Indianapolis, IN, USA
| | - E Gabriela Chiorean
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA.,Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA, USA
| |
Collapse
|
6
|
Faham N, Welm AL. RON Signaling Is a Key Mediator of Tumor Progression in Many Human Cancers. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2017; 81:177-188. [PMID: 28057847 DOI: 10.1101/sqb.2016.81.031377] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
With an increasing body of literature covering RON receptor tyrosine kinase function in different types of human cancers, it is becoming clear that RON has prominent roles in both cancer cells and in the tumor-associated microenvironment. RON not only activates several oncogenic signaling pathways in cancer cells, leading to more aggressive behavior, but also promotes an immunosuppressive, alternatively activated phenotype in macrophages and limits the antitumor immune response. These two unique functions of this oncogene, the strong correlation between RON expression and poor outcomes in cancer, and the high tolerability of a new RON inhibitor make it an exciting therapeutic target, the blocking of which offers an advantage toward improving the survival of cancer patients. Here, we discuss recent findings on the role of RON signaling in cancer progression and its potential in cancer therapy.
Collapse
Affiliation(s)
- Najme Faham
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112
| | - Alana L Welm
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112
| |
Collapse
|
7
|
Zou Y, Howell GM, Humphrey LE, Wang J, Brattain MG. Ron knockdown and Ron monoclonal antibody IMC-RON8 sensitize pancreatic cancer to histone deacetylase inhibitors (HDACi). PLoS One 2013; 8:e69992. [PMID: 23922886 PMCID: PMC3726703 DOI: 10.1371/journal.pone.0069992] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 06/13/2013] [Indexed: 02/06/2023] Open
Abstract
Recepteur d'origine nantais (Ron) is overexpressed in a panel of pancreatic cancer cells and tissue samples from pancreatic cancer patients. Ron can be activated by its ligand macrophage stimulating protein (MSP), thereby activating oncogenic signaling pathways. Crosstalk between Ron and EGFR, c-Met, or IGF-1R may provide a mechanism underlying drug resistance. Thus, targeting Ron may represent a novel therapeutic strategy. IMC-RON8 is the first Ron monoclonal antibody (mAb) entering clinical trial for targeting Ron overexpression. Our studies show IMC-RON8 downmodulated Ron expression in pancreatic cancer cells and significantly blocked MSP-stimulated Ron activation, downstream Akt and ERK phosphorylation, and survivin mRNA expression. IMC-RON8 hindered MSP-induced cell migration and reduced cell transformation. Histone deacetylase inhibitors (HDACi) are reported to target expression of various genes through modification of nucleosome histones and non-histone proteins. Our work shows HDACi TSA and Panobinostat (PS) decreased Ron mRNA and protein expression in pancreatic cancer cells. PS also reduced downstream signaling of pAkt, survivin, and XIAP, as well as enhanced cell apoptosis. Interestingly, PS reduced colony formation in Ron knockdown cells to a greater extent than Ron scramble control cells in colony formation and soft agarose assays. IMC-RON8 could also sensitize pancreatic cancer cells to PS, as reflected by reduced colony numbers and size in combination treatment with IMC-RON8 and PS compared to single treatment alone. The co-treatment further reduced Ron expression and pAkt, and increased PARP cleavage compared to either treatment alone. This study suggests the potential for a novel combination approach which may ultimately be of value in treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Yi Zou
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Gillian M. Howell
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Lisa E. Humphrey
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Jing Wang
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Michael G. Brattain
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| |
Collapse
|
8
|
Ron tyrosine kinase receptor synergises with EGFR to confer adverse features in head and neck squamous cell carcinoma. Br J Cancer 2013; 109:482-92. [PMID: 23799848 PMCID: PMC3721396 DOI: 10.1038/bjc.2013.321] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 05/30/2013] [Accepted: 06/04/2013] [Indexed: 02/04/2023] Open
Abstract
Background: Although EGFR inhibitors have shown some success in the treatment of head and neck squamous cell carcinomas (HNSCCs), the results are not dramatic. Additional molecular targets are urgently needed. We previously showed that the loss of Ron receptor activity significantly slowed squamous tumour growth and progression in a murine model. Based on these data, we hypothesised that Ron expression confers an aggressive phenotype in HNSCCs. Methods: We prospectively collected and evaluated 154 snap-frozen, primary HNSCCs for Ron and EGFR expression/phosphorylation. Biomarker correlation with clinical, pathological and outcome data was performed. The biological responses of HNSCC cell lines to Ron knockdown, its activation and the biochemical interaction between Ron and EGFR were examined. Results: We discovered that 64.3% (99 out of 154) HNSCCs expressed Ron. The carcinomas expressed exclusively mature functional Ron, whereas the adjacent nonmalignant epithelium expressed predominantly nonfunctional Ron precursor. There was no significant association between Ron and sex, tumour differentiation, perineural/vascular invasion or staging. However, patients with Ron+HNSCC were significantly older and more likely to have oropharyngeal tumours. Ron+HNSCC also had significantly higher EGFR expression and correlated strongly with phosphorylated EGFR (pEGFR). Newly diagnosed HNSCC with either Ron/pEGFR or both had lower disease-free survival than those without Ron and pEGFR. Knocking down Ron in SCC9 cells significantly blunted their migratory response to not only the Ron ligand, MSP, but also EGF. Stimulation of Ron in SCC9 cells significantly augmented the growth effect of EGF; the synergistic effect of both growth factors in SCC9 cells was dependent on Ron expression. Activated Ron also interacted with and transactivated EGFR. Conclusion: Ron synergises with EGFR to confer certain adverse features in HNSCCs.
Collapse
|
9
|
Macrophage stimulating protein promotes liver metastases of small cell lung cancer cells by affecting the organ microenvironment. Clin Exp Metastasis 2012; 30:333-44. [PMID: 23011677 DOI: 10.1007/s10585-012-9540-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Accepted: 09/19/2012] [Indexed: 12/12/2022]
Abstract
The organ microenvironment significantly affects the processes of cancer metastasis. Elucidating the molecular mechanisms of interaction between tumor cells and the organ microenvironment is crucial for the development of effective therapeutic strategies to eradicate cancer metastases. Macrophage stimulating protein (MSP), an activator of macrophages, regulates a pleiotropic array of effects, including proliferation, cellular motility, invasiveness, angiogenesis, and resistance to anoikis. However, the role of MSP in cancer metastasis is still largely unknown. In this study, the action of MSP on the production of metastases was determined in a multiple-organ metastasis model. The murine MSP gene was transfected into two human SCLC cell lines, SBC-5 and H1048, to establish transfectants secreting biologically active MSP. MSP gene transduction did not affect cell proliferation and motility in vitro. Intravenously inoculated MSP transfectants produced significantly larger numbers of liver metastases than parental cells or vector control clones, while there were no significant differences in bone or lung metastases among them. Immunohistochemical analyses of liver metastases revealed that tumor-associated microvessel density and tumor-infiltrating macrophages were significantly increased in lesions produced by MSP transfectants. MSP could stimulate the migration of murine macrophages and endothelial cells in vitro. Consequently, MSP may be one of the major determinants that affects the properties of tumor stroma and that produces a permissive microenvironment to promote cancer metastasis.
Collapse
|
10
|
Knockdown of RON inhibits AP-1 activity and induces apoptosis and cell cycle arrest through the modulation of Akt/FoxO signaling in human colorectal cancer cells. Dig Dis Sci 2012; 57:371-80. [PMID: 21901254 DOI: 10.1007/s10620-011-1892-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2011] [Accepted: 08/24/2011] [Indexed: 12/30/2022]
Abstract
BACKGROUND/AIMS Altered Recepteur d'Origine nantais (RON) expression transduces signals inducting invasive growth phenotype that includes cell proliferation, migration, matrix invasion, and protection of apoptosis in human cancer cells. The aims of the current study were to evaluate whether RON affects tumor cell behavior and cellular signaling pathways including activator protein-1 (AP-1) and Akt/forkhead box O (FoxO) in human colorectal cancer cells. METHODS To study the biological role of RON on tumor cell behavior and cellular signaling pathways in human colorectal cancer, we used small interfering RNA (siRNA) to knockdown RON gene expression in human colorectal cancer cell line, DKO-1. RESULTS Knockdown of RON diminished migration, invasion, and proliferation of human colorectal cancer cells. Knockdown of RON decreased AP-1 transcriptional activity and expression of AP-1 target genes. Knockdown of RON activated cleaved caspase-3, -7, -9, and PARP, and down-regulated the expression of Mcl-1, survivin and XIAP, leading to induction of apoptosis. Knockdown of RON induced cell cycle arrest in the G2/M phase of cancer cells by an increase of p27 and a decrease of cyclin D3. Knockdown of RON inhibited the phosphorylation of Akt/FoxO signaling proteins such as Ser473 and Thr308 of Akt and FoxO1/3a. CONCLUSIONS These results indicate that knockdown of RON inhibits AP-1 activity and induces apoptosis and cell cycle arrest through the modulation of Akt/FoxO signaling in human colorectal cancer cells.
