1
|
El Bezawy R, Percio S, Ciniselli CM, De Cesare M, Colella G, Dugo M, Veneroni S, Doldi V, Martini S, Baratti D, Kusamura S, Verderio P, Deraco M, Gandellini P, Zaffaroni N, Zuco V. miR-550a-3p is a prognostic biomarker and exerts tumor-suppressive functions by targeting HSP90AA1 in diffuse malignant peritoneal mesothelioma. Cancer Gene Ther 2022; 29:1394-1404. [PMID: 35352023 PMCID: PMC9576593 DOI: 10.1038/s41417-022-00460-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/10/2022] [Indexed: 11/08/2022]
Abstract
Diffuse malignant peritoneal mesothelioma (DMPM) is a rare and rapidly lethal tumor, poorly responsive to conventional treatments. In this regards, the identification of molecular alterations underlying DMPM onset and progression might be exploited to develop novel therapeutic strategies. Here, we focused on miR-550a-3p, which we found downregulated in 45 DMPM clinical samples compared to normal tissues and whose expression levels were associated with patient outcome. Through a gain-of-function approach using miRNA mimics in 3 DMPM cell lines, we demonstrated the tumor-suppressive role of miR-550a-3p. Specifically, miRNA ectopic expression impaired cell proliferation and invasiveness, enhanced the apoptotic response, and reduced the growth of DMPM xenografts in mice. Antiproliferative and proapoptotic effects were also observed in prostate and ovarian cancer cell lines following miR-550a-3p ectopic expression. miR-550a-3p effects were mediated, at least in part, by the direct inhibition of HSP90AA1 and the consequent downregulation of its target proteins, the levels of which were rescued upon disruption of miRNA-HSP90AA1 mRNA pairing, partially abrogating miR-550a-3p-induced cellular effects. Our results show that miR-550a-3p reconstitution affects several tumor traits, thus suggesting this approach as a potential novel therapeutic strategy for DMPM.
Collapse
Affiliation(s)
- Rihan El Bezawy
- Molecular Pharmacology Unit, Fondazione IRCSS Istituto Nazionale dei Tumori, 20133, Milan, Italy
| | - Stefano Percio
- Molecular Pharmacology Unit, Fondazione IRCSS Istituto Nazionale dei Tumori, 20133, Milan, Italy
| | - Chiara Maura Ciniselli
- Bioinformatics and Biostatistics Unit, Fondazione IRCSS Istituto Nazionale dei Tumori, 20133, Milan, Italy
| | - Michelandrea De Cesare
- Molecular Pharmacology Unit, Fondazione IRCSS Istituto Nazionale dei Tumori, 20133, Milan, Italy
| | - Gennaro Colella
- Molecular Pharmacology Unit, Fondazione IRCSS Istituto Nazionale dei Tumori, 20133, Milan, Italy
| | - Matteo Dugo
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, 20132, Milan, Italy
| | - Silvia Veneroni
- Biomarkers Unit, Fondazione IRCSS Istituto Nazionale dei Tumori, 20133, Milan, Italy
| | - Valentina Doldi
- Molecular Pharmacology Unit, Fondazione IRCSS Istituto Nazionale dei Tumori, 20133, Milan, Italy
| | - Silvia Martini
- Molecular Pharmacology Unit, Fondazione IRCSS Istituto Nazionale dei Tumori, 20133, Milan, Italy
| | - Dario Baratti
- Peritoneal Surface Malignancies Unit, Fondazione IRCSS Istituto Nazionale dei Tumori, 20133, Milan, Italy
| | - Shigeki Kusamura
- Peritoneal Surface Malignancies Unit, Fondazione IRCSS Istituto Nazionale dei Tumori, 20133, Milan, Italy
| | - Paolo Verderio
- Bioinformatics and Biostatistics Unit, Fondazione IRCSS Istituto Nazionale dei Tumori, 20133, Milan, Italy
| | - Marcello Deraco
- Peritoneal Surface Malignancies Unit, Fondazione IRCSS Istituto Nazionale dei Tumori, 20133, Milan, Italy
| | - Paolo Gandellini
- Department of Biosciences, University of Milan, 20133, Milan, Italy
| | - Nadia Zaffaroni
- Molecular Pharmacology Unit, Fondazione IRCSS Istituto Nazionale dei Tumori, 20133, Milan, Italy.
| | - Valentina Zuco
- Molecular Pharmacology Unit, Fondazione IRCSS Istituto Nazionale dei Tumori, 20133, Milan, Italy
| |
Collapse
|
2
|
Zhao J, Luo Z. Discovery of Raf Family Is a Milestone in Deciphering the Ras-Mediated Intracellular Signaling Pathway. Int J Mol Sci 2022; 23:ijms23095158. [PMID: 35563547 PMCID: PMC9101324 DOI: 10.3390/ijms23095158] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 01/27/2023] Open
Abstract
The Ras-Raf-MEK-ERK signaling pathway, the first well-established MAPK pathway, plays essential roles in cell proliferation, survival, differentiation and development. It is activated in over 40% of human cancers owing to mutations of Ras, membrane receptor tyrosine kinases and other oncogenes. The Raf family consists of three isoforms, A-Raf, B-Raf and C-Raf. Since the first discovery of a truncated mutant of C-Raf as a transforming oncogene carried by a murine retrovirus, forty years of extensive studies have provided a wealth of information on the mechanisms underlying the activation, regulation and biological functions of the Raf family. However, the mechanisms by which activation of A-Raf and C-Raf is accomplished are still not completely understood. In contrast, B-Raf can be easily activated by binding of Ras-GTP, followed by cis-autophosphorylation of the activation loop, which accounts for the fact that this isoform is frequently mutated in many cancers, especially melanoma. The identification of oncogenic B-Raf mutations has led to accelerated drug development that targets Raf signaling in cancer. However, the effort has not proved as effective as anticipated, inasmuch as the mechanism of Raf activation involves multiple steps, factors and phosphorylation of different sites, as well as complex interactions between Raf isoforms. In this review, we will focus on the physiological complexity of the regulation of Raf kinases and their connection to the ERK phosphorylation cascade and then discuss the role of Raf in tumorigenesis and the clinical application of Raf inhibitors in the treatment of cancer.
