1
|
Schelbert S, Maurus K, Roth S, Ott G, Kurz KS, Mogler C, Wollenberg B, Linde J, Zamo A, Anagnostopoulos I, Gramlich S, Rosenwald A, Gerhard-Hartmann E. Morphological, immunohistochemical and molecular analysis of follicular dendritic cell sarcomas: L1CAM as a new diagnostic marker. Histopathology 2025. [PMID: 40289262 DOI: 10.1111/his.15458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/23/2025] [Accepted: 04/03/2025] [Indexed: 04/30/2025]
Abstract
AIMS Follicular dendritic cell sarcoma (FDCS) is a rare neoplasm exhibiting morphological and immunophenotypical features of follicular dendritic cells. Given its rarity and broad morphological spectrum, diagnosis can be challenging. Knowledge of the molecular basis of this rare tumour is still limited. To further refine the biological and diagnostic characteristics of these neoplasms, we performed a comprehensive morphological, immunohistochemical and molecular analysis. METHODS AND RESULTS As well as histopathological and immunohistochemical analysis, we performed molecular analysis by next-generation panel sequencing of 15 tissue samples from 13 patients diagnosed with FDCS. In the histomorphological analysis of this FDCS series, we observed a morphological spectrum with a mixture of spindled and epithelioid cells (six of 13), but also cases with predominant epithelioid cytomorphology (seven of 13). We identified the L1 cell adhesion molecule (L1CAM) as a novel immunomarker of FDCS, as it was variably expressed in all cases. Sequencing led to the identification of 170 variants (classes 3, 4 and 5) in 112 genes. The most frequently detected (likely) pathogenic mutations affected NFKBIA (five of 13), leading to activation of nuclear factor kappa B (NFκB) signalling. Notably, deleterious NFKBIA mutations were only found in cases with predominant epithelioid morphology (five of seven). Furthermore, TP53 mutations were detected in two cases with epithelioid morphology and high proliferation rate, and one of these cases relapsed twice. CONCLUSIONS The morphological and genetic landscape of FDCS in this series was heterogeneous. However, in line with previous data, we identified recurrent genetic alterations affecting NFkB signalling. The expression of the adhesion molecule L1CAM might aid in the diagnosis of this uncommon neoplasia.
Collapse
Affiliation(s)
- Selina Schelbert
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Katja Maurus
- Institute of Pathology, University of Würzburg, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, Würzburg, Germany
| | - Sabine Roth
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - German Ott
- Department of Clinical Pathology, Robert-Bosch-Krankenhaus and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
| | - Katrin S Kurz
- Department of Clinical Pathology, Robert-Bosch-Krankenhaus and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
| | - Carolin Mogler
- Institute of Pathology, TUM School of Medicine and Health, Technical University of Munich (TUM), Munich, Germany
| | - Barbara Wollenberg
- Clinic for Otorhinolaryngology, Head and Neck Surgery, MRI TUM, Technical University Munich, Munich, Germany
| | - John Linde
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | - Alberto Zamo
- Institute of Pathology, University of Würzburg, Würzburg, Germany
| | | | - Susanne Gramlich
- Institute of Pathology, University of Würzburg, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, Würzburg, Germany
| | - Andreas Rosenwald
- Institute of Pathology, University of Würzburg, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, Würzburg, Germany
| | - Elena Gerhard-Hartmann
- Institute of Pathology, University of Würzburg, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, Würzburg, Germany
| |
Collapse
|
2
|
Theis T, Kumar S, Shah P, Patel M, Tadmori I, Ayala C, Tschang M, Young W, Schachner M. Depletion of Cell Adhesion Molecule L1 from Microglia and Macrophages Reduces Recovery After Spinal Cord Injury. Int J Mol Sci 2025; 26:3285. [PMID: 40244153 PMCID: PMC11989546 DOI: 10.3390/ijms26073285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 03/27/2025] [Accepted: 03/29/2025] [Indexed: 04/18/2025] Open
Abstract
The young mammalian central nervous system regenerates after spinal cord injury and recovers locomotion, whereas adult mice only show limited recovery that depends on the injury severity, genetic background, and physical therapy. At the molecular level, key regulators that contribute to recovery are cell adhesion molecules, such as L1CAM (L1). At the cell surface, L1 functions as a homotypic receptor that signal-transduces crucial functions in neuronal migration and survival, neurite outgrowth, myelination, formation of synapses, and synaptic plasticity. In the adult central nervous system, L1 is expressed only by neurons. We now show that L1 is unexpectedly also expressed by 26% microglia, freshly isolated from a 7-day-old mouse brain. At postnatal day 21, only 3% of microglia are L1-positive. Using a mouse mutant in which L1 is deleted specifically in monocytes of 10- to 14-week-old mice, functional recovery was reduced up to 4 weeks after injury at lower thoracic spinal levels. Also, NF200-immunoreactive and 5-HT-immunoreactive fibers were found decreased below the injury site as compared to wild-type mice. In conclusion, microglial cells that express L1 stimulate neurite outgrowth in vitro, improve functional recovery after spinal cord injury in adult mice, and increase fiber densities caudal to the lesion site.
