1
|
Kasprzak A, Geltz A. The State-of-the-Art Mechanisms and Antitumor Effects of Somatostatin in Colorectal Cancer: A Review. Biomedicines 2024; 12:578. [PMID: 38540191 PMCID: PMC10968376 DOI: 10.3390/biomedicines12030578] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 03/02/2024] [Accepted: 03/03/2024] [Indexed: 01/03/2025] Open
Abstract
Somatostatin, a somatotropin release inhibiting factor (SST, SRIF), is a widely distributed multifunctional cyclic peptide and acts through a transmembrane G protein-coupled receptor (SST1-SST5). Over the past decades, research has begun to reveal the molecular mechanisms underlying the anticancer activity of this hormonal peptide. Among gastrointestinal tract (GIT) tumors, direct and indirect antitumor effects of SST have been documented best in gastroenteropancreatic neuroendocrine tumors (GEP-NETs) and less well in non-endocrine cancers, including sporadic colorectal cancer (CRC). In the latter, the signaling pathways involved in the antitumor function of SST are primarily MAPK/ERK/AKT and Wnt/β-catenin. Direct (involving the MAPK pathway) and indirect (VEGF production) antiangiogenic effects of SST in CRC have also been described. The anti-inflammatory role of SST in CRC is emphasized, but detailed molecular mechanisms are still being explored. The role of SST in tumor genome/tumor microenvironment (TME)/host's gut microbiome interactions is only partially known. The results of SST analogues (SSAs)' treatment of sporadic CRC in monotherapy in vivo are not spectacular. The current review aims to present the state-of-the-art mechanisms and antitumor activity of endogenous SST and its synthetic analogues in CRC, with particular emphasis on sporadic CRC.
Collapse
Affiliation(s)
- Aldona Kasprzak
- Department of Histology and Embryology, University of Medical Sciences, Swiecicki Street 6, 60-781 Poznań, Poland;
| | | |
Collapse
|
2
|
Sheng W, Zhang C, Mohiuddin TM, Al-Rawe M, Zeppernick F, Falcone FH, Meinhold-Heerlein I, Hussain AF. Multiplex Immunofluorescence: A Powerful Tool in Cancer Immunotherapy. Int J Mol Sci 2023; 24:ijms24043086. [PMID: 36834500 PMCID: PMC9959383 DOI: 10.3390/ijms24043086] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/08/2023] Open
Abstract
Traditional immunohistochemistry (IHC) has already become an essential method of diagnosis and therapy in cancer management. However, this antibody-based technique is limited to detecting a single marker per tissue section. Since immunotherapy has revolutionized the antineoplastic therapy, developing new immunohistochemistry strategies to detect multiple markers simultaneously to better understand tumor environment and predict or assess response to immunotherapy is necessary and urgent. Multiplex immunohistochemistry (mIHC)/multiplex immunofluorescence (mIF), such as multiplex chromogenic IHC and multiplex fluorescent immunohistochemistry (mfIHC), is a new and emerging technology to label multiple biomarkers in a single pathological section. The mfIHC shows a higher performance in cancer immunotherapy. This review summarizes the technologies, which are applied for mfIHC, and discusses how they are employed for immunotherapy research.