Collapse
Affiliation(s)
- Jingtong Zhao
- Queen Mary School, Nanchang University, Nanchang 330031, China;
| | - Zhijun Luo
- Queen Mary School, Nanchang University, Nanchang 330031, China;
- Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University, Nanchang 330031, China
- NCU-QMUL Joint Research Institute of Precision Medical Science, Nanchang 330031, China
- Correspondence:
| |
Collapse
|
3
|
Recent Trends of microRNA Significance in Pediatric Population Glioblastoma and Current Knowledge of Micro RNA Function in Glioblastoma Multiforme. Int J Mol Sci 2020; 21:ijms21093046. [PMID: 32349263 PMCID: PMC7246719 DOI: 10.3390/ijms21093046] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Central nervous system tumors are a significant problem for modern medicine because of their location. The explanation of the importance of microRNA (miRNA) in the development of cancerous changes plays an important role in this respect. The first papers describing the presence of miRNA were published in the 1990s. The role of miRNA has been pointed out in many medical conditions such as kidney disease, diabetes, neurodegenerative disorder, arthritis and cancer. There are several miRNAs responsible for invasiveness, apoptosis, resistance to treatment, angiogenesis, proliferation and immunology, and many others. The research conducted in recent years analyzing this group of tumors has shown the important role of miRNA in the course of gliomagenesis. These particles seem to participate in many stages of the development of cancer processes, such as proliferation, angiogenesis, regulation of apoptosis or cell resistance to cytostatics.
Collapse
|
4
|
TAKAHASHI C, YAZAKI T, SUGIYAMA N, ISHIHAMA Y. Selected Reaction Monitoring of Kinase Activity-Targeted Phosphopeptides. CHROMATOGRAPHY 2019. [DOI: 10.15583/jpchrom.2019.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
| | - Tatsuya YAZAKI
- Graduate School of Pharmaceutical Science, Kyoto University
| | | | | |
Collapse
|
5
|
Li H, Yu Y, Zhao Y, Wu D, Yu X, Lu J, Chen Z, Zhang H, Hu Y, Zhai Y, Su J, Aheman A, De Las Casas A, Jin J, Xu X, Shi Z, Woodfield SE, Vasudevan SA, Agarwal S, Yan Y, Yang J, Foster JH. Small molecule inhibitor agerafenib effectively suppresses neuroblastoma tumor growth in mouse models via inhibiting ERK MAPK signaling. Cancer Lett 2019; 457:129-141. [PMID: 31100410 DOI: 10.1016/j.canlet.2019.05.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Revised: 05/02/2019] [Accepted: 05/08/2019] [Indexed: 12/19/2022]
Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor in early childhood. Despite intensive multimodal therapy, nearly half of children with high-risk disease will relapse with therapy-resistant tumors. Dysregulation of MAPK pathway has been implicated in the pathogenesis of relapsed and refractory NB patients, which underscores the possibility of targeting MAPK signaling cascade as a novel therapeutic strategy. In this study, we found that high expressions of RAF family kinases correlated with advanced tumor stage, high-risk disease, tumor progression, and poor overall survival. Targeted inhibition of RAF family kinases with the novel small molecule inhibitor agerafenib abrogated the activation of ERK MAPK pathway in NB cells. Agerafenib significantly inhibited the cell proliferation and colony formation ability of NB cells in vitro, and its combination with traditional chemotherapy showed a synergistic pro-apoptotic effect. More importantly, agerafenib exhibited a favorable toxicity profile, potently suppressed tumor growth, and prolonged survival in NB mouse models. In conclusion, our preclinical data suggest that agerafenib might be an effective therapeutic agent for NB treatment, both as a single-agent and in combination with chemotherapy.
Collapse
Affiliation(s)
- Hui Li
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA; Department of Cardiothoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, PR China
| | - Yang Yu
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yanling Zhao
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Deanna Wu
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xiaoman Yu
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jiaxiong Lu
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Zhenghu Chen
- Division of Pediatric Surgery, Texas Children's Hospital Department of Surgery, Michael E. DeBakey Department of Surgery, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Huiyuan Zhang
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yongguang Hu
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yuanfen Zhai
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jun Su
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ayinuer Aheman
- Division of Pediatric Surgery, Texas Children's Hospital Department of Surgery, Michael E. DeBakey Department of Surgery, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Augusto De Las Casas
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jingling Jin
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xin Xu
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Zhongcheng Shi
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Sarah E Woodfield
- Division of Pediatric Surgery, Texas Children's Hospital Department of Surgery, Michael E. DeBakey Department of Surgery, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Sanjeev A Vasudevan
- Division of Pediatric Surgery, Texas Children's Hospital Department of Surgery, Michael E. DeBakey Department of Surgery, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Saurabh Agarwal
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yusheng Yan
- Department of Cardiothoracic Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, PR China
| | - Jianhua Yang
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Jennifer H Foster
- Texas Children's Cancer Center, Department of Pediatrics, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
6
|
Kim G, Bhattarai PY, Choi HS. Peptidyl-prolyl cis/trans isomerase NIMA-interacting 1 as a molecular target in breast cancer: a therapeutic perspective of gynecological cancer. Arch Pharm Res 2019; 42:128-139. [PMID: 30684192 DOI: 10.1007/s12272-019-01122-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 01/16/2019] [Indexed: 12/11/2022]
Abstract
Peptidyl-prolyl cis/trans isomerase NIMA-interacting 1 (PIN1) induces conformational and functional changes to numerous key signaling molecules following proline-directed phosphorylation and its deregulation contributes to disease, particularly cancer. PIN1 is overexpressed in breast cancer, promoting cell proliferation and transformation in collaboration with several oncogenic signaling pathways, and is correlated with a poor clinical outcome. PIN1 level is also increased in certain gynecological cancers such as cervical, ovarian, and endometrial cancers. Although women with breast cancer are at risk of developing a second primary gynecological malignancy, particularly of the endometrium and ovary, the common oncogenic signaling pathway mediated by PIN1 has not been noted to date. This review discusses the roles of PIN1 in breast tumorigenesis and gynecological cancer progression, as well as the clinical effect of targeting this enzyme in breast and gynecological cancers.