Collapse
Affiliation(s)
- Thomas Theis
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Suneel Kumar
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, Piscataway, NJ 08844, USA;
| | - Pratiksha Shah
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Mukti Patel
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Iman Tadmori
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Carlos Ayala
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Monica Tschang
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Wise Young
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA; (T.T.); (P.S.); (M.P.); (I.T.); (C.A.); (M.T.); (W.Y.)
| |
Collapse
|
3
|
Yang Z, Wu H, Wang Z, Bian E, Zhao B. The role and application of small extracellular vesicles in glioma. Cancer Cell Int 2024; 24:229. [PMID: 38951882 PMCID: PMC11218314 DOI: 10.1186/s12935-024-03389-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 05/28/2024] [Indexed: 07/03/2024] Open
Abstract
Small extracellular vesicles (sEVs) are cell-derived, nanometer-sized particles enclosed by a lipid bilayer. All kinds of biological molecules, including proteins, DNA fragments, RNA, lipids, and metabolites, can be selectively loaded into sEVs and transmitted to recipient cells that are near and distant. Growing shreds of evidence show the significant biological function and the clinical significance of sEVs in cancers. Numerous recent studies have validated that sEVs play an important role in tumor progression and can be utilized to diagnose, stage, grading, and monitor early tumors. In addition, sEVs have also served as drug delivery nanocarriers and cancer vaccines. Although it is still infancy, the field of basic and translational research based on sEVs has grown rapidly. In this review, we summarize the latest research on sEVs in gliomas, including their role in the malignant biological function of gliomas, and the potential of sEVs in non-invasive diagnostic and therapeutic approaches, i.e., as nanocarriers for drug or gene delivery and cancer vaccines.
Collapse
Affiliation(s)
- Zhihao Yang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, China
- Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, Anhui Province, China
| | - HaoYuan Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, China
- Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, Anhui Province, China
| | - ZhiWei Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, China
- Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, Anhui Province, China
| | - ErBao Bian
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, China.
- Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, Anhui Province, China.
| | - Bing Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, China.
- Cerebral Vascular Disease Research Center, Anhui Medical University, Hefei, 230601, Anhui Province, China.
| |
Collapse
|
4
|
Wang M, Jin F, Tong X. From bench to bedside: The promising value of exosomes in precision medicine for CNS tumors. Heliyon 2024; 10:e32376. [PMID: 38961907 PMCID: PMC11219334 DOI: 10.1016/j.heliyon.2024.e32376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
Exosomes are naturally present extracellular vesicles (EVs) released into the surrounding body fluids upon the fusion of polycystic and plasma membranes. They facilitate intercellular communication by transporting DNA, mRNA, microRNA, long non-coding RNA, circular RNA, proteins, lipids, and nucleic acids. They contribute to the onset and progression of Central Nervous System (CNS) tumors. In addition, they can be used as biomarkers of tumor proliferation, migration, and blood vessel formation, thereby affecting the Tumor Microenvironment (TME). This paper reviews the recent advancements in the diagnosis and treatment of exosomes in various CNS tumors, the promise and challenges of exosomes as natural carriers of CNS tumors, and the therapeutic prospects of exosomes in CNS tumors. Furthermore, we hope this research can contribute to the development of more targeted and effective treatments for central nervous system tumors.