Collapse
|
11
|
Song YA, Park YL, Kim KY, Myung E, Chung CY, Cho SB, Lee WS, Jung YD, Kweon SS, Joo YE. RON is associated with tumor progression via the inhibition of apoptosis and cell cycle arrest in human gastric cancer. Pathol Int 2011; 62:127-36. [PMID: 22243783 DOI: 10.1111/j.1440-1827.2011.02765.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The recepteur d'origine nantais (RON) receptor tyrosine kinase is overexpressed in epithelial cancers, including gastric cancer. The aims of the present study were to evaluate whether RON affects tumor cell behaviors and oncogenic signaling pathways, and to document the relationship of its expression with various clinicopathological parameters in gastric cancer. The biological role of RON in tumor cell behaviors and oncogenic signaling pathways was investigated by using small interfering RNA in gastric cancer cell lines including AGS and MKN28. The expression of RON in gastric cancer tissues was investigated by using reverse transcription polymerase chain reaction and immunohistochemistry. Knockdown of RON suppressed tumor cell migration and invasion in AGS and MKN28, induced apoptosis through modulation of anti-apoptotic and pre-apoptotic genes and induced cell cycle arrest by decreasing cyclin D1, cyclin D3 and CDK4, and by inducing p21 and p27 expression. Signaling cascades, including Akt and mitogen-activated protein kinase (MAPK), were significantly blocked by knockdown of RON. Expression of RON was significantly associated with tumor size, depth of invasion, lymph node metastasis, tumor stage and poor survival. These results indicate that RON is associated with tumor progression via the inhibition of apoptosis and cell cycle arrest in human gastric cancer.
Collapse
Affiliation(s)
- Young-A Song
- Department of Internal Medicine, Chonnam National University Medical School, Gwangju, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Benvenuti S, Lazzari L, Arnesano A, Li Chiavi G, Gentile A, Comoglio PM. Ron kinase transphosphorylation sustains MET oncogene addiction. Cancer Res 2011; 71:1945-55. [PMID: 21212418 DOI: 10.1158/0008-5472.can-10-2100] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Receptors for the scatter factors HGF and MSP that are encoded by the MET and RON oncogenes are key players in invasive growth. Receptor cross-talk between Met and Ron occurs. Amplification of the MET oncogene results in kinase activation, deregulated expression of an invasive growth phenotype, and addiction to MET oncogene signaling (i.e., dependency on sustained Met signaling for survival and proliferation). Here we show that cancer cells addicted to MET also display constitutive activation of the Ron kinase. In human cancer cell lines coexpressing the 2 oncogenes, Ron is specifically transphosphorylated by activated Met. In contrast, Ron phosphorylation is not triggered in cells harboring constitutively active kinase receptors other than Met, including Egfr or Her2. Furthermore, Ron phosphorylation is suppressed by Met-specific kinase inhibitors (PHA-665752 or JNJ-38877605). Last, Ron phosphorylation is quenched by reducing cell surface expression of Met proteins by antibody-induced shedding. In MET-addicted cancer cells, short hairpin RNA-mediated silencing of RON expression resulted in decreased proliferation and clonogenic activity in vitro and tumorigenicity in vivo. Our findings establish that oncogene addiction to MET involves Ron transactivation, pointing to Ron kinase as a target for combinatorial cancer therapy.
Collapse
Affiliation(s)
- Silvia Benvenuti
- Exploratory Research Laboratory, Institute for Cancer Research and Treatment (IRCC), University of Turin Medical School, 10060 Candiolo, Turin, Italy
| | | | | | | | | | | |
Collapse
|
13
|
Germano S, O'Driscoll L. Western blotting analysis as a tool to study receptor tyrosine kinases. Methods Mol Biol 2011; 784:109-121. [PMID: 21898216 DOI: 10.1007/978-1-61779-289-2_8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Receptor tyrosine kinases (RTKs) are involved in critical aspects of cell physiology ranging from cell -survival, proliferation, growth, migration, and differentiation. A tight control of the extent and duration of signals elicited by activated RTKs is crucial for preventing over-stimulation, which can ultimately lead to unrestrained proliferative ability and neoplastic growth. Ligand-induced downregulation of RTKs has emerged as a key negative regulatory mechanism that can accomplish signaling attenuation, by removing activated receptors from the cell surface and committing them to degradation. The ability of RTKs to escape from ligand-induced downregulation has been reported as a recurrent mechanism of oncogenic deregulation in cancer.Western blotting procedures have been extensively proven as straightforward assays to evaluate -protein expression levels and have been widely applied to study RTKs downregulation.
Collapse
Affiliation(s)
- Serena Germano
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland.
| | | |
Collapse
|
14
|
Nikolaidis NM, Gray JK, Gurusamy D, Fox W, Stuart WD, Huber N, Waltz SE. Ron receptor tyrosine kinase negatively regulates TNFalpha production in alveolar macrophages by inhibiting NF-kappaB activity and Adam17 production. Shock 2010; 33:197-204. [PMID: 19487969 DOI: 10.1097/shk.0b013e3181ae8155] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The Ron receptor tyrosine kinase (TK) plays a regulatory role in the inflammatory response to acute lung injury induced by intranasal administration of bacterial LPS. Previously, we have shown that mice with a targeted deletion of the TK signaling domain of the Ron receptor exhibited more severe lung injury in response to intranasal LPS administration as evidenced by an increased leakage of albumin in the lungs and a greater thickening of the alveolar septa compared with wild-type mice. In addition, lung injury in the Ron TK-deficient (TK(-/-)) mice was associated with increased activation of the transcription factor, nuclear factor-kappaB (NF-kappaB), and significantly increased intrapulmonary expression of TNFalpha. TNFalpha, a multifunctional proinflammatory cytokine, is a central mediator in several disease states, including rheumatoid arthritis and sepsis. On the basis of the observation that TNFalpha production is increased in the Ron TK-/- mice and that macrophages are a major source of this cytokine, we hypothesized that the alterations observed in the Ron TK(-/-) mice may be due, in part, to Ron signaling, specifically in alveolar macrophages. To test this hypothesis, we used the wild-type and Ron TK(-/-) primary alveolar macrophages and the murine alveolar macrophage cell line, MH-S, to examine the effects of Ron activation on LPS-induced TNFalpha production and NF-kappaB activity. Here, we reported that Ron is expressed on alveolar macrophages and MH-S cells. Activation of Ron by its ligand, hepatocyte growth factor-like protein, decreases TNFalpha production in alveolar macrophages after LPS challenge. Decreased TNFalpha is associated with hepatocyte growth factor-like protein-induced decreases in NF-kappaB activation and increases in the NF-kappaB inhibitory protein, IkappaB. We also provided the first evidence for Ron as a negative regulator of Adam17, the metalloprotease involved in TNFalpha processing. These results indicate that Ron plays a critical role in regulation of alveolar macrophage signaling and validates this receptor as a target in TNFalpha-mediated pulmonary pathologies.