Collapse
Affiliation(s)
- Garam Kim
- College of Pharmacy, Chosun University, 309 Philmundaero, Dong-gu, Gwangju, 61452, Republic of Korea
| | - Poshan Yugal Bhattarai
- College of Pharmacy, Chosun University, 309 Philmundaero, Dong-gu, Gwangju, 61452, Republic of Korea
| | - Hong Seok Choi
- College of Pharmacy, Chosun University, 309 Philmundaero, Dong-gu, Gwangju, 61452, Republic of Korea.
| |
Collapse
|
7
|
Prediction of sorafenib treatment–related gene expression for hepatocellular carcinoma: preoperative MRI and histopathological correlation. Eur Radiol 2018; 29:2272-2282. [DOI: 10.1007/s00330-018-5882-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/19/2018] [Accepted: 11/09/2018] [Indexed: 01/15/2023]
|
8
|
Lee S, Wottrich S, Bonavida B. Crosstalks between Raf-kinase inhibitor protein and cancer stem cell transcription factors (Oct4, KLF4, Sox2, Nanog). Tumour Biol 2017; 39:1010428317692253. [PMID: 28378634 DOI: 10.1177/1010428317692253] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Raf-kinase inhibitor protein has been reported to inhibit both the Raf/mitogen extracellular signal-regulated kinase/extracellular signal-regulated kinase and nuclear factor kappa-light-chain of activated B cells pathways. It has also been reported in cancers that Raf-kinase inhibitor protein behaves as a metastatic suppressor as well as a chemo-immunosensitizing factor to drug/immune-mediated apoptosis. The majority of cancers exhibit low or no levels of Raf-kinase inhibitor protein. Hence, the activities of Raf-kinase inhibitor protein contrast, in part, to those mediated by several cancer stem cell transcription factors for their roles in resistance and metastasis. In this review, the existence of crosstalks in the signaling pathways between Raf-kinase inhibitor protein and several cancer stem cell transcription factors (Oct4, KLF4, Sox2 and Nanog) was assembled. Oct4 is induced by Lin28, and Raf-kinase inhibitor protein inhibits the microRNA binding protein Lin28. The expression of Raf-kinase inhibitor protein inversely correlates with the expression of Oct4. KLF4 does not interact directly with Raf-kinase inhibitor protein, but rather interacts indirectly via Raf-kinase inhibitor protein's regulation of the Oct4/Sox2/KLF4 complex through the mitogen-activated protein kinase pathway. The mechanism by which Raf-kinase inhibitor protein inhibits Sox2 is via the inhibition of the mitogen-activated protein kinase pathway by Raf-kinase inhibitor protein. Thus, Raf-kinase inhibitor protein's relationship with Sox2 is via its regulation of Oct4. Inhibition of extracellular signal-regulated kinase by Raf-kinase inhibitor protein results in the upregulation of Nanog. The inhibition of Oct4 by Raf-kinase inhibitor protein results in the failure of the heterodimer formation of Oct4 and Sox2 that is necessary to bind to the Nanog promoter for the transcription of Nanog. The findings revealed that there exists a direct correlation between the expression of Raf-kinase inhibitor protein and the expression of each of the above transcription factors. Based on these analyses, we suggest that the expression level of Raf-kinase inhibitor protein may be involved in the regulation of the cancer stem cell phenotype.
Collapse
Affiliation(s)
- SoHyun Lee
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Stephanie Wottrich
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Benjamin Bonavida
- Department of Microbiology, Immunology & Molecular Genetics, David Geffen School of Medicine, Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
9
|
Multiplexing PKA and ERK1&2 kinases FRET biosensors in living cells using single excitation wavelength dual colour FLIM. Sci Rep 2017; 7:41026. [PMID: 28106114 PMCID: PMC5247693 DOI: 10.1038/srep41026] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 12/14/2016] [Indexed: 02/04/2023] Open
Abstract
Monitoring of different signalling enzymes in a single assay using multiplex biosensing provides a multidimensional workspace to elucidate biological processes, signalling pathway crosstalk, and determine precise sequence of events at the single living cell level. In this study, we interrogate the complexity in cAMP/PKA-MAPK/ERK1&2 crosstalk by using multi-parameter biosensing experiments to correlate biochemical activities simultaneously in time and space. Using a single excitation wavelength dual colour FLIM method we are able to detect fluorescence lifetime images of two donors to simultaneously measure PKA and ERK1&2 kinase activities in the same cellular localization by using FRET biosensors. To this end, we excite two FRET donors mTFP1 and LSSmOrange with a 440 nm wavelength and we alleviate spectral bleed-through associated limitations with the very dim-fluorescent acceptor ShadowG for mTFP1 and the red-shifted mKate2 for LSSmOrange. The simultaneous recording of PKA and ERK1&2 kinase activities reveals concomitant EGF-mediated activations of both kinases in HeLa cells. Under these conditions the subsequent Forskolin-induced cAMP release reverses the transient increase of EGF-mediated ERK1&2 kinase activity while reinforcing PKA activation. Here we propose a validated methodology for multiparametric kinase biosensing in living cells using FRET-FLIM.
Collapse
|
10
|
Rose JC, Huang PS, Camp ND, Ye J, Leidal AM, Goreshnik I, Trevillian BM, Dickinson MS, Cunningham-Bryant D, Debnath J, Baker D, Wolf-Yadlin A, Maly DJ. A computationally engineered RAS rheostat reveals RAS-ERK signaling dynamics. Nat Chem Biol 2016; 13:119-126. [PMID: 27870838 PMCID: PMC5161653 DOI: 10.1038/nchembio.2244] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 09/08/2016] [Indexed: 01/07/2023]
Abstract
Synthetic protein switches controlled with user-defined inputs are powerful tools for studying and controlling dynamic cellular processes. To date, these approaches have relied primarily on intermolecular regulation. Here, we report a computationally-guided framework for engineering intramolecular regulation of protein function. We utilize this framework to develop Chemically Inducible Activator of RAS (CIAR), a single-component RAS rheostat that directly activates endogenous RAS in response to a small molecule. Using CIAR, we show that direct RAS activation elicits markedly different RAS/ERK signaling dynamics compared to growth factor stimulation, and that these dynamics differ between cell types. We also found that the clinically-approved RAF inhibitor vemurafenib potently primes cells to respond to direct wild-type RAS activation. These results demonstrate the utility of CIAR for quantitatively interrogating RAS signaling. Finally, we demonstrate the general utility of our approach to design intramolecularly-regulated protein tools by applying this methodology to the Rho Family GEFs.
