201
|
Wang C, Ma HX, Jin MS, Zou YB, Teng YL, Tian Z, Wang HY, Wang YP, Duan XM. Association of matrix metalloproteinase (MMP)-2 and -9 expression with extra-gastrointestinal stromal tumor metastasis. Asian Pac J Cancer Prev 2015; 15:4187-92. [PMID: 24935368 DOI: 10.7314/apjcp.2014.15.10.4187] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Matrix metalloproteinase (MMP)-2 and MMP-9 are important proteases involved in invasion and metastasis of various tumors. Extra-gastrointestinal stromal tumors (EGISTs) are rare neoplasms. This study was performed to assess MMP-2 and MMP-9 expression in EGIST tissue samples for association with clinicopathological data from the patients. Twenty-one surgical EGIST tissue specimens were collected for analysis of MMP-2 and MMP- 9 expression using immunohistochemistry. MMP-2 and MMP-9 proteins were expressed in all of the epithelial cell types of EGISTs, whereas they were only expressed in 75% of the spindle cell type, although there was no statistically significant difference (p>0.05). Expression of MMP-2 and MMP-9 proteins was associated with tumor size, mitotic rate, tumor necrosis, and distant metastasis (p<0.05). MMP-2 expression was linked with MMP-9 levels (p<0.05). However, there was no correlation between MMP-9 expression and age, sex, primary site, or cell morphology in any of these 21 EGIST patients (p>0.05). Moreover, expression of MMP-2 and MMP-9 proteins increased with the degree of EGIST risk. This study provided evidence of an association of MMP-2 and MMP-9 expression with advanced EGIST behavior.
Collapse
Affiliation(s)
- Chao Wang
- Pathological Diagnosis Centre, The First Hospital Affiliated to Bethune Medical College, Jilin University, Changchun, Jilin, China E-mail :
| | | | | | | | | | | | | | | | | |
Collapse
|
202
|
Wu H, Guan S, Sun M, Yu Z, Zhao L, He M, Zhao H, Yao W, Wang E, Jin F, Xiao Q, Wei M. Ano1/TMEM16A Overexpression Is Associated with Good Prognosis in PR-Positive or HER2-Negative Breast Cancer Patients following Tamoxifen Treatment. PLoS One 2015; 10:e0126128. [PMID: 25961581 PMCID: PMC4427473 DOI: 10.1371/journal.pone.0126128] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2014] [Accepted: 03/29/2015] [Indexed: 12/31/2022] Open
Abstract
The calcium-activated chloride channel Ano1 (TMEM16A) is overexpressed in many tumors. Although Ano1 overexpression is found in breast cancer due to 11q13 amplification, it remains unclear whether signaling pathways are involved in Ano1 overexpression during breast cancer tumorigenesis in vivo. Estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) have been known to contribute to breast cancer progression. It is unclear whether Ano1 is associated with clinical outcomes in breast cancer patients with different ER, PR and HER2 status. In the present study, we investigated the Ano1 expression in 431 patients with invasive ductal breast carcinoma and 46 patients with fibroadenoma, using immunohistochemistry, and analyzed the association between Ano1 expression and clinical characteristics and outcomes of breast cancer patients with different ER, PR, and HER2 status. Ano1 was overexpressed in breast cancer compared with fibroadenoma. Ano1 was significantly more associated with breast cancer with the lower clinical stage (stage I or II), or triple-negative status. Mostly importantly, Ano1 overexpression was associated with good prognosis in patients with the PR-positive or HER2-negative status, and in patients following tamoxifen treatment. Multivariate Cox regression analysis showed that Ano1 overexpression was a prognostic factor for longer overall survival in PR-positive or HER2-negative patients, and a predictive factor for longer overall survival in patients following tamoxifen treatment. Our findings suggest that Ano1 may be a potential marker for good prognosis in PR-positive or HER2-negative patients following tamoxifen treatment. The PR and HER2 status defines a subtype of breast cancer in which Ano1 overexpression is associated with good prognosis following tamoxifen treatment.
Collapse
Affiliation(s)
- Huizhe Wu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, P. R. China
| | - Shu Guan
- Department of Breast Surgery, First Hospital of China Medical University, Shenyang, Liaoning, P. R. China
| | - Mingli Sun
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, P. R. China
| | - Zhaojin Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, P. R. China
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, P. R. China
| | - Miao He
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, P. R. China
| | - Haishan Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, P. R. China
| | - Weifan Yao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, P. R. China
| | - Enhua Wang
- Institute of Pathology and Pathophysiology, First Hospital and College of Basic Medical Sciences of China Medical University, Shenyang, Liaoning, P. R. China
| | - Feng Jin
- Department of Breast Surgery, First Hospital of China Medical University, Shenyang, Liaoning, P. R. China
- * E-mail: (MW); (QX); (FJ)
| | - Qinghuan Xiao
- Department of Ion Channel Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, P. R. China
- * E-mail: (MW); (QX); (FJ)
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, P. R. China
- * E-mail: (MW); (QX); (FJ)
| |
Collapse
|
203
|
Tajima S, Ohata A, Koda K, Maruyama Y. Myxoid epithelioid gastrointestinal stromal tumor harboring an unreported PDGFRA mutation: report of a case and review of the literature. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:5821-5829. [PMID: 26191304 PMCID: PMC4503175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 04/26/2015] [Indexed: 06/04/2023]
Abstract
Activating mutations of platelet-derived growth factor receptor α (PDGFRA) are detected in a significant proportion of gastrointestinal stromal tumors (GISTs), in addition to the more frequent mutation in c-kit. GISTs with PDGFRA mutations have been found to have several characteristic morphological features, sometimes allowing to discriminate them from GISTs with c-kit mutations. Among these, epithelioid morphology in tumor cells and tumor-infiltrating mast cells are powerful predictors of PDGFRA mutations. Although myxoid stroma by itself is not so much a reliable predictor of PDGFRA mutation, myxoid stroma in conjunction with epithelioid morphology in tumor cells is a powerful predictor of mutations in this gene. GISTs showing either weak or negative immunoreactivity for c-kit and epithelioid cells with myxoid stroma are called myxoid epithelioid GISTs, which typically show PDGFRA mutation. Herein, we presented a case of a 59-year-old woman with myxoid epithelioid GIST of the stomach. A unique finding in this case was eosinophil infiltration, probably more numerous than mast cells; mast cell infiltration is known to be usually found in myxoid epithelioid GIST. The existence of a similar mechanism in eosinophil and mast cell recruitment via tumor-producing stem cell factor is speculated. Mutational analyses revealed a PDGFRA exon 18 mutation: D842_H845del, D846N. Combined deletion and substitution mutation has been reported in rare instances, but to the best of our knowledge, D846N has not been documented.
Collapse
Affiliation(s)
- Shogo Tajima
- Department of Pathology, Graduate School of Medicine, The University of TokyoTokyo, Japan
- Department of Pathology, Fujieda Municipal General HospitalShizuoka, Japan
| | - Akihiko Ohata
- Department of Gastroenterology, Fujieda Municipal General HospitalShizuoka, Japan
| | - Kenji Koda
- Department of Pathology, Fujieda Municipal General HospitalShizuoka, Japan
| | - Yasuhiko Maruyama
- Department of Gastroenterology, Fujieda Municipal General HospitalShizuoka, Japan
| |
Collapse
|
204
|
Li J, Zhang H, Lu Y, Chen Z, Su K. Presence of PDGFRA and DOG1 mutations in gastrointestinal stromal tumors among Chinese population. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:5721-5726. [PMID: 26191287 PMCID: PMC4503158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 04/13/2015] [Indexed: 06/04/2023]
Abstract
Approximately 15% of gastrointestinal stromal tumors (GIST) do not express KIT mutations and of these about 5 to 7% harbor mutations in PDGFRA. DOG1 was specifically expressed in GISTs. These cases require special attention for PDGFRA and DOG1 mutational status. Hundred cases of GIST were diagnosed between August 2007 and October 2012 at the First Affiliated Hospital of Guangxi Medical University. DNA from tumor tissues and normal adjacent tissues was isolated and amplified for the 22 exons of PDGFRA and 26 exons of DOG1. Each PCR product was sequenced. Amino acid sequences were inferred from DNA and aligned to GenBank reference sequences to determine the position and type of mutations. Overall, 16.0% of the samples had a mutation in PDGFRA, and GISTs with mutations in the DOG1 gene were not found. Of the mutations detected, they were in PDGFRA exon 18 (8 cases, 8%), PDGFRA exon 12 (5 cases, 5%), PDGFRA exon 14 (1 cases, 1.0%), PDGFRA exon 11 (1 cases, 1.0%), and PDGFRA exon 8 (1 cases, 1.0%). Of these, Y392S, L521P and T632K mutant occurred in PDGFRA exon 8, exon 11 and exon 14, respectively. The mutation of PDGFRA has been considered as another causative genetic event as PDGFRA mutations were found in most GISTs lacking a KIT mutation. PDGFRA mutations occurred preferentially in exon 18 and exon 12. Mutations occurring in PDGFRA exon 8 (Y392S), exon 11 (L521P) and exon 14 (T632K) also were first identified. The over-expression of DOG1 was not related to DOG1 gene mutation.
Collapse
Affiliation(s)
- Jiehua Li
- Department of Gastrointestinal and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University Nanning 530021, Guangxi Province, China
| | - Haitian Zhang
- Department of Gastrointestinal and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University Nanning 530021, Guangxi Province, China
| | - Yunfei Lu
- Department of Gastrointestinal and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University Nanning 530021, Guangxi Province, China
| | - Zhibai Chen
- Department of Gastrointestinal and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University Nanning 530021, Guangxi Province, China
| | - Ka Su
- Department of Gastrointestinal and Gland Surgery, The First Affiliated Hospital of Guangxi Medical University Nanning 530021, Guangxi Province, China
| |
Collapse
|
205
|
Lin G, Doyle LA. An update on the application of newly described immunohistochemical markers in soft tissue pathology. Arch Pathol Lab Med 2015; 139:106-21. [PMID: 25549147 DOI: 10.5858/arpa.2014-0488-ra] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT During the last 5 to 10 years, significant progress has been made in the molecular characterization of soft tissue tumors, predominantly with the identification of recurrent translocations or amplification of certain genes in different tumor types. Alongside this, translational efforts have identified many novel and diagnostically useful immunohistochemical markers for many of these tumor types. OBJECTIVE This article reviews a select group of recently described immunohistochemical markers of particular use in the evaluation of mesenchymal neoplasms; the underlying biology of the protein product, practical utility, and limitations of each marker are discussed in detail. DATA SOURCES Literature review, authors' research data, and personal practice experience serve as sources. CONCLUSIONS There are many diagnostically useful immunohistochemical markers to help confirm the diagnosis of many different soft tissue tumor types, some of which have reduced the need for additional, and more costly, studies, such as fluorescence in situ hybridization. However, no one marker is 100% specific for a given tumor, and knowledge of potential pitfalls and overlap in patterns of staining among other tumor types is crucial to ensure the appropriate application of these markers in clinical practice.
