1
|
Sevgen S, Kara G, Kir AS, Şahin A, Boyaci E. A critical review of bioanalytical and clinical applications of solid phase microextraction. J Pharm Biomed Anal 2025; 252:116487. [PMID: 39378761 DOI: 10.1016/j.jpba.2024.116487] [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: 08/09/2024] [Revised: 09/07/2024] [Accepted: 09/26/2024] [Indexed: 10/10/2024]
Abstract
Studying the functions, mechanisms, and effects of drugs and other exogenous compounds on biological systems, together with investigations performed to understand biosystems better, comprises one of the most fascinating areas of research. Although classical sample preparation techniques are dominantly used to infer the relevant information from the investigated system, they fail to meet various imperative requirements, such as being environmentally friendly, applicable in-vivo, and compatible with online analysis. As a chameleon in the analytical toolbox, solid phase microextraction (SPME) is one of the best tools available for studying biological systems in unconventional ways. In this review, SPME is spotlighted, and its capability for bioanalytical applications, including drug analysis, untargeted and targeted metabolomics, in-vivo and clinical studies, is scrutinized based on studies reported in the past five years. In addition, novel extractive phases and instrumental coupling strategies developed to serve bioanalytical research are discussed to give the perspective for state-of-the-art and future developments. The literature assessment showed that SPME could act as a critical tool to investigate in-vivo biological systems and provide information about the elusive portion of the metabolome. Moreover, recently introduced miniaturized SPME probes further improved the low-invasive nature of the sampling and enabled sampling even from a single cell. The coupling of SPME directly to mass spectrometry significantly reduced the total analytical workflow and became one of the promising tools suitable for fast diagnostic purposes and drug analysis. The numerous applications and advancements reported in bioanalysis using SPME show that it will continue to be an indispensable technique in the future.
Collapse
Affiliation(s)
- Sılanur Sevgen
- Department of Chemistry, Middle East Technical University, Ankara 06800, Türkiye
| | - Gökşin Kara
- Department of Chemistry, Middle East Technical University, Ankara 06800, Türkiye
| | - Aysegul Seyma Kir
- Department of Chemistry, Middle East Technical University, Ankara 06800, Türkiye
| | - Alper Şahin
- Department of Chemistry, Middle East Technical University, Ankara 06800, Türkiye
| | - Ezel Boyaci
- Department of Chemistry, Middle East Technical University, Ankara 06800, Türkiye.
| |
Collapse
|
2
|
Odiase P, Ma J, Ranganathan S, Ogunkua O, Turner WB, Marshall D, Ochieng J. The Role of Fetuin-A in Tumor Cell Growth, Prognosis, and Dissemination. Int J Mol Sci 2024; 25:12918. [PMID: 39684629 DOI: 10.3390/ijms252312918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Fetuin-A, also known as alpha-2-Heremans-Schmid-glycoprotein (Ahsg), is a multifunctional molecule with diverse roles in biological processes such as mineralization, tumor growth, and inflammation. This review explores the involvement of Ahsg in various cancers, including liver, breast, prostate, colorectal, brain, osteosarcoma, and lung cancers. In many cancer types, Ahsg promotes tumor growth, invasion, and metastasis through various mechanisms, including cellular adhesion, spreading, chemotaxis, and modulation of cell-growth signaling pathways. Additionally, Ahsg has been implicated in the regulation of inflammatory cytokine production, making it a potential marker of inflammation in cancer. The complex interplay between Ahsg and cancer progression highlights its potential as a diagnostic biomarker and therapeutic target in various cancers. However, further research is needed to fully elucidate the mechanisms of action of Ahsg in cancer and to explore its clinical implications in cancer diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- Peace Odiase
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
| | - Jonathan Ma
- College of Arts and Science, Vanderbilt University, Nashville, TN 37203, USA
| | | | - Olugbemiga Ogunkua
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
| | - Winston B Turner
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
| | - Dana Marshall
- Department of Pathology, Meharry Medical College, Nashville, TN 37208, USA
| | - Josiah Ochieng
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, Meharry Medical College, Nashville, TN 37208, USA
- Department of Biomedical Science, School of Graduate Studies Meharry Medical College, Nashville, TN 37208, USA
| |
Collapse
|
3
|
Xiong F, Zheng Y, Ouyang Y, Song X, Jia S, Wang G, Wang S, Liu Q, Zhao J, Zhang W. Comparison of three methods for collecting interstitial fluid from subcutaneous tissue in mini pigs. MethodsX 2024; 12:102700. [PMID: 38633419 PMCID: PMC11022106 DOI: 10.1016/j.mex.2024.102700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 04/05/2024] [Indexed: 04/19/2024] Open
Abstract
Interstitial fluid, owing to its similarity to blood components and higher sensitivity and specificity, finds widespread application in disease diagnosis and tumor marker detection. However, collecting interstitial fluid, particularly from the deep subcutaneous connective tissue, remains challenging.•This study aimed to compare three different collection methods - push-pull perfusion, multi-filament nylon thread implantation, and tissue centrifugation - for collecting interstitial fluid from the subcutaneous connective tissue layer of mini-pigs. High-performance ion chromatography was employed to analyze the conventional cation components in the samples and compare ion composition analysis between the different methods.•Results indicated that while the distribution of conventional cations in the interstitial fluid collected by the three methods was generally consistent, there were slight variations in the detection rates and concentrations of different ions. Hence, suitable collection methods should be selected based on the ions or collection sites of interest.
Collapse
Affiliation(s)
- Feng Xiong
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Zheng
- Beijing Nuclear Industry Hospital, Beijing, China
| | - Yinggen Ouyang
- Department of Radiochemistry, China Institute of Atomic Energy, Beijing, China
| | - Xiaojing Song
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shuyong Jia
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guangjun Wang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shuyou Wang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qi Liu
- College of Acupuncture and Massage, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jing Zhao
- Department of Radiochemistry, China Institute of Atomic Energy, Beijing, China
| | - Weibo Zhang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
4
|
Mirza MR, Sultan R, Choudhary MI, Tahir M, Larsen MR, Tariq S, Rahman SU. Label-free quantitation of the changes in salivary proteome associated with the chronic consumption of the betel nut ( Areca catechu). Mol Omics 2021; 18:123-132. [PMID: 34851339 DOI: 10.1039/d1mo00391g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Areca nut or betel nut chewing is most frequently used in Pakistan and is associated with a high risk for oral cancer. Until now, however, there has not been any research conducted on the long-term effect(s) of betel nut chewing on the saliva proteome. In the present study, initially, the changes in the saliva proteome associated with betel nut chewing were investigated. Secondly, the analysis was focused on the changes in salivary proteome with respect to prolonged usage of betel nuts. After extraction, the saliva proteins were digested into peptides and these were subsequently analyzed using mass spectrometry. Data are available via ProteomeXchange with identifier PXD029768. Label-free quantitation of saliva samples revealed a total of 12 proteins that were differentially expressed between betel nut addicts (BNAs), and the control group. The study groups were further divided into three subgroups, the BNA-1, BNA-2, and BNA-3 groups, with respect to the extent of consumption of betel nuts in terms of years. The data analysis revealed a more detailed profiling of proteins expressed after five, ten, and more than ten years of betel nut consumption. A total of 30, 17, and 22 proteins were found to be differentially expressed when divided into the BNA-1, BNA-2, and BNA-3 groups. The present study shows that the chronic usage of betel nuts leads to the expression of proteins, such as SPARC1, profilin, and SBSN, which are known to be involved in head and neck cancers.
Collapse
Affiliation(s)
- Munazza Raza Mirza
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences ICCBS), University of Karachi, Karachi-75270, Pakistan.
| | - Rabia Sultan
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences ICCBS), University of Karachi, Karachi-75270, Pakistan.
| | - Muhammad Iqbal Choudhary
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences ICCBS), University of Karachi, Karachi-75270, Pakistan. .,H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan.,Department of Biochemistry, Faculty of Science, King Abdul Aziz University, Jeddah-21412, Saudi Arabia
| | - Muhammad Tahir
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Martin Røssel Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Saria Tariq
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences ICCBS), University of Karachi, Karachi-75270, Pakistan.
| | - Saeed Ur Rahman
- Oral Biology, Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| |
Collapse
|
5
|
Ura B, Di Lorenzo G, Romano F, Monasta L, Mirenda G, Scrimin F, Ricci G. Interstitial Fluid in Gynecologic Tumors and Its Possible Application in the Clinical Practice. Int J Mol Sci 2018; 19:ijms19124018. [PMID: 30545144 PMCID: PMC6321738 DOI: 10.3390/ijms19124018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 11/29/2018] [Indexed: 12/12/2022] Open
Abstract
Gynecologic cancers are an important cause of worldwide mortality. The interstitium consists of solid and fluid phases, situated between the blood vessels and cells. The interstitial fluid (IF), or fluid phase, is an extracellular fluid bathing and surrounding the tissue cells. The TIF (tumor interstitial fluid) is a dynamic fluid rich in lipids, proteins and enzyme-derived substances. The molecules found in the IF may be associated with pathological changes in tissues leading to cancer growth and metastatization. Proteomic techniques have allowed an extensive study of the composition of the TIF as a source of biomarkers for gynecologic cancers. In our review, we analyze the composition of the TIF, its formation process, the sampling methods, the consequences of its accumulation and the proteomic analyses performed, that make TIF valuable for monitoring different types of cancers.