Collapse
Affiliation(s)
- John C Rose
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | - Po-Ssu Huang
- Department of Biochemistry, University of Washington, Seattle, Washington, USA.,Institute for Protein Design, University of Washington, Seattle, Washington, USA
| | - Nathan D Camp
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | - Jordan Ye
- Department of Pathology and Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA
| | - Andrew M Leidal
- Department of Pathology and Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA
| | - Inna Goreshnik
- Department of Biochemistry, University of Washington, Seattle, Washington, USA
| | | | - Miles S Dickinson
- Department of Chemistry, University of Washington, Seattle, Washington, USA
| | | | - Jayanta Debnath
- Department of Pathology and Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, Washington, USA.,Institute for Protein Design, University of Washington, Seattle, Washington, USA.,Howard Hughes Medical Institute, University of Washington, Seattle, Washington, USA
| | | | - Dustin J Maly
- Department of Chemistry, University of Washington, Seattle, Washington, USA.,Department of Biochemistry, University of Washington, Seattle, Washington, USA
| |
Collapse
|
11
|
Faridi U, Dhawan SS, Pal S, Gupta S, Shukla AK, Darokar MP, Sharma A, Shasany AK. Repurposing L-Menthol for Systems Medicine and Cancer Therapeutics? L-Menthol Induces Apoptosis through Caspase 10 and by Suppressing HSP90. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2016; 20:53-64. [PMID: 26760959 DOI: 10.1089/omi.2015.0118] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The objective of the present study was to repurpose L-menthol, which is frequently used in oral health and topical formulations, for cancer therapeutics. In this article, we argue that monoterpenes such as L-menthol might offer veritable potentials in systems medicine, for example, as cheaper anti-cancer compounds. Other monoterpenes such as limonene, perillyl alcohol, and geraniol have been shown to induce apoptosis in various cancer cell lines, but their mechanisms of action are yet to be completely elucidated. Earlier, we showed that L-menthol modulates tubulin polymerization and apoptosis to inhibit cancer cell proliferation. In the present report, we used an apoptosis-related gene microarray in conjunction with proteomics analyses, as well as in silico interpretations, to study gene expression modulation in human adenocarcinoma Caco-2 cell line in response to L-menthol treatment. The microarray analysis identified caspase 10 as the important initiator caspase, instead of caspase 8. The proteomics analyses showed downregulation of HSP90 protein (also corroborated by its low transcript abundance), which in turn indicated inhibition of AKT-mediated survival pathway, release of pro-apoptotic factor BAD from BAD and BCLxL complex, besides regulation of other factors related to apoptosis. Based on the combined microarray, proteomics, and in silico data, a signaling pathway for L-menthol-induced apoptosis is being presented for the first time here. These data and literature analysis have significant implications for "repurposing" L-menthol beyond oral medicine, and in understanding the mode of action of plant-derived monoterpenes towards development of cheaper anticancer drugs in future.
Collapse
Affiliation(s)
- Uzma Faridi
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Sunita S Dhawan
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Shaifali Pal
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Sanchita Gupta
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Ashutosh K Shukla
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Mahendra P Darokar
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Ashok Sharma
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| | - Ajit K Shasany
- Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants , Lucknow, U.P., India
| |
Collapse
|
12
|
Shionoya T, Usuki T, Komenoi S, Isozaki T, Sakai H, Sakane F. Distinct expression and localization of the type II diacylglycerol kinase isozymes δ, η and κ in the mouse reproductive organs. BMC DEVELOPMENTAL BIOLOGY 2015; 15:6. [PMID: 25613821 PMCID: PMC4308931 DOI: 10.1186/s12861-015-0055-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 01/15/2015] [Indexed: 12/30/2022]
Abstract
BACKGROUND We have revealed that the type II diacylglycerol kinases (DGKs) δ, η and κ were expressed in the testis and ovary. However, these enzymes' functions in the reproductive organs remain unknown. RESULTS In this study, we first identified the expression sites of type II DGKs in the mouse reproductive organs in detail. Reverse transcription-polymerase chain reaction and Western blotting confirmed that DGKδ2 (splicing variant 2) but not DGKδ1 (splicing variant 1) and DGKκ were expressed in the testis, ovary and uterus. DGKη1 (splicing variant 1) but not DGKη2 (splicing variant 2) was strongly detected in the ovary and uterus. Interestingly, we found that a new alternative splicing product of the DGKη gene, DGKη3, which lacks exon 26 encoding 31 amino acid residues, was expressed only in the testis. Moreover, we investigated the distribution of type II DGKs in the testis, ovary and uterus through in situ hybridization. DGKδ2 was distributed in the primary spermatocytes of the testis and ovarian follicles. DGKη1 was distributed in the oviductal epithelium of the ovary and the luminal epithelium of the uterus. Intriguingly, DGKη3 was strongly expressed in the secondary spermatocytes and round spermatids of the testis. DGKκ was distributed in the primary and secondary spermatocyte of the testis. CONCLUSION These results indicate that the expression patterns of the type II DGK isoforms δ2, η1, η3 and κ differ from each other, suggesting that these DGK isoforms play specific roles in distinct compartments and developmental stages of the reproductive organs, especially in the processes of spermatogenesis and oocyte maturation.