Collapse
Affiliation(s)
- Wenjie Sheng
- Department of Gynecology and Obstetrics, Medical Faculty, Justus-Liebig-University Giessen, Klinikstr. 33, 35392 Giessen, Germany
| | - Chaoyu Zhang
- Department of Gynecology and Obstetrics, Medical Faculty, Justus-Liebig-University Giessen, Klinikstr. 33, 35392 Giessen, Germany
| | - T. M. Mohiuddin
- Department of Gynecology and Obstetrics, Medical Faculty, Justus-Liebig-University Giessen, Klinikstr. 33, 35392 Giessen, Germany
| | - Marwah Al-Rawe
- Department of Gynecology and Obstetrics, Medical Faculty, Justus-Liebig-University Giessen, Klinikstr. 33, 35392 Giessen, Germany
| | - Felix Zeppernick
- Department of Gynecology and Obstetrics, Medical Faculty, Justus-Liebig-University Giessen, Klinikstr. 33, 35392 Giessen, Germany
| | - Franco H. Falcone
- Institute for Parasitology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - Ivo Meinhold-Heerlein
- Department of Gynecology and Obstetrics, Medical Faculty, Justus-Liebig-University Giessen, Klinikstr. 33, 35392 Giessen, Germany
| | - Ahmad Fawzi Hussain
- Department of Gynecology and Obstetrics, Medical Faculty, Justus-Liebig-University Giessen, Klinikstr. 33, 35392 Giessen, Germany
- Correspondence:
| |
Collapse
|
3
|
Urbiola-Salvador V, Miroszewska D, Jabłońska A, Qureshi T, Chen Z. Proteomics approaches to characterize the immune responses in cancer. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119266. [PMID: 35390423 DOI: 10.1016/j.bbamcr.2022.119266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 03/01/2022] [Accepted: 03/28/2022] [Indexed: 06/14/2023]
Abstract
Despite the dynamic development of cancer research, annually millions of people die of cancer. The human immune system is the major 'guard' against tumor development. Unfortunately, cancer cells have the ability to evade the immune system and continue to grow. The proper understanding of the intricate immune response in tumorigenesis remains the holy grail of cancer immunology and designing effective immunotherapy. To decode the immune responses in cancer, in recent years, proteomics studies have received considerable attention. Proteomics studies focus on the detection and quantification of proteins, which are the effectors of biological functions, and as such, are proven to reflect the cell state more accurately, in comparison to genomic or transcriptomic studies. In this review, we discuss the proteomics studies applied to characterize the immune responses in cancer and tumor immune microenvironment heterogeneity. Further, we describe emerging single-cell proteomics approaches that have the potential to be applied in cancer immunity studies.
Collapse
Affiliation(s)
- Víctor Urbiola-Salvador
- Intercollegiate Faculty of Biotechnology of University of Gdańsk and Medical University of Gdańsk, University of Gdańsk, Poland.
| | - Dominika Miroszewska
- Intercollegiate Faculty of Biotechnology of University of Gdańsk and Medical University of Gdańsk, University of Gdańsk, Poland.
| | - Agnieszka Jabłońska
- Intercollegiate Faculty of Biotechnology of University of Gdańsk and Medical University of Gdańsk, University of Gdańsk, Poland.
| | - Talha Qureshi
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
| | - Zhi Chen
- Intercollegiate Faculty of Biotechnology of University of Gdańsk and Medical University of Gdańsk, University of Gdańsk, Poland; Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
| |
Collapse
|
4
|
Dalal N, Jalandra R, Sharma M, Prakash H, Makharia GK, Solanki PR, Singh R, Kumar A. Omics technologies for improved diagnosis and treatment of colorectal cancer: Technical advancement and major perspectives. Biomed Pharmacother 2020; 131:110648. [PMID: 33152902 DOI: 10.1016/j.biopha.2020.110648] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/09/2020] [Accepted: 08/16/2020] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer (CRC) ranks third among the most commonly occurring cancers worldwide, and it causes half a million deaths annually. Alongside mechanistic study for CRC detection and treatment by conventional techniques, new technologies have been developed to study CRC. These technologies include genomics, transcriptomics, proteomics, and metabolomics which elucidate DNA markers, RNA transcripts, protein and, metabolites produced inside the colon and rectum part of the gut. All these approaches form the omics arena, which presents a remarkable opportunity for the discovery of novel prognostic, diagnostic and therapeutic biomarkers and also delineate the underlying mechanism of CRC causation, which may further help in devising treatment strategies. This review also mentions the latest developments in metagenomics and culturomics as emerging evidence suggests that metagenomics of gut microbiota has profound implications in the causation, prognosis, and treatment of CRC. A majority of bacteria cannot be studied as they remain unculturable, so culturomics has also been strengthened to develop culture conditions suitable for the growth of unculturable bacteria and identify unknown bacteria. The overall purpose of this review is to succinctly evaluate the application of omics technologies in colorectal cancer research for improving the diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Nishu Dalal
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi 110067, India; Department of Environmental Science, Satyawati College, Delhi University, Delhi 110052, India
| | - Rekha Jalandra
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi 110067, India; Department of Zoology, Maharshi Dayanand University, Rohtak 124001, India
| | - Minakshi Sharma
- Department of Zoology, Maharshi Dayanand University, Rohtak 124001, India
| | - Hridayesh Prakash
- Amity Institute of Virology and Immunology, Amity University, Sector 125, Noida 201313, Uttar Pradesh, India
| | - Govind K Makharia
- Department of Gastroenterology and Human Nutrition, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Pratima R Solanki
- Special Centre for Nanoscience, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rajeev Singh
- Department of Environmental Science, Satyawati College, Delhi University, Delhi 110052, India.
| | - Anil Kumar
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi 110067, India.