Collapse
Affiliation(s)
- Blendi Ura
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Giovanni Di Lorenzo
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Federico Romano
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Lorenzo Monasta
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Giuseppe Mirenda
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Federica Scrimin
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
| | - Giuseppe Ricci
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", 34137 Trieste, Italy.
- Department of Medical, Surgery and Health Sciences, University of Trieste, 34137 Trieste, Italy.
| |
Collapse
|
6
|
Haslene-Hox H. Measuring gradients in body fluids - A tool for elucidating physiological processes, diagnosis and treatment of disease. Clin Chim Acta 2018; 489:233-241. [PMID: 30145208 DOI: 10.1016/j.cca.2018.08.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/15/2018] [Accepted: 08/16/2018] [Indexed: 01/03/2023]
Affiliation(s)
- Hanne Haslene-Hox
- SINTEF Industry, Department of biotechnology and nanomedicine, Sem Sælands vei 2A, 7034 Trondheim, Norway.
| |
Collapse
|
7
|
Hou X, Liu R, Huang C, Jiang L, Zhou Y, Chen Q. Cyclophilin A was revealed as a candidate marker for human oral submucous fibrosis by proteomic analysis. Cancer Biomark 2018; 20:345-356. [PMID: 28826174 DOI: 10.3233/cbm-170142] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Oral submucous fibrosis (OSF) is a chronic insidious disease which predisposes to oral cancer. Understanding the molecular markers for OSF is critical for diagnosis and treatment of oral cancer. In this study, the proteins expression profile of OSF tissues was compared to normal mucous tissues by 2 dimensional electrophoresis (2-DE). The 2-DE images were analyzed through cut, spot detection and match analysis using mass spectrometry (MS). Differentially expressed genes were identified as candidates. RT-PCR, Western Blot and immunohistochemistry were performed to validate the difference in expression of the candidates between OSF and normal mucous tissues. The shRNA targeted to the candidates were then transfected by Lipofectamine2000 to the 3T3 cells to study gene function. Cell proliferation and apoptosis were measured by MTT, clonogenic formation, PI and TUNEL staining. From the proteomic analysis, 94 of the 182 selected spots with differential expression were identified by MS analysis and Cyclophilin A (CYPA) was determined to be the OSF-associated protein candidate. The significant differences in expression between OSF and normal tissues were verified and confirmed by RT-PCR, Western blot and Immunohistochemical analysis. Inhibition of CYPA expression by RNA interference suggested its potential activities involved in cell proliferation and apoptosis process. In conclusion, these results indicated a novel molecular mechanism of OSF pathogenesis and demonstrated CYPA as a potential biomarker and gene intervention targets of OSF. These data may help the development for therapeutics of oral cancer.
Collapse
Affiliation(s)
- Xiaohui Hou
- Laboratory of Oral Biomedical Science and Translational Medicine, School of Stomatology, Tongji University, Shanghai 200072, China.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Rui Liu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Canhua Huang
- The State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
| | - Lu Jiang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yu Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| |
Collapse
|
8
|
Innovative methods for biomarker discovery in the evaluation and development of cancer precision therapies. Cancer Metastasis Rev 2018; 37:125-145. [PMID: 29392535 DOI: 10.1007/s10555-017-9710-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The discovery of biomarkers able to detect cancer at an early stage, to evaluate its aggressiveness, and to predict the response to therapy remains a major challenge in clinical oncology and precision medicine. In this review, we summarize recent achievements in the discovery and development of cancer biomarkers. We also highlight emerging innovative methods in biomarker discovery and provide insights into the challenges faced in their evaluation and validation.
Collapse
|
9
|
Costanza B, Turtoi A, Bellahcène A, Hirano T, Peulen O, Blomme A, Hennequière V, Mutijima E, Boniver J, Meuwis MA, Josse C, Koopmansch B, Segers K, Yokobori T, Fahmy K, Thiry M, Coimbra C, Garbacki N, Colige A, Baiwir D, Bours V, Louis E, Detry O, Delvenne P, Nishiyama M, Castronovo V. Innovative methodology for the identification of soluble biomarkers in fresh tissues. Oncotarget 2018. [PMID: 29535834 PMCID: PMC5828218 DOI: 10.18632/oncotarget.24366] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The identification of diagnostic and prognostic biomarkers from early lesions, measurable in liquid biopsies remains a major challenge, particularly in oncology. Fresh human material of high quality is required for biomarker discovery but is often not available when it is totally required for clinical pathology investigation. Hence, all OMICs studies are done on residual and less clinically relevant biological samples. Here after, we present an innovative, simple, and non-destructive, procedure named EXPEL that uses rapid, pressure-assisted, interstitial fluid extrusion, preserving the specimen for full routine clinical pathology investigation. In the meantime, the technique allows a comprehensive OMICs analysis (proteins, metabolites, miRNAs and DNA). As proof of concept, we have applied EXPEL on freshly collected human colorectal cancer and liver metastases tissues. We demonstrate that the procedure efficiently allows the extraction, within a few minutes, of a wide variety of biomolecules holding diagnostic and prognostic potential while keeping both tissue morphology and antigenicity unaltered. Our method enables, for the first time, both clinicians and scientists to explore identical clinical material regardless of its origin and size, which has a major positive impact on translation to the clinic.
Collapse
Affiliation(s)
- Brunella Costanza
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Andrei Turtoi
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Akeila Bellahcène
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Touko Hirano
- Laboratory for Analytical Instruments, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Olivier Peulen
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Arnaud Blomme
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Vincent Hennequière
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Eugene Mutijima
- Department of Pathology, University Hospital (CHU), University of Liège, Liège, Belgium
| | - Jacques Boniver
- Department of Pathology, University Hospital (CHU), University of Liège, Liège, Belgium
| | - Marie-Alice Meuwis
- Gastroenterology Department, University Hospital (CHU), University of Liège, Liège, Belgium
| | - Claire Josse
- Center for Human Genetic, Molecular Haemato-Oncology Unit, UniLab, University Hospital (CHU), University of Liège, Liège, Belgium
| | - Benjamin Koopmansch
- Center for Human Genetic, Molecular Haemato-Oncology Unit, UniLab, University Hospital (CHU), University of Liège, Liège, Belgium
| | - Karin Segers
- Center for Human Genetic, Molecular Haemato-Oncology Unit, UniLab, University Hospital (CHU), University of Liège, Liège, Belgium
| | - Takehiko Yokobori
- Division of Integrated Oncology Research, Research Program for Omics-based Medical Science, Gunma University Initiative for Advanced Research, Gunma, Japan
| | - Karim Fahmy
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| | - Marc Thiry
- Laboratory of Cell Biology, Faculty of Sciences, University of Liège, Liège, Belgium
| | - Carla Coimbra
- Department of Abdominal Surgery, University Hospital (CHU), University of Liège, Liège, Belgium
| | - Nancy Garbacki
- Laboratory of Connective Tissues Biology, GIGA-Cancer, University Hospital, University of Liège, Liège, Belgium
| | - Alain Colige
- Laboratory of Connective Tissues Biology, GIGA-Cancer, University Hospital, University of Liège, Liège, Belgium
| | - Dominique Baiwir
- Mass Spectrometry Laboratory, University of Liège, Liège, Belgium.,GIGA Proteomics Facility, University of Liège, Liège, Belgium
| | - Vincent Bours
- Center for Human Genetic, Molecular Haemato-Oncology Unit, UniLab, University Hospital (CHU), University of Liège, Liège, Belgium
| | - Edouard Louis
- Gastroenterology Department, University Hospital (CHU), University of Liège, Liège, Belgium
| | - Olivier Detry
- Department of Abdominal Surgery, University Hospital (CHU), University of Liège, Liège, Belgium
| | - Philippe Delvenne
- Department of Pathology, University Hospital (CHU), University of Liège, Liège, Belgium
| | - Masahiko Nishiyama
- Division of Integrated Oncology Research, Research Program for Omics-based Medical Science, Gunma University Initiative for Advanced Research, Gunma, Japan.,Department of Molecular Pharmacology and Oncology, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Vincent Castronovo
- Metastasis Research Laboratory, GIGA Cancer, University of Liège, Liège, Belgium
| |
Collapse
|
10
|
Abstract
Tumor interstitial fluid (TIF) surrounds and perfuses bodily tumorigenic tissues and cells, and can accumulate by-products of tumors and stromal cells in a relatively local space. Interstitial fluid offers several important advantages for biomarker and therapeutic target discovery, especially for cancer. Here, we describe the most currently accepted method for recovering TIF from tumor and nonmalignant tissues that was initially performed using breast cancer tissue. TIF recovery is achieved by passive extraction of fluid from small, surgically dissected tissue specimens in phosphate-buffered saline. We also present protocols for hematoxylin and eosin (H&E) staining of snap-frozen and formalin-fixed, paraffin-embedded (FFPE) tumor sections and for proteomic profiling of TIF and matched tumor samples by high-resolution two-dimensional gel electrophoresis (2D-PAGE) to enable comparative analysis of tumor secretome and paired tumor tissue.