Collapse
Affiliation(s)
- Nikolaos M Nikolaidis
- Department of Cancer and Cell Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0521, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Chitra E, Yu SL, Hsiao KN, Shao HY, Sia C, Chen IH, Hsieh SY, Chen JH, Chow YH. Generation and characterization of JSRV envelope transgenic mice in FVB background. Virology 2009; 393:120-6. [DOI: 10.1016/j.virol.2009.07.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Revised: 05/29/2009] [Accepted: 07/22/2009] [Indexed: 01/23/2023]
|
16
|
Eckerich C, Schulte A, Martens T, Zapf S, Westphal M, Lamszus K. RON receptor tyrosine kinase in human gliomas: expression, function, and identification of a novel soluble splice variant. J Neurochem 2009; 109:969-80. [PMID: 19519771 DOI: 10.1111/j.1471-4159.2009.06027.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Malignant gliomas are incurable because of their diffuse infiltration of the surrounding brain. The recepteur d'origine nantais (RON) receptor tyrosine kinase is highly expressed in several epithelial cancer types and mediates tumorigenic, pro-invasive as well as metastatic effects. Analyzing RON expression in human gliomas, we found that different splice variants with known oncogenic activity are expressed in glioblastomas (GBM). In addition, the RON ligand macrophage-stimulating protein (MSP) is secreted by cultured GBM cells. MSP showed no mitogenic effect on GBM cells but displayed significant chemotactic activity for several GBM cell lines. We identified a novel splice variant, RONDelta90, which is generated by a transcript missing exon 6. As a result of a frameshift, translation is terminated in exon 7, resulting in a truncated soluble protein. RONDelta90 transcripts are expressed in normal human brain as well as in low grade astrocytomas but only in approximately 50% of highly malignant astrocytomas. In addition, RONDelta90 is detectable in supernatants of GBM cell lines. We cloned the RONDelta90 cDNA, and purified the recombinant protein from transfected cells. RONDelta90 inhibited MSP-induced phosphorylation of cellular RON and also attenuated basal activation levels. In addition, RONDelta90 inhibited MSP-induced glioma cell migration as well as random motility. To conclude, RONDelta90 is a novel soluble receptor variant with antagonistic activity that may act as a physiological modulator of RON signaling. The expression of several oncogenic RON splice variants in malignant gliomas suggests that these could represent candidate targets for treatment with agents inhibiting RON activity.
Collapse
Affiliation(s)
- Carmen Eckerich
- Department of Neurosurgery, Laboratory for Brain Tumor Biology, University Medical Center Hamburg-Eppendorf, Martinistrasse, Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
17
|
Kobayashi T, Furukawa Y, Kikuchi J, Ito C, Miyata Y, Muto S, Tanaka A, Kusano E. Transactivation of RON receptor tyrosine kinase by interaction with PDGF receptor beta during steady-state growth of human mesangial cells. Kidney Int 2009; 75:1173-1183. [PMID: 19242504 DOI: 10.1038/ki.2009.44] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Although it is well known that platelet-derived growth factor (PDGF) causes mesangial cell proliferation (presumably contributing to progression of glomerular disease), targeted inhibition of the PDGF receptor system has shown only limited efficacy against glomerular diseases. To examine whether this discrepancy is due to the involvement of other pathways, we used phosphorylated receptor tyrosine kinase arrays and found that RON (recepteur d'origine nantais) was phosphorylated while the PDGF receptor was dephosphorylated (thus inactive) in human mesangial cells (HMCs) at the time of cell cycle entry. Further, RON remained active during steady-state growth. Activation of RON was independent of its canonical ligand, macrophage-stimulating protein, but was mediated by transactivation from the PDGF-engaged PDGF receptor. Following stimulation with PDGF we found that the two receptors physically interacted. Knockdown of RON by siRNA increased the number of apoptotic cells without affecting the rate of DNA synthesis, suggesting that RON has anti-apoptotic functions. Immunohistochemical analysis found phosphorylated RON in glomerular lesions of patients with IgA nephropathy but not those with minimal change nephrotic syndrome, a disease not associated with mesangial proliferation. These results suggest that RON is involved in mesangial cell proliferation under both physiological and pathological conditions, and may be a relevant target for therapeutic intervention.
Collapse
Affiliation(s)
- Takahisa Kobayashi
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan; Division of Stem Cell Regulation, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Yusuke Furukawa
- Division of Stem Cell Regulation, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan.
| | - Jiro Kikuchi
- Division of Stem Cell Regulation, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Chiharu Ito
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Yukio Miyata
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Shigeaki Muto
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Akira Tanaka
- Department of Pathology, Jichi Medical University, Shimotsuke, Tochigi, Japan
| | - Eiji Kusano
- Division of Nephrology, Department of Internal Medicine, Jichi Medical University, Shimotsuke, Tochigi, Japan
| |
Collapse
|
18
|
Wagh PK, Peace BE, Waltz SE. Met-related receptor tyrosine kinase Ron in tumor growth and metastasis. Adv Cancer Res 2008; 100:1-33. [PMID: 18620091 DOI: 10.1016/s0065-230x(08)00001-8] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The Ron receptor is a member of the Met family of cell surface receptor tyrosine kinases and is primarily expressed on epithelial cells and macrophages. The biological response of Ron is mediated by binding of its ligand, hepatocyte growth factor-like protein/macrophage stimulating-protein (HGFL). HGFL is primarily synthesized and secreted from hepatocytes as an inactive precursor and is activated at the cell surface. Binding of HGFL to Ron activates Ron and leads to the induction of a variety of intracellular signaling cascades that leads to cellular growth, motility and invasion. Recent studies have documented Ron overexpression in a variety of human cancers including breast, colon, liver, pancreas, and bladder. Moreover, clinical studies have also shown that Ron overexpression is associated with both worse patient outcomes as well as metastasis. Forced overexpression of Ron in transgenic mice leads to tumorigenesis in both the lung and the mammary gland and is associated with metastatic dissemination. While Ron overexpression appears to be a hallmark of many human cancers, the mechanisms by which Ron induces tumorigenesis and metastasis are still unclear. Several strategies are currently being undertaken to inhibit Ron as a potential therapeutic target; current strategies include the use of Ron blocking proteins, small interfering RNA (siRNA), monoclonal antibodies, and small molecule inhibitors. In total, these data suggest that Ron is a critical factor in tumorigenesis and that inhibition of this protein, alone or in combination with current therapies, may prove beneficial in the treatment of cancer patients.
Collapse
Affiliation(s)
- Purnima K Wagh
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH 45267-0558, USA
| | | | | |
Collapse
|
19
|
Abstract
Lung cancer causes more deaths than any other malignancy in the developed world. Advances in surgical techniques and chemotherapy/radiotherapy regimes have produced only minimal improvements in long-term survival. New therapeutic interventions are urgently required. Research has indicated that growth factor signaling may be an important novel target in lung cancer therapy. Preclinical studies have demonstrated the role of extracellular growth factors in lung cancer cell proliferation, metastasis, and resistance to cytotoxic therapy, and have elucidated the key molecular components of growth factor-signaling cascades. This has enabled the development of selective growth factor inhibitors, which have been evaluated in clinical trials and are now an accepted component of advanced lung cancer treatment. Further research is underway to improve the efficacy of this growth factor-targeted therapy. This article will outline the important aspects of this translational research indicating the growth factor-signaling pathways identified in lung cancer, clinical trials of anti-growth factor therapy, and potential future research directions.
Collapse
Affiliation(s)
- Philip S Hodkinson
- University of Edinburgh, MRC Centre for Inflammation Research, Queen's Institute of Medical Research, 47 Little France Crescent, Edinburgh EH16 4TJ, Scotland, UK
| | | | | |
Collapse
|
20
|
|
21
|
Thangasamy A, Rogge J, Ammanamanchi S. Regulation of RON tyrosine kinase-mediated invasion of breast cancer cells. J Biol Chem 2007; 283:5335-43. [PMID: 18165235 DOI: 10.1074/jbc.m706957200] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
RON (recepteur d'origine nantais), a tyrosine kinase receptor for macrophage-stimulating protein (MSP) was implicated in tumor progression. However, it was not investigated how this important oncogene is regulated. We show that MSP promotes invasion of MDA MB 231 and MDA MB 468 but not MCF-7 breast cancer cells. Reverse transcription-PCR and Western analysis indicated the expression of RON message and protein, respectively, in MDA MB 231 and MDA MB 468 cells but not in MCF-7 cells. RON expression correlated with Sp1 expression. Initial analysis of a 1.2-kb and 400-bp RON promoter in MDA MB 231 and MDA MB 468 cells suggested the presence of all the necessary regulatory elements within 400 bp from the transcription start site. Site-directed mutagenesis of the 400-bp RON promoter revealed that the overlapping Sp1 sites at-94 (Sp1-3/4) and Sp1 site at -113 (Sp1-5) are essential for RON gene transcription. Electrophoretic mobility shift assays and chromatin immunoprecipitation analysis indicated that Sp1 binding to these sites is required for RON promoter activity. Ectopic Sp1 expression in Sp1 null SL2 cells confirmed the involvement of these Sp1 sites in the regulation of oncogenic RON tyrosine kinase. Treatment of MDA MB 231 cells with mithramycin A, an inhibitor of Sp1 binding, or siRNA knock-down of Sp1 blocked RON gene expression and MSP-mediated invasion of MDA MB 231 cells. This is the first report demonstrating a clear link between Sp1-dependent RON tyrosine kinase expression and invasion of breast carcinoma cells.