| |
Collapse
|
5
|
Phelps DS, Chinchilli VM, Weisz J, Shearer D, Zhang X, Floros J. Using toponomics to characterize phenotypic diversity in alveolar macrophages from male mice treated with exogenous SP-A1. Biomark Res 2020; 8:5. [PMID: 32082572 PMCID: PMC7020580 DOI: 10.1186/s40364-019-0181-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 12/30/2019] [Indexed: 01/12/2023] Open
Abstract
Background We used the Toponome Imaging System (TIS) to identify “patterns of marker expression”, referred to here as combinatorial molecular phenotypes (CMPs) in alveolar macrophages (AM) in response to the innate immune molecule, SP-A1. Methods We compared 114 AM from male SP-A deficient mice. One group (n = 3) was treated with exogenous human surfactant protein A1 (hSP-A1) and the other with vehicle (n = 3). AM obtained by bronchoalveolar lavage were plated onto slides and analyzed using TIS to study the AM toponome, the spatial network of proteins within intact cells. With TIS, each slide is sequentially immunostained with multiple FITC-conjugated antibodies. Images are analyzed pixel-by-pixel identifying all of the proteins within each pixel, which are then designated as CMPs. CMPs represent organized protein clusters postulated to contribute to specific functions. Results 1) We compared identical CMPs in KO and SP-A1 cells and found them to differ significantly (p = 0.0007). Similarities between pairs of markers in the two populations also differed significantly (p < 0.0001). 2) Focusing on the 20 most abundant CMPs for each cell, we developed a method to generate CMP “signatures” that characterized various groups of cells. Phenotypes were defined as cells exhibiting similar signatures of CMPs. i) AM were extremely diverse and each group contained cells with multiple phenotypes. ii) Among the 114 AM analyzed, no two cells were identical. iii) However, CMP signatures could distinguish among cell subpopulations within and between groups. iv) Some cell populations were enriched with SP-A1 treatment, some were more common without SP-A1, and some seemed not to be influenced by the presence of SP-A1. v) We also found that AM were more diverse in mice treated with SP-A1 compared to those treated with vehicle. Conclusions AM diversity is far more extensive than originally thought. The increased diversity of SP-A1-treated mice points to the possibility that SP-A1 enhances or activates several pathways in the AM to better prepare it for its innate immune functions and other functions shown previously to be affected by SP-A treatment. Future studies may identify key protein(s) responsible for CMP integrity and consequently for a given function, and target it for therapeutic purposes.
Collapse
Affiliation(s)
- David S Phelps
- 1Penn State Center for Host defense, Inflammation, and Lung Disease (CHILD) Research and Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA
| | - Vernon M Chinchilli
- 2Public Health Sciences; and Obstetrics and Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA
| | - Judith Weisz
- 3Obstetrics and Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA
| | - Debra Shearer
- 3Obstetrics and Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA
| | - Xuesheng Zhang
- 1Penn State Center for Host defense, Inflammation, and Lung Disease (CHILD) Research and Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA
| | - Joanna Floros
- 1Penn State Center for Host defense, Inflammation, and Lung Disease (CHILD) Research and Departments of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA.,3Obstetrics and Gynecology, The Pennsylvania State University College of Medicine, Hershey, PA 17033 USA
| |
Collapse
|
6
|
Abstract
Multiplexed imaging platforms to simultaneously detect multiple epitopes in the same tissue section emerged in the last years as very powerful tools to study tumor immune contexture. These revolutionary technologies are providing a deep methodology for tumor evaluation in formalin-fixed and paraffin-embedded (FFPE) to improve the understanding of tumor microenvironment, new targets for treatment, prognostic and predictive biomarkers, and translational studies. Multiplexed imaging platforms allow for the identification of several antigens simultaneously from a single tissue section, core needle biopsies, and tissue microarrays. In recent years, multiplexed imaging has improved the abilities to characterize the different types of cell populations in malignant and non-malignant tissues, and their spatial distribution in relationship to clinical outcomes. Multiplexed technologies associated with digital image analysis software offer a high-quality throughput assay to study cancer specimens at multiple time points before, during and after treatment. The aim of this chapter is to provide a review of multiplexed imaging covering its fundamentals, advantages, disadvantages, and material and methods for staining applied to FFPE tumor tissues and focusing on the use of multiplex immunofluorescence with tyramine signal amplification staining for immune profiling and translational research.