Collapse
Affiliation(s)
- George Lin
- From the Department of Laboratory Medicine, Geisinger Medical Center, Danville, Pennsylvania (Dr Lin); and the Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts (Dr Doyle)
| | | |
Collapse
|
206
|
Zhu JQ, Ou WB. Therapeutic targets in gastrointestinal stromal tumors. World J Transl Med 2015; 4:25-37. [DOI: 10.5528/wjtm.v4.i1.25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Revised: 09/14/2014] [Accepted: 12/01/2014] [Indexed: 02/05/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common type of mesenchymal tumor of the gastrointestinal tract. The tumorigenesis of GISTs is driven by gain-of-function mutations in KIT or platelet-derived growth factor receptor α (PDGFRA), resulting in constitutive activation of the tyrosine kinase and its downstream signaling pathways. Oncogenic KIT or PDGFRA mutations are compelling therapeutic targets for the treatment of GISTs, and the KIT/PDGFRA inhibitor imatinib is the standard of care for patients with metastatic GISTs. However, most GIST patients develop clinical resistance to imatinib and other tyrosine kinase inhibitors. Five mechanisms of resistance have been characterized: (1) acquisition of a secondary point mutation in KIT or PDGFRA; (2) genomic amplification of KIT; (3) activation of an alternative receptor tyrosine kinase; (4) loss of KIT oncoprotein expression; and (5) wild-type GIST. Currently, sunitinib is used as a second-line treatment for patients after imatinib failure, and regorafenib has been approved for patients whose disease is progressing on both imatinib and sunitinib. Phase II/III trials are currently in progress to evaluate novel inhibitors and immunotherapies targeting KIT, its downstream effectors such as phosphatidylinositol 3-kinase, protein kinase B and mammalian target of rapamycin, heat shock protein 90, and histone deacetylase inhibitor. Other candidate targets have been identified, including ETV1, AXL, insulin-like growth factor 1 receptor, KRAS, FAS receptor, protein kinase c theta, ANO1 (DOG1), CDC37, and aurora kinase A. These candidates warrant clinical evaluation as novel therapeutic targets in GIST.
Collapse
|
207
|
Dailey DD, Ehrhart EJ, Duval DL, Bass T, Powers BE. DOG1 is a sensitive and specific immunohistochemical marker for diagnosis of canine gastrointestinal stromal tumors. J Vet Diagn Invest 2015; 27:268-77. [DOI: 10.1177/1040638715578878] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Gastrointestinal stromal tumors (GISTs) and leiomyosarcomas are histologically similar primary neoplasms commonly occurring in the gastrointestinal tract of dogs and humans. Immunohistochemical staining (IHC) is needed to differentiate between these 2 entities and positive reactivity for KIT (cluster of differentiation [CD]117) is regarded as the gold standard for diagnosis of canine GIST. Studies estimate 5–10% of human GISTs stain negative or only weakly positive for KIT and have identified DOG1 (discovered on gastrointestinal stromal tumors protein 1) as a highly sensitive and specific marker for human GISTs. The purpose of this study was to evaluate immunoreactivity of a commercially available DOG1 antibody for use in diagnosis of canine GISTs. Fifty-five primary mesenchymal gastrointestinal tumors with histologic features consistent with GIST or leiomyosarcoma were evaluated via IHC for KIT, DOG1, and desmin. A subset of tumors was additionally evaluated for reactivity for smooth muscle actin (SMA). Thirty-three tumors (60%) were diagnosed as GIST based on positive immunoreactivity for KIT or DOG1 regardless of reactivity for desmin or SMA. Most GISTs (32/33, 97.0%) had similar staining for both KIT and DOG1. DOG1 expression was identified in 2 tumors (1 study tumor and 1 additional tumor) negative for KIT and desmin that had histologic features consistent with KIT-negative, platelet-derived growth factor receptor-alpha (PDGFRA)-mutant human GISTs. Our results suggest that DOG1 has improved specificity and sensitivity to that of KIT for differentiating between canine GISTs and leiomyosarcomas. Inclusion of both DOG1 and KIT IHC in diagnostic panels will improve the accuracy of canine GIST diagnosis.
Collapse
Affiliation(s)
- Deanna D. Dailey
- Veterinary Diagnostic Laboratories (Bass, Ehrhart, Powers), Colorado State University, Fort Collins, CO
- Cell and Molecular Biology Graduate Program (Dailey, Ehrhart, Duval), Colorado State University, Fort Collins, CO
- Departments of Microbiology, Immunology and Pathology (Ehrhart, Powers), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Clinical Sciences (Duval), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Flint Animal Cancer Center (Dailey, Ehrhart, Duval), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - E. J. Ehrhart
- Veterinary Diagnostic Laboratories (Bass, Ehrhart, Powers), Colorado State University, Fort Collins, CO
- Cell and Molecular Biology Graduate Program (Dailey, Ehrhart, Duval), Colorado State University, Fort Collins, CO
- Departments of Microbiology, Immunology and Pathology (Ehrhart, Powers), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Clinical Sciences (Duval), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Flint Animal Cancer Center (Dailey, Ehrhart, Duval), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - Dawn L. Duval
- Veterinary Diagnostic Laboratories (Bass, Ehrhart, Powers), Colorado State University, Fort Collins, CO
- Cell and Molecular Biology Graduate Program (Dailey, Ehrhart, Duval), Colorado State University, Fort Collins, CO
- Departments of Microbiology, Immunology and Pathology (Ehrhart, Powers), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Clinical Sciences (Duval), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Flint Animal Cancer Center (Dailey, Ehrhart, Duval), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - Todd Bass
- Veterinary Diagnostic Laboratories (Bass, Ehrhart, Powers), Colorado State University, Fort Collins, CO
- Cell and Molecular Biology Graduate Program (Dailey, Ehrhart, Duval), Colorado State University, Fort Collins, CO
- Departments of Microbiology, Immunology and Pathology (Ehrhart, Powers), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Clinical Sciences (Duval), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Flint Animal Cancer Center (Dailey, Ehrhart, Duval), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| | - B. E. Powers
- Veterinary Diagnostic Laboratories (Bass, Ehrhart, Powers), Colorado State University, Fort Collins, CO
- Cell and Molecular Biology Graduate Program (Dailey, Ehrhart, Duval), Colorado State University, Fort Collins, CO
- Departments of Microbiology, Immunology and Pathology (Ehrhart, Powers), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Clinical Sciences (Duval), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
- Flint Animal Cancer Center (Dailey, Ehrhart, Duval), College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO
| |
Collapse
|
208
|
Zhao W, Tian B, Wu C, Peng Y, Wang H, Gu WL, Gao FH. DOG1, cyclin D1, CK7, CD117 and vimentin are useful immunohistochemical markers in distinguishing chromophobe renal cell carcinoma from clear cell renal cell carcinoma and renal oncocytoma. Pathol Res Pract 2015; 211:303-7. [DOI: 10.1016/j.prp.2014.12.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2014] [Revised: 12/14/2014] [Accepted: 12/23/2014] [Indexed: 11/29/2022]
|
209
|
Sala-Rabanal M, Yurtsever Z, Nichols CG, Brett TJ. Secreted CLCA1 modulates TMEM16A to activate Ca(2+)-dependent chloride currents in human cells. eLife 2015; 4. [PMID: 25781344 PMCID: PMC4360653 DOI: 10.7554/elife.05875] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 02/12/2015] [Indexed: 12/22/2022] Open
Abstract
Calcium-activated chloride channel regulator 1 (CLCA1) activates calcium-dependent chloride currents; neither the target, nor mechanism, is known. We demonstrate that secreted CLCA1 activates calcium-dependent chloride currents in HEK293T cells in a paracrine fashion, and endogenous TMEM16A/Anoctamin1 conducts the currents. Exposure to exogenous CLCA1 increases cell surface levels of TMEM16A and cellular binding experiments indicate CLCA1 engages TMEM16A on the surface of these cells. Altogether, our data suggest that CLCA1 stabilizes TMEM16A on the cell surface, thus increasing surface expression, which results in increased calcium-dependent chloride currents. Our results identify the first Cl− channel target of the CLCA family of proteins and establish CLCA1 as the first secreted direct modifier of TMEM16A activity, delineating a unique mechanism to increase currents. These results suggest cooperative roles for CLCA and TMEM16 proteins in influencing the physiology of multiple tissues, and the pathology of multiple diseases, including asthma, COPD, cystic fibrosis, and certain cancers. DOI:http://dx.doi.org/10.7554/eLife.05875.001 Many biological processes that are important for our health involve the movement of ions into, and out of, our cells. For example, the flow of chloride ions out of cells controls the production of the sticky mucus that lines our windpipe and other airways. This mucus helps trap pollution and other foreign particles before they reach our lungs, and thus protects the lungs from harm. However in some diseases—such as cystic fibrosis and asthma—excessive amounts of thick mucus are produced; this can lead to breathing difficulties and an increased risk of infection. Proteins belonging to the CLCA protein family were first thought to act as channels that allow chloride ions to flow through cell membranes. Later studies then revealed that these proteins are not channels; instead they trigger the movement of chloride ions across cell membranes by activating other channel proteins. However, the identity of these channel proteins was unknown, and it was unclear how CLCA proteins might activate these channels. Sala-Rabanal, Yurtsever et al. have now shown that a member of the CLCA protein family, called CLCA1, is released from human cells and causes nearby cells to release chloride ions when the channel detects calcium ions. The movement of chloride ions triggered by CLCA1 looked very similar to the way chloride ions flow through a channel protein called TMEM16A, and so Sala-Rabanal, Yurtsever et al. asked whether these two proteins interact. TMEM16A was discovered several years ago, but remains the only calcium-dependent chloride channel known in mammals. Sala-Rabanal, Yurtsever et al. showed that adding CLCA1 to cells caused more TMEM16A channels to appear in the cell surface membrane and thereby increased the flow of chloride ions. The CLCA protein also physically interacted with the chloride channel in the membrane to stabilize it; no other protein has been shown to regulate ion channels in this way before. The findings of Sala-Rabanal, Yurtsever et al. provide a much clearer understanding of how the CLCA protein and the chloride channel work. Both of these proteins are known to contribute to excess mucus production in airway diseases; and both have been linked to cardiovascular diseases and certain cancers. These new findings may therefore also help researchers to target these proteins and develop treatments for these diseases. DOI:http://dx.doi.org/10.7554/eLife.05875.002
Collapse
Affiliation(s)
- Monica Sala-Rabanal
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, United States
| | - Zeynep Yurtsever
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St Louis, United States
| | - Colin G Nichols
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, United States
| | - Tom J Brett
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, United States
| |
Collapse
|
210
|
Hameed M, Corless C, George S, Hornick JL, Kakar S, Lazar AJ, Tang L. Template for Reporting Results of Biomarker Testing of Specimens From Patients With Gastrointestinal Stromal Tumors. Arch Pathol Lab Med 2015; 139:1271-5. [DOI: 10.5858/arpa.2014-0578-cp] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Meera Hameed
- From Surgical Pathology, Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, New York (Drs Hameed and Tang); the Department of Pathology, Oregon Health & Science University, Portland (Dr Corless); the Center for Sarcoma and Bone Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (Dr George); the Department of Pathology, Brigham and Women's Hospital, Boston (Dr Ho
| | | | | | | | | | | | | |
Collapse
|
211
|
Wang J, Thway K. Clear Cell Sarcoma–like Tumor of the Gastrointestinal Tract: An Evolving Entity. Arch Pathol Lab Med 2015; 139:407-412. [DOI: 10.5858/arpa.2013-0547-rs] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Clear cell sarcoma–like tumor of the gastrointestinal tract (CCSLGT) is a rare malignant neoplasm that occurs in the wall of the small bowel, stomach, or large bowel, predominantly in young adults. It is an aggressive neoplasm that frequently presents with metastatic disease and has a high mortality rate. Histologically, it is usually composed of medium-sized primitive ovoid or epithelioid cells with pale or clear cytoplasm that are arranged in sheets or in papillary or alveolar architectures. Clear cell sarcoma–like tumor of the gastrointestinal tract is positive for S100 protein, invariably negative for melanocyte-specific markers and is often also positive for neuroendocrine markers. The etiology of CCSLGT is unknown, but many studies have shown associations with EWSR1-CREB1 gene fusions and, less frequently, with EWSR1-ATF1 fusions. Here, we discuss the current status of CCSLGT, including histologic, immunophenotypic, and molecular findings.