Collapse
|
11
|
Greening DW, Kapp EA, Simpson RJ. The Peptidome Comes of Age: Mass Spectrometry-Based Characterization of the Circulating Cancer Peptidome. Enzymes 2017; 42:27-64. [PMID: 29054270 DOI: 10.1016/bs.enz.2017.08.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Peptides play a seminal role in most physiological processes acting as neurotransmitters, hormones, antibiotics, and immune regulation. In the context of tumor biology, it is hypothesized that endogenous peptides, hormones, cytokines, growth factors, and aberrant degradation of select protein networks (e.g., enzymatic activities, protein shedding, and extracellular matrix remodeling) are fundamental in mediating cancer progression. Analysis of peptides in biological fluids by mass spectrometry holds promise of providing sensitive and specific diagnostic and prognostic information for cancer and other diseases. The identification of circulating peptides in the context of disease constitutes a hitherto source of new clinical biomarkers. The field of peptidomics can be defined as the identification and comprehensive analysis of physiological and pathological peptides. Like proteomics, peptidomics has been advanced by the development of new separation strategies, analytical detection methods such as mass spectrometry, and bioinformatic technologies. Unlike proteomics, peptidomics is targeted toward identifying endogenous protein and peptide fragments, defining proteolytic enzyme substrate specificity, as well as protease cleavage recognition (degradome). Peptidomics employs "top-down proteomics" strategies where mass spectrometry is applied at the proteoform level to analyze intact proteins and large endogenous peptide fragments. With recent advances in prefractionation workflows for separating peptides, mass spectrometry instrumentation, and informatics, peptidomics is an important field that promises to impact on translational medicine. This review covers the current advances in peptidomics, including top-down and imaging mass spectrometry, comprehensive quantitative peptidome analyses (developments in reproducibility and coverage), peptide prefractionation and enrichment workflows, peptidomic data analyses, and informatic tools. The application of peptidomics in cancer biomarker discovery will be discussed.
Collapse
Affiliation(s)
- David W Greening
- La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, Victoria, Australia.
| | - Eugene A Kapp
- Systems Biology & Personalised Medicine Division, Walter & Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Florey Institute of Neuroscience, Parkville, Victoria, Australia; University of Melbourne, Parkville, Victoria, Australia
| | - Richard J Simpson
- La Trobe Institute for Molecular Science (LIMS), La Trobe University, Melbourne, Victoria, Australia.
| |
Collapse
|
12
|
Papaleo E, Gromova I, Gromov P. Gaining insights into cancer biology through exploration of the cancer secretome using proteomic and bioinformatic tools. Expert Rev Proteomics 2017; 14:1021-1035. [PMID: 28967788 DOI: 10.1080/14789450.2017.1387053] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Tumor-associated proteins released by cancer cells and by tumor stroma cells, referred as 'cancer secretome', represent a valuable resource for discovery of potential cancer biomarkers. The last decade was marked by a great increase in number of studies focused on various aspects of cancer secretome including, composition and identification of components externalized by malignant cells and by the components of tumor microenvironment. Areas covered: Here, we provide an overview of achievements in the proteomic analysis of the cancer secretome, elicited through the tumor-associated interstitial fluid recovered from malignant tissues ex vivo or the protein component of conditioned media obtained from cultured cancer cells in vitro. We summarize various bioinformatic tools and approaches and critically appraise their outcomes, focusing on problems and challenges that arise when applied for the analysis of cancer secretomic databases. Expert commentary: Recent achievements in the omics- analysis of structural and metabolic aspects of altered cancer secretome contribute greatly to the various hallmarks of cancer including the identification of clinically significant biomarkers and potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Elena Papaleo
- a Danish Cancer Society Research Center, Computational Biology Laboratory , Copenhagen , Denmark
| | - Irina Gromova
- b Danish Cancer Society Research Center, Genome Integrity Unit, Breast Cancer Biology Group , Copenhagen , Denmark
| | - Pavel Gromov
- b Danish Cancer Society Research Center, Genome Integrity Unit, Breast Cancer Biology Group , Copenhagen , Denmark
| |
Collapse
|
13
|
Composition and Function of the Interstitial Fluid. Protein Sci 2016. [DOI: 10.1201/9781315374307-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
14
|
Gomez-Auli A, Hillebrand LE, Biniossek ML, Peters C, Reinheckel T, Schilling O. Impact of cathepsin B on the interstitial fluid proteome of murine breast cancers. Biochimie 2016; 122:88-98. [DOI: 10.1016/j.biochi.2015.10.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/06/2015] [Indexed: 12/28/2022]
|
15
|
Wagner M, Wiig H. Tumor Interstitial Fluid Formation, Characterization, and Clinical Implications. Front Oncol 2015; 5:115. [PMID: 26075182 PMCID: PMC4443729 DOI: 10.3389/fonc.2015.00115] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 05/06/2015] [Indexed: 12/18/2022] Open
Abstract
The interstitium, situated between the blood and lymph vessels and the cells, consists of a solid or matrix phase and a fluid phase representing the tissue microenvironment. In the present review, we focus on the interstitial fluid phase of solid tumors, the tumor interstitial fluid (TIF), i.e., the fluid bathing the tumor and stroma cells, also including immune cells. This is a component of the internal milieu of a solid tumor that has attracted regained attention. Access to this space may provide important insight into tumor development and therapy response. TIF is formed by transcapillary filtration, and since this fluid is not readily available we discuss available techniques for TIF isolation, results from subsequent characterization and implications of recent findings with respect to fluid filtration and uptake of macromolecular therapeutic agents. There appear to be local gradients in signaling substances from neoplastic tissue to plasma that may provide new understanding of tumor biology. The development of sensitive proteomic technologies has made TIF a valuable source for tumor specific proteins and biomarker candidates. Potential biomarkers will appear locally in high concentrations in tumors and may eventually be found diluted in the plasma. Access to TIF that reliably reflects the local tumor microenvironment enables identification of substances that can be used in early detection and monitoring of disease.
Collapse
Affiliation(s)
- Marek Wagner
- Department of Biomedicine, University of Bergen , Bergen , Norway
| | - Helge Wiig
- Department of Biomedicine, University of Bergen , Bergen , Norway
| |
Collapse
|
16
|
Rizwani W, Fasim A, Sharma D, Reddy DJ, Bin Omar NAM, Singh SS. S137 phosphorylation of profilin 1 is an important signaling event in breast cancer progression. PLoS One 2014; 9:e103868. [PMID: 25084196 PMCID: PMC4118959 DOI: 10.1371/journal.pone.0103868] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 07/02/2014] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Profilins are actin-modulating proteins regulating many intracellular functions based on their multiple and diverse ligand interactions. They have been implicated to play a role in many pathological conditions such as allergies, cardiovascular diseases, muscular atrophy, diabetes, dementia and cancer. Post-translational modifications of profilin 1 can alter its properties and subsequently its function in a cell. In the present study, we identify the importance of phosphorylation of profilin 1 at serine 137 (S137) residue in breast cancer progression. METHODS/PRINCIPAL FINDINGS We found elevated profilin 1 (PFN) in human breast cancer tissues when compared to adjacent normal tissues. Overexpression of wild-type profilin 1 (PFN-WT) in breast cancer MCF7 cells made them more migratory, invasive and adherent independent in comparison to empty vector transfected cells. Mutation in serine phosphorylation site (S137) of profilin 1 (PFN-S137A) significantly abrogated these properties. Mutation affecting actin-binding ability (PFN-R74E) of profilin 1 enhanced its tumorigenic function whereas mutation affecting its poly-L-proline binding function (PFN-H133S) alleviated these mechanisms in breast cancer cells. PFN-WT was found to activate matrix metalloproteinases by zymography, MMP2 and MMP9 in presence of PDBu (phorbol 12, 13 dibutyrate, PI3K agonist) to enhance migration and invasion in MCF7 cells while PFN-S137A did not. Phosphorylation increased migration and invasion in other mutants of profilin 1. Nuclear profilin levels also increased in the presence of PDBu. CONCLUSIONS Previous studies show that profilin could be executing a dual role in cancer by either suppressing or promoting tumorigenesis in a context dependent manner. In this study we demonstrate for the first time that phosphorylation of profilin 1 at serine 137 enhances oncogenic properties in breast cancer cells. Inhibitors targeting profilin 1 phosphorylation directly or indirectly through inhibition of kinases that phosphorylate profilin could be valuable therapeutic agents that can alter its activity and thereby control the progression of cancer.