Collapse
Affiliation(s)
- Amalraj Thangasamy
- Department of Medicine, Division of Hematology and Medical Oncology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
| | | | | |
Collapse
|
22
|
Rampino T, Soccio G, Gregorini M, Guidetti C, Marasà M, Maggio M, Panichi V, Migliori M, Libetta C, Dal Canton A. Neutralization of macrophage-stimulating protein ameliorates renal injury in anti-thy 1 glomerulonephritis. J Am Soc Nephrol 2007; 18:1486-96. [PMID: 17409315 DOI: 10.1681/asn.2006060680] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Macrophage-stimulating protein (MSP) is a scatter factor that causes cell proliferation and migration, and receptor origin nantaise (RON) is its receptor. RON is expressed in macrophages and mesangial cells, and MSP is produced by renal tubular cells. This study investigated whether MSP/RON participate in the pathogenesis of anti-Thy 1 nephritis, a glomerular disease that is characterized by invasion of circulating monocytes into glomeruli and migration and proliferation of mesangial cells. In vivo, renal function and histopathology were studied in rats that had anti-Thy 1 disease and were untreated and treated with a neutralizing anti-MSP antibody. In vitro, whether monocytes express RON and whether MSP has a chemotactic effect on monocytes were studied. In vivo, in anti-Thy 1 disease, MSP was expressed de novo in glomeruli, and neutralization of MSP attenuated the rise in serum creatinine and proteinuria, stopped glomerular neutrophil and monocyte influx, protected from glomerular injury, and lessened mesangial cell overgrowth. In vitro, unstimulated monocytes did not express RON, but the stimulation with LPS induced de novo RON expression. LPS-stimulated monocytes were attracted by MSP. These results demonstrate a pathogenic role of the MSP/RON system in anti-Thy 1 nephritis.
Collapse
Affiliation(s)
- Teresa Rampino
- Unit of Nephrology, Dialysis and Transplantation, Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo and University of Pavia, Pavia, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Various cytokines and soluble growth factors upon interaction with their membrane receptors are responsible for inducing cellular proliferation, differentiation, movement, and protection from anoikis (a planned suicide activated by normal cells in absence of attachment to neighboring cells or extracellular matrix (EMC)). Among those soluble factors a major position is exerted by hepatocyte growth factor (HGF) together with its receptor MET and macrophage-stimulating protein (MSP) in cooperation with its receptor RON.
Collapse
Affiliation(s)
- Silvia Benvenuti
- Division of Molecular Oncology, Institute for Cancer Research and Treatment (IRCC), University of Turin Medical School, Candiolo (Torino), Italy
| | | |
Collapse
|
24
|
O'Toole JM, Rabenau KE, Burns K, Lu D, Mangalampalli V, Balderes P, Covino N, Bassi R, Prewett M, Gottfredsen KJ, Thobe MN, Cheng Y, Li Y, Hicklin DJ, Zhu Z, Waltz SE, Hayman MJ, Ludwig DL, Pereira DS. Therapeutic implications of a human neutralizing antibody to the macrophage-stimulating protein receptor tyrosine kinase (RON), a c-MET family member. Cancer Res 2006; 66:9162-70. [PMID: 16982759 DOI: 10.1158/0008-5472.can-06-0283] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RON is a member of the c-MET receptor tyrosine kinase family. Like c-MET, RON is expressed by a variety of epithelial-derived tumors and cancer cell lines and it is thought to play a functional role in tumorigenesis. To date, antagonists of RON activity have not been tested in vivo to validate RON as a potential cancer target. In this report, we used an antibody phage display library to generate IMC-41A10, a human immunoglobulin G1 (IgG1) antibody that binds with high affinity (ED50 = 0.15 nmol/L) to RON and effectively blocks interaction with its ligand, macrophage-stimulating protein (MSP; IC50 = 2 nmol/L). We found IMC-41A10 to be a potent inhibitor of receptor and downstream signaling, cell migration, and tumorigenesis. It antagonized MSP-induced phosphorylation of RON, mitogen-activated protein kinase (MAPK), and AKT in several cancer cell lines. In HT-29 colon, NCI-H292 lung, and BXPC-3 pancreatic cancer xenograft tumor models, IMC-41A10 inhibited tumor growth by 50% to 60% as a single agent, and in BXPC-3 xenografts, it led to tumor regressions when combined with Erbitux. Western blot analyses of HT-29 and NCI-H292 xenograft tumors treated with IMC-41A10 revealed a decrease in MAPK phosphorylation compared with control IgG-treated tumors, suggesting that inhibition of MAPK activity may be required for the antitumor activity of IMC-41A10. To our knowledge, this is the first demonstration that a RON antagonist and specifically an inhibitory antibody of RON negatively affects tumorigenesis. Another major contribution of this report is an extensive analysis of RON expression in approximately 100 cancer cell lines and approximately 300 patient tumor samples representing 10 major cancer types. Taken together, our results highlight the potential therapeutic usefulness of RON activity inhibition in human cancers.
Collapse
Affiliation(s)
- Jennifer M O'Toole
- Department of Tumor Biology, ImClone Systems, Inc, New York, NY 10014, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Germano S, Barberis D, Santoro MM, Penengo L, Citri A, Yarden Y, Gaudino G. Geldanamycins trigger a novel Ron degradative pathway, hampering oncogenic signaling. J Biol Chem 2006; 281:21710-21719. [PMID: 16740632 DOI: 10.1074/jbc.m602014200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ron, the tyrosine kinase receptor for macrophage-stimulating protein is responsible for proliferation and migration of cells from different tissues. Ron can acquire oncogenic potential by single point mutations in the kinase domain, and dysregulated Ron signaling has been involved in the development of different human cancers. We have previously shown that ligand-activated Ron recruits the negative regulator c-Cbl, which mediates its ubiquitylation and degradation. Here we report that Ron is ubiquitylated also by the U-box E3 ligase C-terminal Hsc70-interacting protein (CHIP), recruited via chaperone intermediates Hsp90 and Hsc70. Gene silencing shows that CHIP activity is necessary to mediate Ron degradation upon cell treatment with Hsp90 inhibitors geldanamycins. The oncogenic Ron(M1254T) receptor escapes from c-Cbl negative regulation but retains a strong association with CHIP. This constitutively active mutant of Ron displays increased sensitivity to geldanamycins, enhanced physical interaction with Hsp90, and more rapid degradation rate. Cell growth and migration, as well as the transforming potential evoked by Ron(M1254T), are abrogated upon Hsp90 inhibition. These data highlight a novel mechanism for Ron degradation and propose Hsp90 antagonists like geldanamycins as suitable pharmacological agents for therapy of cancers where altered Ron signaling is involved.