Collapse
Affiliation(s)
- Mengjie Wang
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
| | - Feng Jin
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Hospital).266042, Qingdao, Shandong, China
| | - Xiaoguang Tong
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin, 300070, China
| |
Collapse
|
5
|
Wang JW, Wang HL, Liu Q, Hu K, Yuan Q, Huang SK, Wan JH. L1CAM expression in either metastatic brain lesion or peripheral blood is correlated with peripheral platelet count in patients with brain metastases from lung cancer. Front Oncol 2022; 12:990762. [DOI: 10.3389/fonc.2022.990762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 10/06/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundSystemic immune-inflammation states across the heterogeneous population of brain metastases from lung cancer are very important, especially in the context of complex brain-immune bidirectional communication. Previous studies from our team and others have shown that the L1 cell adhesion molecule (L1CAM) is deeply involved in the aggressive phenotype, immunosuppressive tumor microenvironment (TME), and metastasis during multiple malignancies, which may lead to an unfavorable outcome. However, little is known about the relationship between the L1CAM expression and the systemic immune-inflammation macroenvironment beyond the TME in brain metastases from lung cancer.MethodsTwo cohorts of patients with brain metastases from lung cancer admitted to the National Cancer Center, Cancer Hospital of Chinese Academy of Medical Sciences, were studied in the present research. The L1CAM expression in cranial metastatic lesions by immunohistochemistry was explored in patients treated with neurosurgical resection, whereas the L1CAM expression in peripheral blood by ELISA was tested in patients treated with non-surgical antitumor management. Furthermore, based on peripheral blood cell counts in the CBC test, six systemic immune-inflammation biomarkers [neutrophil count, lymphocyte count, platelet count, systemic immune-inflammation index (SII), neutrophil-to-lymphocyte ratio (NLR), and platelet-to-lymphocyte ratio] were calculated. Then, the relationship between the L1CAM expression and these systemic immune-inflammation biomarkers was analyzed. In addition, these systemic immune-inflammation biomarkers were also used to compare the systemic immune-inflammation states in two cohorts of patients with brain metastases from lung cancer.ResultsPositive L1CAM expressions in the metastatic brain lesions were accompanied with significantly increased peripheral platelet counts in patients treated with neurosurgical tumor resection (P < 0.05). Similarly, in patients treated with non-surgical antitumor management, L1CAM expressions in the peripheral blood were positively correlated with peripheral platelet counts (P < 0.05). In addition, patients prepared for neurosurgical tumor resection were presented with poorer systemic immune-inflammation states in comparison with the one with non-surgical antitumor management, which was characterized by a significant increase in peripheral neutrophil counts (P < 0.01), SII (P < 0.05), and NLR (P < 0.05) levels.ConclusionThe L1CAM expression in either the metastatic brain lesion or peripheral blood is positively correlated with the peripheral platelet count in patients with brain metastases from lung cancer. In addition, brain metastases that are prepared for neurosurgical tumor resection show poor systemic immune-inflammation states.
Collapse
|
6
|
p53, Pirh2, and L1CAM as Promising Prognostic Biomarkers of Endometrial Carcinoma: An Immunohistochemical and Genetic Study. Appl Immunohistochem Mol Morphol 2022; 30:713-725. [PMID: 36251972 DOI: 10.1097/pai.0000000000001073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 09/12/2022] [Indexed: 11/25/2022]
Abstract
Endometrial cancer (EC) is the most common gynecologic cancer and the current methods for the prediction of its prognosis and treatment response are unfortunately suboptimal. In this study, we evaluated the prognostic value of p53, Pirh2, and L1CAM in 60 cases of EC using immunohistochemistry (IHC) and polymerase chain reaction. TP53 missense mutations result in nuclear accumulation of p53 protein that can be detected as overexpression by IHC. This is in the form of diffuse strong nuclear positivity involving at least at least >50% of the tumor cells as a whole or if >50% of the tumor cells of a discrete geographical areas. Abnormal p53 IHC expression was expressed in 33.3% of the cases and significantly associated with the tumor grade, myometrial invasion (MI), lymphovascular invasion (LVSI), nodal metastasis, and FIGO stage, and the advanced European Society for Medical Oncology (ESMO) risk groups (P<0.001 for each). High IHC Pirh2 expression was noted in 58.3% of the cases, and significantly associated with MI, LVSI, nodal metastasis, FIGO stage, and high-risk group (P<0.001, P=0.011, P=0.010, P=0.024, P=0.005, respectively). There was a significant upregulation of Pirh2 mRNA expression in EC specimens as compared with the control adjacent tissues (P=0.001). Upregulated Pirh2 mRNA expression had a significant association with Pirh2 immunostaining, tumor grade, tumor stage, MI, lymph node involvement, LVSI, and relapse (P<0.001 for each). Positive L1CAM immunoexpression was noted in 26.7% and was significantly associated with grade, MI, LVSI, nodal metastasis, FIGO stage, and high-risk group (P=0.003, P=0.023, P=0.003, P<0.001, P<0.001, P=0.002, respectively). Analysis of follow-up period revealed that EC with abnormal p53 IHC expression, high pirh2 and positive L1CAM expression exhibited a potent relation with tumor relapse, shorter overall survival and disease-specific survival (P<0.001 for each). Mutant p53, high Pirh2, and L1CAM-positive EC are highly aggressive tumors with a shortened survival rate, dismal outcome, and high risk of relapse after the standard protocol of therapy.
Collapse
|
7
|
Wu J, Wu C, Li G. Identifying common driver modules by equilibrating coverage and mutual exclusivity across pan-cancer data. Neurocomputing 2022. [DOI: 10.1016/j.neucom.2022.04.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
8
|
Novel therapeutics and drug-delivery approaches in the modulation of glioblastoma stem cell resistance. Ther Deliv 2022; 13:249-273. [PMID: 35615860 DOI: 10.4155/tde-2021-0086] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is a deadly malignancy with a poor prognosis. An important factor contributing to GBM recurrence is high resistance of GBM cancer stem cells (GSCs). While temozolomide (TMZ), has been shown to consistently extend survival, GSCs grow resistant to TMZ through upregulation of DNA damage repair mechanisms and avoidance of apoptosis. Since a single-drug approach has failed to significantly alter prognosis in the past 15 years, unique approaches such as multidrug combination therapy together with distinctive targeted drug-delivery approaches against cancer stem cells are needed. In this review, a rationale for multidrug therapy using a targeted nanotechnology approach that preferentially target GSCs is proposed with discussion and examples of drugs, nanomedicine delivery systems, and targeting moieties.