Collapse
|
7
|
State-of-the-Art of Profiling Immune Contexture in the Era of Multiplexed Staining and Digital Analysis to Study Paraffin Tumor Tissues. Cancers (Basel) 2019; 11:cancers11020247. [PMID: 30791580 PMCID: PMC6406364 DOI: 10.3390/cancers11020247] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 02/12/2019] [Accepted: 02/14/2019] [Indexed: 02/07/2023] Open
Abstract
Multiplexed platforms for multiple epitope detection have emerged in the last years as very powerful tools to study tumor tissues. These revolutionary technologies provide important visual techniques for tumor examination in formalin-fixed paraffin-embedded specimens to improve the understanding of the tumor microenvironment, promote new treatment discoveries, aid in cancer prevention, as well as allowing translational studies to be carried out. The aim of this review is to highlight the more recent methodologies that use multiplexed staining to study simultaneous protein identification in formalin-fixed paraffin-embedded tumor tissues for immune profiling, clinical research, and potential translational analysis. New multiplexed methodologies, which permit the identification of several proteins at the same time in one single tissue section, have been developed in recent years with the ability to study different cell populations, cells by cells, and their spatial distribution in different tumor specimens including whole sections, core needle biopsies, and tissue microarrays. Multiplexed technologies associated with image analysis software can be performed with a high-quality throughput assay to study cancer specimens and are important tools for new discoveries. The different multiplexed technologies described in this review have shown their utility in the study of cancer tissues and their advantages for translational research studies and application in cancer prevention and treatments.
Collapse
|
8
|
Knight JM, Kim E, Ivanov I, Davidson LA, Goldsby JS, Hullar MAJ, Randolph TW, Kaz AM, Levy L, Lampe JW, Chapkin RS. Comprehensive site-specific whole genome profiling of stromal and epithelial colonic gene signatures in human sigmoid colon and rectal tissue. Physiol Genomics 2016; 48:651-9. [PMID: 27401218 PMCID: PMC5111881 DOI: 10.1152/physiolgenomics.00023.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 07/04/2016] [Indexed: 01/28/2023] Open
Abstract
The strength of associations between various exposures (e.g., diet, tobacco, chemopreventive agents) and colorectal cancer risk may partially depend on the complex interaction between epithelium and stroma across anatomic subsites. Currently, baseline data describing genome-wide coding and long noncoding gene expression profiles in the healthy colon specific to tissue type and location are lacking. Therefore, colonic mucosal biopsies from 10 healthy participants who were enrolled in a clinical study to evaluate effects of lignan supplementation on gut resiliency were used to characterize the site-specific global gene expression signatures associated with stromal vs. epithelial cells in the sigmoid colon and rectum. Using RNA-seq, we demonstrate that tissue type and location patterns of gene expression and upstream regulatory pathways are distinct. For example, consistent with a key role of stroma in the crypt niche, mRNAs associated with immunoregulatory and inflammatory processes (i.e., CXCL14, ANTXR1), smooth muscle contraction (CALD1), proliferation and apoptosis (GLP2R, IGFBP3), and modulation of extracellular matrix (MMP2, COL3A1, MFAP4) were all highly expressed in the stroma. In comparison, HOX genes (HOXA3, HOXD9, HOXD10, HOXD11, and HOXD-AS2, a HOXD cluster antisense RNA 2), and WNT5B expression were also significantly higher in sigmoid colon compared with the rectum. These findings provide strong impetus for considering colorectal tissue subtypes and location in future observational studies and clinical trials designed to evaluate the effects of exposures on colonic health.