Collapse
Affiliation(s)
- Serena Germano
- Department DISCAFF and DFB Center, University of Piemonte Orientale "A. Avogadro," Novara 28100, Italy
| | - Davide Barberis
- Department DISCAFF and DFB Center, University of Piemonte Orientale "A. Avogadro," Novara 28100, Italy
| | - Massimo M Santoro
- Department DISCAFF and DFB Center, University of Piemonte Orientale "A. Avogadro," Novara 28100, Italy
| | - Lorenza Penengo
- Department DISCAFF and DFB Center, University of Piemonte Orientale "A. Avogadro," Novara 28100, Italy
| | - Ami Citri
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yosef Yarden
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Giovanni Gaudino
- Department DISCAFF and DFB Center, University of Piemonte Orientale "A. Avogadro," Novara 28100, Italy.
| |
Collapse
|
26
|
Gunella G, Bardelli C, Amoruso A, Viano I, Balbo P, Brunelleschi S. Macrophage-stimulating protein differently affects human alveolar macrophages from smoker and non-smoker patients: evaluation of respiratory burst, cytokine release and NF-kappaB pathway. Br J Pharmacol 2006; 148:478-89. [PMID: 16633352 PMCID: PMC1751789 DOI: 10.1038/sj.bjp.0706751] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Macrophage activation is a key feature of inflammatory reactions occurring during bacterial infections, immune responses and tissue injury. We previously demonstrated that human macrophages of different origin express the tyrosine kinase receptor recepteur d'origine nantaise, the human receptor for MSP (RON) and produce superoxide anion (O(2)(-)) when challenged with macrophage-stimulating protein (MSP), the endogenous ligand for RON. This study was aimed to evaluate the role of MSP in alveolar macrophages (AM) isolated from healthy volunteers and patients with interstitial lung diseases (sarcoidosis, idiopathic pulmonary fibrosis), either smokers or non-smokers, by evaluating the respiratory burst, cytokine release and nuclear factor-kappa B (NF-kappaB) activation. MSP effects were compared with those induced by known AM stimuli, for example, phorbol myristate acetate, N-formyl-methionyl-leucyl-phenylalanine, lipopolysaccharide.MSP evokes O(2)(-) production, cytokine release and NF-kappaB activation in a concentration-dependent manner. By evaluating the respiratory burst, we demonstrate a significantly increased O(2)(-) production in AM from healthy smokers or smokers with pulmonary fibrosis, as compared to non-smokers, thus suggesting MSP as an enhancer of cigarette smoke toxicity. Besides inducing interleukin-1 beta (IL-1beta) and interleukin-10 (IL-10) production, MSP triggers an enhanced tumor necrosis factor-alpha release, especially in healthy and pulmonary fibrosis smokers. On the contrary, MSP-induced IL-10 release is higher in AM from healthy non-smokers. MSP activates the transcription factor NF-kappaB; this effect is more potent in healthy and fibrosis smokers (2.5-fold increase in p50 subunit translocation). This effect is receptor-mediated, as it is prevented by a monoclonal anti-human MSP antibody. The higher effectiveness of MSP in AM from healthy smokers and patients with pulmonary fibrosis is suggestive of its role in these clinical conditions.
Collapse
Affiliation(s)
- Gabriele Gunella
- Department of Medical Sciences, School of Medicine, University of Piemonte Orientale ‘A. Avogadro', Via Solaroli, 17 – 28100 Novara, Italy
| | - Claudio Bardelli
- Department of Medical Sciences, School of Medicine, University of Piemonte Orientale ‘A. Avogadro', Via Solaroli, 17 – 28100 Novara, Italy
| | - Angela Amoruso
- Department of Medical Sciences, School of Medicine, University of Piemonte Orientale ‘A. Avogadro', Via Solaroli, 17 – 28100 Novara, Italy
| | - Ilario Viano
- Department of Medical Sciences, School of Medicine, University of Piemonte Orientale ‘A. Avogadro', Via Solaroli, 17 – 28100 Novara, Italy
| | - Piero Balbo
- Azienda Ospedaliera ‘Maggiore della Carità', Novara, Italy
| | - Sandra Brunelleschi
- Department of Medical Sciences, School of Medicine, University of Piemonte Orientale ‘A. Avogadro', Via Solaroli, 17 – 28100 Novara, Italy
- IRCAD (Interdisciplinary Research Center on Autoimmune Diseases), University of Piemonte Orientale ‘A. Avogadro', 28100 Novara, Italy
- Author for correspondence:
| |
Collapse
|
27
|
Zalcenstein A, Weisz L, Stambolsky P, Bar J, Rotter V, Oren M. Repression of the MSP/MST-1 gene contributes to the antiapoptotic gain of function of mutant p53. Oncogene 2006; 25:359-69. [PMID: 16170349 DOI: 10.1038/sj.onc.1209061] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Tumor-associated mutant forms of p53 can exert an antiapoptotic gain of function activity, which confers a selective advantage upon tumor cells harboring such mutations. We report that mutant p53 suppresses the expression of the MSP (MST-1/HGFL) gene, encoding the ligand of the receptor tyrosine kinase RON, implicated in a variety of cellular responses. Mutant p53 associates with the MSP gene promoter and represses its transcriptional activity, leading to a decrease in mRNA levels and a subsequent decrease in the levels of secreted MSP protein. Forced downregulation of MSP expression in H1299 cells, derived from a large-cell lung carcinoma, confers increased resistance against etoposide-induced cell death. These antiapoptotic consequences of MSP downregulation seemingly conflict with the well-documented ability of the RON receptor to promote cell survival and tumor progression when aberrantly hyperactive. Yet, they are consistent with the fact that reduced MSP expression was observed in many types of human cancer, including large-cell lung carcinoma. Thus, repression of MSP gene expression by mutant p53 may contribute to oncogenesis in a cell type-specific manner.
Collapse
Affiliation(s)
- A Zalcenstein
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | | | | | | | | | | |
Collapse
|
28
|
Peace BE, Toney-Earley K, Collins MH, Waltz SE. Ron receptor signaling augments mammary tumor formation and metastasis in a murine model of breast cancer. Cancer Res 2005; 65:1285-93. [PMID: 15735014 DOI: 10.1158/0008-5472.can-03-3580] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The tyrosine kinase receptor Ron has been implicated in several types of cancer, including overexpression in human breast cancer. This is the first report describing the effect of Ron signaling on tumorigenesis and metastasis in a mouse model of breast cancer. Mice with a targeted deletion of the Ron tyrosine kinase signaling domain (TK-/-) were crossed to mice expressing the polyoma virus middle T antigen (pMT) under the control of the mouse mammary tumor virus promoter. Both pMT-expressing wild-type control (pMT+/- TK+/+) and pMT+/- TK-/- mice developed mammary tumors and lung metastases. However, a significant decrease in mammary tumor initiation and growth was found in the pMT+/- TK-/- mice compared with controls. An examination of mammary tumors showed that there was a significant decrease in microvessel density, significantly decreased cellular proliferation, and a significant increase in terminal deoxynucleotidyl transferase-mediated nick end labeling-positive staining in mammary tumor cells from the pMT+/- TK-/- mice compared with the pMT+/- TK+/+ mice. Biochemical analyses on mammary tumor lysates showed that whereas both the pMT-expressing TK+/+ and TK-/- tumors have increased Ron expression compared with normal mammary glands, the pMT-expressing TK-/- tumors have deficits in mitogen-activated protein kinase and AKT activation. These results indicate that Ron signaling synergizes with pMT signaling to induce mammary tumor formation, growth, and metastasis. This effect may be mediated in part through the regulation of angiogenesis and through proliferative and cell survival pathways regulated by mitogen-activated protein kinase and AKT.
Collapse
Affiliation(s)
- Belinda E Peace
- Department of Surgery, University of Cincinnati College of Medicine and Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | | | | |
Collapse
|
29
|
Camp ER, Liu W, Fan F, Yang A, Somcio R, Ellis LM. RON, a Tyrosine Kinase Receptor Involved in Tumor Progression and Metastasis. Ann Surg Oncol 2005; 12:273-81. [PMID: 15827676 DOI: 10.1245/aso.2005.08.013] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2004] [Accepted: 11/29/2004] [Indexed: 11/18/2022]
Abstract
Tyrosine kinase receptors mediate many critical cellular functions that contribute to tumor progression and metastasis and thus are potential targets for molecular-based cancer therapy. As has been found for many receptor tyrosine kinases, RON (recepteur d'origine nantais) and its ligand, macrophage-stimulating protein, have recently been implicated in the progression and metastasis of tumors. In in vitro experiments using colon and breast cancer cell lines, overexpression of RON led to increased invasion and migration of cancer cells and prevented apoptosis and anoikis. In addition, transgenic mice engineered to overexpress RON in the lung epithelium developed multiple pulmonary tumors, suggesting a role for RON in tumorigenesis. In human cancer specimens, increased RON expression has been demonstrated in colon, breast, ovarian, and lung tumors. Therefore, therapies designed to inhibit RON activation may hinder critical tumor survival mechanisms and play a role in the treatment of advanced disease.