Collapse
Affiliation(s)
- Takao Shionoya
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan.
| | - Takako Usuki
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan.
| | - Suguru Komenoi
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan.
| | - Takeshi Isozaki
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan.
| | - Hiromichi Sakai
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan.
| | - Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan.
| |
Collapse
|
13
|
Liu Z, Liu Y, Li L, Xu Z, Bi B, Wang Y, Li JY. MiR-7-5p is frequently downregulated in glioblastoma microvasculature and inhibits vascular endothelial cell proliferation by targeting RAF1. Tumour Biol 2014; 35:10177-84. [PMID: 25027403 DOI: 10.1007/s13277-014-2318-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 07/04/2014] [Indexed: 01/15/2023] Open
Abstract
The aberrant expression of microRNAs (miRNAs) is always associated with tumor development and progression. Microvascular proliferation is one of the unique pathologic features of glioblastoma (GBM) . In this study, the microvasculature from GBM or normal brain tissue derived from neurosurgeries was purified and total RNA was isolated from purified microvasculature. The difference of miRNA expression profiles between glioblastoma microvasculature and normal brain capillaries was investigated. It was found that miR-7-5p in GBM microvessels was significantly reduced compared with that in normal brain capillaries. In the in vitro experiments, overexpression of miR-7-5p significantly inhibited human umbilical vein endothelial cell proliferation. Forced expression of miR-7-5p in human umbilical vein endothelial cells in vitro significantly reduced the protein level of RAF1 and repressed the activity of the luciferase, a reporter vector carrying the 3'-untranslated region of RAF1. These findings indicate that RAF1 is one of the miR-7-5p target genes. Furthermore, a significant inverse correlation between miR-7-5p expression and RAF1 protein level in GBM microvasculature was found. These data suggest that miR-7-5p functions as a tumor suppressor gene to regulate GBM microvascular endothelial cell proliferation potentially by targeting the RAF1 oncogene, implicating an important role for miR-7-5p in the pathogenesis of GBM. It may serve as a guide for the antitumor angiogenesis drug development.
Collapse
Affiliation(s)
- Zhiguo Liu
- Department of Neurosurgery, Qilu Hospital of Shandong University, Brain Science Research Institute of Shandong University, Jinan, Shandong, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
14
|
Tilli TM, Bellahcène A, Castronovo V, Gimba ERP. Changes in the transcriptional profile in response to overexpression of the osteopontin-c splice isoform in ovarian (OvCar-3) and prostate (PC-3) cancer cell lines. BMC Cancer 2014; 14:433. [PMID: 24928374 PMCID: PMC4075779 DOI: 10.1186/1471-2407-14-433] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 05/23/2014] [Indexed: 12/16/2022] Open
Abstract
Background Especially in human tumor cells, the osteopontin (OPN) primary transcript is subject to alternative splicing, generating three isoforms termed OPNa, OPNb and OPNc. We previously demonstrated that the OPNc splice variant activates several aspects of the progression of ovarian and prostate cancers. The goal of the present study was to develop cell line models to determine the impact of OPNc overexpression on main cancer signaling pathways and thus obtain insights into the mechanisms of OPNc pro-tumorigenic roles. Methods Human ovarian and prostate cancer cell lines, OvCar-3 and PC-3 cells, respectively, were stably transfected to overexpress OPNc. Transcriptomic profiling was performed on these cells and compared to controls, to identify OPNc overexpression-dependent changes in gene expression levels and pathways by qRT-PCR analyses. Results Among 84 genes tested by using a multiplex real-time PCR Cancer Pathway Array approach, 34 and 16, respectively, were differentially expressed between OvCar-3 and PC-3 OPNc-overexpressing cells in relation to control clones. Differentially expressed genes are included in all main hallmarks of cancer, and several interacting proteins have been identified using an interactome network analysis. Based on marked up-regulation of Vegfa transcript in response to OPNc overexpression, we partially validated the array data by demonstrating that conditioned medium (CM) secreted from OvCar-3 and PC-3 OPNc-overexpressing cells significantly induced endothelial cell adhesion, proliferation and migration, compared to CM secreted from control cells. Conclusions Overall, the present study elucidated transcriptional changes of OvCar-3 and PC-3 cancer cell lines in response to OPNc overexpression, which provides an assessment for predicting the molecular mechanisms by which this splice variant promotes tumor progression features.
Collapse
Affiliation(s)
| | | | | | - Etel R P Gimba
- Coordenação de Pesquisa, Programa de Carcinogênese Molecular, Instituto Nacional de Câncer (INCa)/Programa de Pós Graduação Stricto Sensu em Oncologia do INCa, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
15
|
Li W, Chen L, Li X, Jia X, Feng C, Zhang L, He W, Lv J, He Y, Li W, Qu X, Zhou Y, Shi Y. Cancer-related marketing centrality motifs acting as pivot units in the human signaling network and mediating cross-talk between biological pathways. MOLECULAR BIOSYSTEMS 2013; 9:3026-35. [DOI: 10.1039/c3mb70289h] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
16
|
Ovarian cancer: opportunity for targeted therapy. JOURNAL OF ONCOLOGY 2011; 2012:682480. [PMID: 22235203 PMCID: PMC3253450 DOI: 10.1155/2012/682480] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 09/10/2011] [Accepted: 11/01/2011] [Indexed: 11/30/2022]
Abstract
Ovarian cancer is a common cause of cancer mortality in women with limited treatment effectiveness in advanced stages. The limitation to treatment is largely the result of high rates of cancer recurrence despite chemotherapy and eventual resistance to existing chemotherapeutic agents. The objective of this paper is to review current concepts of ovarian carcinogenesis. We will review existing hypotheses of tumor origin from ovarian epithelial cells, Fallopian tube, and endometrium. We will also review the molecular pathogenesis of ovarian cancer which results in two specific pathways of carcinogenesis: (1) type I low-grade tumor and (2) type II high-grade tumor. Improved understanding of the molecular basis of ovarian carcinogenesis has opened new opportunities for targeted therapy. This paper will also review these potential therapeutic targets and will explore new agents that are currently being investigated.