Collapse
Affiliation(s)
- Jason M Knight
- Department of Electrical Engineering, Texas A&M University, College Station, Texas; Center for Translational Environmental Health Research, Texas A&M University, College Station, Texas
| | - Eunji Kim
- Department of Electrical Engineering, Texas A&M University, College Station, Texas; Center for Translational Environmental Health Research, Texas A&M University, College Station, Texas
| | - Ivan Ivanov
- Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, Texas; Center for Translational Environmental Health Research, Texas A&M University, College Station, Texas
| | - Laurie A Davidson
- Department of Nutrition & Food Science, Texas A&M University, College Station, Texas; Center for Translational Environmental Health Research, Texas A&M University, College Station, Texas
| | - Jennifer S Goldsby
- Department of Nutrition & Food Science, Texas A&M University, College Station, Texas; Center for Translational Environmental Health Research, Texas A&M University, College Station, Texas
| | - Meredith A J Hullar
- Fred Hutchinson Cancer Research Center, Texas A&M University, College Station, Texas; and
| | - Timothy W Randolph
- Fred Hutchinson Cancer Research Center, Texas A&M University, College Station, Texas; and
| | - Andrew M Kaz
- Fred Hutchinson Cancer Research Center, Texas A&M University, College Station, Texas; and Gastroenterology Section, VA Puget Sound Medical Center, Seattle, Washington
| | - Lisa Levy
- Fred Hutchinson Cancer Research Center, Texas A&M University, College Station, Texas; and
| | - Johanna W Lampe
- Fred Hutchinson Cancer Research Center, Texas A&M University, College Station, Texas; and
| | - Robert S Chapkin
- Department of Nutrition & Food Science, Texas A&M University, College Station, Texas; Center for Translational Environmental Health Research, Texas A&M University, College Station, Texas;
| |
Collapse
|
9
|
Abstract
INTRODUCTION LFA-1 is an adhesion molecule which belongs to the β2-integrin family. Overexpression of LFA-1 in hepatic natural killer cells has been associated with increased apoptosis of neoplastic cells in colorectal cancer (CRC); moreover, studies in CRC have linked LFA-1 overexpression in neoplastic cells with vascular intrusion through adhesion to endothelial cells, thus implying a possible role in creation of metastases. AIMS AND METHODS We studied the expression of LFA-1 in a series of 82 patients with CRC. A standard three-step immunohistochemical analysis was performed on formalin-fixed, paraffin-embedded tissue sections. An IgG2a anti-CD11a monoclonal antibody was used. Cases were characterized according to clinicopathological variables including sex, age, tumor localization, size, grade, Dukes stage, wall invasion, and presence of metastatic lymph nodes (mLNs) or distal metastases. RESULTS LFA-1 was expressed at the primary tumor site in 51 cases and 6/33 cases with metastatic lymphnodes. In Dukes D cases (n = 4), only one case was LFA-1(+). LFA-1 expression at the primary tumor site was associated with the absence of metastatic disease and with Dukes B stage. However, in those cases with LFA-1 expression in cancer cells in mLNs, this was associated with its expression at the primary tumor site. CONCLUSION The positive association of LFA-1 expression in mLNs when the primary tumor site is also LFA-1(+) could imply an adaptation advantage of this specific cellular clone to its micro-environment, predisposing it to creation of mLNs, pointing to a role for LFA-1 in creation of mLNs in CRC.
Collapse
|
10
|
Eckhardt J, Ostalecki C, Kuczera K, Schuler G, Pommer AJ, Lechmann M. Murine whole-organ immune cell populations revealed by multi-epitope-ligand cartography. J Histochem Cytochem 2012; 61:125-33. [PMID: 23160665 DOI: 10.1369/0022155412470140] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Multi-epitope-ligand cartography (MELC) is an innovative high-throughput fluorescence microscopy-based method. A tissue section is analyzed through a repeated cycling of (1) incubation with a fluorophore-labeled antibody, (2) fluorescence imaging, and (3) soft bleaching. This method allows staining of the same tissue section with up to 100 fluorescent markers and to analyze their toponomic expression using further image processing and pixel-precise overlay of the corresponding images. In this study, we adapted this method to identify a large panel of murine leukocyte subpopulations in a whole frozen section of a peripheral lymph node. Using the resulting antibody library, we examined non-inflamed versus inflamed tissues of brain and spinal cord in the experimental autoimmune encephalomyelitis (EAE) model. The presence and activity of specific leukocyte subpopulations (different T cell subpopulations, dendritic cells, macrophages, etc.) could be assessed and the cellular localizations and the corresponding activation status in situ were investigated. The results were then correlated with quantitative RT-PCR.
Collapse
|
11
|
Belov L, Zhou J, Christopherson RI. Cell surface markers in colorectal cancer prognosis. Int J Mol Sci 2010; 12:78-113. [PMID: 21339979 PMCID: PMC3039945 DOI: 10.3390/ijms12010078] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2010] [Revised: 12/16/2010] [Accepted: 12/20/2010] [Indexed: 12/14/2022] Open
Abstract
The classification of colorectal cancers (CRC) is currently based largely on histologically determined tumour characteristics, such as differentiation status and tumour stage, i.e., depth of tumour invasion, involvement of regional lymph nodes and the occurrence of metastatic spread to other organs. These are the conventional prognostic factors for patient survival and often determine the requirement for adjuvant therapy after surgical resection of the primary tumour. However, patients with the same CRC stage can have very different disease-related outcomes. For some, surgical removal of early-stage tumours leads to full recovery, while for others, disease recurrence and metastasis may occur regardless of adjuvant therapy. It is therefore important to understand the molecular processes that lead to disease progression and metastasis and to find more reliable prognostic markers and novel targets for therapy. This review focuses on cell surface proteins that correlate with tumour progression, metastasis and patient outcome, and discusses some of the challenges in finding prognostic protein markers in CRC.