Collapse
Affiliation(s)
- E Ramsay Camp
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Unit 444, PO Box 301402, Houston, Texas 77230-1402, USA
| | | | | | | | | | | |
Collapse
|
30
|
Conrotto P, Corso S, Gamberini S, Comoglio PM, Giordano S. Interplay between scatter factor receptors and B plexins controls invasive growth. Oncogene 2004; 23:5131-7. [PMID: 15184888 DOI: 10.1038/sj.onc.1207650] [Citation(s) in RCA: 136] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Met and Ron tyrosine kinases are members of the Scatter Factor Receptor family. Met is the receptor for hepatocyte growth factor while Ron is that for macrophage stimulating protein. On ligand stimulation, activation of these receptors induces 'invasive growth', a complex biological response involved in tissue morphogenesis and, when deregulated, in tumor progression and metastasis. Scatter Factor Receptors share structural homology with Plexins, transmembrane receptors for Semaphorins, a family of ligands originally identified as axon guidance molecules. A physical and functional association between Met and Plexin B1, the prototype of class B Plexin subfamily, has been previously demonstrated. Here, we show that both Met and Ron receptors can interact with each of the three members of class B Plexins, even in the absence of their ligands and that Plexin B1 ligand, Sema 4D, can induce activation of Met and Ron receptors, promoting an invasive response. Furthermore, in some human neoplastic cell lines Plexin B1 is overexpressed, constitutively tyrosine phosphorylated, and associated with Scatter Factor Receptors. These data extend the crosstalk previously described between Met and Plexin B1 to the entire families of Scatter Factor Receptors and class B Plexins and show that interaction with multiple upstream activators can finely tune the invasive growth process both in physiological conditions and in tumor growth and metastatization.
Collapse
Affiliation(s)
- Paolo Conrotto
- Division of Molecular Oncology, Institute for Cancer Research and Treatment (IRCC), University of Torino Medical School, I-10060 Candiolo, Torino, Italy
| | | | | | | | | |
Collapse
|
31
|
Tomida S, Koshikawa K, Yatabe Y, Harano T, Ogura N, Mitsudomi T, Some M, Yanagisawa K, Takahashi T, Osada H, Takahashi T. Gene expression-based, individualized outcome prediction for surgically treated lung cancer patients. Oncogene 2004; 23:5360-70. [PMID: 15064725 DOI: 10.1038/sj.onc.1207697] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Individualized outcome prediction classifiers were successfully constructed through expression profiling of a total of 8644 genes in 50 non-small-cell lung cancer (NSCLC) cases, which had been consecutively operated on within a defined short period of time and followed up for more than 5 years. The resultant classifier of NSCLCs yielded 82% accuracy for forecasting survival or death 5 years after surgery of a given patient. In addition, since two major histologic classes may differ in terms of outcome-related expression signatures, histologic-type-specific outcome classifiers were also constructed. The resultant highly predictive classifiers, designed specifically for nonsquamous cell carcinomas, showed a prediction accuracy of more than 90% independent of disease stage. In addition to the presence of heterogeneities in adenocarcinomas, our unsupervised hierarchical clustering analysis revealed for the first time the existence of clinicopathologically relevant subclasses of squamous cell carcinomas with marked differences in their invasive growth and prognosis. This finding clearly suggests that NSCLCs comprise distinct subclasses with considerable heterogeneities even within one histologic type. Overall, these findings should advance not only our understanding of the biology of lung cancer but also our ability to individualize postoperative therapies based on the predicted outcome.
Collapse
Affiliation(s)
- Shuta Tomida
- Division of Molecular Oncology, Aichi Cancer Center Research Institute, Nagoya 464-8681, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Angeloni D, Danilkovitch-Miagkova A, Miagkov A, Leonard EJ, Lerman MI. The soluble sema domain of the RON receptor inhibits macrophage-stimulating protein-induced receptor activation. J Biol Chem 2003; 279:3726-32. [PMID: 14597639 DOI: 10.1074/jbc.m309342200] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
RON is a receptor tyrosine kinase of the MET family that is involved in cell proliferation, cell survival, and cell motility in both normal and disease states. Macrophage-stimulating protein (MSP) is the RON ligand whose binding to RON causes receptor activation. RON is a trans-membrane heterodimer comprised of one alpha- and one beta-chain originating from a single-chain precursor and held together by several disulfide bonds. The intracellular part of RON contains the kinase domain and regulatory elements. The extracellular region is characterized by the presence of a sema domain (a stretch of approximately 500 amino acids with several highly conserved cysteine residues), a PSI (plexin, semaphorins, integrins) domain, and four immunoglobulin-like folds. Here we show that a soluble, secreted molecule representing the sema domain of RON (referred to as ron-sema) has a dominant negative effect on the ligand-induced receptor activation and is capable of inhibiting RON-dependent signaling pathways and cellular responses. Results suggest that the sema domain of RON participates in ligand binding by the full-length receptor. The ability of ron-sema to suppress growth of MSP-responsive cells in culture, including cancer cells, points to a potential therapeutic use of this molecule, and forced expression of it could potentially be used as a gene therapy tool for treating MSP-dependent types of cancer.
Collapse
Affiliation(s)
- Debora Angeloni
- Laboratory of Immunobiology, National Cancer Institute, National Institutes of Health, Frederick, Maryland 21702-1201, USA
| | | | | | | | | |
Collapse
|
33
|
Rampino T, Gregorini M, Soccio G, Maggio M, Rosso R, Malvezzi P, Collesi C, Dal Canton A. The Ron proto-oncogene product is a phenotypic marker of renal oncocytoma. Am J Surg Pathol 2003; 27:779-85. [PMID: 12766581 DOI: 10.1097/00000478-200306000-00008] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The proto-oncogene product Ron is the receptor for macrophage stimulating protein, a scatter factor that stimulates cell proliferation, prevents apoptosis, and induces an invasive cell phenotype. We investigated the expression of Ron, Ki-67 (proliferation index), p53, and bcl-2 (proapoptotic and antiapoptotic proteins, respectively) in 50 renal tumors (19 clear cell carcinomas, 18 oncocytomas, 7 papillary cell carcinomas, 5 chromophobe cell carcinomas, and 1 carcinoma with sarcomatoid areas). In addition, we studied Ron in normal kidney and in the renal carcinoma cell line Caki-1. Immunostaining and Western blot showed Ron in normal kidney and in all oncocytomas but never in renal cell carcinomas or in Caki-1. In addition, Western blot showed that Ron was expressed in phosphorylated, i.e., active, form. Bcl-2 was strongly expressed in oncocytomas, whereas Ki-67 and p53 were much less expressed in oncocytomas than in carcinomas. These results indicate in Ron a marker that differentiates oncocytoma from the other renal epithelial tumors. We therefore think that Ron may prove to be a new tool for a sound and precise diagnosis of oncocytoma, a benign tumor that cannot always be distinguished from carcinomas at histologic examination. The overexpression of bcl-2, but not p53 in oncocytoma, suggests that the MSP/Ron system sustains the growth of oncocytoma by opposing apoptosis.
Collapse
Affiliation(s)
- Teresa Rampino
- Unit of Nephrology, Dialysis and Transplantation, IRCCS Policlinico San Matteo University, Milano, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Wetzel CC, Degen SJF, Waltz SE. Cis-acting elements in the hepatocyte growth factor-like protein gene regulate kidney and liver-specific expression in mice. DNA Cell Biol 2003; 22:293-301. [PMID: 12941157 DOI: 10.1089/104454903322216644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Previous studies from our laboratory demonstrated that the hepatocyte-specific transcriptional activity of the hepatocyte growth factor-like protein/macrophage stimulating protein (HGFL) promoter is modulated in HepG2 cells by the first 135 base pairs (bp) upstream of the HGFL transcriptional start site. Gel mobility shift and transactivation assays demonstrated that hepatocyte nuclear factor-4 (HNF-4) binds to this region and is responsible, in part, for the liver-specific expression of this gene in HepG2 cells. In an attempt to understand the in vivo mechanism regulating the expression of HGFL, a series of transgenic mice were generated that contained four different regions upstream of the HGFL promoter attached to the coding sequences for chloramphenicol acetyltransferase (CAT). Interestingly, upstream promoter sequences, containing as little as 104 bp upstream of the translational start site, were able to drive reporter expression and protein production specifically in kidney and liver tissue. Strikingly, when the first exon and intron of the HGFL gene was inserted downstream of the 135 bp promoter element, only liver-specific expression was observed. These studies indicate that short sequences upstream of HGFL can drive efficient expression in kidney and liver tissue, and that sequences in the first intron of the HGFL gene contain regulatory elements that direct kidney-specific transcriptional repression in vivo and aid in the proper recapitulation of HGFL expression in mice.