Collapse
|
9
|
Wang JW, Wang SQ, Wu ZY, Liu Q, Yuan Q, Cai HQ, Wan JH. L1 cell adhesion molecule high expression is associated with poor prognosis in surgically resected brain metastases from lung adenocarcinoma. Clinics (Sao Paulo) 2022; 77:100040. [PMID: 35525225 PMCID: PMC9092252 DOI: 10.1016/j.clinsp.2022.100040] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/19/2022] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVES Accurate prognosis assessment across the heterogeneous population of brain metastases is very important, which may facilitate clinical decision-making and appropriate stratification of future clinical trials. Previous studies have shown the L1 Cell Adhesion Molecule (L1CAM) is potentially involved in human malignancies of multiple different samples and unfavorable survival. However, no data of L1CAM are available for the brain metastases from lung adenocarcinoma, especially for the one with neurosurgical resection. METHOD The authors investigated the L1CAM expression in cranial metastatic lesions for patients with brain metastases from lung adenocarcinoma after neurosurgical resection using tissue microarrays that were obtained from the Department of Neurosurgery at the Cancer Hospital of the Chinese Academy of Medical Sciences. Furthermore, the relationship between L1CAM expression and clinic-pathological parameters, including overall survival time, was analyzed to assess the prognostic value of L1CAM. RESULTS L1CAM high expression was found in 62.30% of brain metastases from lung adenocarcinoma and significantly correlated with brain metastasis number (p = 0.028) and Lung-molGPA score (p = 0.042). Moreover, L1CAM expression was an independent predictor of survival for brain metastases after neurosurgical resection in a multivariate analysis. Patients with L1CAM high expression had unfavorable overall survival time (p = 0.016). In addition, the multivariate analysis also showed age and extracranial transfer were also the independent prognostic factors for this type of patient with brain metastases. CONCLUSIONS A subset of brain metastases from lung adenocarcinoma aberrantly expresses L1CAM. L1CAM is a novel independent prognostic factor for brain metastasis from lung adenocarcinoma after neurosurgical resection.
Collapse
Affiliation(s)
- Jia-Wei Wang
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, PR China
| | - Song-Quan Wang
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, PR China
| | - Zhuo-Yi Wu
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, PR China
| | - Qi Liu
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, PR China
| | - Qing Yuan
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, PR China.
| | - Hong-Qing Cai
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, PR China.
| | - Jing-Hai Wan
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, PR China.
| |
Collapse
|
10
|
Marques P, Barry S, Carlsen E, Collier D, Ronaldson A, Grieve J, Dorward N, Mendoza N, Nair R, Muquit S, Grossman AB, Korbonits M. The expression of neural cell adhesion molecule and the microenvironment of pituitary neuroendocrine tumours. J Neuroendocrinol 2021; 33:e13052. [PMID: 34708902 DOI: 10.1111/jne.13052] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 09/09/2021] [Accepted: 10/06/2021] [Indexed: 12/19/2022]
Abstract
The neural cell adhesion molecule (NCAM) has previously been studied in pituitary neuroendocrine tumours (PitNETs), but its role in tumour biology and aggressiveness remains controversial, and its relationship with the tumour microenvironment remains unknown. We aimed to characterise NCAM expression in PitNETs, to correlate this with clinico-pathological features, and to assess the role of various microenvironment components on NCAM expression. NCAM and immune cells were investigated by immunohistochemistry in 16 human non-functioning-PitNETs (NF-PitNETs) and eight somatotrophinomas, including macrophages (CD68, CD163, HLA-DR), cytotoxic (CD8) and T helper (CD4) lymphocytes, regulatory T cells (FOXP3), B cells (CD20), and neutrophils (neutrophil elastase). Five normal pituitaries were included for comparison. The cytokine secretome from these PitNETs and from PitNET-derived tumour-associated fibroblasts (TAFs) were assessed on culture supernatants using a multiplex immunoassay panel. There were no significant NCAM expression differences between PitNETs and normal pituitary, and no difference between types of pituitary tumours (NF-PitNETs vs. somatotrophinomas). There was no association between NCAM expression and different clinico-pathological features, including cavernous sinus invasion and Ki-67, nor with serum hormone levels. NCAM immunoreactivity correlated negatively with PitNET-derived CXCL10 (rho = -0.417; p = .042) and CX3CL1 (rho = -0.423; p = .040) levels. NCAM immunoreactivity was negatively correlated with TAF-derived fibroblast growth factor (FGF)-2 (rho = -0.632; p = .009), but not with other TAF-derived cytokines. Within the PitNET cohort, there were no correlations between NCAM immunoreactivity and immune infiltrates or ratios, although, within NF-PitNETs, NCAM expression was higher in tumours with more FOXP3+ cells. NCAM expression does not differ between PitNETs and normal pituitary, and does not appear to relate to tumour invasiveness or proliferation. However, our data suggest a possible role for cytokines in the modulation of NCAM expression in PitNETs, particularly CXCL10, CX3CL1 and FGF-2, but not for immune cell infiltrates.