Collapse
Affiliation(s)
- Wasia Rizwani
- Department of Biochemistry, Osmania University, Hyderabad, A.P., India
- * E-mail: (WR); (SSS)
| | - Aneesa Fasim
- Department of Biochemistry, Osmania University, Hyderabad, A.P., India
| | - Deepshikha Sharma
- Department of Biochemistry, Osmania University, Hyderabad, A.P., India
| | - Divya J. Reddy
- Department of Biochemistry, Osmania University, Hyderabad, A.P., India
| | | | - Surya S. Singh
- Department of Biochemistry, Osmania University, Hyderabad, A.P., India
- * E-mail: (WR); (SSS)
| |
Collapse
|
17
|
Abstract
The secretome, or secretomics, has recently emerged as a new term to describe the global study of proteins that are secreted by a cell, tissue or organism at any given time or under certain conditions. The secretome constitutes an important class of proteins that control and regulate a multitude of biological and physiological processes, thus making it a clinically relevant source for biomarkers and therapeutic target discoveries. There are several approaches that are being implemented to study such a class of proteins; however, each of these approaches has its advantages and limitations. While genome-wide studies using signal predictions can provide a comprehensive analysis of the secretome, the detection and quantification of the actual secreted proteins in a tissue would be more relevant. The goal of this review is to provide an overview of the methods currently used to analyze such a class of proteins, as well as the challenges encountered during the study of the secretome. The implication of studying the cell secretome together with its clinical relevance will be also covered.
Collapse
Affiliation(s)
- Yetrib Hathout
- Children's National Medical Center, Center for Genetic Medicine, Washington, DC 20010, USA.
| |
Collapse
|
18
|
Gromov P, Gromova I, Olsen CJ, Timmermans-Wielenga V, Talman ML, Serizawa RR, Moreira JM. Tumor interstitial fluid — A treasure trove of cancer biomarkers. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2259-70. [DOI: 10.1016/j.bbapap.2013.01.013] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 01/09/2013] [Accepted: 01/14/2013] [Indexed: 12/11/2022]
|
19
|
Harn HJ, Chen YL, Lin PC, Cheng YL, Lee SC, Chiou TW, Yang HH. Exploration of Potential Tumor Markers for Lung Adenocarcinomas by Two-Dimensional Gel Electrophoresis Coupled with Nano-LC/MS/MS. J CHIN CHEM SOC-TAIP 2013. [DOI: 10.1002/jccs.201000029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
20
|
Lin Q, Tan HT, Lim HSR, Chung MCM. Sieving through the cancer secretome. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2360-71. [PMID: 23376431 DOI: 10.1016/j.bbapap.2013.01.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Revised: 01/03/2013] [Accepted: 01/24/2013] [Indexed: 12/22/2022]
Abstract
Cancer is among the most prevalent and serious health problems worldwide. Therefore, there is an urgent need for novel cancer biomarkers with high sensitivity and specificity for early detection and management of the disease. The cancer secretome, encompassing all the proteins that are secreted by cancer cells, is a promising source of biomarkers as the secreted proteins are most likely to enter the blood circulation. Moreover, since secreted proteins are responsible for signaling and communication with the tumor microenvironment, studying the cancer secretome would further the understanding of cancer biology. Latest developments in proteomics technologies have significantly advanced the study of the cancer secretome. In this review, we will present an overview of the secretome sample preparation process and summarize the data from recent secretome studies of six common cancers with high mortality (breast, colorectal, gastric, liver, lung and prostate cancers). In particular, we will focus on the various platforms that were employed and discuss the clinical applicability of the key findings in these studies. This article is part of a Special Issue entitled: An Updated Secretome.
Collapse
Affiliation(s)
- Qifeng Lin
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, 117597 Singapore
| | | | | | | |
Collapse
|
21
|
Haslene-Hox H, Tenstad O, Wiig H. Interstitial fluid-a reflection of the tumor cell microenvironment and secretome. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:2336-46. [PMID: 23376185 DOI: 10.1016/j.bbapap.2013.01.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 12/27/2012] [Accepted: 01/24/2013] [Indexed: 02/07/2023]
Abstract
The interstitium or interstitial space describes the space outside the blood and lymphatic vessels. It contains two phases; the interstitial fluid (IF) and the extracellular matrix. In this review we focus on the interstitial fluid phase, which is the physical and biochemical microenvironment of the cells, and more specifically that of tumors. IF is created by transcapillary filtration and cleared by lymphatic vessels, and contains substances that are either produced and secreted locally, thus denoted secretome, or brought to the organ by the circulation. The structure of the interstitium is discussed briefly and moreover techniques for IF isolation focusing on those that are relevant for studies of the secretome. Accumulated data show that tumor IF is hypoxic and acidic compared with subcutaneous IF and plasma, and that there are gradients between IF and plasma giving information on where substances are produced and thereby reflecting the local microenvironment. We review recent data on the origin of tissue specific substances, challenges related to isolating a representative secretome and the use of this as a substrate for biomarker identification. Finally we perform a comparative analysis across human tumor types and techniques and show that there is great variation in the results obtained that may at least partially be due to the isolation method used. We conclude that when care is taken in isolation of substrate, analysis of the secretome may give valuable biological insight and result in identification of biomarker candidates. This article is part of a Special Issue entitled: An Updated Secretome.
Collapse
|
22
|
Cierniewski CS, Sobierajska K, Selmi A, Kryczka J, Bednarek R. Thymosin β4 is rapidly internalized by cells and does not induce intracellular Ca2+ elevation. Ann N Y Acad Sci 2013; 1269:44-52. [PMID: 23045969 DOI: 10.1111/j.1749-6632.2012.06685.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Thymosin β4 (Tβ4) is a multifunctional protein that has pleiotropic activities both intracellularly and extracellularly. The mechanisms by which it influences cellular processes such as adhesion, migration, differentiation, or apoptosis are not yet understood. Calcium is a ubiquitous signal molecule that is involved in the regulation of almost all cellular functions. Our data indicate that the release of Ca(2+) from intracellular stores following stimulation of cells with Tβ4 does not occur. Interestingly, Tβ4 becomes rapidly internalized, supporting the concept that it may express its activities via intracellular receptors.
Collapse
Affiliation(s)
- Czeslaw S Cierniewski
- Department of Molecular and Medical Biophysics, Medical University of Lodz, Lodz, Poland.
| | | | | | | | | |
Collapse
|
23
|
Clough GF, Stenken JA, Church MK. High Molecular Weight Targets and Treatments Using Microdialysis. MICRODIALYSIS IN DRUG DEVELOPMENT 2013. [DOI: 10.1007/978-1-4614-4815-0_13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
24
|
Bousette N, Gramolini AO, Kislinger T. Proteomics-based investigations of animal models of disease. Proteomics Clin Appl 2012; 2:638-53. [PMID: 21136864 DOI: 10.1002/prca.200780043] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cells contain a large yet, constant genome, which contains all the coding information necessary to sustain cellular physiology. However, proteins are the end products of genes, and hence dictate the phenotype of cells and tissues. Therefore, proteomics can provide key information for the elucidation of physiological and pathophysiological mechanisms by identifying the protein profile from cells and tissues. The relatively novel techniques used for the study of proteomics thus have the potential to improve diagnostic, prognostic, as well as therapeutic avenues. In this review, we first discuss the benefits of animal models over the use of human samples for the proteomic analysis of human disease. Next, we aim to demonstrate the potential of proteomics in the elucidation of disease mechanisms that may not be possible by other conventional technologies. Following this, we describe the use of proteomics for the analysis of PTM and protein interactions in animal models and their relevance to the study of human disease. Finally, we discuss the development of clinical biomarkers for the early diagnosis of disease via proteomic analysis of animal models. We also discuss the development of standard proteomes and relate how this data will benefit future proteomic research. A comprehensive review of all animal models used in conjunction with proteomics is beyond the scope of this manuscript. Therefore, we aimed to cover a large breadth of topics, which together, demonstrate the potential of proteomics as a powerful tool in biomedical research.
Collapse
Affiliation(s)
- Nicolas Bousette
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada; Heart and Stroke/Richard Lewar Centre of Cardiovascular Excellence, Toronto, Ontario, Canada
| | | | | |
Collapse
|
25
|
Zhu W, Gallo RL, Huang CM. Sampling human indigenous saliva peptidome using a lollipop-like ultrafiltration probe: simplify and enhance peptide detection for clinical mass spectrometry. J Vis Exp 2012:e4108. [PMID: 22895356 DOI: 10.3791/4108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Although human saliva proteome and peptidome have been revealed they were majorly identified from tryptic digests of saliva proteins. Identification of indigenous peptidome of human saliva without prior digestion with exogenous enzymes becomes imperative, since native peptides in human saliva provide potential values for diagnosing disease, predicting disease progression, and monitoring therapeutic efficacy. Appropriate sampling is a critical step for enhancement of identification of human indigenous saliva peptidome. Traditional methods of sampling human saliva involving centrifugation to remove debris may be too time-consuming to be applicable for clinical use. Furthermore, debris removal by centrifugation may be unable to clean most of the infected pathogens and remove the high abundance proteins that often hinder the identification of low abundance peptidome. Conventional proteomic approaches that primarily utilize two-dimensional gel electrophoresis (2-DE) gels in conjugation with in-gel digestion are capable of identifying many saliva proteins. However, this approach is generally not sufficiently sensitive to detect low abundance peptides/proteins. Liquid chromatography-Mass spectrometry (LC-MS) based proteomics is an alternative that can identify proteins without prior 2-DE separation. Although this approach provides higher sensitivity, it generally needs prior sample pre-fractionation and pre-digestion with trypsin, which makes it difficult for clinical use. To circumvent the hindrance in mass spectrometry due to sample preparation, we have developed a technique called capillary ultrafiltration (CUF) probes. Data from our laboratory demonstrated that the CUF probes are capable of capturing proteins in vivo from various microenvironments in animals in a dynamic and minimally invasive manner. No centrifugation is needed since a negative pressure is created by simply syringe withdrawing during sample collection. The CUF probes combined with LC-MS have successfully identified tryptic-digested proteins. In this study, we upgraded the ultrafiltration sampling technique by creating a lollipop-like ultrafiltration (LLUF) probe that can easily fit in the human oral cavity. The direct analysis by LC-MS without trypsin digestion showed that human saliva indigenously contains many peptide fragments derived from various proteins. Sampling saliva with LLUF probes avoided centrifugation but effectively removed many larger and high abundance proteins. Our mass spectrometric results illustrated that many low abundance peptides became detectable after filtering out larger proteins with LLUF probes. Detection of low abundance saliva peptides was independent of multiple-step sample separation with chromatography. For clinical application, the LLUF probes incorporated with LC-MS could potentially be used in the future to monitor disease progression from saliva.