Collapse
|
17
|
Evaluation of the first Ergocalciferol-derived, non hypercalcemic anti-cancer agent MT19c in ovarian cancer SKOV-3 cell lines. Gynecol Oncol 2011; 123:370-8. [DOI: 10.1016/j.ygyno.2011.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Revised: 06/27/2011] [Accepted: 07/01/2011] [Indexed: 01/23/2023]
|
18
|
Zaravinos A, Chatziioannou M, Lambrou GI, Boulalas I, Delakas D, Spandidos DA. Implication of RAF and RKIP Genes in Urinary Bladder Cancer. Pathol Oncol Res 2010; 17:181-90. [DOI: 10.1007/s12253-010-9295-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Accepted: 08/04/2010] [Indexed: 11/29/2022]
|
19
|
Pliarchopoulou K, Pectasides D. Epithelial ovarian cancer: focus on targeted therapy. Crit Rev Oncol Hematol 2010; 79:17-23. [PMID: 20674385 DOI: 10.1016/j.critrevonc.2010.07.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2010] [Revised: 06/26/2010] [Accepted: 07/08/2010] [Indexed: 01/15/2023] Open
Abstract
Ovarian cancer remains the leading cause of gynecological cancer-related mortality in the Western World despite the advances in surgical techniques and chemotherapy regimens over the past three decades. Although response rates and complete responses in advanced disease are >80% and 40-60%, respectively, after first-line treatment with carboplatin and paclitaxel, most of the patients will eventually relapse with a median progression-free survival of 18 months. Currently, research efforts have improved our understanding on the molecular biology of ovarian cancer and novel targeted treatment strategies are likely to contribute to the management of the disease and give the chance to an individualized therapeutic approach.
Collapse
Affiliation(s)
- Kyriaki Pliarchopoulou
- Second Department of Internal Medicine, Propaedeutic Oncology Section, University of Athens, "Attikon" University Hospital, Haidari, Greece.
| | | |
Collapse
|
20
|
Lorusso D, Pietragalla A, Mainenti S, Di Legge A, Amadio G, Scambia G. Emerging drugs for ovarian cancer. Expert Opin Emerg Drugs 2010; 15:635-52. [PMID: 20604741 DOI: 10.1517/14728214.2010.502888] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD Ovarian cancer has the highest mortality of all female reproductive tract cancers, which reflects both the absence of proven ovarian cancer screening tests and the development of drug-resistant cancer cell. Apart from varying the dosages, schedules, mode of delivery and combinations of existing drugs, efforts must continue to identify signaling pathways in tumor cells sufficiently different from normal cells that can be a target for maximizing tumor kill and minimizing toxicity. AREAS COVERED IN THIS REVIEW Some of the most important cellular pathways are analyzed and discussed and the most interesting clinical trials, both closed and ongoing, described. WHAT THE READER WILL GAIN The reader will gain a panoramic vision of all the most active drugs in clinical investigations in ovarian cancer. The reader will also better understand what the unresolved problems of molecular research are and how complicated the process 'from the bench to the bedside' is. TAKE HOME MESSAGE It is only with a strong commitment, cooperation and collaboration from the international ovarian cancer community that significant improvement in patient outcomes can be attained beyond the marginal gains achieved so far.
Collapse
Affiliation(s)
- Domenica Lorusso
- Catholic University of the Sacred Heart, Department of Gynecologic Oncology, Largo Agostino Gemelli 8, IT-00168 Rome.
| | | | | | | | | | | |
Collapse
|
21
|
Kuystermans D, Dunn MJ, Al-Rubeai M. A proteomic study of cMyc improvement of CHO culture. BMC Biotechnol 2010; 10:25. [PMID: 20307306 PMCID: PMC2859402 DOI: 10.1186/1472-6750-10-25] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Accepted: 03/22/2010] [Indexed: 02/07/2023] Open
Abstract
Background The biopharmaceutical industry requires cell lines to have an optimal proliferation rate and a high integral viable cell number resulting in a maximum volumetric recombinant protein product titre. Nutrient feeding has been shown to boost cell number and productivity in fed-batch culture, but cell line engineering is another route one may take to increase these parameters in the bioreactor. The use of CHO-K1 cells with a c-myc plasmid allowing for over-expressing c-Myc (designated cMycCHO) gives a higher integral viable cell number. In this study the differential protein expression in cMycCHO is investigated using two-dimensional gel electrophoresis (2-DE) followed by image analysis to determine the extent of the effect c-Myc has on the cell and the proteins involved to give the new phenotype. Results Over 100 proteins that were differentially expressed in cMycCHO cells were detected with high statistical confidence, of which 41 were subsequently identified by tandem mass spectrometry (LC-MS/MS). Further analysis revealed proteins involved in a variety of pathways. Some examples of changes in protein expression include: an increase in nucleolin, involved in proliferation and known to aid in stabilising anti-apoptotic protein mRNA levels, the cytoskeleton and mitochondrial morphology (vimentin), protein biosysnthesis (eIF6) and energy metabolism (ATP synthetase), and a decreased regulation of all proteins, indentified, involved in matrix and cell to cell adhesion. Conclusion These results indicate several proteins involved in proliferation and adhesion that could be useful for future approaches to improve proliferation and decrease adhesion of CHO cell lines which are difficult to adapt to suspension culture.
Collapse
Affiliation(s)
- Darrin Kuystermans
- School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | |
Collapse
|
22
|
Potentially important microRNA cluster on chromosome 17p13.1 in primary peritoneal carcinoma. Mod Pathol 2009; 22:197-205. [PMID: 18677302 DOI: 10.1038/modpathol.2008.135] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
MicroRNAs are a group of small non-coding RNAs approximately 22 nucleotides in length. Recent work has shown differential expression of mature microRNAs in human cancers. We characterized the alteration in expression of a select group of microRNAs in primary peritoneal carcinoma relative to matched cases of ovarian serous carcinoma. MicroRNA expression was analysed using semi-quantitative stem-loop RT-PCR on a set of 34 formalin-fixed paraffin-embedded samples. Protein expression of p53 and bcl-2 was quantified in the corresponding tissue microarray. We provide definitive evidence that there is downregulation of a select group of microRNAs in tumours meeting Gynaecological Oncology Group criteria for primary peritoneal carcinoma relative to ovarian serous carcinoma. Specifically, we show decreased p53 expression and downregulation of miR-195 and miR-497 from the microRNA cluster site at chromosome 17p13.1 in primary peritoneal carcinoma relative to ovarian serous carcinoma. miR-195 and miR-497 may have potential roles as tumour-suppressor genes in primary peritoneal tumourigenesis.