Collapse
Affiliation(s)
- Pedro Marques
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, UK
- Department of Endocrinology, Hospital de Santa Maria, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
| | - Sayka Barry
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, UK
| | | | - David Collier
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Amy Ronaldson
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Joan Grieve
- The National Hospital for Neurology and Neurosurgery, UCLH, NHS Trust, London, UK
| | - Neil Dorward
- The National Hospital for Neurology and Neurosurgery, UCLH, NHS Trust, London, UK
| | - Nigel Mendoza
- Department of Neurosurgery, Charing Cross Hospital, Imperial College, London, UK
| | - Ramesh Nair
- Department of Neurosurgery, Charing Cross Hospital, Imperial College, London, UK
| | - Samiul Muquit
- Department of Neurosurgery, Derriford Hospital, Plymouth, UK
| | - Ashley B Grossman
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Márta Korbonits
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, William Harvey Research Institute, Queen Mary University of London, London, UK
| |
Collapse
|
11
|
Zeiter D, Vlajnic T, Schötzau A, Heinzelmann-Schwarz V, Montavon C. L1CAM is not a reliable predictor for lymph node metastases in endometrial cancer, but L1CAM positive patients benefit from radiotherapy. J Cancer 2021; 12:6401-6410. [PMID: 34659530 PMCID: PMC8489141 DOI: 10.7150/jca.59283] [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: 02/10/2021] [Accepted: 07/28/2021] [Indexed: 12/24/2022] Open
Abstract
Purpose: Several studies evidenced the potential of L1CAM as a prognostic marker in endometrial cancer. The aim of this study was to investigate whether L1CAM can predict lymph node metastasis and could therefore be used preoperatively to identify patients with low to high-intermediate risk endometrial cancer who would profit from a lymphadenectomy and an adjuvant treatment. To avoid unnecessary morbidity, de-escalating strategies are still required. Methods: Immunohistochemistry for L1CAM was performed on curettage or hysterectomy specimens from 212 patients diagnosed with endometrial cancer who were treated at the University Hospital Basel during 2011-2019. L1CAM expression was correlated with clinicopathological features such as histological subtype, FIGO stage, lymph node metastasis, lymphadenectomy, adjuvant treatment and outcome. Results: Using a cut off ≥10%, L1CAM was positive in 41/212 patients (19.3%) and negative in 171/212 patients (80.7%). L1CAM was associated with high-risk features such as non-endometrioid histology, high tumour grade, and high FIGO stage. There was no significant correlation between L1CAM expression and lymph node metastasis. However, patients with L1CAM positive tumours showed improved disease-specific survival if treated with adjuvant radiotherapy. Conclusion: Although L1CAM expression pointed towards aggressive tumour biology, preoperative L1CAM analysis did not add any substantial predictive information regarding lymph node metastasis in low to high-intermediate risk groups. Therefore, L1CAM status is not suitable to tailor the surgical algorithm for lymph node staging. Nevertheless, our results suggest that L1CAM could be used as a predictive biomarker to select patients who may benefit the most from adjuvant radiotherapy.