Collapse
|
26
|
Wiig H, Swartz MA. Interstitial Fluid and Lymph Formation and Transport: Physiological Regulation and Roles in Inflammation and Cancer. Physiol Rev 2012; 92:1005-60. [PMID: 22811424 DOI: 10.1152/physrev.00037.2011] [Citation(s) in RCA: 478] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The interstitium describes the fluid, proteins, solutes, and the extracellular matrix (ECM) that comprise the cellular microenvironment in tissues. Its alterations are fundamental to changes in cell function in inflammation, pathogenesis, and cancer. Interstitial fluid (IF) is created by transcapillary filtration and cleared by lymphatic vessels. Herein we discuss the biophysical, biomechanical, and functional implications of IF in normal and pathological tissue states from both fluid balance and cell function perspectives. We also discuss analysis methods to access IF, which enables quantification of the cellular microenvironment; such methods have demonstrated, for example, that there can be dramatic gradients from tissue to plasma during inflammation and that tumor IF is hypoxic and acidic compared with subcutaneous IF and plasma. Accumulated recent data show that IF and its convection through the interstitium and delivery to the lymph nodes have many and diverse biological effects, including in ECM reorganization, cell migration, and capillary morphogenesis as well as in immunity and peripheral tolerance. This review integrates the biophysical, biomechanical, and biological aspects of interstitial and lymph fluid and its transport in tissue physiology, pathophysiology, and immune regulation.
Collapse
Affiliation(s)
- Helge Wiig
- Department of Biomedicine, University of Bergen, Bergen, Norway; and Laboratory of Lymphatic and Cancer Bioengineering, Institute of Bioengineering and Swiss Institute for Experimental Cancer Research, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Melody A. Swartz
- Department of Biomedicine, University of Bergen, Bergen, Norway; and Laboratory of Lymphatic and Cancer Bioengineering, Institute of Bioengineering and Swiss Institute for Experimental Cancer Research, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
27
|
Wang LJ, Matoso A, Sciandra KT, Yakirevich E, Sabo E, Zhang Y, Meitner PA, Tavares R, Noble L, Pareek G, DeLellis RA, Resnick MB. Expression of S100A4 in renal epithelial neoplasms. Appl Immunohistochem Mol Morphol 2012; 20:71-6. [PMID: 21691200 DOI: 10.1097/pai.0b013e31821fc8b7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Expression of S100A4 has been associated with progression and poor clinical outcome in a variety of malignancies including those of the breast, pancreas, bladder, and thyroid. To date, the expression of S100A4 protein in renal epithelial neoplasms is poorly understood. In this study, we evaluated the expression of S100A4 protein and mRNA in the nontumoral kidney and renal epithelial neoplasms of different types and correlated its expression with patient outcome. The study population included 155 clear cell renal cell carcinomas (cRCC), 22 papillary renal cell carcinomas (pRCC), 13 chromophobe renal cell carcinomas and 13 oncocytomas. In nontumoral kidney, nuclear and cytoplasmic S100A4 staining was detected in the glomerular epithelium and endothelium, distal tubules and collecting ducts, and loops of Henle. A different expression pattern was noted in the various neoplasms. S100A4 expression was significantly increased in the stromal cells in cRCC (83%) and pRCC (73%) compared with paired nontumoral kidney tissue (P<0.001). There was no increased stromal cell expression of S100A4 in oncocytomas and chromophobe carcinomas. Positive epithelial staining was more common in pRCC (58%) than cRCC (11%) (P=0.01). The level of mRNA detected by reverse transcription-polymerase chain reaction was significantly higher in the tumor as opposed to normal tissue in cRCC but not in the other neoplasms (P=0.03). Multivariate analysis revealed that epithelial S100A4 protein expression is an independent poor prognostic factor along with grade and stage only in cRCC (P<0.01). Although S100A4 protein was expressed in a minority of cRCC, its expression was associated with shorter overall patient survival.
Collapse
Affiliation(s)
- Li J Wang
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI 02903, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Poly(I:C) treatment influences the expression of calreticulin and profilin-1 in a human HNSCC cell line: a proteomic study. Tumour Biol 2012; 33:1201-8. [PMID: 22415225 DOI: 10.1007/s13277-012-0366-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Accepted: 02/15/2012] [Indexed: 12/15/2022] Open
Abstract
Polyinosinic:polycytidylic acid (poly (I:C)) has been formerly known to be an interferon inducer but the mechanism of its action was not revealed until the discovery of Toll-like receptors (TLRs). TLRs are members of transmembrane proteins that recognize conserved molecular motifs of viral and bacterial origin and initiate innate immune response. Recent studies have shown that they are also expressed on tumor cells, but their role in these cells is still not clear. TLR3 recognizes double-stranded RNA (poly (I:C)) and is primarily involved in the defense against viruses. TLR3 ligand binding initiates the activation of transcription factors NF-κB, IRF family members, and AP-1, which can induce wide cascading effect on the cell and consequently activate many cellular processes. Since little is known about TLR3 target genes, we have used the proteomic approach to widen the current knowledge. In this study, we have discovered 15 differentially expressed proteins, mostly connected with protein metabolic processes. Furthermore, we have confirmed by Western blot that calreticulin and profilin-1, proteins which have been shown previously to be involved in processes connected with tumor progression, are differentially expressed after poly(I:C) treatment. By using TLR3 small interfering RNA, we showed that calreticulin expression might be TLR3 dependent, unlike profilin-1.
Collapse
|
29
|
Stastna M, Van Eyk JE. Secreted proteins as a fundamental source for biomarker discovery. Proteomics 2012; 12:722-35. [PMID: 22247067 DOI: 10.1002/pmic.201100346] [Citation(s) in RCA: 136] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Revised: 07/26/2011] [Accepted: 08/10/2011] [Indexed: 12/18/2022]
Abstract
The proteins secreted by various cells (the secretomes) are a potential rich source of biomarkers as they reflect various states of the cells at real time and at given conditions. To have accessible, sufficient and reliable protein markers is desirable as they mark various stages of disease development and their presence/absence can be used for diagnosis, prognosis, risk stratification and therapeutic monitoring. As direct analysis of blood/plasma, a common and noninvasive patient screening method, can be difficult for candidate protein biomarker identification, the alternative/complementary approaches are required, one of them is the analysis of secretomes in cell conditioned media in vitro. As the proteins secreted by cells as a response to various stimuli are most likely secreted into blood/plasma, the identification and pre-selection of candidate protein biomarkers from cell secretomes with subsequent validation of their presence at higher levels in serum/plasma is a promising approach. In this review, we discuss the proteins secreted by three progenitor cell types (smooth muscle, endothelial and cardiac progenitor cells) and two adult cell types (neonatal rat ventrical myocytes and smooth muscle cells) which can be relevant to cardiovascular research and which have been recently published in the literature. We found, at least for secretome studies included in this review, that secretomes of progenitor and adult cells overlap by 48% but the secretomes are very distinct among progenitor cell themselves as well as between adult cells. In addition, we compared secreted proteins to protein identifications listed in the Human Plasma PeptideAtlas and in two reports with cardiovascular-related proteins and we performed the extensive literature search to find if any of these secreted proteins were identified in a biomarker study. As expected, many proteins have been identified as biomarkers in cancer but 18 proteins (out of 62) have been tested as biomarkers in cardiovascular diseases as well.