Collapse
|
23
|
Fan F, Feng L, He J, Wang X, Jiang X, Zhang Y, Wang Z, Chen Y. RKTG sequesters B-Raf to the Golgi apparatus and inhibits the proliferation and tumorigenicity of human malignant melanoma cells. Carcinogenesis 2008; 29:1157-63. [PMID: 18515281 DOI: 10.1093/carcin/bgn119] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Raf kinase trapping to Golgi (RKTG) is a newly characterized negative regulator of the Ras-Raf-mitogen-activated and extracellular signal-regulated kinase kinase (MEK)-extracellular signal-regulated kinase (ERK)-signaling pathway via sequestrating Raf-1 to the Golgi apparatus. Among Raf kinase family members, B-Raf is the most frequently mutated gene in human cancers and an activated B-Raf mutation V600E is associated with >60% of human melanomas. Here, we show that RKTG can also bind and translocate B-Raf to the Golgi apparatus. When overexpressed in A375, a human malignant melanoma cell line with B-Raf(V600E), RKTG inhibits ERK activation, cell proliferation and transformation of A375 cells. In addition, the tumorigenicity of the RKTG-expressing A375 cells is suppressed in nude mice. Consistently, cell proliferation rate was reduced in the tumor xenografts in which RKTG was overexpressed. Collectively, our results suggest that RKTG may play a suppressive role in human melanoma that harbors an oncogenic B-Raf mutation via its antagonistic action on B-Raf.
Collapse
Affiliation(s)
- Fengjuan Fan
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Graduate School of the Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Götz R. Inter-cellular adhesion disruption and the RAS/RAF and beta-catenin signalling in lung cancer progression. Cancer Cell Int 2008; 8:7. [PMID: 18492263 PMCID: PMC2427011 DOI: 10.1186/1475-2867-8-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Accepted: 05/20/2008] [Indexed: 12/30/2022] Open
Abstract
Cadherin cell adhesion molecules play an essential role in creating tight intercellular association and their loss has been correlated with poor prognosis in human cancer. Mutational activation of protein kinases and loss of cell adhesion occur together in human lung adenocarcinoma but how these two pathways interconnect is only poorly understood. Mouse models of human lung adenocarcinoma with oncogene expression targeted to subtypes of lung epithelial cells led to formation of adenomas or adenocarcinomas that lacked metastatic potential. Conditional genetic abrogation of epithelial tumour cell adhesion in mice with benign lung tumours induced by oncogenic RAF kinase has been demonstrated to induce intratumourous vascularization (angiogenic switch), progression to invasive adenocarcinoma and micrometastasis. Importantly, breaking cell adhesion in benign oncogene-driven lung tumour cells activated beta-catenin signalling and induced the expression of several genes that are normally expressed in intestine rather than the lung. I will discuss potential routes to nuclear beta-catenin signalling in cancer and how nuclear beta-catenin may epigenetically alter the plasticity of tumour cells during malignant progression.
Collapse
Affiliation(s)
- Rudolf Götz
- Institut für Medizinische Strahlenkunde und Zellforschung (MSZ), Universität Würzburg, Germany.
| |
Collapse
|
25
|
Dinh P, Harnett P, Piccart-Gebhart MJ, Awada A. New therapies for ovarian cancer: cytotoxics and molecularly targeted agents. Crit Rev Oncol Hematol 2008; 67:103-12. [PMID: 18342536 DOI: 10.1016/j.critrevonc.2008.01.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2007] [Revised: 01/23/2008] [Accepted: 01/30/2008] [Indexed: 12/20/2022] Open
Abstract
Ovarian cancer, although a chemo-sensitive disease, is associated with high morbidity and mortality due to its often-late presentation. Platinums and taxanes have improved the prognosis over recent years but median overall survival is still unacceptably low (24-60 months). Apart from the manipulation of doses, schedules, mode of delivery, and combinations of existing drugs, new cytotoxics and molecularly targeted agents with different mechanisms of action must be evaluated in this patient population. This article will review the most recent clinical trials data pertaining to these new cytotoxic drugs including patupilone, telcyta, and trabectedin, as well as those of small molecules and inhibitors of the EGFR and VEGF receptor families. It will also discuss other potential signal transduction targets worthy of further evaluation in future trials.
Collapse
Affiliation(s)
- Phuong Dinh
- Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium.
| | | | | | | |
Collapse
|
26
|
Banerji U, Sain N, Sharp SY, Valenti M, Asad Y, Ruddle R, Raynaud F, Walton M, Eccles SA, Judson I, Jackman AL, Workman P. An in vitro and in vivo study of the combination of the heat shock protein inhibitor 17-allylamino-17-demethoxygeldanamycin and carboplatin in human ovarian cancer models. Cancer Chemother Pharmacol 2008; 62:769-78. [DOI: 10.1007/s00280-007-0662-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2007] [Accepted: 12/08/2007] [Indexed: 10/22/2022]
|
27
|
Abstract
Over the past 5 years, the Raf kinase family has emerged as a promising target for protein-directed cancer therapy development. The goal of this review is to first provide a concise summary of the data validating Raf proteins as high-interest therapeutic targets. The authors then outline the mode of action of Raf kinases, emphasizing how Raf activities and protein interactions suggest specific approaches to inhibiting Raf. The authors then summarize the set of drugs, antisense reagents and antibodies available or in development for therapeutically targeting Raf or Raf-related proteins, as well as existing strategies combining these and other therapeutic agents. Finally, the authors discuss recent results from systems biology analyses that have the potential to increasingly guide the intelligent selection of combination therapies involving Raf-targeting agents and other therapeutics.
Collapse
|
28
|
Michaloglou C, Vredeveld LCW, Mooi WJ, Peeper DS. BRAF(E600) in benign and malignant human tumours. Oncogene 2007; 27:877-95. [PMID: 17724477 DOI: 10.1038/sj.onc.1210704] [Citation(s) in RCA: 194] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Of the RAF family of protein kinases, BRAF is the only member to be frequently activated by mutation in cancer. A single amino acid substitution (V600E) accounts for the vast majority and results in constitutive activation of BRAF kinase function. Its expression is required to maintain the proliferative and oncogenic characteristics of BRAF(E600)-expressing human tumour cells. Although BRAF(E600) acts as an oncogene in the context of additional genetic lesions, in primary cells it appears to be associated rather with transient stimulation of proliferation. Eventually, BRAF(E600) signalling triggers cell cycle arrest with the hallmarks of cellular senescence, as is illustrated by several recent studies in cultured cells, animal models and benign human lesions. In this review, we will discuss recent advances in our understanding of the role of BRAF(E600) in benign and malignant human tumours and the implications for therapeutic intervention.
