1
|
Liang YY, Khalid K, Le HV, Teo HMV, Raitelaitis M, Gerault MA, Lee JJH, Lyu J, Chan A, Jeyasekharan AD, Tam WL, Nordlund P, Prabhu N. MS CETSA deep functional proteomics uncovers DNA repair programs leading to gemcitabine resistance. Nat Commun 2025; 16:4234. [PMID: 40335468 PMCID: PMC12059070 DOI: 10.1038/s41467-025-59505-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 04/23/2025] [Indexed: 05/09/2025] Open
Abstract
Mechanisms for resistance to cytotoxic cancer drugs are dependent on dynamic changes in the biochemistry of cellular pathways, information which is hard to obtain at the systems level. Here we use a deep functional proteomics implementation of the Cellular Thermal Shift Assay to reveal a range of induced biochemical responses to gemcitabine in resistant and sensitive diffuse large B cell lymphoma cell lines. Initial responses in both, gemcitabine resistant and sensitive cells, reflect known targeted effects by gemcitabine on ribonucleotide reductase and DNA damage responses. However, later responses diverge dramatically where sensitive cells show induction of characteristic CETSA signals for early apoptosis, while resistant cells reveal biochemical modulations reflecting transition through a distinct DNA-damage signaling state, including opening of cell cycle checkpoints and induction of translesion DNA synthesis programs, allowing bypass of damaged DNA-adducts. The results also show the induction of a protein ensemble, labeled the Auxiliary DNA Damage Repair, likely supporting DNA replication at damaged sites that can be attenuated in resistant cells by an ATR inhibitor, thus re-establishing gemcitabine sensitivity and demonstrating ATR as a key signaling node of this response.
Collapse
Affiliation(s)
- Ying Yu Liang
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, 138673, Singapore
| | - Khalidah Khalid
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, 138673, Singapore
| | - Hai Van Le
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, 138673, Singapore
| | - Hui Min Vivian Teo
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, 138672, Singapore
| | - Mindaugas Raitelaitis
- Department of Oncology and Pathology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Marc-Antoine Gerault
- Department of Oncology and Pathology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Jane Jia Hui Lee
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, 138672, Singapore
| | - Jiawen Lyu
- Department of Oncology and Pathology, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Allison Chan
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599, Singapore
| | - Anand Devaprasath Jeyasekharan
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Singapore, 117596, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, 119074, Singapore
| | - Wai Leong Tam
- Department of Oncology and Pathology, Karolinska Institutet, 171 77, Stockholm, Sweden.
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Singapore, 117596, Singapore.
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University Singapore, 14 Medical Drive, Singapore, 117599, Singapore.
| | - Pär Nordlund
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, 138673, Singapore.
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, 138672, Singapore.
| | - Nayana Prabhu
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, 138673, Singapore.
| |
Collapse
|
2
|
Rodríguez-Candela Mateos M, Carpintero-Fernández P, Freijanes PS, Mosquera J, Nebril BA, Mayán MD. Insights into the role of connexins and specialized intercellular communication pathways in breast cancer: Mechanisms and applications. Biochim Biophys Acta Rev Cancer 2024; 1879:189173. [PMID: 39154967 DOI: 10.1016/j.bbcan.2024.189173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/31/2024] [Accepted: 08/14/2024] [Indexed: 08/20/2024]
Abstract
Gap junctions, membrane-based channels comprised of connexin proteins (Cxs), facilitate direct communication among neighbouring cells and between cells and the extracellular space through their hemichannels. The normal human breast expresses various Cxs family proteins, such as Cx43, Cx30, Cx32, Cx46, and Cx26, crucial for proper tissue development and function. These proteins play a significant role in breast cancer development, progression, and therapy response. In primary tumours, there is often a reduction and cytoplasmic mislocalization of Cx43 and Cx26, while metastatic lesions show an upregulation of these and other Cxs. Although existing research predominantly supports the tumour-suppressing role of Cxs in primary carcinomas through channel-dependent and independent functions, controversies persist regarding their involvement in the metastatic process. This review aims to provide an updated perspective on Cxs in human breast cancer, with a specific focus on intrinsic subtypes due to the heterogeneous nature of this disease. Additionally, the manuscript will explore the role of Cxs in immune interactions and novel forms of intercellular communication, such as tunneling nanotubes and extracellular vesicles, within the breast tumour context and tumour microenvironment. Recent findings suggest that Cxs hold potential as therapeutic targets for mitigating metastasis and drug resistance. Furthermore, they may serve as novel biomarkers for cancer prognosis, offering promising avenues for future research and clinical applications.
Collapse
Affiliation(s)
- Marina Rodríguez-Candela Mateos
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Xubias de Arriba, 84, 15006 A Coruña, Spain
| | - Paula Carpintero-Fernández
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Xubias de Arriba, 84, 15006 A Coruña, Spain; CellCOM Research Group, Center for Research in Nanomaterials and Biomedicine (CINBIO), Universidade de Vigo, Edificio Olimpia Valencia, Campus Universitario Lagoas Marcosende, 36310 Vigo, Spain; Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS, Spain
| | - Paz Santiago Freijanes
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Xubias de Arriba, 84, 15006 A Coruña, Spain; Anatomic Pathology Department, Breast Unit, A Coruña University Hospital, SERGAS, A Coruña, Spain
| | - Joaquin Mosquera
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Xubias de Arriba, 84, 15006 A Coruña, Spain; Surgery Department, Breast Unit, A Coruña University Hospital, SERGAS, A Coruña, Spain
| | - Benigno Acea Nebril
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Xubias de Arriba, 84, 15006 A Coruña, Spain; Surgery Department, Breast Unit, A Coruña University Hospital, SERGAS, A Coruña, Spain
| | - María D Mayán
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Servizo Galego de Saúde (SERGAS), Xubias de Arriba, 84, 15006 A Coruña, Spain; CellCOM Research Group, Center for Research in Nanomaterials and Biomedicine (CINBIO), Universidade de Vigo, Edificio Olimpia Valencia, Campus Universitario Lagoas Marcosende, 36310 Vigo, Spain; Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS, Spain.
| |
Collapse
|
3
|
Raji L, Tetteh A, Amin ARMR. Role of c-Src in Carcinogenesis and Drug Resistance. Cancers (Basel) 2023; 16:32. [PMID: 38201459 PMCID: PMC10778207 DOI: 10.3390/cancers16010032] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/12/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024] Open
Abstract
The aberrant transformation of normal cells into cancer cells, known as carcinogenesis, is a complex process involving numerous genetic and molecular alterations in response to innate and environmental stimuli. The Src family kinases (SFK) are key components of signaling pathways implicated in carcinogenesis, with c-Src and its oncogenic counterpart v-Src often playing a significant role. The discovery of c-Src represents a compelling narrative highlighting groundbreaking discoveries and valuable insights into the molecular mechanisms underlying carcinogenesis. Upon oncogenic activation, c-Src activates multiple downstream signaling pathways, including the PI3K-AKT pathway, the Ras-MAPK pathway, the JAK-STAT3 pathway, and the FAK/Paxillin pathway, which are important for cell proliferation, survival, migration, invasion, metastasis, and drug resistance. In this review, we delve into the discovery of c-Src and v-Src, the structure of c-Src, and the molecular mechanisms that activate c-Src. We also focus on the various signaling pathways that c-Src employs to promote oncogenesis and resistance to chemotherapy drugs as well as molecularly targeted agents.
Collapse
Affiliation(s)
| | | | - A. R. M. Ruhul Amin
- Department of Pharmaceutical Sciences, Marshall University School of Pharmacy, Huntington, WV 25755, USA; (L.R.); (A.T.)
| |
Collapse
|
4
|
Zhou M, Zheng M, Zhou X, Tian S, Yang X, Ning Y, Li Y, Zhang S. The roles of connexins and gap junctions in the progression of cancer. Cell Commun Signal 2023; 21:8. [PMID: 36639804 PMCID: PMC9837928 DOI: 10.1186/s12964-022-01009-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/03/2022] [Indexed: 01/15/2023] Open
Abstract
Gap junctions (GJs), which are composed of connexins (Cxs), provide channels for direct information exchange between cells. Cx expression has a strong spatial specificity; however, its influence on cell behavior and information exchange between cells cannot be ignored. A variety of factors in organisms can modulate Cxs and subsequently trigger a series of responses that have important effects on cellular behavior. The expression and function of Cxs and the number and function of GJs are in dynamic change. Cxs have been characterized as tumor suppressors in the past, but recent studies have highlighted the critical roles of Cxs and GJs in cancer pathogenesis. The complex mechanism underlying Cx and GJ involvement in cancer development is a major obstacle to the evolution of therapy targeting Cxs. In this paper, we review the post-translational modifications of Cxs, the interactions of Cxs with several chaperone proteins, and the effects of Cxs and GJs on cancer. Video Abstract.
Collapse
Affiliation(s)
- Mingming Zhou
- Graduate School, Tianjin Medical University, Tianjin, 300070 People’s Republic of China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Nankai University, Tianjin, 300121 People’s Republic of China
| | - Xinyue Zhou
- Graduate School, Tianjin Medical University, Tianjin, 300070 People’s Republic of China
| | - Shifeng Tian
- Graduate School, Tianjin Medical University, Tianjin, 300070 People’s Republic of China
| | - Xiaohui Yang
- Nankai University School of Medicine, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Yidi Ning
- Nankai University School of Medicine, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Yuwei Li
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300121 People’s Republic of China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Nankai University, Tianjin, 300121 People’s Republic of China
| |
Collapse
|
5
|
Kalal BS, Modi PK, Najar MA, Behera SK, Upadhya D, Prasad TSK, Pai VR. Hyperphosphorylation of HDAC2 promotes drug resistance in a novel dual drug resistant mouse melanoma cell line model: an in vitro study. Am J Cancer Res 2021; 11:5881-5901. [PMID: 35018231 PMCID: PMC8727796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 11/22/2021] [Indexed: 06/14/2023] Open
Abstract
Drug-resistant melanoma is very difficult to treat, and a novel approach is needed to overcome resistance. The present study aims at identifying the alternate pathways utilized in the dual drug-resistant mouse melanoma cells (B16F10R) for their survival and proliferation. The dual drug-resistant mouse melanoma, B16F10R, was established by treating the cells with a combination of U0126 (MEK1/2 inhibitor) and LY294002 (PI3K-AKT kinase inhibitor) in a dose-escalating manner till they attained a resistance fold factor of ≥2. The altered phosphoproteome in the B16F10R, as compared to the parental B16F10C, was analyzed using a high-resolution Orbitrap Fusion Tribrid mass spectrometer. Histone deacetylases 2 (HDAC2) was validated for its role in drug resistance by using its inhibitor, valproic acid (VPA). In the B16F10R cells, 363 altered phosphoproteins were identified, among which 126 were hyperphosphorylated, and 137 were hypophosphorylated (1.5-fold change). Pathway analysis shows the altered phosphoproteins are from RNA metabolism and cell cycle proteins. Inhibition of HDAC2 by VPA induces apoptosis in B16F10C and B16F10R. The present study highlights the role of HDAC2, a cell cycle regulator, in the development of resistance to dual drugs in murine melanoma. Therefore, designing leads for targeting HDAC2 along with key signaling pathways may be explored in treatment strategies.
Collapse
Affiliation(s)
- Bhuvanesh Sukhlal Kalal
- Department of Biochemistry, Yenepoya Medical College, Yenepoya (Deemed to be University)Mangaluru, Karnataka, India
| | - Prashant Kumar Modi
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University)Mangaluru, Karnataka, India
| | - Mohd Altaf Najar
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University)Mangaluru, Karnataka, India
| | - Santosh Kumar Behera
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University)Mangaluru, Karnataka, India
| | - Dinesh Upadhya
- Centre for Molecular Neurosciences, Department of Anatomy, Kasturba Medical College, Manipal Academy of Higher Education ManipalUdupi 576104, Karnataka, India
| | | | - Vinitha Ramanath Pai
- Department of Biochemistry, Yenepoya Medical College, Yenepoya (Deemed to be University)Mangaluru, Karnataka, India
| |
Collapse
|
6
|
Panda M, Biswal BK. Cell signaling and cancer: a mechanistic insight into drug resistance. Mol Biol Rep 2019; 46:5645-5659. [PMID: 31280421 DOI: 10.1007/s11033-019-04958-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 06/27/2019] [Indexed: 12/22/2022]
Abstract
Drug resistance is a major setback for advanced therapeutics in multiple cancers. The increasing prevalence of this resistance is a growing concern and bitter headache for the researchers since a decade. Hence, it is essential to revalidate the existing strategies available for cancer treatment and to look after a novel therapeutic approach for target based killing of cancer cells at the genetic level. This review outlines the different mechanisms enabling resistance including drug efflux, drug target alternation, alternative splicing, the release of the extracellular vesicle, tumor heterogeneity, epithelial-mesenchymal transition, tumor microenvironment, the secondary mutation in the receptor, epigenetic alternation, heterodimerization of receptors, amplification of target and amplification of components rather than the target. Furthermore, existing evidence and the role of various signaling pathways like EGFR, Ras, PI3K/Akt, Wnt, Notch, TGF-β, Integrin-ECM signaling in drug resistance are explained. Lastly, the prevention of this resistance by a contemporary therapeutic strategy, i.e., a combination of specific signaling pathway inhibitors and the cocktail of a cancer drug is summarized showing the new treatment strategies.
Collapse
Affiliation(s)
- Munmun Panda
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology, Sundargarh, Rourkela, Odisha, 769008, India
| | - Bijesh K Biswal
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology, Sundargarh, Rourkela, Odisha, 769008, India.
| |
Collapse
|
7
|
Hernández-Guerra M, Hadjihambi A, Jalan R. Gap junctions in liver disease: Implications for pathogenesis and therapy. J Hepatol 2019; 70:759-772. [PMID: 30599172 DOI: 10.1016/j.jhep.2018.12.023] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 12/03/2018] [Accepted: 12/12/2018] [Indexed: 02/07/2023]
Abstract
In the normal liver, cells interact closely through gap junctions. By providing a pathway for the trafficking of low molecular mass molecules, these channels contribute to tissue homeostasis and maintenance of hepatic function. Thus, dysfunction of gap junctions affects a wide variety of liver processes, such as differentiation, cell death, inflammation and fibrosis. In fact, dysfunctional gap junctions have been implicated, for more than a decade, in cholestatic disease, hepatic cancer and cirrhosis. Additionally, in recent years there is an increasing body of evidence that these channels are also involved in other relevant and prevalent liver pathological processes, such as non-alcoholic fatty liver disease, acute liver injury and portal hypertension. In parallel to these new clinical implications the available data include controversial observations. Thus, a comprehensive overview is required to better understand the functional complexity of these pores. This paper will review the most recent knowledge concerning gap junction dysfunction, with a special focus on the role of these channels in the pathogenesis of relevant clinical entities and on potential therapeutic targets that are amenable to modification by drugs.
Collapse
Affiliation(s)
| | | | - Rajiv Jalan
- UCL Institute for Liver and Digestive Health, Royal Free Medical School, London, UK
| |
Collapse
|
8
|
Gap Junction Intercellular Communication Positively Regulates Cisplatin Toxicity by Inducing DNA Damage through Bystander Signaling. Cancers (Basel) 2018; 10:cancers10100368. [PMID: 30279363 PMCID: PMC6210410 DOI: 10.3390/cancers10100368] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 09/27/2018] [Accepted: 09/28/2018] [Indexed: 12/14/2022] Open
Abstract
The radiation-induced bystander effect (RIBE) can increase cellular toxicity in a gap junction dependent manner in unirradiated bystander cells. Recent reports have suggested that cisplatin toxicity can also be mediated by functional gap junction intercellular communication (GJIC). In this study using lung and ovarian cancer cell lines, we showed that cisplatin cytotoxicity is mediated by cellular density. This effect is ablated when GJA1 or Connexin 43 (Cx43) is targeted, a gap junction gene and protein, respectively, leading to cisplatin resistance but only at high or gap junction forming density. We also observed that the cisplatin-mediated bystander effect was elicited as DNA Double Strand Breaks (DSBs) with positive H2AX Ser139 phosphorylation (γH2AX) formation, an indicator of DNA DSBs. These DSBs are not observed when gap junction formation is prevented. We next showed that cisplatin is not the “death” signal traversing the gap junctions by utilizing the cisplatin-GG intrastrand adduct specific antibody. Finally, we also showed that cells deficient in the structure-specific DNA endonuclease ERCC1-ERCC4 (ERCC1-XPF), an important mediator of cisplatin resistance, further sensitized when treated with cisplatin in the presence of gap junction forming density. Taken together, these results demonstrate the positive effect of GJIC on increasing cisplatin cytotoxicity.
Collapse
|
9
|
Samuel P, Mulcahy LA, Furlong F, McCarthy HO, Brooks SA, Fabbri M, Pink RC, Carter DRF. Cisplatin induces the release of extracellular vesicles from ovarian cancer cells that can induce invasiveness and drug resistance in bystander cells. Philos Trans R Soc Lond B Biol Sci 2018; 373:rstb.2017.0065. [PMID: 29158318 DOI: 10.1098/rstb.2017.0065] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2017] [Indexed: 12/14/2022] Open
Abstract
Ovarian cancer has a poor overall survival that is partly caused by resistance to drugs such as cisplatin. Resistance can be acquired as a result of changes to the tumour or due to altered interactions within the tumour microenvironment. Extracellular vesicles (EVs), small lipid-bound vesicles that are loaded with macromolecular cargo and released by cells, are emerging as mediators of communication in the tumour microenvironment. We previously showed that EVs mediate the bystander effect, a phenomenon in which stressed cells can communicate with neighbouring naive cells leading to various effects including DNA damage; however, the role of EVs released following cisplatin treatment has not been tested. Here we show that treatment of cells with cisplatin led to the release of EVs that could induce invasion and increased resistance when taken up by bystander cells. This coincided with changes in p38 and JNK signalling, suggesting that these pathways may be involved in mediating the effects. We also show that EV uptake inhibitors could prevent this EV-mediated adaptive response and thus sensitize cells in vitro to the effects of cisplatin. Our results suggest that preventing pro-tumourigenic EV cross-talk during chemotherapy is a potential therapeutic target for improving outcome in ovarian cancer patients.This article is part of the discussion meeting issue 'Extracellular vesicles and the tumour microenvironment'.
Collapse
Affiliation(s)
- Priya Samuel
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Gipsy Lane, Headington, Oxford, OX3 0BP, UK
| | - Laura Ann Mulcahy
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Gipsy Lane, Headington, Oxford, OX3 0BP, UK
| | - Fiona Furlong
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Susan Ann Brooks
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Gipsy Lane, Headington, Oxford, OX3 0BP, UK
| | - Muller Fabbri
- Children's Center for Cancer and Blood Diseases, Children's Hospital Los Angeles, Los Angeles, CA 90027, USA.,Departments of Pediatrics and Molecular Microbiology & Immunology, University of Southern California-Keck School of Medicine, Norris Comprehensive Cancer Center, Los Angeles, CA 90027, USA
| | - Ryan Charles Pink
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Gipsy Lane, Headington, Oxford, OX3 0BP, UK
| | - David Raul Francisco Carter
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Gipsy Lane, Headington, Oxford, OX3 0BP, UK
| |
Collapse
|
10
|
Aasen T, Johnstone S, Vidal-Brime L, Lynn KS, Koval M. Connexins: Synthesis, Post-Translational Modifications, and Trafficking in Health and Disease. Int J Mol Sci 2018; 19:ijms19051296. [PMID: 29701678 PMCID: PMC5983588 DOI: 10.3390/ijms19051296] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/20/2018] [Accepted: 04/21/2018] [Indexed: 02/06/2023] Open
Abstract
Connexins are tetraspan transmembrane proteins that form gap junctions and facilitate direct intercellular communication, a critical feature for the development, function, and homeostasis of tissues and organs. In addition, a growing number of gap junction-independent functions are being ascribed to these proteins. The connexin gene family is under extensive regulation at the transcriptional and post-transcriptional level, and undergoes numerous modifications at the protein level, including phosphorylation, which ultimately affects their trafficking, stability, and function. Here, we summarize these key regulatory events, with emphasis on how these affect connexin multifunctionality in health and disease.
Collapse
Affiliation(s)
- Trond Aasen
- Translational Molecular Pathology, Vall d'Hebron Institute of Research (VHIR), Autonomous University of Barcelona, CIBERONC, 08035 Barcelona, Spain.
| | - Scott Johnstone
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, P.O. Box 801394, Charlottesville, VI 22908, USA.
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TT, UK.
| | - Laia Vidal-Brime
- Translational Molecular Pathology, Vall d'Hebron Institute of Research (VHIR), Autonomous University of Barcelona, CIBERONC, 08035 Barcelona, Spain.
| | - K Sabrina Lynn
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - Michael Koval
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
11
|
Nazari M, Serrill JD, Wan X, Nguyen MH, Anklin C, Gallegos DA, Smith AB, Ishmael JE, McPhail KL. New Mandelalides Expand a Macrolide Series of Mitochondrial Inhibitors. J Med Chem 2017; 60:7850-7862. [PMID: 28841379 DOI: 10.1021/acs.jmedchem.7b00990] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Mandelalides A-D (1-4) are macrocyclic polyketides known to have an unusual bioactivity profile influenced by compound glycosylation and growth phase of cultured cells. The isolation and characterization of additional natural congeners, mandelalides E-L (5-12), and the supply of synthetic compounds 1 and 12, as well as seco-mandelalide A methyl ester (13), have now facilitated mechanism of action and structure-activity relationship studies. Glycosylated mandelalides are effective inhibitors of aerobic respiration in living cells. Macrolides 1 and 2 inhibit mitochondrial function similar to oligomycin A and apoptolidin A, selective inhibitors of the mammalian ATP synthase (complex V). 1 inhibits ATP synthase activity from isolated mitochondria and triggers caspase-dependent apoptosis in HeLa cells, which are more sensitive to inhibition by 1 in the presence of the glycolysis inhibitor 2-deoxyglucose. Thus, mandelalide cytotoxicity depends on basal metabolic phenotype; cells with an oxidative phenotype are most likely to be inhibited by the mandelalides.
Collapse
Affiliation(s)
- Mohamad Nazari
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University , Corvallis, Oregon 97331, United States
| | - Jeffrey D Serrill
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University , Corvallis, Oregon 97331, United States
| | - Xuemei Wan
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University , Corvallis, Oregon 97331, United States
| | - Minh H Nguyen
- Department of Chemistry, University of Pennsylvania , Philadelphia, Pennsylvania 19104-6323, United States
| | - Clemens Anklin
- Bruker BioSpin , 15 Fortune Drive, Billerica, Massachusetts 01821, United States
| | - David A Gallegos
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University , Corvallis, Oregon 97331, United States
| | - Amos B Smith
- Department of Chemistry, University of Pennsylvania , Philadelphia, Pennsylvania 19104-6323, United States
| | - Jane E Ishmael
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University , Corvallis, Oregon 97331, United States
| | - Kerry L McPhail
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University , Corvallis, Oregon 97331, United States
| |
Collapse
|
12
|
Lin LF, Wu MH, Pidugu VK, Ho IC, Su TL, Lee TC. P-glycoprotein attenuates DNA repair activity in multidrug-resistant cells by acting through the Cbp-Csk-Src cascade. Oncotarget 2017; 8:45072-45087. [PMID: 28178691 PMCID: PMC5542168 DOI: 10.18632/oncotarget.15065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 01/24/2017] [Indexed: 11/25/2022] Open
Abstract
Recent studies have demonstrated that P-glycoprotein (P-gp) expression impairs DNA interstrand cross-linking agent-induced DNA repair efficiency in multidrug-resistant (MDR) cells. To date, the detailed molecular mechanisms underlying how P-gp interferes with Src activation and subsequent DNA repair activity remain unclear. In this study, we determined that the C-terminal Src kinase-binding protein (Cbp) signaling pathway involved in the negative control of Src activation is enhanced in MDR cells. We also demonstrated that cells that ectopically express P-gp exhibit reduced activation of DNA damage response regulators, such as ATM, Chk2, Braca1 and Nbs1 and hence attenuated DNA double-strand break repair capacity and become more susceptible than vector control cells to DNA interstrand cross-linking (ICL) agents. Moreover, we demonstrated that P-gp can not only interact with Cbp and Src but also enhance the formation of inhibitory C-terminal Src kinase (Csk)-Cbp complexes that reduce phosphorylation of the Src activation residue Y416 and increase phosphorylation of the Src negative regulatory residue Y527. Notably, suppression of Cbp expression in MDR cells restores cisplatin-induced Src activation, improves DNA repair capacity, and increases resistance to ICL agents. Ectopic expression of Cbp attenuates cisplatin-induced Src activation and increases the susceptibility of cells to ICL agents. Together, the current results indicate that P-gp inhibits DNA repair activity by modulating Src activation via Cbp-Csk-Src cascade. These results suggest that DNA ICL agents are likely to have therapeutic potential against MDR cells with P-gp-overexpression.
Collapse
Affiliation(s)
- Li-Fang Lin
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Ming-Hsi Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Vijaya Kumar Pidugu
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan.,Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University, Academia Sinica, Taipei 11529, Taiwan
| | - I-Ching Ho
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Tsann-Long Su
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Te-Chang Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan.,Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University, Academia Sinica, Taipei 11529, Taiwan.,Institute of Pharmacology, National Yang-Ming University, Taipei 11221, Taiwan
| |
Collapse
|
13
|
Huang K, Chen J, Yang MS, Tang YJ, Pan F. Inhibition of Src by microRNA-23b increases the cisplatin sensitivity of chondrosarcoma cells. Cancer Biomark 2017; 18:231-239. [DOI: 10.3233/cbm-160102] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
14
|
Wong P, Laxton V, Srivastava S, Chan YWF, Tse G. The role of gap junctions in inflammatory and neoplastic disorders (Review). Int J Mol Med 2017; 39:498-506. [PMID: 28098880 PMCID: PMC5360388 DOI: 10.3892/ijmm.2017.2859] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 12/23/2016] [Indexed: 12/29/2022] Open
Abstract
Gap junctions are intercellular channels made of connexin proteins, mediating both electrical and biochemical signals between cells. The ability of gap junction proteins to regulate immune responses, cell proliferation, migration, apoptosis and carcinogenesis makes them attractive therapeutic targets for treating inflammatory and neoplastic disorders in different organ systems. Alterations in gap junction profile and expression levels are observed in hyperproliferative skin disorders, lymphatic vessel diseases, inflammatory lung diseases, liver injury and neoplastic disorders. It is now recognized that the therapeutic effects mediated by traditional pharmacological agents are dependent upon gap junction communication and may even act by influencing gap junction expression or function. Novel strategies for modulating the function or expression of connexins, such as the use of synthetic mimetic peptides and siRNA technology are considered.
Collapse
Affiliation(s)
- Pui Wong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Victoria Laxton
- Intensive Care Department, Royal Brompton and Harefield NHS Foundation Trust, London SW3 6NP
| | | | - Yin Wah Fiona Chan
- School of Biological Sciences, University of Cambridge, Cambridge CB2 1AG, UK
| | - Gary Tse
- Department of Medicine and Therapeutics
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, SAR, P.R. China
| |
Collapse
|
15
|
Sun Y, Zhang W, Chen Y, Ma Q, Wei J, Liu Q. Identifying anti-cancer drug response related genes using an integrative analysis of transcriptomic and genomic variations with cell line-based drug perturbations. Oncotarget 2017; 7:9404-19. [PMID: 26824188 PMCID: PMC4891048 DOI: 10.18632/oncotarget.7012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 01/01/2016] [Indexed: 01/18/2023] Open
Abstract
Background Clinical responses to anti-cancer therapies often only benefit a defined subset of patients. Predicting the best treatment strategy hinges on our ability to effectively translate genomic data into actionable information on drug responses. Results To achieve this goal, we compiled a comprehensive collection of baseline cancer genome data and drug response information derived from a large panel of cancer cell lines. This data set was applied to identify the signature genes relevant to drug sensitivity and their resistance by integrating CNVs and the gene expression of cell lines with in vitro drug responses. We presented an efficient in-silico pipeline for integrating heterogeneous cell line data sources with the simultaneous modeling of drug response values across all the drugs and cell lines. Potential signature genes correlated with drug response (sensitive or resistant) in different cancer types were identified. Using signature genes, our collaborative filtering-based drug response prediction model outperformed the 44 algorithms submitted to the DREAM competition on breast cancer cells. The functions of the identified drug response related signature genes were carefully analyzed at the pathway level and the synthetic lethality level. Furthermore, we validated these signature genes by applying them to the classification of the different subtypes of the TCGA tumor samples, and further uncovered their in vivo implications using clinical patient data. Conclusions Our work may have promise in translating genomic data into customized marker genes relevant to the response of specific drugs for a specific cancer type of individual patients.
Collapse
Affiliation(s)
- Yi Sun
- Department of Central Laboratory, Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Wei Zhang
- Department of Central Laboratory, Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yunqin Chen
- R & D Information, AstraZeneca, Shanghai, China
| | - Qin Ma
- Department of Plant Science, South Dakota State University, Brookings, SD, USA
| | - Jia Wei
- R & D Information, AstraZeneca, Shanghai, China
| | - Qi Liu
- Department of Central Laboratory, Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
16
|
Nazari M, Serrill JD, Sikorska J, Ye T, Ishmael JE, McPhail KL. Discovery of Mandelalide E and Determinants of Cytotoxicity for the Mandelalide Series. Org Lett 2016; 18:1374-7. [DOI: 10.1021/acs.orglett.6b00308] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Mohamad Nazari
- Department
of Pharmaceutical Sciences, Oregon State University, 203 Pharmacy
Building, Corvallis, Oregon 97331-4003, United States
| | - Jeffrey D. Serrill
- Department
of Pharmaceutical Sciences, Oregon State University, 203 Pharmacy
Building, Corvallis, Oregon 97331-4003, United States
| | - Justyna Sikorska
- Department
of Pharmaceutical Sciences, Oregon State University, 203 Pharmacy
Building, Corvallis, Oregon 97331-4003, United States
| | - Tao Ye
- Laboratory
of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School Xili, Nanshan District, Shenzhen 518055, China
| | - Jane E. Ishmael
- Department
of Pharmaceutical Sciences, Oregon State University, 203 Pharmacy
Building, Corvallis, Oregon 97331-4003, United States
| | - Kerry L. McPhail
- Department
of Pharmaceutical Sciences, Oregon State University, 203 Pharmacy
Building, Corvallis, Oregon 97331-4003, United States
| |
Collapse
|
17
|
Kong H, Liu X, Yang L, Qi K, Zhang H, Zhang J, Huang Z, Wang H. All-trans retinoic acid enhances bystander effect of suicide gene therapy in the treatment of breast cancer. Oncol Rep 2015; 35:1868-74. [PMID: 26717879 DOI: 10.3892/or.2015.4535] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Accepted: 11/24/2015] [Indexed: 11/05/2022] Open
Abstract
All-trans retinoic acid (ATRA) has been shown to enhance the expression of connexin 43 (Cx43) and the bystander effect (BSE) in suicide gene therapy. These in turn improve effects of suicide gene therapies for several tumor types. However, whether ATRA can improve BSE remains unclear in suicide gene therapy for breast cancer. In the present study, MCF-7, human breast cancer cells were treated with ATRA in combination with a VEGFP-TK/CD gene suicide system developed by our group. We found that this combination enhances the efficiency of cell killing and apoptosis of breast cancer by strengthening the BSE in vitro. ATRA also promotes gap junction intercellular communication (GJIC) in MCF-7 cells by upregulation of the connexin 43 mRNA and protein in MCF-7 cells. These results indicate that enhancement of GJIC by ATRA in suicide gene system might serve as an attractive and cost-effective strategy of therapy for breast cancer cells.
Collapse
Affiliation(s)
- Heng Kong
- Department of Thyroid and Breast Surgery, Shenzhen Key Laboratory for Endogenous Infection, Shenzhen Nanshan District People's Hospital (The Sixth People's Hospital of Shenzhen), Shenzhen, Guangdong 518052, P.R. China
| | - Xia Liu
- Department of Human Resource, Shenzhen Nanshan District People's Hospital, Shenzhen, Guangdong 518052, P.R. China
| | - Liucheng Yang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Ke Qi
- Department of Thyroid and Breast Surgery, Shenzhen Key Laboratory for Endogenous Infection, Shenzhen Nanshan District People's Hospital (The Sixth People's Hospital of Shenzhen), Shenzhen, Guangdong 518052, P.R. China
| | - Haoyun Zhang
- Department of Thyroid and Breast Surgery, Shenzhen Key Laboratory for Endogenous Infection, Shenzhen Nanshan District People's Hospital (The Sixth People's Hospital of Shenzhen), Shenzhen, Guangdong 518052, P.R. China
| | - Jingwen Zhang
- Clinical Laboratory, Shenzhen Nanshan District People's Hospital, Shenzhen, Guangdong 518052, P.R. China
| | - Zonghai Huang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Hongxian Wang
- Department of Thyroid and Breast Surgery, Shenzhen Key Laboratory for Endogenous Infection, Shenzhen Nanshan District People's Hospital (The Sixth People's Hospital of Shenzhen), Shenzhen, Guangdong 518052, P.R. China
| |
Collapse
|
18
|
Gao K, Chi Y, Zhang X, Zhang H, Li G, Sun W, Takeda M, Yao J. A novel TXNIP-based mechanism for Cx43-mediated regulation of oxidative drug injury. J Cell Mol Med 2015; 19:2469-80. [PMID: 26154105 PMCID: PMC4594688 DOI: 10.1111/jcmm.12641] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 05/28/2015] [Indexed: 01/28/2023] Open
Abstract
Gap junctions (GJs) play an important role in the regulation of cell response to many drugs. However, little is known about their mechanisms. Using an in vitro model of cytotoxicity induced by geneticin (G418), we explored the potential signalling mechanisms involved. Incubation of cells with G418 resulted in cell death, as indicated by the change in cell morphology, loss of cell viability and activation of caspase-3. Before the onset of cell injury, G418 induced reactive oxygen species (ROS) generation, activated oxidative sensitive kinase P38 and caused a shift of connexin 43 (Cx43) from non-phosphorylated form to hyperphosphorylated form. These changes were largely prevented by antioxidants, suggesting an implication of oxidative stress. Downregulation of Cx43 with inhibitors or siRNA suppressed the expression of thioredoxin-interacting protein (TXNIP), activated Akt and protected cells against the toxicity of G418. Further analysis revealed that inhibition of TXNIP with siRNA activated Akt and reproduced the protective effect of Cx43-inhibiting agents, whereas suppression of Akt sensitized cells to the toxicity of G418. Furthermore, interference of TXNIP/Akt also affected puromycin- and adriamycin-induced cell injury. Our study thus characterized TXNIP as a presently unrecognized molecule implicated in the regulatory actions of Cx43 on oxidative drug injury. Targeting Cx43/TXNIP/Akt signalling cascade might be a promising approach to modulate cell response to drugs.
Collapse
Affiliation(s)
- Kun Gao
- Department of Molecular Signaling, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan.,Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yuan Chi
- Department of Molecular Signaling, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Xiling Zhang
- Department of Molecular Signaling, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Hui Zhang
- Department of Molecular Signaling, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Gang Li
- Department of Urology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan.,Department of Urology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Wei Sun
- Department of Nephrology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Masayuki Takeda
- Department of Urology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Jian Yao
- Department of Molecular Signaling, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Chuo, Yamanashi, Japan
| |
Collapse
|
19
|
Zhang K, Wang X, Wang H. Effect and mechanism of Src tyrosine kinase inhibitor sunitinib on the drug-resistance reversal of human A549/DDP cisplatin-resistant lung cancer cell line. Mol Med Rep 2014; 10:2065-72. [PMID: 25109654 DOI: 10.3892/mmr.2014.2440] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 05/09/2014] [Indexed: 11/05/2022] Open
Abstract
The development of tumor cell drug resistance is the primary reason for treatment failure in lung cancer chemotherapy. Therefore, overcoming multidrug resistance is currently an urgent issue to be addressed in lung cancer treatment. Sunitinib is a tyrosine kinase inhibitor with confirmed inhibitory effects on tumor growth and metastasis; however, the effects of sunitinib and mechanisms of action in lung cancer multidrug resistance are yet to be determined. The present study was designed to examine the effects of sunitinib and the mechanisms underlying lung cancer multidrug resistance. It was observed that sunitinib was able to improve the sensitivity of A549/DDP lung cancer cells to cisplatin, enhance tumor apoptosis, arrest the cell cycle in G0/G1 phase, upregulate intracellular Rh-123 content, downregulate the expression of P-glycoprotein, multidrug resistance protein 1, multidrug resistance-associated protein 1, lung resistance protein, glutathione-S-transferase, ERCC1, survivin and Bcl-2 in tumor cells, phosphorylation of AKT and extracellular signal-regulated kinase (ERK), glutathione activity, and transcriptional activity of nuclear factor-κB, Twist, Snail and AP-1. The results demonstrated that sunitinib was able to reverse the multidrug resistance of A549/DDP lung cancer cells, which was possibly associated with the downregulation of multidrug resistance-associated gene expression and the inhibition of AKT and ERK phosphorylation.
Collapse
Affiliation(s)
- Ke Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Xian Wang
- Department of Thoracic Surgery, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| | - Hongyan Wang
- Department of Thoracic Surgery, The First Affiliated Hospital, Henan University of Science and Technology, Luoyang, Henan 471003, P.R. China
| |
Collapse
|
20
|
Comparison of Intracellular Stress Response of NCI-H526 Small Cell Lung Cancer (SCLC) Cells to Platinum(II) Cisplatin and Platinum(IV) Oxoplatin. Cancers (Basel) 2014; 6:1487-99. [PMID: 25006835 PMCID: PMC4190551 DOI: 10.3390/cancers6031487] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 06/30/2014] [Accepted: 07/02/2014] [Indexed: 12/18/2022] Open
Abstract
In attempts to develop an orally applicable platinum-based drug, platinum(IV) drugs which exhibit higher in vivo stability compared to the platinum(II) drug cisplatin were formulated. The first such chemotherapeutic agent, namely satraplatin, failed to receive approval. In the present work, we checked the initial cellular stress response of the chemosensitive NCI-H526 small cell lung cancer (SCLC) cells by determination of the relative phosphorylation of 46 specific phosphorylation sites of 38 selected proteins in a six hours response to cisplatin (platinum(II)) or oxoplatin (platinum(IV)), respectively. Oxoplatin is considered as prodrug of cisplatin, although several findings point to differences in intracellular effects. Cisplatin induced hyperphosphorylation of p38α MAPK and AMPKα1, whereas oxoplatin treatment resulted in increased phosphorylation of a large number of signaling proteins involved in stress response/drug resistance, including JNK, GSK-3α, AMPKα1, src kinases, STATs, CHK-2 and especially focal adhesion kinase (FAK). Cisplatin exerts markedly higher cytotoxicity upon four hours short-term exposure in comparison to oxoplatin and, correspondingly, the extended initial stress response to the platinum(IV) drug oxoplatin thus is expected to increase clinical drug resistance. Induction of a substantial stress response to any prodrug of a platinum-based compound may likewise limit the effectivity of its active metabolite(s), such contributing to the failure of selected derivatized platinum complexes.
Collapse
|
21
|
Liu T, Hu W, Dalton HJ, Choi HJ, Huang J, Kang Y, Pradeep S, Miyake T, Song JH, Wen Y, Lu C, Pecot CV, Bottsford-Miller J, Zand B, Jennings NB, Ivan C, Gallick GE, Baggerly KA, Hangauer DG, Coleman RL, Frumovitz M, Sood AK. Targeting SRC and tubulin in mucinous ovarian carcinoma. Clin Cancer Res 2013; 19:6532-43. [PMID: 24100628 PMCID: PMC3852199 DOI: 10.1158/1078-0432.ccr-13-1305] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
PURPOSE To investigate the antitumor effects of targeting Src and tubulin in mucinous ovarian carcinoma. EXPERIMENTAL DESIGN The in vitro and in vivo effects and molecular mechanisms of KX-01, which inhibits Src pathway and tubulin polymerization, were examined in mucinous ovarian cancer models. RESULTS In vitro studies using RMUG-S and RMUG-L cell lines showed that KX-01 inhibited cell proliferation, induced apoptosis, arrested the cell cycle at the G2-M phase, and enhanced the cytotoxicity of oxaliplatin in the KX-01-sensitive cell line, RMUG-S. In vivo studies showed that KX-01 significantly decreased tumor burden in RMUG-S and RMUG-L mouse models relative to untreated controls, and the effects were greater when KX-01 was combined with oxaliplatin. KX-01 alone and in combination with oxaliplatin significantly inhibited tumor growth by reducing cell proliferation and inducing apoptosis in vivo. PTEN knock-in experiments in RMUG-L cells showed improved response to KX-01. Reverse phase protein array analysis showed that in addition to blocking downstream molecules of Src family kinases, KX-01 also activated acute stress-inducing molecules. CONCLUSION Our results showed that targeting both the Src pathway and tubulin with KX-01 significantly inhibited tumor growth in preclinical mucinous ovarian cancer models, suggesting that this may be a promising therapeutic approach for patients with mucinous ovarian carcinoma.
Collapse
Affiliation(s)
- Tao Liu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of General Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Wei Hu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Heather J. Dalton
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Hyun Jin Choi
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jie Huang
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yu Kang
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Gynecology, Obstetrics and Gynecology, Hospital of Fudan University, Shanghai 20001, P.R. China
| | - Sunila Pradeep
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Takahito Miyake
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jian H. Song
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yunfei Wen
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chunhua Lu
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chad V. Pecot
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Justin Bottsford-Miller
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Behrouz Zand
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nicholas B Jennings
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Cristina Ivan
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gary E. Gallick
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Keith A Baggerly
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - David G. Hangauer
- Kinex Pharmaceuticals LLC, New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY 14203, USA
| | - Robert L. Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael Frumovitz
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Anil K. Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
22
|
Choi S, Kim S, Lee J, Lim H, Kim Y, Tian C, So H, Park R, Choung Y.H. Gingko biloba extracts protect auditory hair cells from cisplatin-induced ototoxicity by inhibiting perturbation of gap junctional intercellular communication. Neuroscience 2013; 244:49-61. [DOI: 10.1016/j.neuroscience.2013.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2012] [Revised: 03/30/2013] [Accepted: 04/01/2013] [Indexed: 12/23/2022]
|
23
|
Lv J, Tian Y. [Effect of Src tyrosine kinase inhibition on the drug-resistance as well as MDR1 and LRP expression of the human cis-platinum-resistant lung cancer cell line A549/DDP]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2013; 15:501-6. [PMID: 22989452 PMCID: PMC5999856 DOI: 10.3779/j.issn.1009-3419.2012.09.01] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND The aim of this study is to investigate the effect of Src tyrosine kinase inhibition on the drug-resistance as well as the expression of multidrug resistance 1 (MDR1) and lung resistance-related protein (LRP) of the human cis-platinum-resistant lung cancer cell line A549/DDP. METHODS 4-Anilinoquirazoline was used to inhibit Src tyrosine kinase activity in A549/DDP. Western blot analysis was used to detect the Src tyrosine kinase activity. CellTiter-Glo assay was used to detect the drug sensitivity of tumor cells. Flow cytometry was used to detect the intracellular Rh-123 content. Western blot and real-time PCR assay were used to detect the expression of tumor MDR1 and LRP. RESULTS 4-Anilinoquirazoline can down-regulate the cellular Src tyrosine kinase activity in A549/DDP. After treatment with 2.5 μM and 10 μM of 4-anilinoquirazoline, the cells became more sensitive to the drug and the reversal folds (RFs) of tumor cell sensitivity to the drug were 1.59- and 2.10-fold, respectively. The intracellular content of Rh-123 improved by 1.21- and 1.59-fold, respectively. The mRNA levels of MDR1 were 53.8% and 27.5% of the control, respectively. The mRNA level of LRP was 59.3% and 21.4% of the control, respectively. The expression of MDR1 and LRP protein significantly decreased. CONCLUSIONS The inhibition of Src tyrosine kinase activity in A549/DDP cells can reverse multi-drug resistance and increase the sensitivity of the cells to the drug. The mechanism may be related to the down-regulation of cellular MDR1 and LRP.
Collapse
Affiliation(s)
- Jie Lv
- Department of Laboratory Medicine, Rizhao People's Hospital, Rizhao 276826, China.
| | | |
Collapse
|
24
|
Shishido SN, Nguyen TA. Gap junction enhancer increases efficacy of cisplatin to attenuate mammary tumor growth. PLoS One 2012; 7:e44963. [PMID: 23028705 PMCID: PMC3441663 DOI: 10.1371/journal.pone.0044963] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 08/10/2012] [Indexed: 02/05/2023] Open
Abstract
Cisplatin treatment has an overall 19% response rate in animal models with malignant tumors. Increasing gap junction activity in tumor cells provides the targets to enhance antineoplastic therapies. Previously, a new class of substituted quinolines (PQs) acts as gap junction enhancer, ability to increase the gap junctional intercellular communication, in breast cancer cells. We examined the effect of combinational treatment of PQs and antineoplastic drugs in an animal model, showing an increase in efficacy of antineoplastic drugs via the enhancement of gap junctions. Mice were implanted with estradiol-17ß (1.7 mg/pellet) before the injection of 1×107 T47D breast cancer cells subcutaneously into the inguinal region of mammary fat pad. Animals were treated intraperitoneally with DMSO (control), cisplatin (3.5 mg/kg), PQ (25 mg/kg), or a combining treatment of cisplatin and PQ. Cisplatin alone decreased mammary tumor growth by 85% while combinational treatment of cisplatin and PQ1 or PQ7 showed an additional reduction of 77% and 22% of tumor growth after 7 treatments at every 2 days, respectively. Histological results showed a significant increase of gap junction proteins, Cx43 and Cx26, in PQ-treated tissues compared to control or cisplatin. Furthermore, evidence of highly stained caspase 3 in tumors of combinational treatment (PQ and cisplatin) was seen compared to cisplatin alone. We have showed for the first time an increase in the efficacy of antineoplastic drugs through a combinational treatment with PQs, a specific class of gap junction enhancers.
Collapse
Affiliation(s)
- Stephanie N. Shishido
- Departments of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, Kansas, United States of America
| | - Thu A. Nguyen
- Departments of Diagnostic Medicine/Pathobiology, Kansas State University, Manhattan, Kansas, United States of America
- * E-mail:
| |
Collapse
|
25
|
Kaye S, Aamdal S, Jones R, Freyer G, Pujade-Lauraine E, de Vries EGE, Barriuso J, Sandhu S, Tan DSW, Hartog V, Kuenen B, Ruijter R, Kristensen GB, Nyakas M, Barrett S, Burke W, Pietersma D, Stuart M, Emeribe U, Boven E. Phase I study of saracatinib (AZD0530) in combination with paclitaxel and/or carboplatin in patients with solid tumours. Br J Cancer 2012; 106:1728-34. [PMID: 22531637 PMCID: PMC3364128 DOI: 10.1038/bjc.2012.158] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: As a prelude to combination studies aimed at resistance reversal, this dose-escalation/dose-expansion study investigated the selective Src kinase inhibitor saracatinib (AZD0530) in combination with carboplatin and/or paclitaxel. Methods: Patients with advanced solid tumours received saracatinib once-daily oral tablets in combination with either carboplatin AUC 5 every 3 weeks (q3w), paclitaxel 175 mg m−2 q3w, paclitaxel 80 mg m−2 every 1 week (q1w), or carboplatin AUC 5 plus paclitaxel 175 mg m−2 q3w. The primary endpoint was safety/tolerability. Results: A total of 116 patients received saracatinib 125 (N=20), 175 (N=44), 225 (N=40), 250 (N=9), or 300 mg (N=3). There were no clear dose-related trends within each chemotherapy regimen group in number or severity of adverse events (AEs). However, combining all groups, the occurrence of grade ⩾3 asthenic AEs (all causality) was dose-related (125 mg, 10% 175 mg, 20% ⩾225 mg, 33%), and grade ⩾3 neutropenia occurred more commonly at doses ⩾225 mg. There was no evidence that saracatinib affected exposure to carboplatin or paclitaxel, or vice versa. Objective responses were seen in 5 out of 44 patients (11%) receiving carboplatin plus paclitaxel q3w, and 5 out of 24 (21%) receiving paclitaxel q1w. Conclusion: Saracatinib doses up to 175 mg with paclitaxel with/without carboplatin showed acceptable toxicity in most patients, and are suitable for further trials.
Collapse
Affiliation(s)
- S Kaye
- Drug Development Unit, Royal Marsden Hospital, Downs Road, Sutton, Surrey SM2 5PT, UK.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Ding Y, Nguyen TA. Gap Junction Enhancer Potentiates Cytotoxicity of Cisplatin in Breast Cancer Cells. ACTA ACUST UNITED AC 2012; 4:371-378. [PMID: 25045421 DOI: 10.4172/1948-5956.1000170] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cisplatin is one of the most widely used anti-cancer drugs due to its ability to damage DNA and induce apoptosis. However, increasing reports of side effects and drug resistance indicate the limitation of cisplatin in cancer therapeutics. Recent studies showed that inhibition of gap junctions diminishes the cytotoxic effect and contributes to drug resistance. Therefore, identification of molecules that counteract gap junctional inhibition without decreasing the anti-cancer effect of cisplatin could be used in combinational treatment, potentiating cisplatin efficacy and preventing resistance. This study investigates the effects of combinational treatment of cisplatin and PQ1, a gap junction enhancer, in T47D breast cancer cells. Our results showed that combinational treatment of PQ1 and cisplatin increased gap junctional intercellular communication (GJIC) as well as expressions of connexins (Cx26, Cx32 and Cx43), and subsequently decreased cell viability. Ki67, a proliferation marker, was decreased by 75% with combinational treatment. Expressions of pro-apoptotic factors (cleaved caspase-3/-8/-9 and bax) were increased by the combinational treatment with PQ1 and cisplatin; whereas, the pro-survival factor, bcl-2, was decreased by the combinational treatment. Our study demonstrates for the first time that the combinational treatment with gap junction enhancers can counteract cisplatin induced inhibition of gap junctional intercellular communication and reduction of connexin expression, thereby increasing the efficacy of cisplatin in cancer cells.
Collapse
Affiliation(s)
- Ying Ding
- Department of Biochemistry, Kansas State University, Manhattan, Kansas, USA
| | - Thu Annelise Nguyen
- Department of Diagnostic Medicine/ Pathobiology, Kansas State University, Manhattan, Kansas, USA
| |
Collapse
|
27
|
Abstract
Genetic interactions are functional crosstalk among different genetic loci that lead to phenotypic changes, such as health or viability alterations. A disease or lethal phenotype that results from the combined effects of gene mutations at different loci is termed a synthetic sickness or synthetic lethality, respectively. Studies of genetic interaction have provided insight on the relationships among biochemical processes or pathways. Cancer results from genetic interactions and is a major focus of current studies in genetic interactions. Various basic and translational cancer studies have explored the concept of genetic interactions, including studies of the mechanistic characterization of genes, drug discovery, biomarker identification and the rational design of combination therapies. This review discusses the implications of genetic interactions in the development of personalized cancer therapies, the identification of treatment-responsive genes, the delineation of mechanisms of chemoresistance and the rational design of combined therapeutic strategies to overcome drug resistance.
Collapse
|
28
|
Matsuo K, Nishimura M, Bottsford-Miller JN, Huang1 J, Komurov K, Armaiz-Pena GN, Shahzad MMK, Stone RL, Roh JW, Sanguino AM, Lu C, Im DD, Rosenshien NB, Sakakibara A, Nagano T, Yamasaki M, Enomoto T, Kimura T, Ram PT, Schmeler KM, Gallick GE, Wong KK, Frumovitz M, Sood AK. Targeting SRC in mucinous ovarian carcinoma. Clin Cancer Res 2011; 17:5367-78. [PMID: 21737505 PMCID: PMC4028171 DOI: 10.1158/1078-0432.ccr-10-3176] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE Mucinous ovarian carcinomas have a distinct clinical pattern compared with other subtypes of ovarian carcinoma. Here, we evaluated (i) stage-specific clinical significance of mucinous ovarian carcinomas in a large cohort and (ii) the functional role of Src kinase in preclinical models of mucinous ovarian carcinoma. EXPERIMENTAL DESIGN A total of 1,302 ovarian cancer patients including 122 (9.4%) cases of mucinous carcinoma were evaluated for survival analyses. Biological effects of Src kinase inhibition were tested using dasatinib-based therapy in a novel orthotopic mucinous ovarian cancer model (RMUG-S-ip2). RESULTS Patients with advanced-stage mucinous ovarian cancer had significantly worse survival than those with serous histology: median overall survival, 1.67 versus 3.41 years, P = 0.002; median survival time after recurrence of 0.53 versus 1.66 years, P < 0.0001. Among multiple ovarian cancer cell lines, RMUG-S-ip2 mucinous ovarian cancer cells showed the highest Src kinase activity. Moreover, oxaliplatin treatment induced phosphorylation of Src kinase. This induced activity by oxaliplatin therapy was inhibited by concurrent administration of dasatinib. Targeting Src with dasatinib in vivo showed significant antitumor effects in the RMUG-S-ip2 model but not in the serous ovarian carcinoma (SKOV3-TR) model. Combination therapy of oxaliplatin with dasatinib further showed significant effects on reducing cell viability, increasing apoptosis, and in vivo antitumor effects in the RMUG-S-ip2 model. CONCLUSIONS Our results suggest that poor survival of women with mucinous ovarian carcinoma is associated with resistance to cytotoxic therapy. Targeting Src kinase with a combination of dasatinib and oxaliplatin may be an attractive approach for this disease.
Collapse
Affiliation(s)
- Koji Matsuo
- Department of Gynecologic Oncology and Reproductive Medicine, MD-Anderson Cancer Center, University of Texas, Houston, TX, USA
- Department of Obstetrics and Gynecology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Masato Nishimura
- Department of Gynecologic Oncology and Reproductive Medicine, MD-Anderson Cancer Center, University of Texas, Houston, TX, USA
- Department of Obstetrics and Gynecology, University of Tokushima, Japan
| | - Justin N. Bottsford-Miller
- Department of Gynecologic Oncology and Reproductive Medicine, MD-Anderson Cancer Center, University of Texas, Houston, TX, USA
| | | | - Kakajan Komurov
- Systems Biology, MD-Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Guillermo N. Armaiz-Pena
- Department of Gynecologic Oncology and Reproductive Medicine, MD-Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Mian M. K. Shahzad
- Department of Gynecologic Oncology and Reproductive Medicine, MD-Anderson Cancer Center, University of Texas, Houston, TX, USA
- Department of Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Rebecca L. Stone
- Department of Gynecologic Oncology and Reproductive Medicine, MD-Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Ju Won Roh
- Department of Gynecologic Oncology and Reproductive Medicine, MD-Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Angela M. Sanguino
- Department of Gynecologic Oncology and Reproductive Medicine, MD-Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Chunhua Lu
- Department of Gynecologic Oncology and Reproductive Medicine, MD-Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Dwight D. Im
- The Gynecologic Oncology Center, Mercy Medical Center, Baltimore, MD, USA
| | - Neil B. Rosenshien
- The Gynecologic Oncology Center, Mercy Medical Center, Baltimore, MD, USA
| | - Atsuko Sakakibara
- Department of Obstetrics and Gynecology, Kitano Hospital, Osaka, Japan
| | - Tadayoshi Nagano
- Department of Obstetrics and Gynecology, Kitano Hospital, Osaka, Japan
| | - Masato Yamasaki
- Department of Obstetrics and Gynecology, Osaka Rosai Hospital, Osaka, Japan
| | - Takayuki Enomoto
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tadashi Kimura
- Department of Obstetrics and Gynecology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Prahlad T. Ram
- Systems Biology, MD-Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Kathleen M. Schmeler
- Department of Gynecologic Oncology and Reproductive Medicine, MD-Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Gary E. Gallick
- Cancer Biology, MD-Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Kwong K. Wong
- Department of Gynecologic Oncology and Reproductive Medicine, MD-Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Michael Frumovitz
- Department of Gynecologic Oncology and Reproductive Medicine, MD-Anderson Cancer Center, University of Texas, Houston, TX, USA
| | - Anil K. Sood
- Department of Gynecologic Oncology and Reproductive Medicine, MD-Anderson Cancer Center, University of Texas, Houston, TX, USA
- Cancer Biology, MD-Anderson Cancer Center, University of Texas, Houston, TX, USA
- Center for RNA Interference and non-Coding RNA, University of Texas, Houston, TX, USA
| |
Collapse
|
29
|
Eberle KE, Sansing HA, Szaniszlo P, Resto VA, Berrier AL. Carcinoma matrix controls resistance to cisplatin through talin regulation of NF-kB. PLoS One 2011; 6:e21496. [PMID: 21720550 PMCID: PMC3123362 DOI: 10.1371/journal.pone.0021496] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 05/30/2011] [Indexed: 12/31/2022] Open
Abstract
Extracellular matrix factors within the tumor microenvironment that control resistance to chemotherapeutics are poorly understood. This study focused on understanding matrix adhesion pathways that control the oral carcinoma response to cisplatin. Our studies revealed that adhesion of HN12 and JHU012 oral carcinomas to carcinoma matrix supported tumor cell proliferation in response to treatment with cisplatin. Proliferation in response to 30 µM cisplatin was not observed in HN12 cells adherent to other purified extracellular matrices such as Matrigel, collagen I, fibronectin or laminin I. Integrin β1 was important for adhesion to carcinoma matrix to trigger proliferation after treatment with cisplatin. Disruption of talin expression in HN12 cells adherent to carcinoma matrix increased cisplatin induced proliferation. Pharmacological inhibitors were used to determine signaling events required for talin deficiency to regulate cisplatin induced proliferation. Pharmacological inhibition of NF-kB reduced proliferation of talin-deficient HN12 cells treated with 30 µM cisplatin. Nuclear NF-kB activity was assayed in HN12 cells using a luciferase reporter of NF-kB transcriptional activity. Nuclear NF-kB activity was similar in HN12 cells adherent to carcinoma matrix and collagen I when treated with vehicle DMSO. Following treatment with 30 µM cisplatin, NF-kB activity is maintained in cells adherent to carcinoma matrix whereas NF-kB activity is reduced in collagen I adherent cells. Expression of talin was sufficient to trigger proliferation of HN12 cells adherent to collagen I following treatment with 1 and 30 µM cisplatin. Talin overexpression was sufficient to trigger NF-kB activity following treatment with cisplatin in carcinoma matrix adherent HN12 cells in a process disrupted by FAK siRNA. Thus, adhesions within the carcinoma matrix create a matrix environment in which exposure to cisplatin induces proliferation through the function of integrin β1, talin and FAK pathways that regulate NF-kB nuclear activity.
Collapse
Affiliation(s)
- Karen E. Eberle
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center-New Orleans, New Orleans, Louisiana, United States of America
| | - Hope A. Sansing
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center-New Orleans, New Orleans, Louisiana, United States of America
| | - Peter Szaniszlo
- Department of Otolaryngology, University of Texas Medical Branch Cancer Center, University of Texas Medical Branch Health, Galveston, Texas, United States of America
| | - Vicente A. Resto
- Department of Otolaryngology, University of Texas Medical Branch Cancer Center, University of Texas Medical Branch Health, Galveston, Texas, United States of America
| | - Allison L. Berrier
- Department of Oral and Craniofacial Biology, School of Dentistry, Louisiana State University Health Sciences Center-New Orleans, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
30
|
Kandouz M, Batist G. Gap junctions and connexins as therapeutic targets in cancer. Expert Opin Ther Targets 2010; 14:681-92. [PMID: 20446866 DOI: 10.1517/14728222.2010.487866] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
IMPORTANCE OF THE FIELD Connexins (Cxs) and gap junctional intercellular communications (GJICs) play roles in cancer development, growth and metastasis. Experimental studies suggest that targeting Cxs may be a novel technique, either to inhibit tumor cell growth directly or to sensitize to various therapies. AREAS COVERED IN THIS REVIEW A brief introduction to the role of Cxs in cancer. The focus is mainly on data available in the literature regarding therapeutic aspects. WHAT THE READER WILL GAIN This article reviews the various strategies that take advantage of gap junctions and connexins to eliminate cancer cells, including use of the bystander effect (BE) in gene therapy, the effect of connexins on chemosensitization, the role of apoptotic processes and interactions with the microenvironment. Attempts to restore connexin expression at the transcriptional and post-transcriptional levels are described, as well as promising strategies recently explored. The potential and limitations of the approaches are discussed. TAKE HOME MESSAGE Connexins have multiple facets, singly, in hemichannel complexes, in gap junctions or interacting with different proteins. The regulation of their expression is not fully resolved and selective manipulation of Cxs expression is therefore a challenge. Although the therapeutic potential of connexins is undeniable, more effort is needed to study the regulation and functions of these proteins.
Collapse
Affiliation(s)
- Mustapha Kandouz
- Wayne State University, Department of Pathology, 5101 Cass Avenue, Chemistry Building, Detroit, Michigan 48202, USA.
| | | |
Collapse
|
31
|
Wang Q, You T, Yuan D, Han X, Hong X, He B, Wang L, Tong X, Tao L, Harris AL. Cisplatin and oxaliplatin inhibit gap junctional communication by direct action and by reduction of connexin expression, thereby counteracting cytotoxic efficacy. J Pharmacol Exp Ther 2010; 333:903-11. [PMID: 20215407 DOI: 10.1124/jpet.109.165274] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Cisplatin [cis-diamminedichloroplatinum(II)]/oxaliplatin [1,2-diamminocyclohexane(trans-1)oxolatoplatinum(II)] toxicity is enhanced by functional gap junctions between treated cells, implying that inhibition of gap junctions may decrease cytotoxic activity of these platinum-based agents. This study investigates the effect of gap junction modulation by cisplatin/oxaliplatin on cytotoxicity in a transformed cell line. The effects were explored using junctional channels expressed in transfected HeLa cells and purified hemichannels. Junctional channels showed a rapid, dose-dependent decrease in dye coupling with exposure to cisplatin/oxaliplatin. With longer exposure, both compounds also decreased connexin expression. Both compounds inhibit the activity of purified connexin hemichannels, over the same concentration range that they inhibit junctional dye permeability, demonstrating that inhibition occurs by direct interaction of the drugs with connexin protein. Cisplatin/oxaliplatin reduced the clonogenic survival of HeLa cells at low density and high density in a dose-dependent manner, but to a greater degree at high density, consistent with a positive effect of gap junctional intercellular communication (GJIC) on cytotoxicity. Reduction of GJIC by genetic or pharmacological means decreased cisplatin/oxaliplatin toxicity. At low cisplatin/oxaliplatin concentrations, where effects on connexin channels are minimal, the toxicity increased with increased cell density. However, higher concentrations strongly inhibited GJIC, and this counteracted the enhancing effect of greater cell density on toxicity. The present results indicate that inhibition of GJIC by cisplatin/oxaliplatin decreases their cytotoxicity. Direct inhibition of GJIC and reduction of connexin expression by cisplatin/oxaliplatin may thereby compromise the effectiveness of these compounds and be a factor in the development of resistance to this class of chemotherapeutic agents.
Collapse
Affiliation(s)
- Qin Wang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Lieu C, Kopetz S. The SRC family of protein tyrosine kinases: a new and promising target for colorectal cancer therapy. Clin Colorectal Cancer 2010; 9:89-94. [PMID: 20378502 PMCID: PMC3091503 DOI: 10.3816/ccc.2010.n.012] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Aberrant activation of the Src family of tyrosine kinases has been implicated in the development and progression of colorectal cancer (CRC). As a result, Src inhibitors are now being studied as possible therapeutic agents to treat metastatic disease. In this review, we discuss the effects of aberrant Src activation in CRC, Src as a target of single-agent drug therapy, and Src as a target of combination therapy with epidermal growth factor receptor inhibition and cytotoxic chemotherapy. The greatest potential for clinically relevant benefit most likely lies in combination regimens. Further evaluation with biomarkers will continue to define the molecular phenotype of patients with CRC who will benefit the most from Src-based therapy.
Collapse
Affiliation(s)
- Christopher Lieu
- Department of Gastrointestinal Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA
| | | |
Collapse
|