Collapse
Affiliation(s)
- Larissa Belov
- School of Molecular Bioscience, University of Sydney, Sydney, NSW 2006, Australia; E-Mails: (J.Z.); (R.I.C.)
| | | | | |
Collapse
|
12
|
Sung MK, Bae YJ. Linking obesity to colorectal cancer: application of nutrigenomics. Biotechnol J 2010; 5:930-41. [PMID: 20715079 DOI: 10.1002/biot.201000165] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diet is one of the most affective environmental factors in cancer development. Due to complicated nature of the diet, it has been very difficult to provide clear explanations for the role of dietary components in carcinogenesis. However, as high-throughput omics techniques became available, researchers are now able to analyze large sets of gene transcripts, proteins, and metabolites to identify molecules involved in disease development. Bioinformatics uses these data to perform network analyses and suggest possible interactions between metabolic processes and environmental factors. Obesity is known as one of the most closely related risk factors of colorectal cancer (CRC). Metabolic disturbances due to a positive energy balance may trigger and accelerate CRC development. In this review, we have summarized reports on genes, proteins and metabolites that are related to either obesity or CRC, and suggested candidate molecules linking obesity and CRC based on currently available literature. Possible application of bioinformatics for a large scale network analysis in studying cause-effect relationship between dietary components and CRC are suggested.
Collapse
Affiliation(s)
- Mi-Kyung Sung
- Department of Food and Nutrition, Sookmyung Women's University, Seoul, South Korea.
| | | |
Collapse
|
13
|
Berndt U, Philipsen L, Bartsch S, Hu Y, Röcken C, Bertram W, Hämmerle M, Rösch T, Sturm A. Comparative Multi-Epitope-Ligand-Cartography reveals essential immunological alterations in Barrett's metaplasia and esophageal adenocarcinoma. Mol Cancer 2010; 9:177. [PMID: 20604962 PMCID: PMC2909181 DOI: 10.1186/1476-4598-9-177] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2010] [Accepted: 07/06/2010] [Indexed: 12/21/2022] Open
Abstract
Background Barrett's esophagus (BE) is caused by gastroesophageal reflux with consecutive mucosal inflammation, predisposing patients to the development of esophageal adenocarcinoma (EAC). We investigated changes in T cell-related mucosal combinatorial molecular protein patterns in both diseases using the novel Multi-Epitope-Ligand-Cartography, a unique robotic whole-cell imaging technology that simultaneously visualizes dozens of proteins in structurally intact tissues and correlates cellular localization of proteins with function. Results Biopsies were taken during endoscopy from BE, EAC, and normal control tissue, and proteomic microscopy was performed on 32 different epitopes. When the significance level was set to p < 0.0005 and the search depth to five antibody combinations, controls and BE can be differentiated by 63, controls and EAC by 3222, and BE from EAC by 1521 distinct protein combinations. For example, the number of activated apoptotic naïve and memory T cells was significantly increased only in BE, whereas the number of activated apoptotic helper and regulatory T cells was significantly elevated in BE and EAC. In contrast, the number of activated apoptotic cytotoxic T cells was significantly elevated only in EAC. Confirming different pathways in BE and EAC, the number of T lymphocytes with p53 expression and downregulation of bcl2 expression (CD3+p53+Bcl2-NfkB-) was significantly increased in EAC compared to BE and controls. Interestingly, the number of precursor T cells (CD7+) was significantly elevated only in EAC. These cells lack Bax and caspase-8, suggesting impaired apoptosis in the early stages of T cell differentiation. Conclusion Proteomic analysis showed for the first time that proteins, which are critically involved in the mucosal immune system of the esophagus, are distinctly expressed in BE and EAC, whereas others are comparably altered in both diseases, suggesting that many pathogenic events might be shared by both diseases. Topological proteomic analysis, therefore, helps us to understand the different pathogenic events in the underlying disease pathways.
Collapse
Affiliation(s)
- Uta Berndt
- Department of Medicine, Division of Gastroenterology and Hepatology, Charité-Campus Virchow Clinic, Universitätsmedizin Berlin, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Pierre S, Scholich K. Toponomics: studying protein-protein interactions and protein networks in intact tissue. MOLECULAR BIOSYSTEMS 2010; 6:641-7. [PMID: 20237641 DOI: 10.1039/b910653g] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The function of a protein is determined on several levels including the genome, transcriptome, proteome, and the recently introduced toponome. The toponome describes the topology of all proteins, protein complexes and protein networks which constitute and influence the microenvironment of a given protein. It has long been known that cellular function or dysfunction of proteins strongly depends on their microenvironment and even small changes in protein arrangements can dramatically alter their activity/function. Thus, deciphering the topology of the multi-dimensional networks which control normal and disease-related pathways will give a better understanding of the mechanisms underlying disease development. While various powerful proteomic tools allow simultaneous quantification of proteins, only a limited number of techniques are available to visualize protein networks in intact cells and tissues. This review discusses a novel approach to map and decipher functional molecular networks of proteins in intact cells or tissues. Multi-epitope-ligand-cartography (MELC) is an imaging technology that identifies and quantifies protein networks at the subcellular level of morphologically-intact specimens. This immunohistochemistry-based method allows serial visualization and biomathematical analysis of up to 100 cellular components using fluorescence-labelled tags. The resulting toponome maps, simultaneously ranging from the subcellular to the supracellular scale, have the potential to provide the basis for a mathematical description of the dynamic topology of protein networks, and will complement current proteomic data to enhance the understanding of physiological and pathophysiological cell functions.
Collapse
Affiliation(s)
- Sandra Pierre
- Institut für Klinische Pharmakologie, Klinikum der Johann Wolfgang Goethe-Universität, Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | | |
Collapse
|
15
|
Decaestecker C, Lopez XM, D'Haene N, Roland I, Guendouz S, Duponchelle C, Berton A, Debeir O, Salmon I. Requirements for the valid quantification of immunostains on tissue microarray materials using image analysis. Proteomics 2009; 9:4478-94. [PMID: 19670370 DOI: 10.1002/pmic.200800936] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Antibody-based proteomics applied to tissue microarray (TMA) technology provides a very efficient means of visualizing and locating antigen expression in large collections of normal and pathological tissue samples. To characterize antigen expression on TMAs, the use of image analysis methods avoids the effects of human subjectivity evidenced in manual microscopical analysis. Thus, these methods have the potential to significantly enhance both precision and reproducibility. Although some commercial systems include tools for the quantitative evaluation of immunohistochemistry-stained images, there exists no clear agreement on best practices to allow for correct and reproducible quantification results. Our study focuses on practical aspects regarding (i) image acquisition (ii) segmentation of staining and counterstaining areas and (iii) extraction of quantitative features. We illustrate our findings using a commercial system to quantify different immunohistochemistry markers targeting proteins with different expression patterns (cytoplasmic, nuclear or membranous) in colon cancer or brain tumor TMAs. Our investigations led us to identify several steps that we consider essential for standardizing computer-assisted immunostaining quantification experiments. In addition, we propose a data normalization process based on reference materials to be able to compare measurements between studies involving different TMAs. In conclusion, we recommend certain critical prerequisites that commercial or in-house systems should satisfy in order to permit valid immunostaining quantification.
Collapse
Affiliation(s)
- Christine Decaestecker
- Laboratory of Image Synthesis and Analysis (LISA), Faculty of Applied Sciences, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Linke B, Pierre S, Coste O, Angioni C, Becker W, Maier TJ, Steinhilber D, Wittpoth C, Geisslinger G, Scholich K. Toponomics Analysis of Drug-Induced Changes in Arachidonic Acid-Dependent Signaling Pathways during Spinal Nociceptive Processing. J Proteome Res 2009; 8:4851-9. [DOI: 10.1021/pr900106v] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Bona Linke
- Pharmazentrum Frankfurt, ZAFES, Institute of Clinical Pharmacology, Klinikum der Goethe-Universität Frankfurt, Germany, Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Germany, and MelTec GmbH&Co KG, Magdeburg, Germany
| | - Sandra Pierre
- Pharmazentrum Frankfurt, ZAFES, Institute of Clinical Pharmacology, Klinikum der Goethe-Universität Frankfurt, Germany, Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Germany, and MelTec GmbH&Co KG, Magdeburg, Germany
| | - Ovidiu Coste
- Pharmazentrum Frankfurt, ZAFES, Institute of Clinical Pharmacology, Klinikum der Goethe-Universität Frankfurt, Germany, Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Germany, and MelTec GmbH&Co KG, Magdeburg, Germany
| | - Carlo Angioni
- Pharmazentrum Frankfurt, ZAFES, Institute of Clinical Pharmacology, Klinikum der Goethe-Universität Frankfurt, Germany, Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Germany, and MelTec GmbH&Co KG, Magdeburg, Germany
| | - Wiebke Becker
- Pharmazentrum Frankfurt, ZAFES, Institute of Clinical Pharmacology, Klinikum der Goethe-Universität Frankfurt, Germany, Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Germany, and MelTec GmbH&Co KG, Magdeburg, Germany
| | - Thorsten Jürgen Maier
- Pharmazentrum Frankfurt, ZAFES, Institute of Clinical Pharmacology, Klinikum der Goethe-Universität Frankfurt, Germany, Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Germany, and MelTec GmbH&Co KG, Magdeburg, Germany
| | - Dieter Steinhilber
- Pharmazentrum Frankfurt, ZAFES, Institute of Clinical Pharmacology, Klinikum der Goethe-Universität Frankfurt, Germany, Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Germany, and MelTec GmbH&Co KG, Magdeburg, Germany
| | - Claus Wittpoth
- Pharmazentrum Frankfurt, ZAFES, Institute of Clinical Pharmacology, Klinikum der Goethe-Universität Frankfurt, Germany, Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Germany, and MelTec GmbH&Co KG, Magdeburg, Germany
| | - Gerd Geisslinger
- Pharmazentrum Frankfurt, ZAFES, Institute of Clinical Pharmacology, Klinikum der Goethe-Universität Frankfurt, Germany, Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Germany, and MelTec GmbH&Co KG, Magdeburg, Germany
| | - Klaus Scholich
- Pharmazentrum Frankfurt, ZAFES, Institute of Clinical Pharmacology, Klinikum der Goethe-Universität Frankfurt, Germany, Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Germany, and MelTec GmbH&Co KG, Magdeburg, Germany
| |
Collapse
|
17
|
Pierre S, Maeurer C, Coste O, Becker W, Schmidtko A, Holland S, Wittpoth C, Geisslinger G, Scholich K. Toponomics analysis of functional interactions of the ubiquitin ligase PAM (Protein Associated with Myc) during spinal nociceptive processing. Mol Cell Proteomics 2008; 7:2475-85. [PMID: 18753128 DOI: 10.1074/mcp.m800201-mcp200] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Protein associated with Myc (PAM) is a giant E3 ubiquitin ligase of 510 kDa. Although the role of PAM during neuronal development is well established, very little is known about its function in the regulation of synaptic strength. Here we used multiepitope ligand cartography (MELC) to study protein network profiles associated with PAM during the modulation of synaptic strength. MELC is a novel imaging technology that utilizes biomathematical tools to describe protein networks after consecutive immunohistochemical visualization of up to 100 proteins on the same sample. As an in vivo model to modulate synaptic strength we used the formalin test, a common model for acute and inflammatory pain. MELC analysis was performed with 37 different antibodies or fluorescence tags on spinal cord slices and led to the identification of 1390 PAM-related motifs that distinguish untreated and formalin-treated spinal cords. The majority of these motifs related to ubiquitin-dependent processes and/or the actin cytoskeleton. We detected an intermittent colocalization of PAM and ubiquitin with TSC2, a known substrate of PAM, and the glutamate receptors mGluR5 and GLUR1. Importantly these complexes were detected exclusively in the presence of F-actin. A direct PAM/F-actin interaction was confirmed by colocalization and cosedimentation. The binding of PAM toward F-actin varied strongly between the PAM splice forms found in rat spinal cords. PAM did not ubiquitylate actin or alter actin polymerization and depolymerization. However, F-actin decreased the ubiquitin ligase activity of purified PAM. Because PAM activation is known to involve its translocation, the binding of PAM to F-actin may serve to control its subcellular localization as well as its activity. Taken together we show that defining protein network profiles by topological proteomics analysis is a useful tool to identify previously unknown protein/protein interactions that underlie synaptic processes.
Collapse
Affiliation(s)
- Sandra Pierre
- Pharmazentrum Frankfurt, ZAFES, Institute of Clinical Pharmacology, Klinikum der Goethe-Universität, 60590 Frankfurt, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Toponomics: visualizing cellular protein networks in health and disease A single picture is worth more than a thousand words! J Cutan Pathol 2008; 35:791-3. [DOI: 10.1111/j.1600-0560.2008.01106.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|