Collapse
Affiliation(s)
- Jayson Wang
- From the Department of Histopathology, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Khin Thway
- From the Department of Histopathology, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
212
|
Li F, Huynh H, Li X, Ruddy DA, Wang Y, Ong R, Chow P, Qiu S, Tam A, Rakiec DP, Schlegel R, Monahan JE, Huang A. FGFR-Mediated Reactivation of MAPK Signaling Attenuates Antitumor Effects of Imatinib in Gastrointestinal Stromal Tumors. Cancer Discov 2015; 5:438-51. [PMID: 25673643 DOI: 10.1158/2159-8290.cd-14-0763] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 02/05/2015] [Indexed: 01/19/2023]
Abstract
UNLABELLED Activating mutations in either KIT or PDGFRA are present in approximately 90% of gastrointestinal stromal tumors (GIST). Although treatment with the KIT and PDGFR inhibitor imatinib can control advanced disease in about 80% of GIST patients, the beneficial effect is not durable. Here, we report that ligands from the FGF family reduced the effectiveness of imatinib in GIST cells, and FGF2 and FGFR1 are highly expressed in all primary GIST samples examined. The combination of KIT and FGFR inhibition showed increased growth inhibition in imatinib-sensitive GIST cell lines and improved efficacy in patient-derived GIST xenografts. In addition, inhibition of MAPK signaling by imatinib was not sustained in GIST cells. An ERK rebound occurred through activation of FGF signaling, and was repressed by FGFR1 inhibition. Downregulation of Sprouty proteins played a role in the imatinib-induced feedback activation of FGF signaling in GIST cells. SIGNIFICANCE We here show that FGFR-mediated reactivation of the MAPK pathway attenuates the antiproliferation effects of imatinib in GISTs. The imatinib-induced ERK rebound can be repressed by the FGFR inhibitor BGJ398, and combined KIT and FGFR inhibition leads to increased efficacy in vitro and in patient-derived xenografts.
Collapse
Affiliation(s)
- Fang Li
- Oncology Translational Research, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts.
| | - Hung Huynh
- Laboratory of Molecular Endocrinology, Division of Molecular and Cellular Research, National Cancer Centre, Singapore
| | - Xiaoyan Li
- Oncology Translational Research, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - David A Ruddy
- Oncology Translational Research, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - Youzhen Wang
- Oncology Translational Research, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - Richard Ong
- Laboratory of Molecular Endocrinology, Division of Molecular and Cellular Research, National Cancer Centre, Singapore
| | - Pierce Chow
- Laboratory of Molecular Endocrinology, Division of Molecular and Cellular Research, National Cancer Centre, Singapore
| | - Shumei Qiu
- Oncology Translational Research, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - Angela Tam
- Oncology Translational Research, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - Daniel P Rakiec
- Oncology Translational Research, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - Robert Schlegel
- Oncology Translational Research, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - John E Monahan
- Oncology Translational Research, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts
| | - Alan Huang
- Oncology Translational Research, Novartis Institutes for BioMedical Research, Cambridge, Massachusetts.
| |
Collapse
|
213
|
Abstract
Gastrointestinal stromal tumors (GISTs) were originally thought to harbor either KIT or platelet-derived growth factor receptor A (PDGFRA) mutations only. However, more recent discoveries have highlighted additional, less common oncogenic driver mutations including NF1, BRAF, and succinate dehydrogenase (SDH) mutations. Genotyping GISTs has become more important since not all genotypes respond equally to FDA-approved tyrosine kinase inhibitors. GIST is a paradigm for personalized cancer therapy. Recent studies demonstrate how immunohistochemistry can be used both to diagnose GIST and to screen for specific mutations. DOG1 is particularly useful in the diagnosis of KIT-negative GIST, including tumors with PDGFRA mutations, which can also potentially be identified by immunohistochemistry for PDGFRA. SDHB immunohistochemistry is useful in characterizing GISTs with SDHA-D mutations, whereas SDHA immunohistochemistry is able to identify SDHA mutant GISTs.
Collapse
Affiliation(s)
- Brian P Rubin
- Robert J. Tomsich Pathology Institute, Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio; Department of Molecular Genetics, Lerner Research Institute, Taussig Cancer Center, Cleveland Clinic, NE20, 9500 Euclid Ave, Cleveland, Ohio.
| | - Michael C Heinrich
- Department of Oncology, Veterans Affairs Portland Heatlh Care System and OHSU Knight Cancer Institute, Portland, Oregon
| |
Collapse
|
214
|
Kakkar A, Mathur SR, Jain D, Iyer VK, Nalwa A, Sharma MC. Utility of DOG1 Immunomarker in Fine Needle Aspirates of Gastrointestinal Stromal Tumor. Acta Cytol 2015; 59:61-7. [PMID: 25632981 DOI: 10.1159/000370057] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 11/21/2014] [Indexed: 12/28/2022]
Abstract
BACKGROUND Gastrointestinal stromal tumor (GIST) is the most common mesenchymal tumor of the gastrointestinal tract. DOG1 is a sensitive and specific immunohistochemical marker for the diagnosis of GIST. To date, no study has reported the utility of DOG1 immunocytochemistry on aspirate smears. METHODS Aspirates with a cytological diagnosis of GIST were retrieved. DOG1 immunocytochemistry was performed on aspirates with adequate material. RESULTS 23 cases were included (11 primary, 2 recurrent, 10 metastatic). Primary tumors were most frequently located in the stomach; most metastatic tumors were in the liver. Tumor cells were arranged in cohesive clusters with high cellularity. Cells were spindled, had a low N:C ratio, and a moderate amount of cytoplasm, which was elongated and tapering. Characteristic nuclear features included elongated nuclei with blunt or tapering ends, fine chromatin, mild anisonucleosis, and longitudinal grooves. The mitotic count was low, including in metastatic tumors. DOG1 immunopositivity was noted in 57% of the cases examined. Histopathology was available in 5 cases, all diagnosed as GIST. CONCLUSION Cytology is a sensitive investigative modality for the preoperative diagnosis and confirmation of metastasis of GISTs. In ambiguous cases, DOG1 immunocytochemistry can serve as a valuable adjunct. Cytologic assessment, however, cannot predict malignant potential of GISTs as even metastatic tumors display bland nuclear features.
Collapse
Affiliation(s)
- Aanchal Kakkar
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | | | | | | | | | | |
Collapse
|
215
|
Standardization of Positive Controls in Diagnostic Immunohistochemistry. Appl Immunohistochem Mol Morphol 2015; 23:1-18. [DOI: 10.1097/pai.0000000000000163] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
216
|
Atta IS, Al Qahtani FN. DOG1, alpha-amylase and p63 expression in acinic cell carcinoma of salivary gland; immunohistochemical, clinical and radiological study. Histol Histopathol 2015. [DOI: 10.7243/2055-091x-2-13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
217
|
Quatrième édition de la classification OMS des tumeurs des tissus mous. Ann Pathol 2015; 35:71-85. [DOI: 10.1016/j.annpat.2014.11.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 11/13/2014] [Indexed: 12/11/2022]
|
218
|
Gordon BL, Finnerty BM, Aronova A, Fahey TJ. Genomic medicine for cancer diagnosis. J Surg Oncol 2015; 111:24-30. [DOI: 10.1002/jso.23778] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Affiliation(s)
- Benjamin L. Gordon
- Research Fellow, Department of Surgery; Weill Cornell Medical College/New York Presbyterian Hospital; NY New York
| | - Brendan M. Finnerty
- Research Fellow, Department of Surgery; Weill Cornell Medical College/New York Presbyterian Hospital; NY New York
| | - Anna Aronova
- Research Fellow, Department of Surgery; Weill Cornell Medical College/New York Presbyterian Hospital; NY New York
| | - Thomas J. Fahey
- Chief of Endocrine Surgery and Professor of Surgery, Department of Surgery; Weill Cornell Medical College/New York Presbyterian Hospital; NY New York
| |
Collapse
|
219
|
Peretti M, Angelini M, Savalli N, Florio T, Yuspa SH, Mazzanti M. Chloride channels in cancer: Focus on chloride intracellular channel 1 and 4 (CLIC1 AND CLIC4) proteins in tumor development and as novel therapeutic targets. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1848:2523-31. [PMID: 25546839 DOI: 10.1016/j.bbamem.2014.12.012] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Revised: 12/05/2014] [Accepted: 12/11/2014] [Indexed: 02/07/2023]
Abstract
In recent decades, growing scientific evidence supports the role of ion channels in the development of different cancers. Both potassium selective pores and chloride permeabilities are considered the most active channels during tumorigenesis. High rate of proliferation, active migration, and invasiveness into non-neoplastic tissues are specific properties of neoplastic transformation. All these actions require partial or total involvement of chloride channel activity. In this context, this class of membrane proteins could represent valuable therapeutic targets for the treatment of resistant tumors. However, this encouraging premise has not so far produced any valid new channel-targeted antitumoral molecule for cancer treatment. Problematic for drug design targeting ion channels is their vital role in normal cells for essential physiological functions. By targeting these membrane proteins involved in pathological conditions, it is inevitable to cause relevant side effects in healthy organs. In light of this, a new protein family, the chloride intracellular channels (CLICs), could be a promising class of therapeutic targets for its intrinsic individualities: CLIC1 and CLIC4, in particular, not only are overexpressed in specific tumor types or their corresponding stroma but also change localization and function from hydrophilic cytosolic to integral transmembrane proteins as active ionic channels or signal transducers during cell cycle progression in certain cases. These changes in intracellular localization, tissue compartments, and channel function, uniquely associated with malignant transformation, may offer a unique target for cancer therapy, likely able to spare normal cells. This article is part of a special issue itled "Membrane Channels and Transporters in Cancers."
Collapse
Affiliation(s)
- Marta Peretti
- Department of Life Science, University of Milan, Milano I-20133, Italy
| | - Marina Angelini
- Department of Life Science, University of Milan, Milano I-20133, Italy
| | - Nicoletta Savalli
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90075, USA
| | - Tullio Florio
- Sezione di Farmacologia, Dipartimento di Medicina Interna and Centro di Eccellenza per la Ricerca Biomedica (CEBR), University of Genova, Genova, Italy
| | - Stuart H Yuspa
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, NCI, Bethesda, MD 20892, USA
| | - Michele Mazzanti
- Department of Life Science, University of Milan, Milano I-20133, Italy.
| |
Collapse
|
220
|
Sui Y, Sun M, Wu F, Yang L, Di W, Zhang G, Zhong L, Ma Z, Zheng J, Fang X, Ma T. Inhibition of TMEM16A expression suppresses growth and invasion in human colorectal cancer cells. PLoS One 2014; 9:e115443. [PMID: 25541940 PMCID: PMC4277312 DOI: 10.1371/journal.pone.0115443] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 11/23/2014] [Indexed: 12/12/2022] Open
Abstract
Metastasis leads to poor prognosis in colorectal cancer patients, and there is a growing need for new therapeutic targets. TMEM16A (ANO1, DOG1 or TAOS2) has recently been identified as a calcium-activated chloride channel (CaCC) and is reported to be overexpressed in several malignancies; however, its expression and function in colorectal cancer (CRC) remains unclear. In this study, we found expression of TMEM16A mRNA and protein in high-metastatic-potential SW620, HCT116 and LS174T cells, but not in primary HCT8 and SW480 cells, using RT-PCR, western blotting and immunofluorescence labeling. Patch-clamp recordings detected CaCC currents regulated by intracellular Ca(2+) and voltage in SW620 cells. Knockdown of TMEM16A by short hairpin RNAs (shRNA) resulted in the suppression of growth, migration and invasion of SW620 cells as detected by MTT, wound-healing and transwell assays. Mechanistically, TMEM16A depletion was accompanied by the dysregulation of phospho-MEK, phospho-ERK1/2 and cyclin D1 expression. Flow cytometry analysis showed that SW620 cells were inhibited from the G1 to S phase of the cell cycle in the TMEM16A shRNA group compared with the control group. In conclusion, our results indicate that TMEM16A CaCC is involved in growth, migration and invasion of metastatic CRC cells and provide evidence for TMEM16A as a potential drug target for treating metastatic colorectal carcinoma.
Collapse
Affiliation(s)
- Yujie Sui
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Meiyan Sun
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Fei Wu
- Department of Gynecology and Obstetrics, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Longfei Yang
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Weihua Di
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Guizhen Zhang
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Lili Zhong
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Zhiming Ma
- Department of General Surgery, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Jinhao Zheng
- Department of General Surgery, Jilin University Bethune Second Hospital, Changchun, P. R. China
| | - Xuedong Fang
- Department of General Surgery, Jilin University Bethune Second Hospital, Changchun, P. R. China
- Department of General Surgery, China-Japan Friendship Hospital of Jilin University, Changchun, P. R. China
- * E-mail: (XDF); (THM)
| | - Tonghui Ma
- Key Laboratory for Molecular and Chemical Genetics of Critical Human Diseases of Jilin Province, Jilin University Bethune Second Hospital, Changchun, P. R. China
- College of Basic Medical Sciences, Dalian Medical University, Dalian, P. R. China
- * E-mail: (XDF); (THM)
| |
Collapse
|
221
|
Kodigepalli KM, Bowers K, Sharp A, Nanjundan M. Roles and regulation of phospholipid scramblases. FEBS Lett 2014; 589:3-14. [PMID: 25479087 DOI: 10.1016/j.febslet.2014.11.036] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/22/2014] [Accepted: 11/24/2014] [Indexed: 02/06/2023]
Abstract
Phospholipid scramblase activity is involved in the collapse of phospholipid (PL) asymmetry at the plasma membrane leading to externalization of phosphatidylserine. This activity is crucial for initiation of the blood coagulation cascade and for recognition/elimination of apoptotic cells by macrophages. Efforts to identify gene products associated with this activity led to the characterization of PL scramblase (PLSCR) and XKR family members which contribute to phosphatidylserine exposure in response to apoptotic stimuli. Meanwhile, TMEM16 family members were identified to externalize phosphatidylserine in response to elevated calcium in Scott syndrome platelets, which is critical for activation of the coagulation cascade. Herein, we report their mechanisms of gene regulation, molecular functions independent of their scrambling activity, and their potential roles in pathogenic conditions.
Collapse
Affiliation(s)
- Karthik M Kodigepalli
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, United States
| | - Kiah Bowers
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, United States
| | - Arielle Sharp
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, United States
| | - Meera Nanjundan
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL 33620, United States.
| |
Collapse
|
222
|
Raherinantenaina F, Rasoaherinomenjanahary F, Francine AT, Rambel AH, Rantomalala HYH, Samison LH, Ratsimba HNR. [Gastrointestinal stromal tumor of the ileum with metastatic relapse developed in the mesentery]. Pan Afr Med J 2014; 18:18. [PMID: 25360202 PMCID: PMC4213519 DOI: 10.11604/pamj.2014.18.18.4119] [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: 03/02/2014] [Accepted: 05/01/2014] [Indexed: 11/16/2022] Open
Abstract
Nous rapportons le cas d'un homme de 29 ans admis pour une masse hypogastrique très douloureuse. La laparotomie exploratrice réalisée en urgence permettait de mettre en évidence l'origine et la localisation iléale de la masse tumorale. Le traitement chirurgical consistait en une tumorectomie complète avec respect des marges carcinologiques. Les suites opératoires étaient simples et l'examen histologique confirmait la nature stromale de la tumeur iléale réséquée. Les marges de résection passaient en tissus sains. Le patient était perdu de vue et n'ayant reçu aucun traitement adjuvant. Un an plus tard, il est revenu pour ballonnement et masse pelviens d’évolution rapidement progressive et dont l'exploration chirurgicale révélait l'existence d'une tumeur mésentérique. Une tumorectomie était réalisée mais incomplète à cause d'une perte sanguine avec instabilité hémodynamique induite par l'exérèse tumorale. Après analyse histologique et immunohistochimique de la pièce opératoire, le diagnostic d'une tumeur stromale était confirmé. Il s'agissait d'une rechute métastatique à localisation mésentérique d'une tumeur stromale digestive d'origine iléale à fort potentiel malin. La réduction tumorale suivie d'un complément thérapeutique par l'Imatinib (glivec®) permettaient d'obtenir un résultat satisfaisant. Avec un recul de 12 mois, le patient était asymptomatique et aucune récidive locale ni de métastase à distance n'a été observée.
Collapse
Affiliation(s)
- Fanomezantsoa Raherinantenaina
- Service de Chirurgie Générale et Vasculaire, CHUJRA (Centre Hospitalo-universitaire Joseph Ravoahangy Andrianavalona), Antananarivo, Madagascar
| | | | | | | | | | | | - Hery Nirina Rakoto Ratsimba
- Service de Chirurgie Générale et Vasculaire, CHUJRA (Centre Hospitalo-universitaire Joseph Ravoahangy Andrianavalona), Antananarivo, Madagascar
| |
Collapse
|
223
|
Bartley AN, Hamilton SR. Select biomarkers for tumors of the gastrointestinal tract: present and future. Arch Pathol Lab Med 2014; 139:457-68. [PMID: 25333834 DOI: 10.5858/arpa.2014-0189-ra] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT Advances in molecular biomarkers of the gastrointestinal tract have contributed to a decline in the incidence of and mortality from diseases of the gastrointestinal tract. The discovery and clinical validation of new biomarkers are important to personalized cancer therapy, and numerous clinical trials are currently ongoing to help identify individualized therapy affecting these biomarkers and molecular mechanisms they represent. Distinct molecular pathways leading to cancers of the colorectum, esophagus, stomach, small bowel, and pancreas have been identified. Using biomarkers in these pathways to direct patient care, including selection of proper molecular testing for identification of actionable mutations and reporting the results of these biomarkers to guide clinicians and genetic counselors, is paramount. OBJECTIVE To examine and review select clinically actionable biomarkers of the colon, esophagus, stomach, small bowel, and pancreas, including present and future biomarkers with relevant clinical trials. DATA SOURCES Extensive literature review and practical and consultation experience of the authors. CONCLUSIONS Although numerous biomarkers have been identified and are currently guiding patient therapy, few have shown evidence of clinical utility in the management of patients with gastrointestinal cancers. Inconsistent results and discordant proposed algorithms for testing were identified throughout the literature; however, the potential for biomarkers to improve outcomes for patients with gastrointestinal cancer remains high. Continued advances through high-quality studies are needed.
Collapse
Affiliation(s)
- Angela N Bartley
- From Molecular Diagnostics, Department of Pathology, St. Joseph Mercy Hospital, Ypsilanti, Michigan (Dr Bartley); and the Division of Pathology and Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas (Dr Hamilton)
| | | |
Collapse
|
224
|
Abstract
The fourth edition of the World Health Organization (WHO) Classification of Tumours of Soft Tissue and Bone was published in February 2013, and serves to provide an updated classification scheme and reproducible diagnostic criteria for pathologists. Given the relative rarity of soft tissue tumours and the rapid rate of immunohistochemical and genetic/molecular developments (not infrequently facilitating recognition of new tumour entities), this updated text edited by a consensus group is important for both practising pathologists and oncologists. The 2013 WHO classification includes several changes in soft tissue tumour classification, including several new entities (e.g., pseudomyogenic haemangioendothelioma, haemosiderotic fibrolipomatous tumour, and acral fibromyxoma), three newly included sections for gastrointestinal stromal tumours, nerve sheath tumours, and undifferentiated/unclassified soft tissue tumours, respectively, various 'reclassified' tumours, and a plethora of new genetic and molecular data for established tumour types that facilitate better definition and are useful as diagnostic tools. This article briefly outlines these updates based on the 2013 WHO classification of soft tissue tumours.
Collapse
|
225
|
Sanders KM, Ward SM, Koh SD. Interstitial cells: regulators of smooth muscle function. Physiol Rev 2014; 94:859-907. [PMID: 24987007 DOI: 10.1152/physrev.00037.2013] [Citation(s) in RCA: 338] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Smooth muscles are complex tissues containing a variety of cells in addition to muscle cells. Interstitial cells of mesenchymal origin interact with and form electrical connectivity with smooth muscle cells in many organs, and these cells provide important regulatory functions. For example, in the gastrointestinal tract, interstitial cells of Cajal (ICC) and PDGFRα(+) cells have been described, in detail, and represent distinct classes of cells with unique ultrastructure, molecular phenotypes, and functions. Smooth muscle cells are electrically coupled to ICC and PDGFRα(+) cells, forming an integrated unit called the SIP syncytium. SIP cells express a variety of receptors and ion channels, and conductance changes in any type of SIP cell affect the excitability and responses of the syncytium. SIP cells are known to provide pacemaker activity, propagation pathways for slow waves, transduction of inputs from motor neurons, and mechanosensitivity. Loss of interstitial cells has been associated with motor disorders of the gut. Interstitial cells are also found in a variety of other smooth muscles; however, in most cases, the physiological and pathophysiological roles for these cells have not been clearly defined. This review describes structural, functional, and molecular features of interstitial cells and discusses their contributions in determining the behaviors of smooth muscle tissues.
Collapse
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sang Don Koh
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| |
Collapse
|
226
|
Unexpected DOG-1 immunoreactivity in sarcomatous carcinoma of the liver: a diagnostic pitfall. Pathology 2014; 46:572-4. [PMID: 25158815 DOI: 10.1097/pat.0000000000000154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
227
|
Quezada N, Acevedo F, Marambio A, León F, Galindo H, Roa JC, Jarufe N. Complete pathological response to Imatinib mesylate in an extraintestinal gastrointestinal stromal tumor. Int J Surg Case Rep 2014; 5:681-5. [PMID: 25194604 PMCID: PMC4189531 DOI: 10.1016/j.ijscr.2014.05.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Accepted: 05/14/2014] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION Gastrointestinal stromal tumors (GIST) are the most frequent mesenchymal tumors of the digestive tract. Extraintestinal locations (EGIST) have been described showing similar pattern of immunohistochemical markers than GIST. Inhibitors of tyrosine kinases such as Imatinib or Sunitinib are the mainstay treatment in the management of advanced or metastatic GIST. Complete pathological response to these agents is an extremely rare event, especially in the case of EGIST due to its more aggressive behavior reported. PRESENTATION OF CASE Here we describe the case of a 61 years old woman, with an advanced GIST, who was operated after 10 months of Imatinib mesylate. The biopsy demonstrated the extra intestinal location of the tumor and a complete pathological response was confirmed. DISCUSSION Complete pathological response to Imatinib is a rare event. To our knowledge, this is the first report of complete response in an EGIST. New clinical, radiological and metabolic criteria of tumoral response to neoadjuvant treatment are revised. CONCLUSION EGIST complete pathological response to Imatinib can be achieved. However, recommendation of systematic neoadjuvant therapy with Imatinib remains investigational and more studies are warranted in the future.
Collapse
Affiliation(s)
- Nicolás Quezada
- Digestive Surgery Department, School of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 350, Patio Interior, Santiago 8320000, Chile
| | - Francisco Acevedo
- Hematology-Oncology Department, School of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 350, Patio Interior, Santiago 8320000, Chile
| | - Andrés Marambio
- Digestive Surgery Department, School of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 350, Patio Interior, Santiago 8320000, Chile
| | - Felipe León
- Digestive Surgery Department, School of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 350, Patio Interior, Santiago 8320000, Chile
| | - Hector Galindo
- Hematology-Oncology Department, School of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 350, Patio Interior, Santiago 8320000, Chile
| | - Juan Carlos Roa
- Pathology Department, School of Medicine. Pontificia Universidad Católica de Chile, Marcoleta 350, Patio Interior, Santiago 8320000, Chile
| | - Nicolás Jarufe
- Digestive Surgery Department, School of Medicine, Pontificia Universidad Católica de Chile, Marcoleta 350, Patio Interior, Santiago 8320000, Chile.
| |
Collapse
|
228
|
Blair PJ, Rhee PL, Sanders KM, Ward SM. The significance of interstitial cells in neurogastroenterology. J Neurogastroenterol Motil 2014; 20:294-317. [PMID: 24948131 PMCID: PMC4102150 DOI: 10.5056/jnm14060] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 06/06/2014] [Accepted: 06/07/2014] [Indexed: 12/21/2022] Open
Abstract
Smooth muscle layers of the gastrointestinal tract consist of a heterogeneous population of cells that include enteric neurons, several classes of interstitial cells of mesenchymal origin, a variety of immune cells and smooth muscle cells (SMCs). Over the last number of years the complexity of the interactions between these cell types has begun to emerge. For example, interstitial cells, consisting of both interstitial cells of Cajal (ICC) and platelet-derived growth factor receptor alpha-positive (PDGFRα(+)) cells generate pacemaker activity throughout the gastrointestinal (GI) tract and also transduce enteric motor nerve signals and mechanosensitivity to adjacent SMCs. ICC and PDGFRα(+) cells are electrically coupled to SMCs possibly via gap junctions forming a multicellular functional syncytium termed the SIP syncytium. Cells that make up the SIP syncytium are highly specialized containing unique receptors, ion channels and intracellular signaling pathways that regulate the excitability of GI muscles. The unique role of these cells in coordinating GI motility is evident by the altered motility patterns in animal models where interstitial cell networks are disrupted. Although considerable advances have been made in recent years on our understanding of the roles of these cells within the SIP syncytium, the full physiological functions of these cells and the consequences of their disruption in GI muscles have not been clearly defined. This review gives a synopsis of the history of interstitial cell discovery and highlights recent advances in structural, molecular expression and functional roles of these cells in the GI tract.
Collapse
Affiliation(s)
- Peter J Blair
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA; and
| | - Poong-Lyul Rhee
- Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA; and
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA; and
| |
Collapse
|
229
|
Blair PJ, Rhee PL, Sanders KM, Ward SM. The significance of interstitial cells in neurogastroenterology. J Neurogastroenterol Motil 2014. [PMID: 24948131 DOI: 10.5056/jnm140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Smooth muscle layers of the gastrointestinal tract consist of a heterogeneous population of cells that include enteric neurons, several classes of interstitial cells of mesenchymal origin, a variety of immune cells and smooth muscle cells (SMCs). Over the last number of years the complexity of the interactions between these cell types has begun to emerge. For example, interstitial cells, consisting of both interstitial cells of Cajal (ICC) and platelet-derived growth factor receptor alpha-positive (PDGFRα(+)) cells generate pacemaker activity throughout the gastrointestinal (GI) tract and also transduce enteric motor nerve signals and mechanosensitivity to adjacent SMCs. ICC and PDGFRα(+) cells are electrically coupled to SMCs possibly via gap junctions forming a multicellular functional syncytium termed the SIP syncytium. Cells that make up the SIP syncytium are highly specialized containing unique receptors, ion channels and intracellular signaling pathways that regulate the excitability of GI muscles. The unique role of these cells in coordinating GI motility is evident by the altered motility patterns in animal models where interstitial cell networks are disrupted. Although considerable advances have been made in recent years on our understanding of the roles of these cells within the SIP syncytium, the full physiological functions of these cells and the consequences of their disruption in GI muscles have not been clearly defined. This review gives a synopsis of the history of interstitial cell discovery and highlights recent advances in structural, molecular expression and functional roles of these cells in the GI tract.
Collapse
Affiliation(s)
- Peter J Blair
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - Poong-Lyul Rhee
- Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| |
Collapse
|
230
|
Sözütek D, Yanık S, Akkoca AN, Sözütek A, Özdemir ZT, Avşar ÇU, Günaldı M, Şahin B, Doron F. Diagnostic and prognostic roles of DOG1 and Ki-67, in GIST patients with localized or advanced/metastatic disease. Int J Clin Exp Med 2014; 7:1914-1922. [PMID: 25126201 PMCID: PMC4132165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 06/27/2014] [Indexed: 06/03/2023]
Abstract
AIM Gastrointestinal stromal tumor is the most common mesenchymal neoplasia in the gastrointestinal tract and has a broad spectrum of pathological patterns and also clinical features changing from benign to malignant. Although the well-characterized parameters to predict the outcome have been the size and the mitotic index of the tumor in the patients with early-staged disease, bulky recurrent or metastatic tumor, resistance to medical treatment and mutation analysis are the prognostic factors for advanced stage-GIST. The aim of this study is to investigate new and more practical tissue markers, such as DOG1 and Ki-67 to specify the GIST diagnosis and also to predict the outcome in GIST patients with both localized and advanced staged disease. METHODS For the last 14 years, from 1999 to 2013, 111 patients with a histopathological GIST diagnosis from the hospital files were enrolled to the study. In their parafin-embedded tissue samples, DOG1 and Ki-67 expressions were evaluated with immunohistochemisty by two independent pathologists from Cukurova University Medical Faculty. Patients were divided into two groups, the patients with localized disease treated by surgery and the patients with advanced/metastatic disease. DOG1 and Ki-67 expressions were corelated with other diagnostic and prognostic histopathological markers and also the clinical outcome in these two group of patients. RESULTS The specificity and the sensitivity of DOG1 in GIST diagnosis was found 94 and 43%, respectively. DOG1 expression was especially important in the diagnosis of c-kit negative cases. Although Ki-67 was not found a statistically significant prognostic factor for overall survival, it was strongly corelated with mitotic index which is a well-known standart prognostic factor for localized disease. DISCUSSION DOG1 seems to be an important diagnostic tool for clinically suspected GIST diagnosis in both advanced or early staged patients whose tumours are c-kit expression negative. On the other hand, Ki-67 can be a stronger candidate for prognostic factor instead of mitotic index to identify the proliferative cells out of mitotic phase but this statement needs be prospectively validated on studies with large number of patients.
Collapse
Affiliation(s)
- Didem Sözütek
- Department of Internal Medicine, Iskenderun State HospitalHatay, Turkey
| | - Serdar Yanık
- Department of Patology, Iskenderun State HospitalHatay, Turkey
| | | | - Alper Sözütek
- Department of General Surgeons and Gastroenterology, Mersin State HospitalMersin, Turkey
| | - Zeynep Tuba Özdemir
- Department of Internal Medicine, Bozok University Medical SchoolYozgat, Turkey
| | - Çiğdem Usul Avşar
- Department of Medical Oncology, Çorlu Statae HospitalTekirdağ, Turkey
| | - Meral Günaldı
- Department of Medical Oncology Dr. Sadi Konuk Research Hospitalİstanbul, Turkey
| | - Berksoy Şahin
- Department of Medical Oncology, Cukurova University Medical School, Internal MedicineAdana, Turkey
| | - Figen Doron
- Department of Patology, Cukurova University Medical SchoolAdana, Turkey
| |
Collapse
|
231
|
Kataoka TR, Yamashita N, Furuhata A, Hirata M, Ishida T, Nakamura I, Hirota S, Haga H, Katsuyama E. An inflammatory myofibroblastic tumor exhibiting immunoreactivity to KIT: a case report focusing on a diagnostic pitfall. World J Surg Oncol 2014; 12:186. [PMID: 24938355 PMCID: PMC4083143 DOI: 10.1186/1477-7819-12-186] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 06/12/2014] [Indexed: 01/01/2023] Open
Abstract
Inflammatory myofibroblastic tumors (IMTs) and gastrointestinal stromal tumors (GISTs) are both spindle cell tumors, and occur rarely in the wall of the urinary bladder. In general, immunostaining allows differentiation of IMTs and GISTs. Most IMTs are positive for anaplastic lymphoma kinase (ALK) and negative for KIT, whereas most GISTs are ALK-negative and KIT-positive. Here, we describe a case of a spindle cell tumor in the wall of the urinary bladder. The spindle cells were positive for both ALK and KIT, and it was thus difficult to determine whether the tumor was an IMT or a GIST. We eventually diagnosed an IMT, because ALK gene rearrangement was confirmed by fluorescent in-situ hybridization. Cytoplasmic staining for KIT and the absence of other GIST markers, including DOG1 and platelet-derived growth factor α, indicated that the tumor was not a GIST. Therefore, IMTs should be included in the differential diagnosis of spindle cell tumors, even those that are KIT-positive.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hironori Haga
- Department of Diagnostic Pathology, Kyoto University Hospital, Sakyo-ku, Kyoto 606-8507, Japan.
| | | |
Collapse
|
232
|
Peng MF, Li K, Wang C, Zhu XY, Yang Z, Zhang GH, Wang PH, Wang YH, Tang LJ, Zhang L. Therapeutic effect and mechanism of electroacupuncture at Zusanli on plasticity of interstitial cells of Cajal: a study of rat ileum. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 14:186. [PMID: 24908398 PMCID: PMC4096531 DOI: 10.1186/1472-6882-14-186] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 05/21/2014] [Indexed: 12/13/2022]
Abstract
Background Electroacupuncture (EA) is one of the techniques of acupuncture and is believed to be an effective alternative and complementary treatment in many disorders. The aims of this study were to investigate the effects and mechanisms of EA at acupoint Zusanli (ST36) on the plasticity of interstitial cells of Cajal (ICCs) in partial bowel obstruction. Methods A Sprague Dawley rat model of partial bowel obstruction was established and EA was conducted at Zusanli (ST36) and Yinglingquan (SP9) in test and control groups, respectively. Experiments were performed to study the effects and mechanisms of EA at Zusanli on intestinal myoelectric activity, distribution and alteration of ICCs, expression of inflammatory mediators, and c-Kit expression. Results 1) EA at Zusanli somewhat improved slow wave amplitude and frequency in the partial obstruction rats. 2) EA at Zusanli significantly stimulated the recovery of ICC networks and numbers. 3) the pro-inflammatory mediator TNF-α and NO activity were significantly reduced after EA at Zusanli, However, no significant changes were observed in the anti-inflammatory mediator IL-10 activity. 4) EA at Zusanli re-expressed c-Kit protein. However, EA at the control acupoint, SP9, significantly improved slow wave frequency and amplitude, but had no effect on ICC or inflammatory mediators. Conclusions We concluded that EA at Zusanli might have a therapeutic effect on ICC plasticity, and that this effect might be mediated via a decrease in pro-inflammatory mediators and through the c-Kit signaling pathway, but that the relationship between EA at different acupoints and myoelectric activity needs further study.
Collapse
|
233
|
|
234
|
Berglund E, Akcakaya P, Berglund D, Karlsson F, Vukojević V, Lee L, Bogdanović D, Lui WO, Larsson C, Zedenius J, Fröbom R, Bränström R. Functional role of the Ca²⁺-activated Cl⁻ channel DOG1/TMEM16A in gastrointestinal stromal tumor cells. Exp Cell Res 2014; 326:315-25. [PMID: 24825187 DOI: 10.1016/j.yexcr.2014.05.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 05/02/2014] [Accepted: 05/03/2014] [Indexed: 01/12/2023]
Abstract
DOG1, a Ca(2+)-activated Cl(-) channel (CaCC), was identified in 2004 to be robustly expressed in gastrointestinal stromal tumors (GIST). It was rapidly included as a tumor marker in routine diagnostics, but the functional role remained unknown. CaCCs are important regulators of normal physiological functions, but also implicated in tumorigenesis, cancer progression, metastasis, cell migration, apoptosis, proliferation and viability in several malignancies. We therefore investigated whether DOG1 plays a role in the three latter in GIST by utilizing in vitro cell model systems. Confocal microscopy identified different subcellular localizations of DOG1 in imatinib-sensitive and imatinib-resistant cells. Electrophysiological studies confirmed that DOG1-specific pharmacological agents possess potent activating and inhibiting properties. Proliferation assays showed small effects up to 72 h, and flow cytometric analysis of adherent cells with 7-AAD/Annexin V detected no pharmacological effects on viable GIST cells. However, inhibition of DOG1 conveyed pro-apoptotic effects among early apoptotic imatinib-resistant cells. In conclusion, DOG1 generates Cl(-) currents in GIST that can be regulated pharmacologically, with small effects on cell viability and proliferation in vitro. Inhibition of DOG1 might act pro-apoptotic on some early apoptotic GIST cell populations. Further studies are warranted to fully illuminate the function of DOG1 and its potential as therapeutic target.
Collapse
Affiliation(s)
- Erik Berglund
- Endocrine and Sarcoma Surgery Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Breast and Endocrine Surgery, Karolinska University Hospital, Stockholm, Sweden.
| | - Pinar Akcakaya
- Department of Oncology-Pathology, Karolinska Institutet, Cancer Center Karolinska, Stockholm, Sweden
| | - David Berglund
- Section for Transplantation Surgery, Department of Surgical Sciences, Uppsala University Hospital, Uppsala, Sweden
| | - Fredrik Karlsson
- Endocrine and Sarcoma Surgery Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Breast and Endocrine Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Vladana Vukojević
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Linkiat Lee
- Department of Oncology-Pathology, Karolinska Institutet, Cancer Center Karolinska, Stockholm, Sweden
| | - Darko Bogdanović
- Endocrine and Sarcoma Surgery Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Weng-Onn Lui
- Department of Oncology-Pathology, Karolinska Institutet, Cancer Center Karolinska, Stockholm, Sweden
| | - Catharina Larsson
- Department of Oncology-Pathology, Karolinska Institutet, Cancer Center Karolinska, Stockholm, Sweden
| | - Jan Zedenius
- Endocrine and Sarcoma Surgery Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Breast and Endocrine Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Robin Fröbom
- Endocrine and Sarcoma Surgery Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Robert Bränström
- Endocrine and Sarcoma Surgery Unit, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Breast and Endocrine Surgery, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
235
|
Choi EJ, Yun JA, Jabeen S, Jeon EK, Won HS, Ko YH, Kim SY. Prognostic significance of TMEM16A, PPFIA1, and FADD expression in invasive ductal carcinoma of the breast. World J Surg Oncol 2014; 12:137. [PMID: 24886289 PMCID: PMC4028009 DOI: 10.1186/1477-7819-12-137] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 04/20/2014] [Indexed: 01/01/2023] Open
Abstract
Background 11q13 region is a frequently amplified locus in human malignancies. Among the genes located in this region, FADD is one of the alleged driving genes. Because amplification is not generally confined to a single gene and amplified genes may not show increased expression, we need to evaluate clinical significance of changes occurring in 11q13 region to understand their roles in carcinogenesis. Therefore, we screened expressions of FADD and closely located genes (PPFIA1 and TMEM16A) and evaluated the expressions to find clinical significance in invasive ductal carcinoma of the breast. Methods Ninety-eight cases of invasive ductal carcinoma of the breast were collected. Using archival tissues resected from the cases, we built a tissue microarray and used it in immunohistochemistry. We evaluated the association of FADD, PPFIA1, and TMEM16A expression scores with clinicopathological parameters, including disease-free survival. Results FADD expression was associated with T stage (P = 0.046). The combined score of FADD, PPFIA1, and TMEM16A gene expressions was associated with perineural invasion (P = 0.022). Although individual gene expressions of TMEM16A, FADD, and PPFIA1 failed to show significant association with disease-free survival, combined gene expression scores did show association with disease-free survival (P = 0.034). Conclusions FADD, TMEM16A, and PPFIA1 gene expressions as a whole were associated with disease-free survival in breast cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Su Young Kim
- Department of Pathology, The Catholic University of Korea, School of Medicine, Seochogu Banpodaero 222, Seoul 137-701, South Korea.
| |
Collapse
|
236
|
Hoffmann EK, Holm NB, Lambert IH. Functions of volume-sensitive and calcium-activated chloride channels. IUBMB Life 2014; 66:257-67. [PMID: 24771413 DOI: 10.1002/iub.1266] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 03/31/2014] [Accepted: 03/31/2014] [Indexed: 01/23/2023]
Abstract
The review describes molecular and functional properties of the volume regulated anion channel and Ca(2+)-dependent Cl(-) channels belonging to the anoctamin family with emphasis on physiological importance of these channels in regulation of cell volume, cell migration, cell proliferation, and programmed cell death. Finally, we discuss the role of Cl(-) channels in various diseases.
Collapse
Affiliation(s)
- Else Kay Hoffmann
- Department of Biology, University of Copenhagen, 13 Universitetsparken, Copenhagen Ø, Denmark
| | | | | |
Collapse
|
237
|
Abstract
TMEM16 proteins, also known as anoctamins, are involved in a variety of functions that include ion transport, phospholipid scrambling, and regulation of other membrane proteins. The first two members of the family, TMEM16A (anoctamin-1, ANO1) and TMEM16B (anoctamin-2, ANO2), function as Ca2+-activated Cl- channels (CaCCs), a type of ion channel that plays important functions such as transepithelial ion transport, smooth muscle contraction, olfaction, phototransduction, nociception, and control of neuronal excitability. Genetic ablation of TMEM16A in mice causes impairment of epithelial Cl- secretion, tracheal abnormalities, and block of gastrointestinal peristalsis. TMEM16A is directly regulated by cytosolic Ca2+ as well as indirectly by its interaction with calmodulin. Other members of the anoctamin family, such as TMEM16C, TMEM16D, TMEM16F, TMEM16G, and TMEM16J, may work as phospholipid scramblases and/or ion channels. In particular, TMEM16F (ANO6) is a major contributor to the process of phosphatidylserine translocation from the inner to the outer leaflet of the plasma membrane. Intriguingly, TMEM16F is also associated with the appearance of anion/cation channels activated by very high Ca2+ concentrations. Furthermore, a TMEM16 protein expressed in Aspergillus fumigatus displays both ion channel and lipid scramblase activity. This finding suggests that dual function is an ancestral characteristic of TMEM16 proteins and that some members, such as TMEM16A and TMEM16B, have evolved to a pure channel function. Mutations in anoctamin genes (ANO3, ANO5, ANO6, and ANO10) cause various genetic diseases. These diseases suggest the involvement of anoctamins in a variety of cell functions whose link with ion transport and/or lipid scrambling needs to be clarified.
Collapse
|
238
|
Stromal signatures in endometrioid endometrial carcinomas. Mod Pathol 2014; 27:631-9. [PMID: 24263966 DOI: 10.1038/modpathol.2013.131] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 05/29/2013] [Accepted: 05/30/2013] [Indexed: 02/06/2023]
Abstract
The pattern of myometrial invasion in endometrioid endometrial carcinomas varies considerably; ie, from widely scattered glands and cell nests, often associated with a fibromyxoid stromal reaction (desmoplasia) and/or a lymphocytic infiltrate, to invasive glands with little or no stromal response. Recently, two distinct stromal signatures derived from a macrophage response (colony-stimulating factor 1, CSF1) and a fibroblastic response (desmoid-type fibromatosis, DTF) were identified in breast carcinomas and correlated with clinicopathologic features including outcome. In this study, we explored whether these stromal signatures also apply to endometrioid carcinomas and how their expression patterns correlated with morphologic changes. We studied the stromal signatures both by immunohistochemistry and in situ hybridization in 98 primary endometrioid carcinomas with (87 cases) and without (11 cases) myometrial invasion as well as in the corresponding regional lymph nodes metatases of 9 myoinvasive tumors. Desmoplasia correlated positively with the DTF expression signature. Likewise, mononuclear infiltrates were found in the stroma of tumors expressing CSF1. Twenty-four out of eighty-seven (27%) myoinvasive endometrioid carcinomas were positive for the macrophage signature and thirteen out of eighty-seven (15%) expressed the fibroblast signature. Eleven additional cases were positive for both DTF and CSF1 signatures (11/87; 13%). However, over half of the cases (39/87; 45%) and the majority of the non-myoinvasive tumors (8/11; 73%) failed to express any of the two stromal signatures. The macrophage response (CSF1) was associated with higher tumor grade, lymphovascular invasion, and PIK3CA mutations (P<0.05). There was a concordance in the expression of the CSF1 signature in the primary tumors and their corresponding lymph node metastases. This study is the first characterization of stromal signatures in endometrioid carcinomas. Our findings shed new light on the relationship between genetically different endometrioid carcinomas and various stromal responses. Preservation of the CSF1 macrophage stromal response in the metastases leds support to targeting the CSF1 pathway in endometrioid endometrial carcinomas.
Collapse
|
239
|
Qu Z, Yao W, Yao R, Liu X, Yu K, Hartzell C. The Ca(2+) -activated Cl(-) channel, ANO1 (TMEM16A), is a double-edged sword in cell proliferation and tumorigenesis. Cancer Med 2014; 3:453-61. [PMID: 24639373 PMCID: PMC4101736 DOI: 10.1002/cam4.232] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Revised: 02/13/2014] [Accepted: 02/21/2014] [Indexed: 01/11/2023] Open
Abstract
Since anoctamin 1 ANO1 (TMEM16A) was found to be a molecular component of Ca2+-activated Cl− channels, its role in tumorigenesis has gained attention at a fast pace. ANO1 overexpression frequently occurs in the cancer tissues along with 11q13 chromosome amplification. Poor prognosis of many types of cancers has been closely correlated with ANO1 gene amplification and protein overexpression. ANO1 is now considered an excellent biomarker for certain cancers. Recent research suggests that it is the channel function of ANO1 that is involved in the tumorigenesis. However, how the overexpression of the functional ANO1 causes malignant transformation of tissues via signaling pathways, for example, MAPK remains to be investigated. Clarification of the reasons in future will avail to make ANO1 as a target for cancer treatment.
Collapse
Affiliation(s)
- Zhiqiang Qu
- Medical Research Center, Affiliated Hospital, Qingdao University, Qingdao, 266555, China; State Key Physiological Discipline, Qingdao University, Qingdao, 266555, China
| | | | | | | | | | | |
Collapse
|
240
|
Zhang ZH, Feng GW, Liu ZF, Qiao L, Zhang T, Gao C, Xu Y. A young man with primary prostatic extra-gastrointestinal stromal tumor: a rare case report and review of the literature. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:1764-1770. [PMID: 24817976 PMCID: PMC4014260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 03/10/2014] [Indexed: 06/03/2023]
Abstract
Mounting evidence demonstrates the presence of extragastrointestinal stromal tumor (EGIST) which originates from tissues outside the gastrointestinal (GI) tract and shares overlapping immunohistological features with gastrointestinal stromal tumor (GIST). GIST emanating from prostate is extremely rare. To our knowledge, there are only 3 definitely reported cases of primary prostatic EGIST. Herein, we report a case of prostatic EGIST in 31-year-old man with low urinary tract symptoms who was initially misdiagnosed as sarcoma of prostate. Imaging studies assist in determining the origin and location of EGIST. Immunohistochemical assessment (DOG-1, CD117, and CD34) helps in differentiating such lesion from other stromal tumors and in addressing an appropriate and optimal therapeutic strategy.
Collapse
Affiliation(s)
- Zhi-Hong Zhang
- Department of Urology, Second Hospital of Tianjin Medical University, Tianjin Institute of Urology Tianjin, China
| | - Guo-Wei Feng
- Department of Urology, Second Hospital of Tianjin Medical University, Tianjin Institute of Urology Tianjin, China
| | - Zhi-Fei Liu
- Department of Urology, Second Hospital of Tianjin Medical University, Tianjin Institute of Urology Tianjin, China
| | - Lei Qiao
- Department of Urology, Second Hospital of Tianjin Medical University, Tianjin Institute of Urology Tianjin, China
| | - Tao Zhang
- Department of Urology, Second Hospital of Tianjin Medical University, Tianjin Institute of Urology Tianjin, China
| | - Chao Gao
- Department of Urology, Second Hospital of Tianjin Medical University, Tianjin Institute of Urology Tianjin, China
| | - Yong Xu
- Department of Urology, Second Hospital of Tianjin Medical University, Tianjin Institute of Urology Tianjin, China
| |
Collapse
|
241
|
Liu S, Yu Q, Han W, Qi L, Zu X, Zeng F, Xie Y, Liu J. Primary gastrointestinal stromal tumor of the prostate: A case report and literature review. Oncol Lett 2014; 7:1925-1929. [PMID: 24932260 PMCID: PMC4049718 DOI: 10.3892/ol.2014.1968] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 01/30/2014] [Indexed: 12/25/2022] Open
Abstract
Extragastrointestinal stromal tumors (EGISTs) are relatively rare soft tissue neoplasms arising from the extra gastrointestinal tract. The current study presents a case of primary EGIST of the prostate observed in a 55-year-old male. Imaging studies showed a 10×10.5×9.5-cm prostate mass. On histological observation, the mass was separated from the rectum serosa and exhibited a high mitotic count (8/50 high-power fields). The results of immunohistochemical staining showed positive immunoreactivity for cluster of differentiation (CD)117 (c-kit), CD34 and DOG1 in the tumor. On mutation analysis, loss of heterozygosity of the c-kit gene was observed in the prostatic EGIST; however, the platelet-derived growth factor receptor-α (PDGFRA) gene was considered to be normal. Therefore, as EGIST of the prostate is rare, there is a requirement for the confirmation of the diagnosis to be based on immunohistochemistry and mutation analysis (of c-kit and PDGFRA).
Collapse
Affiliation(s)
- Sulai Liu
- Department of Urology, Xiangya Hospital, The Central South University, Changsha, Hunan 410008, P.R. China ; Department of Urology, The Affiliated Tumor Hospital of Xiangya Medical School, The Central South University, Changsha, Hunan 410013, P.R. China
| | - Qiuxia Yu
- Department of Rheumatology, Ningbo Medical Center Li Huili Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Weiqing Han
- Department of Urology, The Affiliated Tumor Hospital of Xiangya Medical School, The Central South University, Changsha, Hunan 410013, P.R. China
| | - Lin Qi
- Department of Urology, Xiangya Hospital, The Central South University, Changsha, Hunan 410008, P.R. China
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital, The Central South University, Changsha, Hunan 410008, P.R. China
| | - Fuhua Zeng
- Department of Urology, The Affiliated Tumor Hospital of Xiangya Medical School, The Central South University, Changsha, Hunan 410013, P.R. China
| | - Yu Xie
- Department of Urology, The Affiliated Tumor Hospital of Xiangya Medical School, The Central South University, Changsha, Hunan 410013, P.R. China
| | - Jingshi Liu
- Department of Anesthesia, The Affiliated Tumor Hospital of Xiangya Medical School, The Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
242
|
Bill A, Hall ML, Borawski J, Hodgson C, Jenkins J, Piechon P, Popa O, Rothwell C, Tranter P, Tria S, Wagner T, Whitehead L, Gaither LA. Small molecule-facilitated degradation of ANO1 protein: a new targeting approach for anticancer therapeutics. J Biol Chem 2014; 289:11029-11041. [PMID: 24599954 PMCID: PMC4036244 DOI: 10.1074/jbc.m114.549188] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
ANO1, a calcium-activated chloride channel, is highly expressed and amplified in human cancers and is a critical survival factor in these cancers. The ANO1 inhibitor CaCCinh-A01 decreases proliferation of ANO1-amplified cell lines; however, the mechanism of action remains elusive. We explored the mechanism behind the inhibitory effect of CaCCinh-A01 on cell proliferation using a combined experimental and in silico approach. We show that inhibition of ANO1 function is not sufficient to diminish proliferation of ANO1-dependent cancer cells. We report that CaCCinh-A01 reduces ANO1 protein levels by facilitating endoplasmic reticulum-associated, proteasomal turnover of ANO1. Washout of CaCCinh-A01 rescued ANO1 protein levels and resumed cell proliferation. Proliferation of newly derived CaCCinh-A01-resistant cell pools was not affected by CaCCinh-A01 as compared with the parental cells. Consistently, CaCCinh-A01 failed to reduce ANO1 protein levels in these cells, whereas ANO1 currents were still inhibited by CaCCinh-A01, indicating that CaCCinh-A01 inhibits cell proliferation by reducing ANO1 protein levels. Furthermore, we employed in silico methods to elucidate novel biological functions of ANO1 inhibitors. Specifically, we derived a pharmacophore model to describe inhibitors capable of promoting ANO1 degradation and report new inhibitors of ANO1-dependent cell proliferation. In summary, our data demonstrate that inhibition of the channel activity of ANO1 is not sufficient to inhibit ANO1-dependent cell proliferation, indicating that the role of ANO1 in cancer only partially depends on its function as a channel. Our results provide an impetus for gaining a deeper understanding of ANO1 modulation in cells and introduce a new targeting approach for antitumor therapy in ANO1-amplified cancers.
Collapse
Affiliation(s)
- Anke Bill
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139
| | - Michelle Lynn Hall
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139
| | - Jason Borawski
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139
| | - Catherine Hodgson
- Novartis Institutes for Biomedical Research, Horsham, West Sussex, RH12 5AB, United Kingdom, and
| | - Jeremy Jenkins
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139
| | - Philippe Piechon
- the Novartis Institutes for Biomedical Research, Basel CH-4002, Switzerland
| | - Oana Popa
- Novartis Institutes for Biomedical Research, Horsham, West Sussex, RH12 5AB, United Kingdom, and
| | | | - Pamela Tranter
- Novartis Institutes for Biomedical Research, Horsham, West Sussex, RH12 5AB, United Kingdom, and
| | - Scott Tria
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139
| | - Trixie Wagner
- the Novartis Institutes for Biomedical Research, Basel CH-4002, Switzerland
| | - Lewis Whitehead
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139
| | - L Alex Gaither
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139,.
| |
Collapse
|
243
|
Nuclear expression of STAT6 distinguishes solitary fibrous tumor from histologic mimics. Mod Pathol 2014; 27:390-5. [PMID: 24030747 DOI: 10.1038/modpathol.2013.164] [Citation(s) in RCA: 484] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 07/09/2013] [Accepted: 07/14/2013] [Indexed: 12/17/2022]
Abstract
Solitary fibrous tumor (SFT) is composed of spindled to ovoid cells in a patternless architecture with prominent stromal collagen and hemangiopericytoma-like vessels. Some tumors show hypercellularity, nuclear atypia, and significant mitotic activity; the latter feature in particular often portends an aggressive clinical course. SFT can sometimes be difficult to distinguish from other benign mesenchymal tumors and sarcomas. The most characteristic (albeit nonspecific) immunohistochemical finding in SFT is CD34 expression. A NAB2-STAT6 gene fusion, resulting in a chimeric protein in which a repressor domain of NGFI-A binding protein 2 (EGR1 binding protein 2) (NAB2) is replaced with a carboxy-terminal transactivation domain from signal transducer and activator of transcription 6, interleukin-4 induced (STAT6), was recently identified as a consistent finding in SFT. However, as these genes are located in close proximity on 12q13, this fusion can only rarely be detected by conventional chromosomal banding or fluorescence in situ hybridization analysis. Nuclear expression of the carboxy terminal part of STAT6 is a consistent finding in SFT of the meninges (so-called 'meningeal hemangiopericytoma'). We investigated STAT6 expression by immunohistochemistry in SFTs and other soft tissue tumors arising outside the central nervous system to validate the diagnostic utility of this novel marker. Whole-tissue sections of 231 tumors were evaluated, including 60 cases of SFT as well as other benign and malignant mesenchymal neoplasms and sarcomatoid mesotheliomas. Fifty-nine of 60 SFT cases (98%) showed nuclear expression of STAT6, which was usually diffuse and intense. All other tumor types were negative for STAT6, except for three dedifferentiated liposarcomas and one deep fibrous histiocytoma, which showed weak staining. In conclusion, STAT6 is a highly sensitive and almost perfectly specific immunohistochemical marker for SFT and can be helpful to distinguish this tumor type from histologic mimics.
Collapse
|
244
|
Grubb S, Poulsen KA, Juul CA, Kyed T, Klausen TK, Larsen EH, Hoffmann EK. TMEM16F (Anoctamin 6), an anion channel of delayed Ca(2+) activation. ACTA ACUST UNITED AC 2014; 141:585-600. [PMID: 23630341 PMCID: PMC3639583 DOI: 10.1085/jgp.201210861] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Members of the TMEM16 (Anoctamin) family of membrane proteins have been shown to be essential constituents of the Ca2+-activated Cl− channel (CaCC) in many cell types. In this study, we have investigated the electrophysiological properties of mouse TMEM16F. Heterologous expression of TMEM16F in HEK293 cells resulted in plasma membrane localization and an outwardly rectifying ICl,Ca that was activated with a delay of several minutes. Furthermore, a significant Na+ current was activated, and the two permeabilities were correlated according to PNa = 0.3 PCl. The current showed an EC50 of 100 µM intracellular free Ca2+ concentration and an Eisenman type 1 anion selectivity sequence of PSCN > PI > PBr > PCl > PAsp. The mTMEM16F-associated ICl,Ca was abolished in one mutant of the putative pore region (R592E) but retained in two other mutants (K616E and R636E). The mutant K616E had a lower relative permeability to iodide, and the mutant R636E had an altered anion selectivity sequence (PSCN = PI = PBr = PCl > PAsp). Our data provide evidence that TMEM16F constitutes a Ca2+-activated anion channel or a pore-forming subunit of an anion channel with properties distinct from TMEM16A.
Collapse
Affiliation(s)
- Søren Grubb
- Department of Biology, University of Copenhagen, DK-2100 Copenhagen Ø, Denmark
| | | | | | | | | | | | | |
Collapse
|
245
|
|
246
|
Reichardt P, Morosi C, Wardelmann E, Gronchi A. Gastrointestinal stromal tumors: evolving role of the multidisciplinary team approach in management. Expert Rev Anticancer Ther 2014; 12:1053-68. [DOI: 10.1586/era.12.48] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
247
|
Novel uses of immunohistochemistry in the diagnosis and classification of soft tissue tumors. Mod Pathol 2014; 27 Suppl 1:S47-63. [PMID: 24384853 DOI: 10.1038/modpathol.2013.177] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 07/30/2013] [Indexed: 12/13/2022]
Abstract
Immunohistochemistry plays a key role in the diagnosis of soft tissue tumors. Until recently, however, the primary purpose of immunohistochemistry in this context was simply to attempt to demonstrate a line of differentiation. Unfortunately, most traditional markers (predominantly directed against cytoplasmic determinants) show relatively limited specificity. Over the last decade or so, much more specific immunohistochemical markers for soft tissue tumors have been developed. This review will provide an update of some of the most useful new diagnostic markers, which are significantly changing clinical practice for surgical pathologists, separated into three general categories: (1) lineage-restricted transcription factors, (2) protein correlates of molecular alterations, and (3) diagnostic markers identified by gene expression profiling.
Collapse
|
248
|
Skipworth JRA, Fanshawe AEE, West MJ, Al-Bahrani A. Perforation as a rare presentation of gastric gastrointestinal stromal tumours: a case report and review of the literature. Ann R Coll Surg Engl 2014; 96:96E-100E. [PMID: 24417854 PMCID: PMC5137650 DOI: 10.1308/003588414x13824511650010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2012] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION Gastrointestinal stromal tumours (GISTs) are the most common connective tissue neoplasms of the gastrointestinal tract, the most common clinical presentation of which is with abdominal pain or gastrointestinal bleeding. METHODS We describe a case of a perforated gastric GIST as well as reviewing the relevant published literature. RESULTS A 51-year-old woman presented to the acute assessment unit with a 1-day history of severe epigastric pain on a background of longstanding reflux symptoms. Radiological investigation demonstrated a perforated mass in the gastric antrum and the patient subsequently underwent an emergency distal gastrectomy. She recovered well postoperatively and was discharged home. Her condition remains stable six months following surgery. Histological analysis revealed the perforated lesion to be a GIST. A PubMed search suggests that this is the first English report to describe a perforated gastric GIST. Six further published reports (written in English or with an English abstract) describing the presentation of small bowel GISTs with perforation are reviewed. CONCLUSIONS We present the first English report of a perforated gastric GIST. More common presentations include abdominal pain and gastrointestinal bleeding. Although rare, GISTs should be considered in the differential diagnoses of perforated gastrointestinal masses.
Collapse
|
249
|
Corless CL. Gastrointestinal stromal tumors: what do we know now? Mod Pathol 2014; 27 Suppl 1:S1-16. [PMID: 24384849 DOI: 10.1038/modpathol.2013.173] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 06/15/2013] [Accepted: 06/17/2013] [Indexed: 12/15/2022]
Abstract
Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the GI tract, arising from the interstitial cells of Cajal, primarily in the stomach and small intestine. They manifest a wide range of morphologies, from spindle cell to epithelioid, but are immunopositive for KIT (CD117) and/or DOG1 in essentially all cases. Although most tumors are localized at presentation, up to half will recur in the abdomen or spread to the liver. The growth of most GISTs is driven by oncogenic mutations in either of two receptor tyrosine kinases: KIT (75% of cases) or PDGFRA (10%). Treatment with tyrosine kinase inhibitors (TKIs) such as imatinib, sunitinib, and regorafenib is effective in controlling unresectable disease; however, drug resistance caused by secondary KIT or PDGFRA mutations eventually develops in 90% of cases. Adjuvant therapy with imatinib is commonly used to reduce the likelihood of disease recurrence after primary surgery, and for this reason assessing the prognosis of newly resected tumors is one of the most important roles for pathologists. Approximately 15% of GISTs are negative for mutations in KIT and PDGFRA. Recent studies of these so-called wild-type GISTs have uncovered a number of other oncogenic drivers, including mutations in neurofibromatosis type I, RAS genes, BRAF, and subunits of the succinate dehydrogenase complex. Routine genotyping is strongly recommended for optimal management of GISTs, as the type and dose of TKI used for treatment is dependent on the mutation identified.
Collapse
Affiliation(s)
- Christopher L Corless
- Department of Pathology (L471) and Knight Diagnostic Laboratories, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
250
|
Shackelford RE, Abdelbaqi MQ, Almhanna K, Meredith K, Coppola D. Molecular Pathology and Diagnostics in Esophago-gastric Cancer. MOLECULAR PATHOLOGY AND DIAGNOSTICS OF CANCER 2014:177-210. [DOI: 10.1007/978-94-007-7192-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|