Collapse
Affiliation(s)
- C Michaloglou
- Division of Molecular Genetics, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
29
|
Mukherjee B, Das T, Ghosh S, Datta S. Changes in the antioxidant defense and hepatic drug metabolizing enzyme and isoenzyme levels, 8-hydroxydeoxyguanosine formation and expressions of c-raf.1 and insulin-like growth factor II genes during the stages of development of hepatocellular carcinoma in rats. Eur J Cancer Prev 2007; 16:363-71. [PMID: 17554210 DOI: 10.1097/01.cej.0000236254.01608.33] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
This is an extensive study in a defined initiation-promotion hepatocellular carcinoma model of hepatocarcinogenesis (in rats) in which many important marker enzymes and isoenzymes and 8-hydroxydeoxyguanosine formation have been studied together with two very important cellular proliferating genes, insulin-like growth factor II and c-raf.1, known for their role in hepatocellular cancer development. Experiments were carried out on hepatic tissues of male Sprague-Dawley rats. Variations in different enzyme/isoenzyme activities/contents/expression pattern and 8-hydroxydeoxyguanosine-positive cells were studied. Insulin-like growth factor II and c-raf.1 gene expressions were monitored. A direct shift with increase in size and numbers of lesions was found to occur in different experimental groups. In this study, glutathione peroxidase (1.14 and 1.46-fold) and reduced triphosphopyridine nucleotide (TPNH)-cytochrome-c-reductase (1.94 and 2.94-fold) activities, cytochrome b5 (1.57 and 3.28-fold) and P-450 contents (1.45 and 1.22-fold), glutathione content (1.27 and 1.45-fold) and superoxide dismutase and catalase (1.16 and 1.39-fold) activities in group A animals were found to be lower than those in initiation and promotion studies, respectively. 8-Hydroxydeoxyguanosine-positive nuclei count showed that oxidative damage of nuclear DNA enhanced with the progress of the disease. The insulin-like growth factor II expression was found to be predominant in hepatocellular carcinoma and in early preneoplastic lesions. Unlike insulin-like growth factor II, c-raf.1 expression was located in the late basophilic lesions associated with hepatocellular carcinoma. During the various stages of the development of hepatocellular carcinoma, the enzymes played a significant role in metabolizing carcinogens and thereby scavenging various toxic metabolites or free radicals produced. A sequence of cellular changes starting from the appearance of glycogen storage foci to basophilic foci leading to hepatocellular carcinoma via mixed cell foci varied the activity/content or expression pattern of the enzymes and isoenzymes and in 8-hydroxydeoxyguanosine formation. It has been established that c-raf.1-induced signaling pathways activated by insulin-like growth factor II is implicated in the late stage of development of cancer.
Collapse
Affiliation(s)
- Biswajit Mukherjee
- Department of Pharmaceutical Technology, Jadavpur University, Kolkata, West Bengal, India.
| | | | | | | |
Collapse
|
30
|
Leicht DT, Balan V, Kaplun A, Singh-Gupta V, Kaplun L, Dobson M, Tzivion G. Raf kinases: function, regulation and role in human cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1773:1196-212. [PMID: 17555829 PMCID: PMC1986673 DOI: 10.1016/j.bbamcr.2007.05.001] [Citation(s) in RCA: 206] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2006] [Revised: 05/01/2007] [Accepted: 05/02/2007] [Indexed: 12/25/2022]
Abstract
The Ras-Raf-MAPK pathway regulates diverse physiological processes by transmitting signals from membrane based receptors to various nuclear, cytoplasmic and membrane-bound targets, coordinating a large variety of cellular responses. Function of Raf family kinases has been shown to play a role during organism development, cell cycle regulation, cell proliferation and differentiation, cell survival and apoptosis and many other cellular and physiological processes. Aberrations along the Ras-Raf-MAPK pathway play an integral role in various biological processes concerning human health and disease. Overexpression or activation of the pathway components is a common indicator in proliferative diseases such as cancer and contributes to tumor initiation, progression and metastasis. In this review, we focus on the physiological roles of Raf kinases in normal and disease conditions, specifically cancer, and the current thoughts on Raf regulation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Guri Tzivion
- To whom correspondence should be addressed: Karmanos Cancer Institute, Wayne State University, 4100 John R., HWCRC 716, Detroit, MI 48201, Tel: 313-576-8311, Fax: 313-576-8308, E-mail:
| |
Collapse
|
31
|
Abstract
Ovarian cancer is a heterogeneous disease with extensive cytogenetic and molecular heterogeneity including aneuploidy, chromosomal alterations, mutations and overexpression as well as a natural propensity to disseminate and spread, making it difficult to diagnose at an early stage. Insights into the molecular mechanisms operative in cancer development, progression and metastasis have uncovered a wide array of targets for therapeutic intervention. In the absence of a common driving oncogene in ovarian cancer, single targeted therapy for this disease is unlikely to yield significant clinical benefit. Tailored approaches that combine molecular targeting agents with cytotoxic regimens hold great promise when used in primary treatment, during consolidation and maintenance therapy, and in the treatment of persistent or recurrent disease. The most promising treatment strategies are those that target the drivers of tumorigenesis and enhance the activity of cytotoxic agents. Receptor tyrosine kinases, non-receptor tyrosine kinases, serine/threonine kinases, transferases, proteases and deacetylases are among the relevant molecular markers and targets for ovarian cancer that are discussed. Collaboration, coordination, creativity and aggressive outreach to patients and their advocates are essential for success in running the concurrent trials with multiple clinical end points and embedded translational research that are needed to evaluate the array of promising targeted therapeutics and combinations. Validated biomarkers, surrogate specimens and end points, and additional clinically relevant in vitro and in vivo models for ovarian cancer are needed to facilitate the drug development and evaluation process, and ultimately to make meaningful improvements in the diagnosis, prevention and management of ovarian cancer.
Collapse
|