Collapse
Affiliation(s)
- Deborah Zeiter
- Gynaecological Cancer Centre, Hospital for Women, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Tatjana Vlajnic
- Institute of Pathology, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Andreas Schötzau
- Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Viola Heinzelmann-Schwarz
- Gynaecological Cancer Centre, Hospital for Women, University Hospital Basel and University of Basel, 4031 Basel, Switzerland.,Ovarian Cancer Research, Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Céline Montavon
- Gynaecological Cancer Centre, Hospital for Women, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| |
Collapse
|
12
|
Dräger O, Metz K, Busch M, Dünker N. Role of L1CAM in retinoblastoma tumorigenesis: identification of novel therapeutic targets. Mol Oncol 2021; 16:957-981. [PMID: 34228897 PMCID: PMC8847994 DOI: 10.1002/1878-0261.13054] [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: 03/11/2021] [Revised: 05/11/2021] [Accepted: 07/05/2021] [Indexed: 11/08/2022] Open
Abstract
The study presented focuses on the role of the neuronal cell adhesion molecule L1 cell adhesion molecule (L1CAM) in retinoblastoma (RB), the most common malignant intraocular childhood tumor. L1CAM is differentially expressed in a variety of human cancers and has been suggested as a promising therapeutic target. We likewise observed differential expression patterns for L1CAM in RB cell lines and patient samples. The two proteases involved in ectodomain shedding of L1CAM (L1CAM sheddases: ADAM10 and ADAM17) were likewise differentially expressed in the RB cell lines investigated, and an involvement in L1CAM processing in RB cells could be verified. We also identified ezrin, galectin-3, and fibroblast growth factor basic as L1CAM signaling target genes in RB cells. Lentiviral L1CAM knockdown induced apoptosis and reduced cell viability, proliferation, growth, and colony formation capacity of RB cells, whereas L1CAM-overexpressing RB cells displayed the opposite effects. Chicken chorioallantoic membrane assays revealed that L1CAM depletion decreases the tumorigenic and migration potential of RB cells in vivo. Moreover, L1CAM depletion decreased viability and tumor growth of etoposide-resistant RB cell lines upon etoposide treatment in vitro and in vivo. Thus, L1CAM and its processing sheddases are potential novel targets for future therapeutic RB approaches.
Collapse
Affiliation(s)
- Oliver Dräger
- Institute of Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, Medical Faculty, Germany
| | - Klaus Metz
- Institute of Pathology, University of Duisburg-Essen, Medical Faculty, Germany
| | - Maike Busch
- Institute of Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, Medical Faculty, Germany
| | - Nicole Dünker
- Institute of Anatomy II, Department of Neuroanatomy, University of Duisburg-Essen, Medical Faculty, Germany
| |
Collapse
|
13
|
Nam JK, Kim AR, Choi SH, Kim JH, Choi KJ, Cho S, Lee JW, Cho HJ, Kwon YW, Cho J, Kim KS, Kim J, Lee HJ, Lee TS, Bae S, Hong HJ, Lee YJ. An antibody against L1 cell adhesion molecule inhibits cardiotoxicity by regulating persistent DNA damage. Nat Commun 2021; 12:3279. [PMID: 34078883 PMCID: PMC8172563 DOI: 10.1038/s41467-021-23478-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 05/03/2021] [Indexed: 02/04/2023] Open
Abstract
Targeting the molecular pathways underlying the cardiotoxicity associated with thoracic irradiation and doxorubicin (Dox) could reduce the morbidity and mortality associated with these anticancer treatments. Here, we find that vascular endothelial cells (ECs) with persistent DNA damage induced by irradiation and Dox treatment exhibit a fibrotic phenotype (endothelial-mesenchymal transition, EndMT) correlating with the colocalization of L1CAM and persistent DNA damage foci. We demonstrate that treatment with the anti-L1CAM antibody Ab417 decreases L1CAM overexpression and nuclear translocation and persistent DNA damage foci. We show that in whole-heart-irradiated mice, EC-specific p53 deletion increases vascular fibrosis and the colocalization of L1CAM and DNA damage foci, while Ab417 attenuates these effects. We also demonstrate that Ab417 prevents cardiac dysfunction-related decrease in fractional shortening and prolongs survival after whole-heart irradiation or Dox treatment. We show that cardiomyopathy patient-derived cardiovascular ECs with persistent DNA damage show upregulated L1CAM and EndMT, indicating clinical applicability of Ab417. We conclude that controlling vascular DNA damage by inhibiting nuclear L1CAM translocation might effectively prevent anticancer therapy-associated cardiotoxicity.
Collapse
Affiliation(s)
- Jae-Kyung Nam
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
- Laboratory of Biochemistry, Division of Life Sciences, Korea University, Seoul, Korea
| | - A-Ram Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Seo-Hyun Choi
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
- Department of Surgery, Memorial Sloan Kettering Cancer Center, NY, USA
| | - Ji-Hee Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
- Laboratory of Biochemistry, Division of Life Sciences, Korea University, Seoul, Korea
| | - Kyu Jin Choi
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Seulki Cho
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, Korea
| | - Jae Won Lee
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Hyun-Jai Cho
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Yoo-Wook Kwon
- Cardiovascular Center & Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Jaeho Cho
- Department of Radiation Oncology, Yonsei University College of Medicine, Seoul, Korea
| | - Kwang Seok Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Joon Kim
- Laboratory of Biochemistry, Division of Life Sciences, Korea University, Seoul, Korea
| | - Hae-June Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Tae Sup Lee
- Division of RI Convergence Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Sangwoo Bae
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Hyo Jeong Hong
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, Korea.
- Scripps Korea Antibody Institute, Chuncheon, Korea.
| | - Yoon-Jin Lee
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea.
| |
Collapse
|
14
|
Del Favero G, Zeugswetter M, Kiss E, Marko D. Endoplasmic Reticulum Adaptation and Autophagic Competence Shape Response to Fluid Shear Stress in T24 Bladder Cancer Cells. Front Pharmacol 2021; 12:647350. [PMID: 34012396 PMCID: PMC8126838 DOI: 10.3389/fphar.2021.647350] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 02/17/2021] [Indexed: 12/26/2022] Open
Abstract
Accumulation of xenobiotics and waste metabolites in the urinary bladder is constantly accompanied by shear stress originating from the movement of the luminal fluids. Hence, both chemical and physical cues constantly modulate the cellular response in health and disease. In line, bladder cells have to maintain elevated mechanosensory competence together with chemical stress response adaptation potential. However, much of the molecular mechanisms sustaining this plasticity is currently unknown. Taking this as a starting point, we investigated the response of T24 urinary bladder cancer cells to shear stress comparing morphology to functional performance. T24 cells responded to the shear stress protocol (flow speed of 0.03 ml/min, 3 h) by significantly increasing their surface area. When exposed to deoxynivalenol-3-sulfate (DON-3-Sulf), bladder cells increased this response in a concentration-dependent manner (0.1-1 µM). DON-3-Sulf is a urinary metabolite of a very common food contaminant mycotoxin (deoxynivalenol, DON) and was already described to enhance proliferation of cancer cells. Incubation with DON-3-Sulf also caused the enlargement of the endoplasmic reticulum (ER), decreased the lysosomal movement, and increased the formation of actin stress fibers. Similar remodeling of the endoplasmic reticulum and area spread after shear stress were observed upon incubation with the autophagy activator rapamycin (1-100 nM). Performance of experiments in the presence of chloroquine (chloroquine, 30 μM) further contributed to shed light on the mechanistic link between adaptation to the biomechanical stimulation and ER stress response. At the molecular level, we observed that ER reshaping was linked to actin organization, with the two components mutually regulating each other. Indeed, we identified in the ER stress-cytoskeletal rearrangement an important axis defining the physical/chemical response potential of bladder cells and created a workflow for further investigation of urinary metabolites, food constituents, and contaminants, as well as for pharmacological profiling.
Collapse
Affiliation(s)
- Giorgia Del Favero
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria.,Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Michael Zeugswetter
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Endre Kiss
- Core Facility Multimodal Imaging, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - Doris Marko
- Department of Food Chemistry and Toxicology, Faculty of Chemistry, University of Vienna, Vienna, Austria
| |
Collapse
|
15
|
Guo M, Gong H, Nie D, Li Z. High L1CAM expression predicts poor prognosis of patients with endometrial cancer: A systematic review and meta-analysis. Medicine (Baltimore) 2021; 100:e25330. [PMID: 33787629 PMCID: PMC8021316 DOI: 10.1097/md.0000000000025330] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 02/19/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUD Previous studies have reported that the levels of L1 cell adhesion molecule (L1CAM) indicate poor prognosis of patients with various solid tumors. However, the prognostic significance of L1CAM in endometrial cancer has remained controversial. Herein, we conducted a systematic review and meta-analysis to evaluate the prognostic value of L1CAM in endometrial cancer. METHODS All studies related to the association between L1CAM expression and clinical characteristics of endometrial cancer were identified by searching the PubMed, MEDLINE, EMBASE, and Web of Science databases. Primary outcomes of the meta-analysis were the hazard ratios (HRs) for overall survival (OS) and disease-free survival (DFS). Secondary outcomes were odds ratios (ORs) for clinicopathological characteristics. Publication bias and sensitivity analysis were conducted to ensure reliability of the results. RESULTS Overall, 17 studies encompassing 7146 patients were eligible for the meta-analysis. Results showed L1CAM overexpression to be significantly associated with decreased overall survival (HR = 2.87, 95% CI; 1.81-4.55, P < .001) and disease-free survival (HR = 3.32, 95% CI; 1.99-5.55, P < .001) in patients with endometrial cancer. High L1CAM expression was also related to adverse clinicopathological characteristics. CONCLUSION This systematic review demonstrated that high L1CAM expression is correlated with poor survival outcomes and adverse clinicopathological parameters in patients with endometrial cancer.
Collapse
Affiliation(s)
- Min Guo
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University
- Department of Gynecology, University Hospital, University of Electronic Science and Technology of China
| | - Han Gong
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University
| | - Dan Nie
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Southwest Medical University
| | - Zhengyu Li
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University
| |
Collapse
|
16
|
Li J, Jiang ZZ, Li YY, Tang WT, Yin J, Long XP. LncRNA CHRF promotes TGF-β1 induced EMT in alveolar epithelial cells by inhibiting miR-146a up-regulating L1CAM expression. Exp Lung Res 2021; 47:198-209. [PMID: 33754922 DOI: 10.1080/01902148.2021.1891354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
PURPOSE Idiopathic pulmonary fibrosis (IPF) is a type of progressive lung fibrosis disease. The survival time of diagnosed IPF patients is often only 2 years. Currently much evidence showed that the epithelial-mesenchymal transition (EMT) process is the main cause of the occurrence and development of IPF. LncRNA cardiac hypertrophy related factor (CHRF) was reported to be related with IPF development. Here we explored the functions and regulatory mechanisms of CHRF on EMT in IPF. MATERIALS AND METHODS A549 cells were treated with transforming growth factor-β1 (TGF-β1) for 48 h to construct IPF cell model. CHRF and miR-146a expression were quantified using qPCR. The expression of L1 cell adhesion molecule (L1CAM) and EMT related indicators (E-cadherin, Vimentin, Slug and N-cadherin) were detected by qPCR and western blot. Dual luciferase reporter experiment was conducted to prove the molecular interaction of miR-146a and L1CAM, as well as CHRF and miR-146a. RESULTS CHRF and L1CAM expression were significantly upregulated and promoted the EMT process in A549 after treatment of TGF-β1. MiR-146a was obviously down-regulated, and knockdown of CHRF inhibited the EMT process by up-regulating miR-146a, in A549 after treatment of TGF-β1. Meanwhile, overexpression of miR-146a inhibited EMT process via targeting L1CAM. In addition, L1CAM overexpression eliminated the inhibitory effect of sh-CHRF on the EMT process. CONCLUSIONS These results provided evidence that CHRF promoted EMT process in A549 after treatment of TGF-β1, which proposed a new insight for depth understanding the pathological mechanisms of IPF.
Collapse
Affiliation(s)
- Ju Li
- Graduate School of University of South China, Hengyang, Hunan Province, China.,Department of Emergency, Third People's Hospital of Longgang District, Shenzhen, Guangdong Province, China
| | - Zhen-Zhu Jiang
- Graduate School of University of South China, Hengyang, Hunan Province, China.,Department of Emergency, Third People's Hospital of Longgang District, Shenzhen, Guangdong Province, China
| | - You-You Li
- Graduate School of University of South China, Hengyang, Hunan Province, China.,Pulmonary and Critical Care Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan Province, China
| | - Wen-Ting Tang
- Graduate School of University of South China, Hengyang, Hunan Province, China.,Pulmonary and Critical Care Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan Province, China
| | - Jing Yin
- Graduate School of University of South China, Hengyang, Hunan Province, China.,Pulmonary and Critical Care Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan Province, China
| | - Xiao-Ping Long
- Pulmonary and Critical Care Medicine, The First Affiliated Hospital of University of South China, Hengyang, Hunan Province, China
| |
Collapse
|
17
|
Ghaemmaghami AB, Mahjoubin-Tehran M, Movahedpour A, Morshedi K, Sheida A, Taghavi SP, Mirzaei H, Hamblin MR. Role of exosomes in malignant glioma: microRNAs and proteins in pathogenesis and diagnosis. Cell Commun Signal 2020; 18:120. [PMID: 32746854 PMCID: PMC7397575 DOI: 10.1186/s12964-020-00623-9] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023] Open
Abstract
Malignant gliomas are the most common and deadly type of central nervous system tumors. Despite some advances in treatment, the mean survival time remains only about 1.25 years. Even after surgery, radiotherapy and chemotherapy, gliomas still have a poor prognosis. Exosomes are the most common type of extracellular vesicles with a size range of 30 to 100 nm, and can act as carriers of proteins, RNAs, and other bioactive molecules. Exosomes play a key role in tumorigenesis and resistance to chemotherapy or radiation. Recent evidence has shown that exosomal microRNAs (miRNAs) can be detected in the extracellular microenvironment, and can also be transferred from cell to cell via exosome secretion and uptake. Therefore, many recent studies have focused on exosomal miRNAs as important cellular regulators in various physiological and pathological conditions. A variety of exosomal miRNAs have been implicated in the initiation and progression of gliomas, by activating and/or inhibiting different signaling pathways. Exosomal miRNAs could be used as therapeutic agents to modulate different biological processes in gliomas. Exosomal miRNAs derived from mesenchymal stem cells could also be used for glioma treatment. The present review summarizes the exosomal miRNAs that have been implicated in the pathogenesis, diagnosis and treatment of gliomas. Moreover, exosomal proteins could also be involved in glioma pathogenesis. Exosomal miRNAs and proteins could also serve as non-invasive biomarkers for prognosis and disease monitoring. Video Abstract.
Collapse
Affiliation(s)
- Amir B. Ghaemmaghami
- grid.17063.330000 0001 2157 2938Department of Psychology, Behaviour, Genetics and Neurobiology Program, University of Toronto, Toronto, