1
|
Nalla LV, Kanukolanu A, Yeduvaka M, Gajula SNR. Advancements in Single-Cell Proteomics and Mass Spectrometry-Based Techniques for Unmasking Cellular Diversity in Triple Negative Breast Cancer. Proteomics Clin Appl 2025; 19:e202400101. [PMID: 39568435 PMCID: PMC11726282 DOI: 10.1002/prca.202400101] [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: 10/11/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is an aggressive and complex subtype of breast cancer characterized by a lack of targeted treatment options. Intratumoral heterogeneity significantly drives disease progression and complicates therapeutic responses, necessitating advanced analytical approaches to understand its underlying biology. This review aims to explore the advancements in single-cell proteomics and their application in uncovering cellular diversity in TNBC. It highlights innovations in sample preparation, mass spectrometry-based techniques, and the potential for integrating proteomics into multi-omics platforms. METHODS The review discusses the combination of improved sample preparation methods and cutting-edge mass spectrometry techniques in single-cell proteomics. It emphasizes the challenges associated with protein analysis, such as the inability to amplify proteins akin to transcripts, and examines strategies to overcome these limitations. RESULTS Single-cell proteomics provides a direct link to phenotype and cell behavior, complementing transcriptomic approaches and offering new insights into the mechanisms driving TNBC. The integration of advanced techniques has enabled deeper exploration of cellular heterogeneity and disease mechanisms. CONCLUSION Despite the challenges, single-cell proteomics holds immense potential to evolve into a high-throughput and scalable multi-omics platform. Addressing existing hurdles will enable deeper biological insights, ultimately enhancing the diagnosis and treatment of TNBC.
Collapse
Affiliation(s)
- Lakshmi Vineela Nalla
- Department of Pharmacology, GITAM School of PharmacyGITAM (Deemed to be University)VisakhapatnamAndhra PradeshIndia
| | - Aarika Kanukolanu
- Department of Pharmaceutical Analysis, GITAM School of PharmacyGITAM (Deemed to be University)VisakhapatnamAndhra PradeshIndia
| | - Madhuri Yeduvaka
- Department of Pharmacology, GITAM School of PharmacyGITAM (Deemed to be University)VisakhapatnamAndhra PradeshIndia
| | - Siva Nageswara Rao Gajula
- Department of Pharmaceutical Analysis, GITAM School of PharmacyGITAM (Deemed to be University)VisakhapatnamAndhra PradeshIndia
| |
Collapse
|
2
|
Yan Z, Huang H, Wang Q, Kong Y, Liu X. Function and mechanism of action of the TRPV1 channel in the development of triple-negative breast cancer. Acta Biochim Biophys Sin (Shanghai) 2024; 56:957-962. [PMID: 38734935 PMCID: PMC11322878 DOI: 10.3724/abbs.2024068] [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: 11/26/2023] [Accepted: 03/25/2024] [Indexed: 05/13/2024] Open
Abstract
Transient receptor potential channel subfamily vanilloid 1 (TRPV1) is a member of the transient receptor potential family of nonselective cationic transmembrane channel proteins that are involved in the regulation of calcium homeostasis. It is expressed in various tumor types and has been implicated in the regulation of cancer growth, metastasis, apoptosis, and cancer-related pain. TRPV1 is highly expressed in triple-negative breast cancer (TNBC), and both its agonists and antagonists may exert anti-cancer effects. In this review, we provide an overview of the effect of TRPV1 on TNBC development and its influence on immunotherapy in an attempt to facilitate the development of future treatment strategies.
Collapse
Affiliation(s)
- Ziling Yan
- />Pathology Departmentthe First Affiliated Hospital of Shenzhen UniversityShenzhen Second People’s HospitalShenzhen518035China
| | - Haihui Huang
- />Pathology Departmentthe First Affiliated Hospital of Shenzhen UniversityShenzhen Second People’s HospitalShenzhen518035China
| | - Qianqian Wang
- />Pathology Departmentthe First Affiliated Hospital of Shenzhen UniversityShenzhen Second People’s HospitalShenzhen518035China
| | - Yanjie Kong
- />Pathology Departmentthe First Affiliated Hospital of Shenzhen UniversityShenzhen Second People’s HospitalShenzhen518035China
| | - Xia Liu
- />Pathology Departmentthe First Affiliated Hospital of Shenzhen UniversityShenzhen Second People’s HospitalShenzhen518035China
| |
Collapse
|
3
|
Punetha A, Kotiya D. Advancements in Oncoproteomics Technologies: Treading toward Translation into Clinical Practice. Proteomes 2023; 11:2. [PMID: 36648960 PMCID: PMC9844371 DOI: 10.3390/proteomes11010002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
Proteomics continues to forge significant strides in the discovery of essential biological processes, uncovering valuable information on the identity, global protein abundance, protein modifications, proteoform levels, and signal transduction pathways. Cancer is a complicated and heterogeneous disease, and the onset and progression involve multiple dysregulated proteoforms and their downstream signaling pathways. These are modulated by various factors such as molecular, genetic, tissue, cellular, ethnic/racial, socioeconomic status, environmental, and demographic differences that vary with time. The knowledge of cancer has improved the treatment and clinical management; however, the survival rates have not increased significantly, and cancer remains a major cause of mortality. Oncoproteomics studies help to develop and validate proteomics technologies for routine application in clinical laboratories for (1) diagnostic and prognostic categorization of cancer, (2) real-time monitoring of treatment, (3) assessing drug efficacy and toxicity, (4) therapeutic modulations based on the changes with prognosis and drug resistance, and (5) personalized medication. Investigation of tumor-specific proteomic profiles in conjunction with healthy controls provides crucial information in mechanistic studies on tumorigenesis, metastasis, and drug resistance. This review provides an overview of proteomics technologies that assist the discovery of novel drug targets, biomarkers for early detection, surveillance, prognosis, drug monitoring, and tailoring therapy to the cancer patient. The information gained from such technologies has drastically improved cancer research. We further provide exemplars from recent oncoproteomics applications in the discovery of biomarkers in various cancers, drug discovery, and clinical treatment. Overall, the future of oncoproteomics holds enormous potential for translating technologies from the bench to the bedside.
Collapse
Affiliation(s)
- Ankita Punetha
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers University, 225 Warren St., Newark, NJ 07103, USA
| | - Deepak Kotiya
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 900 South Limestone St., Lexington, KY 40536, USA
| |
Collapse
|
4
|
Bett F, Brown S, Dong A, Christian M, Ajala S, Santiago K, Albin S, Marz A, Deo M. Optical Deformation of Biological Cells using Dual-Beam Laser Tweezer. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2022; 2022:17-20. [PMID: 36085603 DOI: 10.1109/embc48229.2022.9871373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Optical tweezer is a non-contact tool to trap and manipulate microparticles such as biological cells using coherent light beams. In this study, we utilized a dual-beam optical tweezer, created using two counterpropagating and slightly divergent laser beams to trap and deform biological cells. Human embryonic kidney 293 (HEK-293) and breast cancer (SKBR3) cells were used to characterize their membrane elasticity by optically stretching in the dual-beam optical tweezer. It was observed that the extent of deformation in both cell types increases with increasing optical trapping power. The SKBR3 cells exhibited greater percentage deformation than that of HEK-293 cells for a given trapping power. Our results demonstrate that the dual-beam optical tweezer provides measures of cell elasticity that can distinguish between various cell types. The non-contact optical cell stretching can be effectively utilized in disease diagnosis such as cancer based on the cell elasticity measures.
Collapse
|
5
|
Li Y, Kong X, Wang Z, Xuan L. Recent advances of transcriptomics and proteomics in triple-negative breast cancer prognosis assessment. J Cell Mol Med 2022; 26:1351-1362. [PMID: 35150062 PMCID: PMC8899180 DOI: 10.1111/jcmm.17124] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 11/18/2021] [Accepted: 12/01/2021] [Indexed: 12/24/2022] Open
Abstract
Triple-negative breast cancer (TNBC), a heterogeneous tumour that lacks the expression of oestrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2), is often characterized by aggressiveness and tends to recur or metastasize. TNBC lacks therapeutic targets compared with other subtypes and is not sensitive to endocrine therapy or targeted therapy except chemotherapy. Therefore, identifying the prognostic characteristics and valid therapeutic targets of TNBC could facilitate early personalized treatment. Due to the rapid development of various technologies, researchers are increasingly focusing on integrating 'big data' and biological systems, which is referred to as 'omics', as a means of resolving it. Transcriptomics and proteomics analyses play an essential role in exploring prospective biomarkers and potential therapeutic targets for triple-negative breast cancers, which provides a powerful engine for TNBC's therapeutic discovery when combined with complementary information. Here, we review the recent progress of TNBC research in transcriptomics and proteomics to identify possible therapeutic goals and improve the survival of patients with triple-negative breast cancer. Also, researchers may benefit from this article to catalyse further analysis and investigation to decipher the global picture of TNBC cancer.
Collapse
Affiliation(s)
- Yuan Li
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhongzhao Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lixue Xuan
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
6
|
Gwark S, Ahn HS, Yeom J, Yu J, Oh Y, Jeong JH, Ahn JH, Jung KH, Kim SB, Lee HJ, Gong G, Lee SB, Chung IY, Kim HJ, Ko BS, Lee JW, Son BH, Ahn SH, Kim K, Kim J. Plasma Proteome Signature to Predict the Outcome of Breast Cancer Patients Receiving Neoadjuvant Chemotherapy. Cancers (Basel) 2021; 13:6267. [PMID: 34944885 PMCID: PMC8699627 DOI: 10.3390/cancers13246267] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 12/31/2022] Open
Abstract
The plasma proteome of 51 non-metastatic breast cancer patients receiving neoadjuvant chemotherapy (NCT) was prospectively analyzed by high-resolution mass spectrometry coupled with nano-flow liquid chromatography using blood drawn at the time of diagnosis. Plasma proteins were identified as potential biomarkers, and their correlation with clinicopathological variables and survival outcomes was analyzed. Of 51 patients, 20 (39.2%) were HR+/HER2-, five (9.8%) were HR+/HER2+, five (9.8%) were HER2+, and 21 (41.2%) were triple-negative subtype. During a median follow-up of 52.0 months, there were 15 relapses (29.4%) and eight deaths (15.7%). Four potential biomarkers were identified among differentially expressed proteins: APOC3 had higher plasma concentrations in the pathological complete response (pCR) group, whereas MBL2, ENG, and P4HB were higher in the non-pCR group. Proteins statistically significantly associated with survival and capable of differentiating low- and high-risk groups were MBL2 and P4HB for disease-free survival, P4HB for overall survival, and MBL2 for distant metastasis-free survival (DMFS). In the multivariate analysis, only MBL2 was a consistent risk factor for DMFS (HR: 9.65, 95% CI 2.10-44.31). The results demonstrate that the proteomes from non-invasive sampling correlate with pCR and survival in breast cancer patients receiving NCT. Further investigation may clarify the role of these proteins in predicting prognosis and thus their therapeutic potential for the prevention of recurrence.
Collapse
Affiliation(s)
- Sungchan Gwark
- Department of Surgery, Ewha Womans University Mokdong Hospital, Ewha Womans University College of Medicine, Seoul 07985, Korea;
| | - Hee-Sung Ahn
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (H.-S.A.); (J.Y.); (Y.O.)
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea;
| | - Jeonghun Yeom
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea;
| | - Jiyoung Yu
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (H.-S.A.); (J.Y.); (Y.O.)
| | - Yumi Oh
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (H.-S.A.); (J.Y.); (Y.O.)
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505, Korea
| | - Jae Ho Jeong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (J.H.J.); (J.-H.A.); (K.H.J.); (S.-B.K.)
| | - Jin-Hee Ahn
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (J.H.J.); (J.-H.A.); (K.H.J.); (S.-B.K.)
| | - Kyung Hae Jung
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (J.H.J.); (J.-H.A.); (K.H.J.); (S.-B.K.)
| | - Sung-Bae Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (J.H.J.); (J.-H.A.); (K.H.J.); (S.-B.K.)
| | - Hee Jin Lee
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (H.J.L.); (G.G.)
| | - Gyungyub Gong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (H.J.L.); (G.G.)
| | - Sae Byul Lee
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (S.B.L.); (I.Y.C.); (H.J.K.); (B.S.K.); (J.W.L.); (B.H.S.); (S.H.A.)
| | - Il Yong Chung
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (S.B.L.); (I.Y.C.); (H.J.K.); (B.S.K.); (J.W.L.); (B.H.S.); (S.H.A.)
| | - Hee Jeong Kim
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (S.B.L.); (I.Y.C.); (H.J.K.); (B.S.K.); (J.W.L.); (B.H.S.); (S.H.A.)
| | - Beom Seok Ko
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (S.B.L.); (I.Y.C.); (H.J.K.); (B.S.K.); (J.W.L.); (B.H.S.); (S.H.A.)
| | - Jong Won Lee
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (S.B.L.); (I.Y.C.); (H.J.K.); (B.S.K.); (J.W.L.); (B.H.S.); (S.H.A.)
| | - Byung Ho Son
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (S.B.L.); (I.Y.C.); (H.J.K.); (B.S.K.); (J.W.L.); (B.H.S.); (S.H.A.)
| | - Sei Hyun Ahn
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (S.B.L.); (I.Y.C.); (H.J.K.); (B.S.K.); (J.W.L.); (B.H.S.); (S.H.A.)
| | - Kyunggon Kim
- Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea; (H.-S.A.); (J.Y.); (Y.O.)
- Convergence Medicine Research Center, Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea;
- Department of Biomedical Sciences, University of Ulsan College of Medicine, Seoul 05505, Korea
- Clinical Proteomics Core Laboratory, Convergence Medicine Research Center, Asan Medical Center, Seoul 05505, Korea
- Bio-Medical Institute of Technology, Asan Medical Center, Seoul 05505, Korea
| | - Jisun Kim
- Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea; (S.B.L.); (I.Y.C.); (H.J.K.); (B.S.K.); (J.W.L.); (B.H.S.); (S.H.A.)
| |
Collapse
|
7
|
Datta A, Deng S, Gopal V, Yap KCH, Halim CE, Lye ML, Ong MS, Tan TZ, Sethi G, Hooi SC, Kumar AP, Yap CT. Cytoskeletal Dynamics in Epithelial-Mesenchymal Transition: Insights into Therapeutic Targets for Cancer Metastasis. Cancers (Basel) 2021; 13:1882. [PMID: 33919917 PMCID: PMC8070945 DOI: 10.3390/cancers13081882] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/08/2021] [Accepted: 04/12/2021] [Indexed: 12/14/2022] Open
Abstract
In cancer cells, a vital cellular process during metastasis is the transformation of epithelial cells towards motile mesenchymal cells called the epithelial to mesenchymal transition (EMT). The cytoskeleton is an active network of three intracellular filaments: actin cytoskeleton, microtubules, and intermediate filaments. These filaments play a central role in the structural design and cell behavior and are necessary for EMT. During EMT, epithelial cells undergo a cellular transformation as manifested by cell elongation, migration, and invasion, coordinated by actin cytoskeleton reorganization. The actin cytoskeleton is an extremely dynamic structure, controlled by a balance of assembly and disassembly of actin filaments. Actin-binding proteins regulate the process of actin polymerization and depolymerization. Microtubule reorganization also plays an important role in cell migration and polarization. Intermediate filaments are rearranged, switching to a vimentin-rich network, and this protein is used as a marker for a mesenchymal cell. Hence, targeting EMT by regulating the activities of their key components may be a potential solution to metastasis. This review summarizes the research done on the physiological functions of the cytoskeleton, its role in the EMT process, and its effect on multidrug-resistant (MDR) cancer cells-highlight some future perspectives in cancer therapy by targeting cytoskeleton.
Collapse
Affiliation(s)
- Arpita Datta
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (A.D.); (S.D.); (V.G.); (K.C.-H.Y.); (C.E.H.); (M.L.L.); (M.S.O.); (S.C.H.)
| | - Shuo Deng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (A.D.); (S.D.); (V.G.); (K.C.-H.Y.); (C.E.H.); (M.L.L.); (M.S.O.); (S.C.H.)
| | - Vennila Gopal
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (A.D.); (S.D.); (V.G.); (K.C.-H.Y.); (C.E.H.); (M.L.L.); (M.S.O.); (S.C.H.)
| | - Kenneth Chun-Hong Yap
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (A.D.); (S.D.); (V.G.); (K.C.-H.Y.); (C.E.H.); (M.L.L.); (M.S.O.); (S.C.H.)
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore;
| | - Clarissa Esmeralda Halim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (A.D.); (S.D.); (V.G.); (K.C.-H.Y.); (C.E.H.); (M.L.L.); (M.S.O.); (S.C.H.)
| | - Mun Leng Lye
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (A.D.); (S.D.); (V.G.); (K.C.-H.Y.); (C.E.H.); (M.L.L.); (M.S.O.); (S.C.H.)
| | - Mei Shan Ong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (A.D.); (S.D.); (V.G.); (K.C.-H.Y.); (C.E.H.); (M.L.L.); (M.S.O.); (S.C.H.)
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117593, Singapore;
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore;
- Cancer Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Shing Chuan Hooi
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (A.D.); (S.D.); (V.G.); (K.C.-H.Y.); (C.E.H.); (M.L.L.); (M.S.O.); (S.C.H.)
- Cancer Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore;
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117593, Singapore;
- Cancer Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| | - Celestial T. Yap
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (A.D.); (S.D.); (V.G.); (K.C.-H.Y.); (C.E.H.); (M.L.L.); (M.S.O.); (S.C.H.)
- Cancer Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| |
Collapse
|
8
|
Proteomic Differences in Feline Fibrosarcomas Grown Using Doxorubicin-Sensitive and -Resistant Cell Lines in the Chick Embryo Model. Int J Mol Sci 2018; 19:ijms19020576. [PMID: 29443940 PMCID: PMC5855798 DOI: 10.3390/ijms19020576] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 02/09/2018] [Accepted: 02/12/2018] [Indexed: 12/19/2022] Open
Abstract
Proteomic analyses are rapid and powerful tools that are used to increase the understanding of cancer pathogenesis, discover cancer biomarkers and predictive markers, and select and monitor novel targets for cancer therapy. Feline injection-site sarcomas (FISS) are aggressive skin tumours with high recurrence rates, despite treatment with surgery, radiotherapy, and chemotherapy. Doxorubicin is a drug of choice for soft tissue sarcomas, including FISS. However, multidrug resistance is one of the major causes of chemotherapy failure. The main aim of the present study was to identify proteins that differentiate doxorubicin-resistant from doxorubicin-sensitive FISS using two-dimensional gel electrophoresis (2DE), followed by matrix-assisted laser desorption ionisation time-of-flight mass spectrometry (MALDI-TOF MS) analysis. Using the three-dimensional (3D) preclinical in ovo model, which resembles features of spontaneous fibrosarcomas, three significantly (p ≤ 0.05) differentially expressed proteins were identified in tumours grown from doxorubicin-resistant fibrosarcoma cell lines (FFS1 and FFS3) in comparison to the doxorubicin-sensitive one (FFS5): Annexin A5 (ANXA5), Annexin A3 (ANXA3), and meiosis-specific nuclear structural protein 1 (MNS1). Moreover, nine other proteins were significantly differentially expressed in tumours grown from the high doxorubicin-resistant cell line (FFS1) in comparison to sensitive one (FFS5). This study may be the first proteomic fingerprinting of FISS reported, identifying potential candidates for specific predictive biomarkers and research targets for doxorubicin-resistant FISS.
Collapse
|
9
|
Golubnitschaja O, Filep N, Yeghiazaryan K, Blom HJ, Hofmann-Apitius M, Kuhn W. Multi-omic approach decodes paradoxes of the triple-negative breast cancer: lessons for predictive, preventive and personalised medicine. Amino Acids 2017; 50:383-395. [PMID: 29249020 DOI: 10.1007/s00726-017-2524-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 12/05/2017] [Indexed: 10/24/2022]
Abstract
Breast cancer epidemic in the early twenty-first century results in around two million new cases and half-a-million of the disease-related deaths registered annually worldwide. A particularly dramatic situation is attributed to some specific patient subgroups such as the triple-negative breast cancer (TNBC). TNBC is a particularly aggressive type of breast cancer lacking clear diagnostic approach and targeted therapies. Consequently, more than 50% of the TNBC patients die of the metastatic BC within the first 6 months of the diagnosis. In the current study we have hypothesised that multi-omic approach utilising blood samples may lead to discovery of a unique molecular signature of the TNBC subtype. The results achieved demonstrate, indeed, multi-omics as highly promising approach that could be of great clinical utility for development of predictive diagnosis, targeted prevention and treatments tailored to the person-overall advancing the management of the TNBC.
Collapse
Affiliation(s)
- Olga Golubnitschaja
- Department of Radiology, Rheinische Friedrich-Wilhelms-Universität Bonn, Sigmund-Freud-Str 25, 53105, Bonn, Germany. .,Breast Cancer Research Centre, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany. .,Centre for Integrated Oncology, Cologne-Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany.
| | - Nora Filep
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, Germany
| | - Kristina Yeghiazaryan
- Department of Radiology, Rheinische Friedrich-Wilhelms-Universität Bonn, Sigmund-Freud-Str 25, 53105, Bonn, Germany.,Breast Cancer Research Centre, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany.,Centre for Integrated Oncology, Cologne-Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Henricus Johannes Blom
- Laboratory of Clinical Biochemistry and Metabolism, Department of General Pediatrics, Adolescent Medicine and Neonatology, University Medical Centre Freiburg, Freiburg, Germany
| | - Martin Hofmann-Apitius
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Sankt Augustin, Germany
| | - Walther Kuhn
- Breast Cancer Research Centre, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany.,Centre for Integrated Oncology, Cologne-Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany.,Centre for Obstetrics and Gynaecology, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| |
Collapse
|
10
|
Desai S, Barai A, Bukhari AB, De A, Sen S. α-Actinin-4 confers radioresistance coupled invasiveness in breast cancer cells through AKT pathway. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1865:196-208. [PMID: 29055790 DOI: 10.1016/j.bbamcr.2017.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 10/09/2017] [Accepted: 10/13/2017] [Indexed: 12/18/2022]
Abstract
Acquired radioresistance accompanied with increased metastatic potential is a major hurdle in effective radiotherapy of breast cancers. However, the nature of their inter-dependence and the underlying mechanism remains largely intangible. By employing radioresistant (RR) cell lines, we herein demonstrate that MCF-7 RR cells display phenotypic and molecular alterations evocative of epithelial to mesenchymal transition (EMT) with increased traction forces and membrane ruffling culminating in boosted invasiveness. We then show that these changes can be attributed to overexpression of alpha-actinin-4 (ACTN4), with ACTN4 knockdown near-completely abrogating both radioresistance and EMT-associated changes. We further found that in MCF-7 RR cells, ACTN4 mediates the observed effects by activating AKT, and downstream AKT/GSK3β signalling. Though ACTN4 plays a similar role in mediating radioresistance and invasiveness in MDA-MB-231 RR cells, co-immunoprecipitation studies reveal that these changes are effected through increased association with AKT and not by overexpression of AKT. Taken together, our study identifies ACTN4/AKT/GSK3β as a novel pathway regulating radioresistance coupled invasion which can be further explored to improve the radiotherapeutic gain.
Collapse
Affiliation(s)
- Sejal Desai
- Biosciences and Bioengineering Department, IIT Bombay, Mumbai, India
| | - Amlan Barai
- Biosciences and Bioengineering Department, IIT Bombay, Mumbai, India
| | | | - Abhijit De
- ACTREC, Tata Memorial Centre, Kharghar, Navi Mumbai, India.
| | - Shamik Sen
- Biosciences and Bioengineering Department, IIT Bombay, Mumbai, India.
| |
Collapse
|
11
|
Thuwajit C, Thuwajit P, Jamjantra P, Pairojkul C, Wongkham S, Bhudhisawasdi V, Ono J, Ohta S, Fujimoto K, Izuhara K. Clustering of patients with intrahepatic cholangiocarcinoma based on serum periostin may be predictive of prognosis. Oncol Lett 2017; 14:623-634. [PMID: 28693214 PMCID: PMC5494708 DOI: 10.3892/ol.2017.6250] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/03/2017] [Indexed: 12/13/2022] Open
Abstract
An effective serum biomarker may improve cholangiocarcinoma (CCA) management. Periostin (PN) has been demonstrated to be associated with aggressive CCA. The current study evaluated PN in blood serum for its diagnostic and prognostic potential in patients with CCA. Sera of 68 patients with CCA were collected prior to treatment, and PN levels were measured using an ELISA. Sera from 50 normal controls, 6 patients with benign liver diseases, 2 with hepatocellular carcinoma and 21 with breast cancer were analyzed. Immunohistochemistry of PN in CCA tissues was also investigated. The data were analyzed using the Mann-Whitney U test, Kaplan-Meier log rank tests, Cox proportional hazard regression models and Fisher's exact tests. The median serum PN level in patients with CCA was significantly increased compared with that in healthy controls, patients with benign liver diseases and patients with breast cancer (all P<0.05). Using an optimal threshold value of 94 ng/ml PN, the diagnostic values for CCA compared with other conditions demonstrated a sensitivity level of 0.38 [95% confidence interval (CI), 0.27-0.51], specificity of 0.90 (95% CI, 0.81-0.96), accuracy of 0.66 (95% CI, 0.58-0.74), positive predictive value of 0.76 (95% CI, 0.59-0.89) and negative predictive value of 0.63 (95% CI, 0.53-0.72) (P<0.001). Furthermore, PN stain in stromal fibroblasts in CCA tissues was associated with serum PN levels (P=0.001), and patients with CCA were classified as low (≤94 ng/ml) or high PN (>94 ng/ml) accordingly. High serum and tissue PN levels were significantly associated with reduced survival rate (P<0.001 and P=0.033, respectively). Serum PN was an independent prognostic factor with a hazard ratio of 3.197 (P=0.001). In conclusion, serum PN may be used to divide patients with intrahepatic CCA into high and low PN groups. Elevated serum PN may be utilized as a marker of poor prognosis in patients with CCA.
Collapse
Affiliation(s)
- Chanitra Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-Noi, Bangkok 10700, Thailand
| | - Peti Thuwajit
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-Noi, Bangkok 10700, Thailand
| | - Pranisa Jamjantra
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok-Noi, Bangkok 10700, Thailand
| | - Chawalit Pairojkul
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Khon Kaen 40002, Thailand
| | - Sopit Wongkham
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Khon Kaen 40002, Thailand
| | | | - Junya Ono
- Research and Development Unit, Shino-Test Corporation, Minami-Ku, Sagamihara, Kanagawa 252-0331, Japan
| | - Shoichiro Ohta
- Department of Biomolecular Sciences, Saga Medical School, Saga 849-8501, Japan
| | - Kiminori Fujimoto
- Department of Radiology, Kurume University School of Medicine and Center for Diagnostic Imaging, Kurume University Hospital, Kurume, Fukuoka 830-0011, Japan
| | - Kenji Izuhara
- Department of Biomolecular Sciences, Saga Medical School, Saga 849-8501, Japan
| |
Collapse
|
12
|
Bhardwaj G, Dörr M, Sappa PK, Ameling S, Dhople V, Steil L, Klingel K, Empen K, Beug D, Völker U, Felix SB, Hammer E. Endomyocardial proteomic signature corresponding to the response of patients with dilated cardiomyopathy to immunoadsorption therapy. J Proteomics 2017; 150:121-129. [DOI: 10.1016/j.jprot.2016.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 08/28/2016] [Accepted: 09/07/2016] [Indexed: 02/07/2023]
|
13
|
Miah S, Banks CAS, Adams MK, Florens L, Lukong KE, Washburn MP. Advancement of mass spectrometry-based proteomics technologies to explore triple negative breast cancer. MOLECULAR BIOSYSTEMS 2016; 13:42-55. [PMID: 27891540 PMCID: PMC5173390 DOI: 10.1039/c6mb00639f] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Understanding the complexity of cancer biology requires extensive information about the cancer proteome over the course of the disease. The recent advances in mass spectrometry-based proteomics technologies have led to the accumulation of an incredible amount of such proteomic information. This information allows us to identify protein signatures or protein biomarkers, which can be used to improve cancer diagnosis, prognosis and treatment. For example, mass spectrometry-based proteomics has been used in breast cancer research for over two decades to elucidate protein function. Breast cancer is a heterogeneous group of diseases with distinct molecular features that are reflected in tumour characteristics and clinical outcomes. Compared with all other subtypes of breast cancer, triple-negative breast cancer is perhaps the most distinct in nature and heterogeneity. In this review, we provide an introductory overview of the application of advanced proteomic technologies to triple-negative breast cancer research.
Collapse
Affiliation(s)
- Sayem Miah
- Stowers Institute for Medical Research, 1000 E. 50th St, Kansas City, MO 64110, USA. and Department of Biochemistry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Charles A S Banks
- Stowers Institute for Medical Research, 1000 E. 50th St, Kansas City, MO 64110, USA.
| | - Mark K Adams
- Stowers Institute for Medical Research, 1000 E. 50th St, Kansas City, MO 64110, USA.
| | - Laurence Florens
- Stowers Institute for Medical Research, 1000 E. 50th St, Kansas City, MO 64110, USA.
| | - Kiven E Lukong
- Department of Biochemistry, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Michael P Washburn
- Stowers Institute for Medical Research, 1000 E. 50th St, Kansas City, MO 64110, USA. and Departments of Pathology & Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| |
Collapse
|
14
|
Song D, Yue L, Zhang J, Ma S, Zhao W, Guo F, Fan Y, Yang H, Liu Q, Zhang D, Xia Z, Qin P, Jia J, Yue M, Yu J, Zheng S, Yang F, Wang J. Diagnostic and prognostic significance of serum apolipoprotein C-I in triple-negative breast cancer based on mass spectrometry. Cancer Biol Ther 2016; 17:635-47. [PMID: 27260686 DOI: 10.1080/15384047.2016.1156262] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Women with triple-negative breast cancer (TNBC) have poor prognosis because of the aggressive nature of the tumor, delayed diagnosis and non-specific symptoms in the early stages. Identification of novel specific TNBC serum biomarkers for screening and therapeutic purposes therefore remains an urgent clinical requirement.We obtained serum samples from a total of 380 recruited individuals split into mining and testing sets, with the aim of screening for reliable protein biomarkers from TNBC and non-TNBC (NTNBC) sera. Samples were assessed using mass spectrometry, followed by receiver operating characteristic (ROC), survival and hazard function curve as well as multivariate Cox regression analyses to ascertain the potential of the protein constituents as diagnostic and prognostic biomarkers for TNBC.We identified upregulated apolipoprotein C-I (apoC-I) with a validated positive effect on TNBC tumorigenesis, with confirmation in an independent test set and minimization of systematic bias by pre-analytical parameters. The apoC-I protein had superior diagnostic ability in distinguishing between TNBC and NTNBC cases. Moreover, the protein presented a more robust potential prognostic factor for TNBC than NTNBC. The apoC-I protein identified in this study presents an effective novel diagnostic and prognostic biomarker for TNBC, indicating that measurement of the peak intensity at 7785 Da in serum samples could facilitate improved early detection and estimation of postoperative survival prognosis for TNBC.
Collapse
Affiliation(s)
- Dongjian Song
- a Department of Pediatric Surgery , First Affiliated Hospital, Zhengzhou University , Zhengzhou , PR China.,b Institute of Clinical Medicine, First Affiliated Hospital, Zhengzhou University , Zhengzhou , PR China
| | - Lifang Yue
- c Department of Ultrasonography , Third Affiliated Hospital, Zhengzhou University , Zhengzhou , PR China
| | - Junjie Zhang
- a Department of Pediatric Surgery , First Affiliated Hospital, Zhengzhou University , Zhengzhou , PR China
| | - Shanshan Ma
- d School of Life Science , Zhengzhou University , Zhengzhou , PR China
| | - Wei Zhao
- a Department of Pediatric Surgery , First Affiliated Hospital, Zhengzhou University , Zhengzhou , PR China
| | - Fei Guo
- a Department of Pediatric Surgery , First Affiliated Hospital, Zhengzhou University , Zhengzhou , PR China
| | - Yingzhong Fan
- a Department of Pediatric Surgery , First Affiliated Hospital, Zhengzhou University , Zhengzhou , PR China
| | - Heying Yang
- a Department of Pediatric Surgery , First Affiliated Hospital, Zhengzhou University , Zhengzhou , PR China
| | - Qiuliang Liu
- a Department of Pediatric Surgery , First Affiliated Hospital, Zhengzhou University , Zhengzhou , PR China
| | - Da Zhang
- a Department of Pediatric Surgery , First Affiliated Hospital, Zhengzhou University , Zhengzhou , PR China
| | - Ziqiang Xia
- a Department of Pediatric Surgery , First Affiliated Hospital, Zhengzhou University , Zhengzhou , PR China
| | - Pan Qin
- a Department of Pediatric Surgery , First Affiliated Hospital, Zhengzhou University , Zhengzhou , PR China
| | - Jia Jia
- a Department of Pediatric Surgery , First Affiliated Hospital, Zhengzhou University , Zhengzhou , PR China
| | - Ming Yue
- a Department of Pediatric Surgery , First Affiliated Hospital, Zhengzhou University , Zhengzhou , PR China
| | - Jiekai Yu
- e Institute of Cancer, Second Affiliated Hospital, Zhejiang University , Hangzhou , PR China
| | - Shu Zheng
- e Institute of Cancer, Second Affiliated Hospital, Zhejiang University , Hangzhou , PR China
| | - Fuquan Yang
- f Proteomic Platform , Institute of Biophysics, Chinese Academy of Sciences , Beijing , PR China
| | - Jiaxiang Wang
- a Department of Pediatric Surgery , First Affiliated Hospital, Zhengzhou University , Zhengzhou , PR China
| |
Collapse
|
15
|
Kurono S, Kaneko Y, Matsuura N, Oishi H, Noguchi S, Kim SJ, Tamaki Y, Aikawa T, Kotsuma Y, Inaji H, Matsuura S. Identification of potential breast cancer markers in nipple discharge by protein profile analysis using two-dimensional nano-liquid chromatography/nanoelectrospray ionization-mass spectrometry. Proteomics Clin Appl 2016; 10:605-13. [PMID: 26970563 DOI: 10.1002/prca.201500016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 02/01/2016] [Accepted: 03/07/2016] [Indexed: 12/11/2022]
Abstract
PURPOSE This research aimed to establish a diagnostic technique for breast cancer using nipple discharge (ND), with the objective of preventive diagnosis. ND has been proposed as a source of secreted proteomes that reflect early pathological changes in the ductal-lobular epithelial microenvironment, and could thus provide breast-specific cancer biomarkers that could be accessed noninvasively as a new clinical diagnostic technique. EXPERIMENTAL DESIGN Minute amounts of ND from patients with and without breast cancer (n = 19 and 12, respectively) were collected at the hospital and kept frozen until just before use. They were analyzed using high-pH RP peptide fractionations/low-pH RP 2D nano-LC/ESI-MS/MS. Biomarker candidates were also investigated using Western blot analysis and sandwich ELISA on ND and/or sera. RESULTS We found distinct tendencies in protein expression and three candidate breast cancer biomarkers (carbonic anhydrase 2, catalase, and peroxiredoxin-2) whose levels differed significantly between ND specimens from patients with and without breast cancer. CONCLUSIONS AND CLINICAL RELEVANCE These tendencies in protein expression and markers provide new ways to identify breast cancer patients. Therefore, RP/RP 2D LC/MS/MS analyses of ND and the above three markers are supported as a new breast cancer diagnostic technique.
Collapse
Affiliation(s)
- Sadamu Kurono
- Laboratory of Molecular Signature Analysis, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Laboratory Chemicals Division, Wako Pure Chemical Industries, Ltd, Osaka, Japan
| | - Yuka Kaneko
- Laboratory of Molecular Signature Analysis, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita, Osaka, Japan.,Laboratory Chemicals Division, Wako Pure Chemical Industries, Ltd, Osaka, Japan
| | - Nariaki Matsuura
- Department of Molecular Pathology, Osaka University Graduate School of Medicine and Health Science, Suita, Osaka, Japan.,Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Haruki Oishi
- Laboratory of Molecular Signature Analysis, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Shinzaburo Noguchi
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Seung Jin Kim
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Yasuhiro Tamaki
- Department of Breast Surgery, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Takao Aikawa
- Osaka Center for Cancer and Cardiovascular Diseases Prevention, Osaka, Japan
| | | | - Hideo Inaji
- Department of Breast Surgery, Osaka Medical Center for Cancer and Cardiovascular Diseases, Osaka, Japan
| | - Shuji Matsuura
- Laboratory of Molecular Signature Analysis, Division of Health Sciences, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
16
|
Vergara D, Simeone P, Franck J, Trerotola M, Giudetti A, Capobianco L, Tinelli A, Bellomo C, Fournier I, Gaballo A, Alberti S, Salzet M, Maffia M. Translating epithelial mesenchymal transition markers into the clinic: Novel insights from proteomics. EUPA OPEN PROTEOMICS 2016; 10:31-41. [PMID: 29900098 PMCID: PMC5988589 DOI: 10.1016/j.euprot.2016.01.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 11/30/2015] [Accepted: 01/05/2016] [Indexed: 12/15/2022]
Abstract
The growing understanding of the molecular mechanisms underlying epithelial-to-mesenchymal transition (EMT) may represent a potential source of clinical markers. Despite EMT drivers have not yet emerged as candidate markers in the clinical setting, their association with established clinical markers may improve their specificity and sensitivity. Mass spectrometry-based platforms allow analyzing multiple samples for the expression of EMT candidate markers, and may help to diagnose diseases or monitor treatment efficiently. This review highlights proteomic approaches applied to elucidate the differences between epithelial and mesenchymal tumors and describes how these can be used for target discovery and validation.
Collapse
Affiliation(s)
- Daniele Vergara
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Monteroni, 73100 Lecce, Italy.,Laboratory of Clinical Proteomic, "Giovanni Paolo II" Hospital, ASL-Lecce, Italy
| | - Pasquale Simeone
- Research Centre on Aging (Ce.S.I), Unit of Cytomorphology, "University G. d'Annunzio" Foundation, 66100 Chieti, Italy.,Department of Medicine and Aging Science, School of Medicine and Health Science, University "G. d'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy
| | - Julien Franck
- U1192 INSERM, Laboratoire PRISM: Protéomique, Réponse Inflammatoire, Spectrométrie de Masse, Université Lille 1, Villeneuve D'Ascq, France
| | - Marco Trerotola
- Unit of Cancer Pathology, CeSI, Foundation University 'G. d'Annunzio', Chieti, Italy.,Department of Medical, Oral and Biotechnological Sciences, University "G. D'Annunzio" of Chieti-Pescara, 66100 Chieti, Italy
| | - Anna Giudetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Monteroni, 73100 Lecce, Italy
| | - Loredana Capobianco
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Monteroni, 73100 Lecce, Italy
| | - Andrea Tinelli
- Department of Gynecology and Obstetrics, Division of Experimental Endoscopic Surgery, Imaging, Minimally Invasive Therapy and Technology, "Vito Fazzi" Hospital, ASL-Lecce, Italy.,International Translational Medicine and Biomodelling Research Group, Department of Applied Mathematics, Moscow Institute of Physics and Technology (MIPT), State University, Moscow, Russia
| | - Claudia Bellomo
- Department of Medical Biochemistry and Microbiology and Ludwig Institute for Cancer Research, Science for Life Laboratory, Biomedical Center, Uppsala University, Box 582, SE 75 123 Uppsala, Sweden
| | - Isabelle Fournier
- U1192 INSERM, Laboratoire PRISM: Protéomique, Réponse Inflammatoire, Spectrométrie de Masse, Université Lille 1, Villeneuve D'Ascq, France
| | - Antonio Gaballo
- CNR NANOTEC-Institute of Nanotechnology, Polo di Nanotecnologia c/o Campus Ecotekne, via Monteroni, 73100 Lecce, Italy
| | - Saverio Alberti
- Unit of Cancer Pathology, CeSI, Foundation University 'G. d'Annunzio', Chieti, Italy
| | - Michel Salzet
- U1192 INSERM, Laboratoire PRISM: Protéomique, Réponse Inflammatoire, Spectrométrie de Masse, Université Lille 1, Villeneuve D'Ascq, France
| | - Michele Maffia
- Department of Biological and Environmental Sciences and Technologies, University of Salento, via Monteroni, 73100 Lecce, Italy.,Laboratory of Clinical Proteomic, "Giovanni Paolo II" Hospital, ASL-Lecce, Italy
| |
Collapse
|
17
|
Pendharkar N, Gajbhiye A, Taunk K, RoyChoudhury S, Dhali S, Seal S, Mane A, Abhang S, Santra MK, Chaudhury K, Rapole S. Quantitative tissue proteomic investigation of invasive ductal carcinoma of breast with luminal B HER2 positive and HER2 enriched subtypes towards potential diagnostic and therapeutic biomarkers. J Proteomics 2015; 132:112-30. [PMID: 26642762 DOI: 10.1016/j.jprot.2015.11.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 10/16/2015] [Accepted: 11/26/2015] [Indexed: 02/09/2023]
Abstract
Worldwide, breast cancer is one of the frequently diagnosed cancers in women with high mortality if not diagnosed at early stage. Although biomarker discoveries through various proteomic approaches have been studied in breast cancer, a limited number of studies have explored the invasive ductal carcinoma with Luminal B HER2 positive (LB) and HER2 enriched (HE) subtypes. The present study employed the complementary quantitative proteomic approaches to find a panel of markers that could discriminate LB and HE subtypes as well as early (ES) and late stages (LS) of these subtypes. A total of 67 and 68 differentially expressed proteins were identified by DIGE for the subtype and stage wise categories, respectively. Multivariate statistical analysis was employed to identify the set of most significant proteins, which could discriminate between these two subtypes and also early and late stages under study. Immunoblotting and MRM based validation in a separate cohort of samples confirmed that panel of biosignatures for LB are APOA1, GELS, HS90B, EF1A1, NHRF1 and PRDX3 and for HE are PRDX1, CATD, CALR, ATPB and CH60. For the diagnosis of early and late stages the potential markers are TPM4, CATD, PRDX3, ANXA3, HSPB1 and CALR, TRFE, GELS, CH60, CAPG, NHRF1, 1433G, GRP78 respectively.
Collapse
Affiliation(s)
- Namita Pendharkar
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune 411007, MH, India; B. J. Medical College, Sassoon Hospital, Pune 411001, MH, India
| | - Akshada Gajbhiye
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune 411007, MH, India
| | - Khushman Taunk
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune 411007, MH, India
| | - Sourav RoyChoudhury
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur 721302, WB, India
| | - Snigdha Dhali
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune 411007, MH, India
| | | | - Anupama Mane
- Grant Medical Foundation, Ruby Hall Clinic, Pune 411001, MH, India
| | | | - Manas K Santra
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune 411007, MH, India
| | - Koel Chaudhury
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur 721302, WB, India
| | - Srikanth Rapole
- Proteomics Lab, National Centre for Cell Science, Ganeshkhind, Pune 411007, MH, India.
| |
Collapse
|
18
|
Proteomic analysis reveals that the protective effects of ginsenoside Rb1 are associated with the actin cytoskeleton in β-amyloid-treated neuronal cells. J Ginseng Res 2015; 40:278-84. [PMID: 27616904 PMCID: PMC5005364 DOI: 10.1016/j.jgr.2015.09.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 09/16/2015] [Accepted: 09/22/2015] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND The ginsenoside Rb1 (Rb1) is the most abundant compound in the root of Panax ginseng. Recent studies have shown that Rb1 has a neuroprotective effect. However, the mechanisms underlying this effect are still unknown. METHODS We used stable isotope labeling with amino acids in cell culture, combined with quantitative mass spectrometry, to explore a potential protective mechanism of Rb1 in β-amyloid-treated neuronal cells. RESULTS A total of 1,231 proteins were commonly identified from three replicate experiments. Among these, 40 proteins were significantly changed in response to Rb1 pretreatment in β-amyloid-treated neuronal cells. Analysis of the functional enrichments and protein interactions of altered proteins revealed that actin cytoskeleton proteins might be linked to the regulatory mechanisms of Rb1. The CAP1, CAPZB, TOMM40, and DSTN proteins showed potential as molecular target proteins for the functional contribution of Rb1 in Alzheimer's disease (AD). CONCLUSION Our proteomic data may provide new insights into the protective mechanisms of Rb1 in AD.
Collapse
|
19
|
White MJV, Roife D, Gomer RH. Galectin-3 Binding Protein Secreted by Breast Cancer Cells Inhibits Monocyte-Derived Fibrocyte Differentiation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:1858-67. [PMID: 26136428 PMCID: PMC4530092 DOI: 10.4049/jimmunol.1500365] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/02/2015] [Indexed: 12/26/2022]
Abstract
To metastasize, tumor cells often need to migrate through a layer of collagen-containing scar tissue which encapsulates the tumor. A key component of scar tissue and fibrosing diseases is the monocyte-derived fibrocyte, a collagen-secreting profibrotic cell. To test the hypothesis that invasive tumor cells may block the formation of the fibrous sheath, we determined whether tumor cells secrete factors that inhibit monocyte-derived fibrocyte differentiation. We found that the human metastatic breast cancer cell line MDA-MB-231 secretes activity that inhibits human monocyte-derived fibrocyte differentiation, whereas less aggressive breast cancer cell lines secrete less of this activity. Purification indicated that Galectin-3 binding protein (LGALS3BP) is the active factor. Recombinant LGALS3BP inhibits monocyte-derived fibrocyte differentiation, and immunodepletion of LGALS3BP from MDA-MB 231 conditioned media removes the monocyte-derived fibrocyte differentiation-inhibiting activity. LGALS3BP inhibits the differentiation of monocyte-derived fibrocytes from wild-type mouse spleen cells, but not from SIGN-R1(-/-) mouse spleen cells, suggesting that CD209/SIGN-R1 is required for the LGALS3BP effect. Galectin-3 and galectin-1, binding partners of LGALS3BP, potentiate monocyte-derived fibrocyte differentiation. In breast cancer biopsies, increased levels of tumor cell-associated LGALS3BP were observed in regions of the tumor that were invading the surrounding stroma. These findings suggest LGALS3BP and galectin-3 as new targets to treat metastatic cancer and fibrosing diseases.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/pharmacology
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/pharmacology
- Breast Neoplasms/genetics
- Breast Neoplasms/immunology
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Carrier Proteins/genetics
- Carrier Proteins/metabolism
- Carrier Proteins/pharmacology
- Cell Adhesion Molecules/metabolism
- Cell Differentiation/drug effects
- Cell Line, Tumor
- Culture Media, Conditioned
- Female
- Galectin 1/metabolism
- Galectin 3/metabolism
- Glycoproteins/genetics
- Glycoproteins/metabolism
- Glycoproteins/pharmacology
- Humans
- Lectins, C-Type/metabolism
- Mice
- Monocytes/cytology
- Monocytes/drug effects
- Monocytes/metabolism
- RNA, Messenger/genetics
- Receptors, Cell Surface/metabolism
- Recombinant Proteins/pharmacology
Collapse
Affiliation(s)
- Michael J V White
- Department of Biology, Texas A&M University, College Station, TX 77843; and
| | - David Roife
- Department of Surgical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX 77030
| | - Richard H Gomer
- Department of Biology, Texas A&M University, College Station, TX 77843; and
| |
Collapse
|
20
|
Sobral-Leite M, Wesseling J, Smit VTHBM, Nevanlinna H, van Miltenburg MH, Sanders J, Hofland I, Blows FM, Coulson P, Patrycja G, Schellens JHM, Fagerholm R, Heikkilä P, Aittomäki K, Blomqvist C, Provenzano E, Ali HR, Figueroa J, Sherman M, Lissowska J, Mannermaa A, Kataja V, Kosma VM, Hartikainen JM, Phillips KA, Couch FJ, Olson JE, Vachon C, Visscher D, Brenner H, Butterbach K, Arndt V, Holleczek B, Hooning MJ, Hollestelle A, Martens JWM, van Deurzen CHM, van de Water B, Broeks A, Chang-Claude J, Chenevix-Trench G, Easton DF, Pharoah PDP, García-Closas M, de Graauw M, Schmidt MK. Annexin A1 expression in a pooled breast cancer series: association with tumor subtypes and prognosis. BMC Med 2015; 13:156. [PMID: 26137966 PMCID: PMC4489114 DOI: 10.1186/s12916-015-0392-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 06/04/2015] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Annexin A1 (ANXA1) is a protein related with the carcinogenesis process and metastasis formation in many tumors. However, little is known about the prognostic value of ANXA1 in breast cancer. The purpose of this study is to evaluate the association between ANXA1 expression, BRCA1/2 germline carriership, specific tumor subtypes and survival in breast cancer patients. METHODS Clinical-pathological information and follow-up data were collected from nine breast cancer studies from the Breast Cancer Association Consortium (BCAC) (n = 5,752) and from one study of familial breast cancer patients with BRCA1/2 mutations (n = 107). ANXA1 expression was scored based on the percentage of immunohistochemical staining in tumor cells. Survival analyses were performed using a multivariable Cox model. RESULTS The frequency of ANXA1 positive tumors was higher in familial breast cancer patients with BRCA1/2 mutations than in BCAC patients, with 48.6 % versus 12.4 %, respectively; P <0.0001. ANXA1 was also highly expressed in BCAC tumors that were poorly differentiated, triple negative, EGFR-CK5/6 positive or had developed in patients at a young age. In the first 5 years of follow-up, patients with ANXA1 positive tumors had a worse breast cancer-specific survival (BCSS) than ANXA1 negative (HRadj = 1.35; 95 % CI = 1.05-1.73), but the association weakened after 10 years (HRadj = 1.13; 95 % CI = 0.91-1.40). ANXA1 was a significant independent predictor of survival in HER2+ patients (10-years BCSS: HRadj = 1.70; 95 % CI = 1.17-2.45). CONCLUSIONS ANXA1 is overexpressed in familial breast cancer patients with BRCA1/2 mutations and correlated with poor prognosis features: triple negative and poorly differentiated tumors. ANXA1 might be a biomarker candidate for breast cancer survival prediction in high risk groups such as HER2+ cases.
Collapse
Affiliation(s)
- Marcelo Sobral-Leite
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Programa de Farmacologia, Instituto Nacional do Câncer (INCA), Rio de Janeiro, RJ, Brazil.
| | - Jelle Wesseling
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Division of Diagnostic Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Vincent T H B M Smit
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Heli Nevanlinna
- University of Helsinki, Helsinki, Finland.
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital, Helsinki, Finland.
| | | | - Joyce Sanders
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Ingrid Hofland
- Core Facility Molecular Pathology and Biobanking, Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Fiona M Blows
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK.
| | - Penny Coulson
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK.
| | | | - Jan H M Schellens
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Department of Pharmacoepidemiology & Clinical Pharmacology, Utrecht Institute of Pharmaceutical Sciences (UIPS), Utrecht, The Netherlands.
| | - Rainer Fagerholm
- University of Helsinki, Helsinki, Finland.
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital, Helsinki, Finland.
| | - Päivi Heikkilä
- University of Helsinki, Helsinki, Finland.
- Department of Pathology, Helsinki University Central Hospital, Helsinki, Finland.
| | - Kristiina Aittomäki
- University of Helsinki, Helsinki, Finland.
- Department of Clinical Genetics, Helsinki University Central Hospital, Helsinki, Finland.
| | - Carl Blomqvist
- University of Helsinki, Helsinki, Finland.
- Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland.
| | - Elena Provenzano
- Cancer Research UK Cambridge Institute Oncology, University of Cambridge, Cambridge, UK.
- Department of Histopathology, Addenbrooke's Hospital, Cambridge University Hospital NHS Foundation Trust, Cambridge, UK.
| | - Hamid Raza Ali
- Cancer Research UK Cambridge Institute Oncology, University of Cambridge, Cambridge, UK.
- Department of Pathology, University of Cambridge, Cambridge, UK.
| | - Jonine Figueroa
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA.
| | - Mark Sherman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA.
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD, USA.
| | - Jolanta Lissowska
- Department of Cancer Epidemiology and Prevention, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland.
| | - Arto Mannermaa
- School of Medicine, Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, Kuopio, Finland.
- Cancer Center of Eastern Finland, University of Eastern Finland, Kuopio, Finland.
- Imaging Center, Department of Clinical Pathology, Kuopio University Hospital, Kuopio, Finland.
| | - Vesa Kataja
- Cancer Center, Kuopio University Hospital, Kuopio, Finland.
- Jyväskylä Central Hospital, Jyväskylä, Finland.
| | - Veli-Matti Kosma
- School of Medicine, Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, Kuopio, Finland.
- Cancer Center of Eastern Finland, University of Eastern Finland, Kuopio, Finland.
- Imaging Center, Department of Clinical Pathology, Kuopio University Hospital, Kuopio, Finland.
| | - Jaana M Hartikainen
- School of Medicine, Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, Kuopio, Finland.
- Cancer Center of Eastern Finland, University of Eastern Finland, Kuopio, Finland.
- Imaging Center, Department of Clinical Pathology, Kuopio University Hospital, Kuopio, Finland.
| | - Kelly-Anne Phillips
- Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Australia.
- Centre for Molecular, Environmental, Genetic and Analytic Epidemiology, School of Population Health, The University of Melbourne, Melbourne, Australia.
- Department of Medicine, St Vincent's Hospital, The University of Melbourne, Melbourne, Australia.
| | - Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.
| | - Janet E Olson
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA.
| | - Celine Vachon
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA.
| | - Daniel Visscher
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Division of Preventive Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Katja Butterbach
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Volker Arndt
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | | | - Maartje J Hooning
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
| | - Antoinette Hollestelle
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
| | - John W M Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
| | | | - Bob van de Water
- Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| | - Annegien Broeks
- Core Facility Molecular Pathology and Biobanking, Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, Unit of Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | | | - Douglas F Easton
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK.
| | - Paul D P Pharoah
- Centre for Cancer Genetic Epidemiology, Department of Oncology, University of Cambridge, Cambridge, UK.
| | - Montserrat García-Closas
- Division of Genetics and Epidemiology, Institute of Cancer Research, London, UK.
- Breakthrough Breast Cancer Centre, London, UK.
| | - Marjo de Graauw
- Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| | - Marjanka K Schmidt
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
- Division of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Plesmanlaan 121, 1066, CX, Amsterdam, The Netherlands.
| |
Collapse
|
21
|
Quantitative iTRAQ LC-MS/MS proteomics reveals the proteome profiles of DF-1 cells after infection with subgroup J Avian leukosis virus. BIOMED RESEARCH INTERNATIONAL 2015; 2015:395307. [PMID: 25632391 PMCID: PMC4302370 DOI: 10.1155/2015/395307] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 12/09/2014] [Accepted: 12/17/2014] [Indexed: 12/18/2022]
Abstract
Avian leukosis virus subgroup J (ALV-J) is an avian oncogenic retrovirus that can induce various clinical tumors and has caused severe economic losses in China. To improve our understanding of the host cellular responses to virus infection and the pathogenesis of ALV-J infection, we applied isobaric tags for relative and absolute quantification (iTRAQ) labeling coupled with multidimensional liquid chromatography-tandem mass spectrometry to detect the protein changes in DF-1 cells infected and mock-infected with ALV-J. A total of 75 cellular proteins were significantly changed, including 33 upregulated proteins and 42 downregulated proteins. The reliability of iTRAQ-LC MS/MS was confirmed via real-time PCR. Most of these proteins were related to the physiological functions of metabolic processes, biosynthetic processes, responses to stimuli, protein binding, signal transduction, cell cytoskeleton, and so forth. We also found some proteins that play important roles in apoptosis and oncogenicity. The differentially expressed proteins identified may provide valuable information to elucidate the pathogenesis of virus infection and virus-host interactions.
Collapse
|
22
|
Gasparini P, Cascione L, Fassan M, Lovat F, Guler G, Balci S, Irkkan C, Morrison C, Croce CM, Shapiro CL, Huebner K. microRNA expression profiling identifies a four microRNA signature as a novel diagnostic and prognostic biomarker in triple negative breast cancers. Oncotarget 2014; 5:1174-1184. [PMID: 24632568 PMCID: PMC4012726 DOI: 10.18632/oncotarget.1682] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 01/19/2014] [Indexed: 12/13/2022] Open
Abstract
Triple Negative Breast Cancers (TNBC) is a heterogeneous disease at the molecular and clinical level with poor outcome. Molecular subclassification of TNBCs is essential for optimal use of current therapies and for development of new drugs. microRNAs (miRNA) are widely recognized as key players in cancer progression and drug resistance; investigation of their involvement in a TNBC cohort may reveal biomarkers for diagnosis and prognosis of TNBC. Here we stratified a large TNBC cohort into Core Basal (CB, EGFR and/or CK5, 6 positive) and five negative (5NP) if all markers are negative. We determined the complete miRNA expression profile and found a subset of miRNAs specifically deregulated in the two subclasses.We identified a 4-miRNA signature given by miR-155, miR-493, miR-30e and miR-27a expression levels, that allowed subdivision of TNBCs not only into CB and 5NP subgroups (sensitivity 0.75 and specificity 0.56; AUC=0.74) but also into high risk and low risk groups. We tested the diagnostic and prognostic performances of both the 5 IHC marker panel and the 4-miRNA expression signatures, which clearly identify worse outcome patients in the treated and untreated subcohorts. Both signatures have diagnostic and prognostic value, predicting outcomes of patient treatment with the two most commonly used chemotherapy regimens in TNBC: anthracycline or anthracycline plus taxanes. Further investigations of the patients’ overall survival treated with these regimens show that regardless of IHC group subdivision, taxanes addition did not benefit patients, possibly due to miRNA driven taxanes resistance. TNBC subclassification based on the 5 IHC markers and on the miR-155, miR-493, miR-30e, miR-27a expression levels are powerful diagnostic tools. Treatment choice and new drug development should consider this new subtyping and miRNA expression signature in planning low toxicity, maximum efficacy therapies.
Collapse
Affiliation(s)
- Pierluigi Gasparini
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Luciano Cascione
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, Ohio, USA
- IOR, Institute of Oncology Research, Bellinzona, Switzerland
| | - Matteo Fassan
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, Ohio, USA
- ARC-NET Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Francesca Lovat
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Gulnur Guler
- Department of Pathology, Hacettepe University, Ankara,Turkey
| | - Serdar Balci
- Department of Pathology, Yildirim Beyazit University, Ankara Ataturk Research and Training Hospital, Ankara, Turkey
| | - Cigdem Irkkan
- Dr. Abdurrahman Yurtaslan Ankara Oncology Training and Research Hospital of the Ministry of Health
| | - Carl Morrison
- Department of Pathology, Roswell Park Cancer Institute, S606 Basic Science Building, Elm and Carlton Streets Buffalo, NY
| | - Carlo M. Croce
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Charles L. Shapiro
- Division of Medical Oncology and the Breast Program James Cancer Hospital and Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Kay Huebner
- Department of Molecular Virology, Immunology and Medical Genetics, Ohio State University Wexner Medical Center and Comprehensive Cancer Center, Columbus, Ohio, USA
| |
Collapse
|
23
|
Zhang H, Ghai P, Wu H, Wang C, Field J, Zhou GL. Mammalian adenylyl cyclase-associated protein 1 (CAP1) regulates cofilin function, the actin cytoskeleton, and cell adhesion. J Biol Chem 2013; 288:20966-20977. [PMID: 23737525 DOI: 10.1074/jbc.m113.484535] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
CAP (adenylyl cyclase-associated protein) was first identified in yeast as a protein that regulates both the actin cytoskeleton and the Ras/cAMP pathway. Although the role in Ras signaling does not extend beyond yeast, evidence supports that CAP regulates the actin cytoskeleton in all eukaryotes including mammals. In vitro actin polymerization assays show that both mammalian and yeast CAP homologues facilitate cofilin-driven actin filament turnover. We generated HeLa cells with stable CAP1 knockdown using RNA interference. Depletion of CAP1 led to larger cell size and remarkably developed lamellipodia as well as accumulation of filamentous actin (F-actin). Moreover, we found that CAP1 depletion also led to changes in cofilin phosphorylation and localization as well as activation of focal adhesion kinase (FAK) and enhanced cell spreading. CAP1 forms complexes with the adhesion molecules FAK and Talin, which likely underlie the cell adhesion phenotypes through inside-out activation of integrin signaling. CAP1-depleted HeLa cells also had substantially elevated cell motility as well as invasion through Matrigel. In summary, in addition to generating in vitro and in vivo evidence further establishing the role of mammalian CAP1 in actin dynamics, we identified a novel cellular function for CAP1 in regulating cell adhesion.
Collapse
Affiliation(s)
- Haitao Zhang
- From the Department of Biological Sciences and; Molecular Biosciences Program, Arkansas State University, State University, Arkansas 72467
| | - Pooja Ghai
- From the Department of Biological Sciences and; Molecular Biosciences Program, Arkansas State University, State University, Arkansas 72467
| | - Huhehasi Wu
- From the Department of Biological Sciences and
| | - Changhui Wang
- the Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, China, and
| | - Jeffrey Field
- the Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104.
| | - Guo-Lei Zhou
- From the Department of Biological Sciences and; Molecular Biosciences Program, Arkansas State University, State University, Arkansas 72467,.
| |
Collapse
|
24
|
Abstract
Alpha-actinins (ACTNs) were originally identified as cytoskeletal proteins which cross-link filamentous actin to establish cytoskeletal architect that protects cells from mechanical stress and controls cell movement. Notably, unlike other ACTNs, alpha-actinin 4 (ACTN4) displays unique characteristics in signaling transduction, nuclear translocation, and gene expression regulation. Initial reports indicated that ACTN4 is part of the breast cancer cell motile apparatus and is highly expressed in the nucleus. These results imply that ACTN4 plays a role in breast cancer tumorigenesis. While several observations in breast cancer and other cancers support this hypothesis, little direct evidence links the tumorigenic phenotype with ACTN4-mediated pathological mechanisms. Recently, several studies have demonstrated that in addition to its role in coordinating cytoskeleton, ACTN4 interacts with signaling mediators, chromatin remodeling factors, and transcription factors including nuclear receptors. Thus, ACTN4 functions as a versatile promoter for breast cancer tumorigenesis and appears to be an ideal drug target for future therapeutic development.
Collapse
Affiliation(s)
- Kuo-Sheng Hsu
- Department of Biochemistry, School of Medicine, Case Western Reserve University-CWRU, The Comprehensive Cancer Center of CWRU, Cleveland, Ohio, USA
| | | |
Collapse
|
25
|
Comparability of differential proteomics data generated from paired archival fresh-frozen and formalin-fixed samples by GeLC-MS/MS and spectral counting. J Proteomics 2012; 77:561-76. [PMID: 23043969 DOI: 10.1016/j.jprot.2012.09.033] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 09/17/2012] [Accepted: 09/22/2012] [Indexed: 11/22/2022]
Abstract
In this study, a Veterinary Department repository composed by paired formalin-fixed paraffin-embedded (FFPE) and fresh-frozen (FrFr) sets of the same tissues, routinely archived in the typical conditions of a clinical setting, was exploited to perform a comparative evaluation of the results generated by GeLC-MS/MS (1-DE followed by in-gel digestion and LC-MS/MS) and spectral counting with the two types of archival samples. Therefore, two parallel differential proteomic studies were performed using 3 canine mammary carcinomas and 3 normal controls in a paired fashion (6 FrFr and 6 FFPE in total). As a result, the FrFr and FFPE differential proteomic datasets exhibited fair consistency in differential expression trends, according to protein molecular function, cellular localization, networks, and pathways. However, FFPE samples were globally slightly less informative, especially concerning the high-MW subproteome. As a further investigation, new insights into the molecular aspects of protein fixation and retrieval were obtained. In conclusion, archival FFPE samples can be reliably used for differential proteomics studies employing a spectral counting GeLC-MS/MS approach, although some typical biases need to be taken into account, and FrFr specimens (when available) should still be considered as the gold standard for clinical proteomics.
Collapse
|
26
|
Li X, Wan L, Geng J, Wu CL, Bai X. Aldehyde Dehydrogenase 1A1 Possesses Stem-Like Properties and Predicts Lung Cancer Patient Outcome. J Thorac Oncol 2012; 7:1235-45. [DOI: 10.1097/jto.0b013e318257cc6d] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|