Collapse
Affiliation(s)
- Miroslava Stastna
- Johns Hopkins Bayview Proteomics Center, Department of Medicine, Division of Cardiology, School of Medicine, Johns Hopkins University, Baltimore, MD 21224, USA.
| | | |
Collapse
|
30
|
Zoidakis J, Makridakis M, Zerefos PG, Bitsika V, Esteban S, Frantzi M, Stravodimos K, Anagnou NP, Roubelakis MG, Sanchez-Carbayo M, Vlahou A. Profilin 1 is a potential biomarker for bladder cancer aggressiveness. Mol Cell Proteomics 2011; 11:M111.009449. [PMID: 22159600 DOI: 10.1074/mcp.m111.009449] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Of the most important clinical needs for bladder cancer (BC) management is the identification of biomarkers for disease aggressiveness. Urine is a "gold mine" for biomarker discovery, nevertheless, with multiple proteins being in low amounts, urine proteomics becomes challenging. In the present study we applied a fractionation strategy of urinary proteins based on the use of immobilized metal affinity chromatography for the discovery of biomarkers for aggressive BC. Urine samples from patients with non invasive (two pools) and invasive (two pools) BC were subjected to immobilized metal affinity chromatography fractionation and eluted proteins analyzed by 1D-SDS-PAGE, band excision and liquid chromatography tandem MS. Among the identified proteins, multiple corresponded to proteins with affinity for metals and/or reported to be phosphorylated and included proteins with demonstrated association with BC such as MMP9, fibrinogen forms, and clusterin. In agreement to the immobilized metal affinity chromatography results, aminopeptidase N, profilin 1, and myeloblastin were further found to be differentially expressed in urine from patients with invasive compared with non invasive BC and benign controls, by Western blot or Elisa analysis, nevertheless exhibiting high interindividual variability. By tissue microarray analysis, profilin 1 was found to have a marked decrease of expression in the epithelial cells of the invasive (T2+) versus high risk non invasive (T1G3) tumors with occasional expression in stroma; importantly, this pattern strongly correlated with poor prognosis and increased mortality. The functional relevance of profilin 1 was investigated in the T24 BC cells where blockage of the protein by the use of antibodies resulted in decreased cell motility with concomitant decrease in actin polymerization. Collectively, our study involves the application of a fractionation method of urinary proteins and as one main result of this analysis reveals the association of profilin 1 with BC paving the way for its further investigation in BC stratification.
Collapse
Affiliation(s)
- Jerome Zoidakis
- Biotechnology Division, Biomedical Research Foundation, Academy of Athens, Greece
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Zhu P, Bowden P, Zhang D, Marshall JG. Mass spectrometry of peptides and proteins from human blood. MASS SPECTROMETRY REVIEWS 2011; 30:685-732. [PMID: 24737629 DOI: 10.1002/mas.20291] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Revised: 12/09/2009] [Accepted: 01/19/2010] [Indexed: 06/03/2023]
Abstract
It is difficult to convey the accelerating rate and growing importance of mass spectrometry applications to human blood proteins and peptides. Mass spectrometry can rapidly detect and identify the ionizable peptides from the proteins in a simple mixture and reveal many of their post-translational modifications. However, blood is a complex mixture that may contain many proteins first expressed in cells and tissues. The complete analysis of blood proteins is a daunting task that will rely on a wide range of disciplines from physics, chemistry, biochemistry, genetics, electromagnetic instrumentation, mathematics and computation. Therefore the comprehensive discovery and analysis of blood proteins will rank among the great technical challenges and require the cumulative sum of many of mankind's scientific achievements together. A variety of methods have been used to fractionate, analyze and identify proteins from blood, each yielding a small piece of the whole and throwing the great size of the task into sharp relief. The approaches attempted to date clearly indicate that enumerating the proteins and peptides of blood can be accomplished. There is no doubt that the mass spectrometry of blood will be crucial to the discovery and analysis of proteins, enzyme activities, and post-translational processes that underlay the mechanisms of disease. At present both discovery and quantification of proteins from blood are commonly reaching sensitivities of ∼1 ng/mL.
Collapse
Affiliation(s)
- Peihong Zhu
- Department of Chemistry and Biology, Ryerson University, 350 Victoria Street, Toronto, Ontario, Canada M5B 2K3
| | | | | | | |
Collapse
|
32
|
Comparison of alternative extraction methods for secretome profiling in human hepatocellular carcinoma cells. SCIENCE CHINA-LIFE SCIENCES 2011; 54:34-8. [DOI: 10.1007/s11427-010-4122-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Accepted: 08/30/2010] [Indexed: 12/25/2022]
|
33
|
Affiliation(s)
| | - Junying Yuan
- Department of Cell Biology, Harvard Medical School, 240 Longwood Ave. Boston, MA 02115
| |
Collapse
|
34
|
Makridakis M, Vlahou A. Secretome proteomics for discovery of cancer biomarkers. J Proteomics 2010; 73:2291-305. [PMID: 20637910 DOI: 10.1016/j.jprot.2010.07.001] [Citation(s) in RCA: 203] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Revised: 06/14/2010] [Accepted: 07/05/2010] [Indexed: 12/11/2022]
|
35
|
Interstitial fluid: the overlooked component of the tumor microenvironment? FIBROGENESIS & TISSUE REPAIR 2010; 3:12. [PMID: 20653943 PMCID: PMC2920231 DOI: 10.1186/1755-1536-3-12] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Accepted: 07/23/2010] [Indexed: 01/01/2023]
Abstract
Background The interstitium, situated between the blood and lymph vessels and the cells, consists of a solid or matrix phase and a fluid phase, together constituting the tissue microenvironment. Here we focus on the interstitial fluid phase of tumors, i.e., the fluid bathing the tumor and stromal cells. Novel knowledge on this compartment may provide important insight into how tumors develop and how they respond to therapy. Results We discuss available techniques for interstitial fluid isolation and implications of recent findings with respect to transcapillary fluid balance and uptake of macromolecular therapeutic agents. By the development of new methods it is emerging that local gradients exist in signaling substances from neoplastic tissue to plasma. Such gradients may provide new insight into the biology of tumors and mechanistic aspects linked to therapy. The emergence of sensitive proteomic technologies has made the interstitial fluid compartment in general and that of tumors in particular a highly valuable source for tissue-specific proteins that may serve as biomarker candidates. Potential biomarkers will appear locally at high concentrations in the tissue of interest and will eventually appear in the plasma, where they are diluted. Conclusions Access to fluid that reliably reflects the local microenvironment enables us to identify substances that can be used in early detection and monitoring of disease.
Collapse
|
36
|
Liu JM, Garcia-Alvarez MC, Bignon J, Kusinski M, Kuzdak K, Riches A, Wdzieczak-Bakala J. Overexpression of the natural tetrapeptide acetyl-N-ser-asp-lys-pro derived from thymosin beta4 in neoplastic diseases. Ann N Y Acad Sci 2010; 1194:53-9. [PMID: 20536450 DOI: 10.1111/j.1749-6632.2010.05488.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The natural tetrapeptide acetyl-ser-asp-lys-pro (AcSDKP) is formed in vivo by enzymatic cleavage of the N terminus of thymosin beta4 by prolyl oligopeptidase (POP). Recently, AcSDKP was shown to promote angiogenesis. Because of the critical role of neovascularization in cancer development, the levels of AcSDKP and POP activity in a number of different malignant tissues were investigated. Our studies revealed that AcSDKP levels were markedly elevated in neoplastic diseases including hematologic malignancies and solid neoplasms. Consistent with this finding, the enhanced activity of POP was also detected in all analyzed specimens of cancer tissues. Both these novel findings are in concert with the previously reported overexpression of thymosin beta4 in a large variety of malignant tumors and with its potential role in cancerogenesis. The physiological relevance of these findings awaits further studies; however, our first results strongly suggest a key role for AcSDKP in the pathogenesis of cancer.
Collapse
Affiliation(s)
- Jian-Miao Liu
- Centre de Recherche de Gif, Institut de Chimie des Substances Naturelles, CNRS, Gif-sur-Yvette, France
| | | | | | | | | | | | | |
Collapse
|
37
|
Okada S, List EO, Sankaran S, Kopchick JJ. Plasma Protein Biomarkers Correlated with the Development of Diet-Induced Type 2 Diabetes in Mice. Clin Proteomics 2010; 6:6-17. [PMID: 20625478 PMCID: PMC2899708 DOI: 10.1007/s12014-009-9040-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
INTRODUCTION: Early detection, assessment of disease progression, and application of an appropriate therapeutic intervention are all important for the care of patients with type 2 diabetes. Currently, however, there is no simple test for early detection of type 2 diabetes. Established diagnostic tests for the disease including oral glucose tolerance, fasting blood glucose, and hemoglobin A1c are relatively late markers where the disease has already progressed. Since blood is in direct contact with many tissues, we hypothesized that pathological tissue changes are likely to be reflected in proteomic profiles of plasma. METHODS: Mice were reared either on regular chow or a high-fat diet at weaning and several physiological responses (i.e., weight, fasting plasma glucose and insulin, and glucose tolerance) were monitored at regular time intervals. Plasma was collected at regular intervals for proteomic analysis by two-dimensional gel electrophoresis and subsequent mass spectrometry. RESULTS: Onset of hyperinsulinemia with corresponding glucose intolerance was observed in 2 weeks and fasting blood glucose levels rose significantly after 4 weeks on the high-fat diet. Many proteins were found to exist in multiple forms (isoforms). Levels of some isoforms including plasma retinol binding protein, transthyretin, Apolipoprotein A1, and kininogen showed significant changes as early as 4 weeks which coincided with the very early development of glucose intolerance. CONCLUSIONS: These results show that a proteomic approach to study the development of type 2 diabetes may uncover unknown early post-translationally modified diagnostic and/or therapeutic protein targets.
Collapse
Affiliation(s)
- Shigeru Okada
- Edison Biotechnology Institute, Konneker Research Laboratories, Ohio University, The Ridges, Bldg. 25, Athens, OH 45701-2979, USA, Department of Pediatrics, College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Edward O. List
- Edison Biotechnology Institute, Konneker Research Laboratories, Ohio University, The Ridges, Bldg. 25, Athens, OH 45701-2979, USA
| | - Sudha Sankaran
- Edison Biotechnology Institute, Konneker Research Laboratories, Ohio University, The Ridges, Bldg. 25, Athens, OH 45701-2979, USA
| | - John J. Kopchick
- Edison Biotechnology Institute, Konneker Research Laboratories, Ohio University, The Ridges, Bldg. 25, Athens, OH 45701-2979, USA, Department of Biomedical Science, College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| |
Collapse
|
38
|
Cierniewski CS, Papiewska-Pajak I, Malinowski M, Sacewicz-Hofman I, Wiktorska M, Kryczka J, Wysocki T, Niewiarowska J, Bednarek R. Thymosin β4 regulates migration of colon cancer cells by a pathway involving interaction with Ku80. Ann N Y Acad Sci 2010; 1194:60-71. [PMID: 20536451 DOI: 10.1111/j.1749-6632.2010.05480.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
39
|
Huinink KD, Lambooij B, Jansen-van Zelm K, Cremers TIFH, van Oeveren W, Bakker PL, Venema K, Westerink BHC, Korf J. Microfiltration sampling in rats and in cows: toward a portable device for continuous glucocorticoidhormone sampling. Analyst 2010; 135:390-6. [DOI: 10.1039/b921629d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
40
|
Williams D, Ackloo S, Zhu P, Bowden P, Evans KR, Addison CL, Lock C, Marshall JG. Precipitation and selective extraction of human serum endogenous peptides with analysis by quadrupole time-of-flight mass spectrometry reveals posttranslational modifications and low-abundance peptides. Anal Bioanal Chem 2009; 396:1223-47. [PMID: 20033139 DOI: 10.1007/s00216-009-3345-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2009] [Revised: 11/23/2009] [Accepted: 11/24/2009] [Indexed: 11/30/2022]
Abstract
The endogenous peptides of human serum may have regulatory functions, have been associated with physiological states, and their modifications may reveal some mechanisms of disease. In order to correlate levels of specific peptides with disease alongside internal standards, the polypeptides must first be reliably extracted and identified. Endogenous blood peptides can be effectively enriched by precipitation of the serum with organic solvents followed by selective extraction of peptides using aqueous solutions modified with organic solvents. Polypeptides on filter paper were assayed with Coomasie brilliant blue binding. The polypeptides were resolved by detergent tricine polyacrylamide electrophoresis and visualized by diamine silver staining. Peptides in the extracts were collected by C18 and analyzed by matrix-assisted laser desorption/ionization and liquid chromatography-electrospray ionization-tandem mass spectrometry (MS/MS) quadrupole time-of-flight MS/MS. Peptides were resolved as multiple isotopic peaks in MS mode with mass deviation of 0.1 Da or less and similar accuracy for fragments. The sensitivity of MS and MS/MS analysis was estimated to be in the picomolar range or less. The peptide composition of the extracts was dependent on solvent formulation. Multiple peptides from apolipoproteins, complement proteins, coagulation factors, and many others were identified by X!Tandem with high mass accuracy of peptide ions and fragments from collision-induced dissociation. Many previously unreported posttranslational modifications of peptides including phosphorylations, oxidations, glycosylations, and others were detected with high mass accuracy and may be of clinical importance. About 4,630 redundant peptides were identified with 99% confidence separately, and together some 1,251 distinct proteins were identified with 99% confidence or greater using the Paragon algorithm.
Collapse
Affiliation(s)
- Declan Williams
- Department of Chemistry and Biology, Faculty of Engineering and Applied Science, Ryerson University, 350 Victoria Street, Toronto, ON, M5B 2K3, Canada
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Ding J, List EO, Okada S, Kopchick JJ. Perspective: proteomic approach to detect biomarkers of human growth hormone. Growth Horm IGF Res 2009; 19:399-407. [PMID: 19501004 PMCID: PMC2760539 DOI: 10.1016/j.ghir.2009.04.018] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/03/2009] [Indexed: 10/20/2022]
Abstract
Several serum biomarkers for recombinant human growth hormone (rhGH) have been established, however, none alone or in combination have generate a specific, sensitive, and reproducible 'kit' for the detection of rhGH abuse. Thus, the search for additional GH specific biomarkers continues. In this review, we focus on the use of proteomics in general and two-dimensional electrophoresis (2-DE) in particular for the discovery of new GH induced serum biomarkers. Also, we review some of the protocols involved in 2-DE. Finally, the possibility of tissues other than blood for biomarker discovery is discussed.
Collapse
Affiliation(s)
- Juan Ding
- Edison Biotechnology Institute, College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, USA
- Molecular and Cellular Biology Program, College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, USA
- Department of Biological Sciences, College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, USA
| | - Edward O. List
- Edison Biotechnology Institute, College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, USA
| | - Shigeru Okada
- Edison Biotechnology Institute, College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, USA
- Department of Biomedical Sciences, College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, USA
| | - John J. Kopchick
- Edison Biotechnology Institute, College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, USA
- Molecular and Cellular Biology Program, College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, USA
- Department of Biomedical Sciences, College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, USA
| |
Collapse
|
42
|
Hood BL, Stewart NA, Conrads TP. Development of High-Throughput Mass Spectrometry–Based Approaches for Cancer Biomarker Discovery and Implementation. Clin Lab Med 2009; 29:115-38. [DOI: 10.1016/j.cll.2009.01.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
43
|
Xiao Z, Blonder J, Zhou M, Veenstra TD. Proteomic analysis of extracellular matrix and vesicles. J Proteomics 2009; 72:34-45. [DOI: 10.1016/j.jprot.2008.11.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2008] [Revised: 11/12/2008] [Accepted: 11/13/2008] [Indexed: 12/21/2022]
|
44
|
Palmblad M, Tiss A, Cramer R. Mass spectrometry in clinical proteomics - from the present to the future. Proteomics Clin Appl 2009; 3:6-17. [PMID: 21136932 DOI: 10.1002/prca.200800090] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Indexed: 05/31/2025]
Abstract
MS is an important analytical tool in clinical proteomics, primarily in the disease-specific discovery, identification and characterisation of proteomic biomarkers and patterns. MS-based proteomics is increasingly used in clinical validation and diagnostic method development. The latter departs from the typical application of MS-based proteomics by exchanging some of the high performance of analysis for the throughput, robustness and simplicity required for clinical diagnostics. Although conventional MS-based proteomics has become an important field in clinical applications, some of the most recent MS technologies have not yet been extensively applied in clinical proteomics. In this review, we will describe the current state of MS in clinical proteomics and look to the future of this field.
Collapse
Affiliation(s)
- Magnus Palmblad
- Biomolecular Mass Sepctrometry Unit, Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | |
Collapse
|
45
|
Warder SE, Tucker LA, McLoughlin SM, Strelitzer TJ, Meuth JL, Zhang Q, Sheppard GS, Richardson PL, Lesniewski R, Davidsen SK, Bell RL, Rogers JC, Wang J. Discovery, identification, and characterization of candidate pharmacodynamic markers of methionine aminopeptidase-2 inhibition. J Proteome Res 2008; 7:4807-20. [PMID: 18828628 DOI: 10.1021/pr800388p] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The catalytic activity of methionine aminopeptidase-2 (MetAP2) has been pharmacologically linked to cell growth, angiogenesis, and tumor progression, making this an attractive target for cancer therapy. An assay for monitoring specific protein changes in response to MetAP2 inhibition, allowing pharmacokinetic (PK)/pharmacodynamic (PD) models to be established, could dramatically improve clinical decision-making. Candidate MetAP2-specific protein substrates were discovered from undigested cell culture-derived proteomes by MALDI-/SELDI-MS profiling and a biochemical method using (35)S-Met labeled protein lysates. Substrates were identified either as intact proteins by FT-ICR-MS or applying in-gel protease digestions followed by LC-MS/MS. The combination of these approaches led to the discovery of novel MetAP2-specific substrates including thioredoxin-1 (Trx-1), SH3 binding glutamic acid rich-like protein (SH3BGRL), and eukaryotic elongation factor-2 (eEF2). These studies also confirmed glyceraldehye 3-phosphate dehydrogenase (GAPDH) and cyclophillin A (CypA) as MetAP2 substrates. Additional data in support of these proteins as MetAP2-specific substrates were provided by in vitro MetAP1/MetAP2 enzyme assays with the corresponding N-terminal derived peptides and 1D/2D Western analyses of cellular and tissue lysates. FT-ICR-MS characterization of all intact species of the 18 kDa substrate, CypA, enabled a SELDI-MS cell-based assay to be developed for correlating N-terminal processing and inhibition of proliferation. The MetAP2-specific protein substrates discovered in this study have diverse properties that should facilitate the development of reagents for testing in preclinical and clinical environments.