Collapse
Affiliation(s)
- Cynthia C Wetzel
- College of Medicine, University of Cincinnati Medical Center, Cincinnati, OH 45267-0555, USA
| | | | | |
Collapse
|
35
|
Angeloni D, Duh FM, Moody M, Dean M, Zabarovsky ER, Sentchenko V, Braga E, Lerman MI. C to A single nucleotide polymorphism in intron 18 of the human MST1R (RON) gene that maps at 3p21.3. Mol Cell Probes 2003; 17:55-7. [PMID: 12788025 DOI: 10.1016/s0890-8508(02)00115-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The MST1R (RON) gene, that maps at 3p21.3, encodes a protein tyrosine kinase receptor comprised of an extra-cellular domain that contains the ligand binding pocket and an intracellular region where the kinase domain is located. It controls cell survival and motility programs related to invasive growth. With the single strand conformation polymorphism (SSCP) method, a C to A nucleotide polymorphism (SNP) was found in intron 18 of the gene. The SNP has a frequency of 0.28 among African-American, 0.25 among Caucasian CEPH and 0.09 among Asian healthy individuals. During these studies, an alternatively spliced cDNA of MST1R, lacking exon 19, was also found that may result from this change.
Collapse
Affiliation(s)
- Debora Angeloni
- Laboratory of Immunobiology, National Cancer Institute at Frederick, Frederick, MD 21702-1201, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Chen YQ, Zhou YQ, Fisher JH, Wang MH. Targeted expression of the receptor tyrosine kinase RON in distal lung epithelial cells results in multiple tumor formation: oncogenic potential of RON in vivo. Oncogene 2002; 21:6382-6. [PMID: 12214279 DOI: 10.1038/sj.onc.1205783] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2001] [Revised: 06/06/2002] [Accepted: 06/18/2002] [Indexed: 11/08/2022]
Abstract
RON, a member of the MET proto-oncogene family, has been implicated in the progression of certain epithelial cancers. The purpose of this study was to determine the oncogenic potential of RON in vivo in lung epithelial cells. Transgenic mice were established using surfactant protein C promoter to express human RON in the distal lung epithelial cells. These mice were born normal but developed multiple lung tumors with distinct morphology and growth patterns. Tumors appeared as a single mass in the lung around 2 months of age and gradually developed into multiple nodules located mostly in the peripheral portions of the lung. A transition from early adenomas to later adenocarcinomas was observed. Morphologically, tumors were characterized as cuboidal epithelial cells with a type II cell phenotype, grew along the alveolar walls, and projected into the alveolar septa. RON was highly expressed and constitutively activated in tumors. These results indicate that overexpression of human wild-type RON causes the formation of lung tumors with unique biological characteristics in vivo. This model provides opportunities to study the role of RON in the pathogenesis of lung tumors and to elucidate the mechanisms underlying this distinct lung tumor.
Collapse
Affiliation(s)
- Yi-Qing Chen
- Division of Pulmonary Sciences and Critical Care, Department of Medicine, University of Colorado School of Medicine, CU Cancer Center, and Denver Health Medical Center, Denver, Colorado, CO 80204, USA
| | | | | | | |
Collapse
|
37
|
Rampino T, Collesi C, Gregorini M, Maggio M, Soccio G, Guallini P, Canton AD. Macrophage-stimulating protein is produced by tubular cells and activates mesangial cells. J Am Soc Nephrol 2002; 13:649-657. [PMID: 11856768 DOI: 10.1681/asn.v133649] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Until now, hepatocytes have been the only known cell source of macrophage-stimulating protein (MSP), and tissue macrophages have been the cells on which the biologic effects of MSP have been proved. To extend the understanding of the biologic meaning of MSP, it was investigated whether MSP operates in the kidney. MSP protein was evaluated by Western blot in supernatant of cultured human tubular cells (HK2) and human mesangial cells (HMC). MSP mRNA was investigated in HK2 by reverse transcription-polymerase chain reaction (RT-PCR). The expression of the MSP receptor, RON, was evaluated in HMC and HK2 by Western blot. RON mRNA was investigated in HMC by RT-PCR. The expression of MSP and RON in normal human renal tissue was studied by immunohistochemistry. HMC were stimulated with recombinant MSP (rMSP) and HK2 supernatant to study cell growth, migration, and the capacity to invade an artificial collagen matrix and synthesize interleukin-6 (IL-6). HK2 produced MSP and expressed RON in a form that was phosphorylated by rMSP. HMC expressed RON but did not produce MSP. MSP in HK2 supernatant and rMSP induced in HMC phosphorylation of RON, growth, migration, invasion, and IL-6 synthesis. In normal human kidney, tubules expressed MSP and RON. These results indicate a novel field of operation for MSP and suggest a pathogenic role of the MSP/RON system in renal disease. In fact, MSP released by tubular cells may recruit monocytes/macrophages in inflammatory tubulointerstitial disorders. In addition, MSP either circulating or as paracrine product may sustain glomerular mesangioproliferative disease.
Collapse
Affiliation(s)
- Teresa Rampino
- *Unit of Nephrology, Dialysis and Transplant, I. R. C. C. S. Policlinico San Matteo and University, Pavia, Italy; and Institute for Cancer Research and Treatment, Torino, Italy
| | - Chiara Collesi
- *Unit of Nephrology, Dialysis and Transplant, I. R. C. C. S. Policlinico San Matteo and University, Pavia, Italy; and Institute for Cancer Research and Treatment, Torino, Italy
| | - Marilena Gregorini
- *Unit of Nephrology, Dialysis and Transplant, I. R. C. C. S. Policlinico San Matteo and University, Pavia, Italy; and Institute for Cancer Research and Treatment, Torino, Italy
| | - Milena Maggio
- *Unit of Nephrology, Dialysis and Transplant, I. R. C. C. S. Policlinico San Matteo and University, Pavia, Italy; and Institute for Cancer Research and Treatment, Torino, Italy
| | - Grazia Soccio
- *Unit of Nephrology, Dialysis and Transplant, I. R. C. C. S. Policlinico San Matteo and University, Pavia, Italy; and Institute for Cancer Research and Treatment, Torino, Italy
| | - Paola Guallini
- *Unit of Nephrology, Dialysis and Transplant, I. R. C. C. S. Policlinico San Matteo and University, Pavia, Italy; and Institute for Cancer Research and Treatment, Torino, Italy
| | - Antonio Dal Canton
- *Unit of Nephrology, Dialysis and Transplant, I. R. C. C. S. Policlinico San Matteo and University, Pavia, Italy; and Institute for Cancer Research and Treatment, Torino, Italy
| |
Collapse
|
38
|
Brunelleschi S, Penengo L, Lavagno L, Santoro C, Colangelo D, Viano I, Gaudino G. Macrophage stimulating protein (MSP) evokes superoxide anion production by human macrophages of different origin. Br J Pharmacol 2001; 134:1285-95. [PMID: 11704649 PMCID: PMC1573047 DOI: 10.1038/sj.bjp.0704356] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. Macrophage Stimulating Protein (MSP), a serum factor related to Hepatocyte Growth Factor, was originally discovered to stimulate chemotaxis of murine resident peritoneal macrophages. MSP is the ligand for Ron, a member of the Met subfamily of tyrosine kinase receptors. The effects of MSP on human macrophages and the role played in human pathophysiology have long been elusive. 2. We show here that human recombinant MSP (hrMSP) evokes a dose-dependent superoxide anion production in human alveolar and peritoneal macrophages as well as in monocyte-derived macrophages, but not in circulating human monocytes. Consistently, the mature Ron protein is expressed by the MSP responsive cells but not by the unresponsive monocytes. The respiratory burst evoked by hrMSP is quantitatively higher than the one induced by N-formylmethionyl-leucyl-phenylalanine and similar to phorbol myristate acetate-evoked one. 3. To investigate the mechanisms involved in NADPH oxidase activation, leading to superoxide anion production, different signal transduction inhibitors were used. By using the non selective tyrosine kinase inhibitor genistein, the selective c-Src inhibitor PP1, the tyrosine phosphatase inhibitor sodium orthovanadate, the phosphatidylinositol 3-kinase inhibitor wortmannin, the p38 inhibitor SB203580, the MEK inhibitor PD098059, we demonstrate that hrMSP-evoked superoxide production is mediated by tyrosine kinase activity, requires the activation of Src but not of PI 3-kinase. We also show that MAP kinase and p38 signalling pathways are involved. 4. These results clearly indicate that hrMSP induces the respiratory burst in human macrophages but not in monocytes, suggesting for the MSP/Ron complex a role of activator as well as of possible marker for human mature macrophages.
Collapse
Affiliation(s)
- S Brunelleschi
- Department of Medical Sciences, University of Piemonte Orientale A. Avogadro, Via Solaroli, 17 - 28100 NOVARA, Italy.
| | | | | | | | | | | | | |
Collapse
|
39
|
Peace BE, Hughes MJ, Degen SJ, Waltz SE. Point mutations and overexpression of Ron induce transformation, tumor formation, and metastasis. Oncogene 2001; 20:6142-51. [PMID: 11593422 DOI: 10.1038/sj.onc.1204836] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2000] [Revised: 07/03/2001] [Accepted: 07/16/2001] [Indexed: 12/14/2022]
Abstract
The receptor tyrosine kinase Ron is a member of the receptor family that includes the proto-oncogene Met and the avian oncogene Sea. The interaction of Ron with its ligand, known as hepatocyte growth factor-like protein (HGFL) or macrophage stimulating protein (MSP), induces crucial cellular responses including invasive growth, proliferation, cell scattering, and branching morphogenesis. Based on the homology and functional similarities between Met and Ron it was hypothesized that Ron may be important in tumor formation and metastasis. To test this hypothesis, wild-type mouse Ron and three mutant forms of Ron containing mutations similar to those found in the Met gene in human hereditary papillary renal carcinoma (HPRC), were expressed in NIH3T3 cells. A transformed phenotype was produced in cell lines expressing either wild-type Ron or the mutated Ron proteins. Further, these cell lines displayed oncogenic potential by exhibiting increased proliferation and constitutive phosphorylation of Ron. These cell lines were also tested for the ability to form solid tumors. Cells expressing wild-type Ron and the three proteins with single amino acid substitutions were highly tumorigenic in vivo. In a model of experimental metastasis, two of the cell lines with altered Ron protein formed highly aggressive tumors in the lungs. These results suggest that Ron may be an aggressive oncogene when either overexpressed or when activated by mutation.
Collapse
Affiliation(s)
- B E Peace
- Division of Developmental Biology, Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | | | | | | |
Collapse
|
40
|
Stella MC, Vercelli A, Repici M, Follenzi A, Comoglio PM. Macrophage stimulating protein is a novel neurotrophic factor. Mol Biol Cell 2001; 12:1341-52. [PMID: 11359926 PMCID: PMC34588 DOI: 10.1091/mbc.12.5.1341] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Macrophage stimulating protein (MSP), also known as hepatocyte growth factor-like, is a soluble cytokine that belongs to the family of the plasminogen-related growth factors (PRGFs). PRGFs are alpha/beta heterodimers that bind to transmembrane tyrosine kinase receptors. MSP was originally isolated as a chemotactic factor for peritoneal macrophages. Through binding to its receptor, encoded by the RON gene, it stimulates dissociation of epithelia and works as an inflammatory mediator by repressing the production of nitric oxide (NO). Here, we identify a novel role for MSP in the central nervous system. As a paradigm to analyze this function we chose the hypoglossal system of adult mice. We demonstrate in vivo that either administration of exogenous MSP or transplantation of MSP-producing cells at the proximal stump of the resected nerve is sufficient to prevent motoneuron atrophy upon axotomy. We also show that the MSP gene is expressed in the tongue, the target of the hypoglossal nerve, and that MSP induces biosynthesis of Ron receptor in the motoneuron somata. Finally, we show that MSP suppresses NO production in the injured hypoglossal nuclei. Together, these data suggest that MSP is a novel neurotrophic factor for cranial motoneurons and, by regulating the production of NO, may have a role in brain plasticity and regeneration.
Collapse
Affiliation(s)
- M C Stella
- Institute for Cancer Research and Treatment, IRCC, University of Torino Medical School, 10060 Candiolo, Torino, Italy.
| | | | | | | | | |
Collapse
|
41
|
Follenzi A, Bakovic S, Gual P, Stella MC, Longati P, Comoglio PM. Cross-talk between the proto-oncogenes Met and Ron. Oncogene 2000; 19:3041-9. [PMID: 10871856 DOI: 10.1038/sj.onc.1203620] [Citation(s) in RCA: 150] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Scatter Factors control a complex genetic program known as 'invasive growth'. HGF (Scatter factor 1) and MSP (Scatter Factor 2) bind to tyrosine kinase receptors encoded by the proto-oncogenes MET and RON. Using the appropriate 'kinase inactive' mutant receptors, we show that ligand-induced activation of Met results in transphosphorylation of Ron, and vice versa. Transphosphorylation is direct, as it occurs in Met or Ron receptors lacking the docking sites for signal transducers. Phosphate groups are transferred to the tyrosine phosphorylation sites responsible both for kinase up-regulation (Met: Y1234/Y1235 and Ron: Y1238/Y1239) and for generation of signal transducer docking sites (Met: Y1349/Y1356 and Ron Y1353/Y1360). The transphosphorylation specifically takes place for the receptor subfamily, as it is not observed between Met or Ron and ErbB1, ErbB2 or TrkA. Cross-linking experiments show that non-covalent Met-Ron complexes are present on the cell surface, before ligand-induced dimerization. Co-expression of a kinase inactive Ron receptor with naturally-occurring oncogenic Met mutants suppresses the transforming phenotype, suggesting a dominant negative role for the inefficient kinase partner. These data show that, while specific for their ligands, scatter factor receptors cross-talk and cooperate in intracellular signaling.
Collapse
Affiliation(s)
- A Follenzi
- Institute for Cancer Research and Treatment (IRCC), University of Torino, School of Medicine 10060, Candiolo, Italy
| | | | | | | | | | | |
Collapse
|
42
|
Affiliation(s)
- E J Leonard
- Laboratory of Immunobiology, NCI-Frederick Cancer Research and Development Center, Maryland 21702, USA
| | | |
Collapse
|
43
|
Rozengurt E. Autocrine loops, signal transduction, and cell cycle abnormalities in the molecular biology of lung cancer. Curr Opin Oncol 1999; 11:116-22. [PMID: 10188076 DOI: 10.1097/00001622-199903000-00007] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Lung cancers produce a variety of mitogenic neuropeptides and growth factors and express receptors for these signaling peptides leading to autocrine and paracrine loops that stimulate tumor cell proliferation and migration and promote neovascularization. The effects of these autocrine and paracrine factors are mediated by a variety of intracellular signal transduction pathways that ultimately converge on the regulation of cell cycle proteins whose expression and activity are abnormal in lung cancer cells. During the past year, further advances have been made in unraveling autocrine loops, signal transduction pathways, and cell cycle abnormalities in both small cell lung carcinoma (SCLC) and in non-SCLC. In addition, the hunt for novel tumor suppressor genes has continued. As our understanding of the fundamental cell and molecular biology of lung cancer increases, novel possibilities for translational research are emerging for improving the diagnosis, prognosis, and treatment of the disease.
Collapse
Affiliation(s)
- E Rozengurt
- Department of Medicine, University of California, Los Angeles 90095-1786, USA
| |
Collapse
|