Collapse
Affiliation(s)
- Scott E Warder
- Advanced Technology and Cancer Research, Global Pharmaceutical Research and Development, Abbott Laboratories, 100 Abbott Park Road, Abbott Park, Illinois 60064-6202, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Hu S, Arellano M, Boontheung P, Wang J, Zhou H, Jiang J, Elashoff D, Wei R, Loo JA, Wong DT. Salivary proteomics for oral cancer biomarker discovery. Clin Cancer Res 2008; 14:6246-52. [PMID: 18829504 PMCID: PMC2877125 DOI: 10.1158/1078-0432.ccr-07-5037] [Citation(s) in RCA: 381] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE This study aims to explore the presence of informative protein biomarkers in the human saliva proteome and to evaluate their potential for detection of oral squamous cell carcinoma (OSCC). EXPERIMENTAL DESIGN Whole saliva samples were collected from patients (n = 64) with OSCC and matched healthy subjects (n = 64). The proteins in pooled whole saliva samples of patients with OSCC (n = 16) and matched healthy subjects (n = 16) were profiled using shotgun proteomics based on C4 reversed-phase liquid chromatography for prefractionation, capillary reversed-phase liquid chromatography with quadruple time-of-flight mass spectrometry, and Mascot sequence database searching. Immunoassays were used for validation of the candidate biomarkers on a new group of OSCC (n = 48) and matched healthy subjects (n = 48). Receiver operating characteristic analysis was exploited to evaluate the diagnostic value of discovered candidate biomarkers for OSCC. RESULTS Subtractive proteomics revealed several salivary proteins at differential levels between the OSCC patients and matched control subjects. Five candidate biomarkers were successfully validated using immunoassays on an independent set of OSCC patients and matched healthy subjects. The combination of these candidate biomarkers yielded a receiver operating characteristic value of 93%, sensitivity of 90%, and specificity of 83% in detecting OSCC. CONCLUSION Patient-based saliva proteomics is a promising approach to searching for OSCC biomarkers. The discovery of these new targets may lead to a simple clinical tool for the noninvasive diagnosis of oral cancer. Long-term longitudinal studies with large populations of individuals with oral cancer and those who are at high risk of developing oral cancer are needed to validate these potential biomarkers.
Collapse
Affiliation(s)
- Shen Hu
- Oral Biology and Medicine Division and Dental Research Institute, School of Dentistry, University of California at Los Angeles, Los Angeles, California
| | - Martha Arellano
- Oral Biology and Medicine Division and Dental Research Institute, School of Dentistry, University of California at Los Angeles, Los Angeles, California
| | - Pinmanee Boontheung
- Department of Chemistry and Biochemistry, University of California at Los Angeles, Los Angeles, California
| | - Jianghua Wang
- Oral Biology and Medicine Division and Dental Research Institute, School of Dentistry, University of California at Los Angeles, Los Angeles, California
| | - Hui Zhou
- Oral Biology and Medicine Division and Dental Research Institute, School of Dentistry, University of California at Los Angeles, Los Angeles, California
| | - Jiang Jiang
- Oral Biology and Medicine Division and Dental Research Institute, School of Dentistry, University of California at Los Angeles, Los Angeles, California
| | - David Elashoff
- Department of Biostatistics, School of Public Health, University of California at Los Angeles, Los Angeles, California
| | - Roger Wei
- Oral Biology and Medicine Division and Dental Research Institute, School of Dentistry, University of California at Los Angeles, Los Angeles, California
| | - Joseph A. Loo
- Department of Chemistry and Biochemistry, University of California at Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, California
- Department of Biological Chemistry, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California
| | - David T. Wong
- Oral Biology and Medicine Division and Dental Research Institute, School of Dentistry, University of California at Los Angeles, Los Angeles, California
- Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, California
- Division of Head and Neck Surgery/Otolaryngology, School of Medicine, University of California at Los Angeles, Los Angeles, California
- Henry Samueli School of Engineering and Applied Science, University of California at Los Angeles, Los Angeles, California
| |
Collapse
|
47
|
Xue H, Lu B, Lai M. The cancer secretome: a reservoir of biomarkers. J Transl Med 2008; 6:52. [PMID: 18796163 PMCID: PMC2562990 DOI: 10.1186/1479-5876-6-52] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2008] [Accepted: 09/17/2008] [Indexed: 12/17/2022] Open
Abstract
Biomarkers are pivotal for cancer detection, diagnosis, prognosis and therapeutic monitoring. However, currently available cancer biomarkers have the disadvantage of lacking specificity and/or sensitivity. Developing effective cancer biomarkers becomes a pressing and permanent need. The cancer secretome, the totality of proteins released by cancer cells or tissues, provides useful tools for the discovery of novel biomarkers. The focus of this article is to review the recent advances in cancer secretome analysis. We aim to elaborate the approaches currently employed for cancer secretome studies, as well as its applications in the identification of biomarkers and the clarification of carcinogenesis mechanisms. Challenges encountered in this newly emerging field, including sample preparation, in vivo secretome analysis and biomarker validation, are also discussed. Further improvements on strategies and technologies will continue to drive forward cancer secretome research and enable development of a wealth of clinically valuable cancer biomarkers.
Collapse
Affiliation(s)
- Hua Xue
- Department of Pathology, School of Medicine, Zhejiang University, PR China.
| | | | | |
Collapse
|
48
|
Huang CP, Liu YT, Nakatsuji T, Shi Y, Gallo RR, Lin SB, Huang CM. Proteomics integrated with Escherichia coli vector-based vaccines and antigen microarrays reveals the immunogenicity of a surface sialidase-like protein of Propionibacterium acnes. Proteomics Clin Appl 2008; 2:1234-45. [PMID: 21136919 PMCID: PMC3070417 DOI: 10.1002/prca.200780103] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2007] [Indexed: 11/06/2022]
Abstract
Proteomics is a powerful tool for the identification of proteins, which provides a basis for rational vaccine design. However, it is still a highly technical and time-consuming task to examine a protein's immunogenicity utilizing traditional approaches. Here, we present a platform for effectively evaluating protein immunogenicity and antibody detection. A tetanus toxin C fragment (Tet-c) was used as a representative antigen to establish this platform. A cell wall-anchoring sialidase-like protein (SLP) of Propionibacterium acnes was utilized to assess the efficacy of this platform. We constructed an Escherichia coli vector-based vaccine by overexpressing Tet-c or SLP in E. coli and utilized an intact particle of E. coli itself as a vaccine (E. coli Tet-c or SLP vector). After ultraviolet (UV) irradiation, the E. coli vector-based vaccines were administered intranasally into imprinting control region mice without adding exogenous adjuvants. For antibody detection, we fabricated antigen microarrays by printing with purified recombinant proteins including Tet-c and SLP. Our results demonstrated that detectable antibodies were elicited in mice 6 weeks after intranasal administration of UV-irradiated E. coli vector-based vaccines. The antibody production of Tet-c and SLP was significantly elevated after boosting. Notably, the platform with main benefits of using E. coli itself as a vaccine carrier provides a critical template for applied proteomics aimed at screening novel vaccine targets. In addition, the novel immunogenic SLP potentially serves as an antigen candidate for the development of vaccines targeting P. acnes-associated diseases.
Collapse
Affiliation(s)
- Cheng-Po Huang
- Moores Cancer Center, University of California, San Diego, CA, USA
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taiwan
| | - Yu-Tsueng Liu
- Moores Cancer Center, University of California, San Diego, CA, USA
| | - Teruaki Nakatsuji
- VA San Diego Healthcare Center, University of California, San Diego, CA, USA
- Department of Medicine, Division of Dermatology, University of California, San Diego, CA, USA
| | - Yang Shi
- The Burnham Institute for Medical Research, La Jolla, CA, USA
| | - Richard R. Gallo
- VA San Diego Healthcare Center, University of California, San Diego, CA, USA
- Department of Medicine, Division of Dermatology, University of California, San Diego, CA, USA
| | - Shwu-Bin Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taiwan
| | - Chun-Ming Huang
- Moores Cancer Center, University of California, San Diego, CA, USA
- VA San Diego Healthcare Center, University of California, San Diego, CA, USA
- Department of Medicine, Division of Dermatology, University of California, San Diego, CA, USA
- La Jolla Institute for Molecular Medicine, San Diego, CA, USA
| |
Collapse
|
49
|
Chenau J, Michelland S, Seve M. Le sécrétome : définitions et intérêt biomédical. Rev Med Interne 2008; 29:606-8. [DOI: 10.1016/j.revmed.2007.12.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2007] [Accepted: 12/09/2007] [Indexed: 01/15/2023]
|
50
|
Lanoix J, Paramithiotis E. Secretory vesicle analysis for discovery of low abundance plasma biomarkers. ACTA ACUST UNITED AC 2008; 2:475-85. [DOI: 10.1517/17530059.2.5.475] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|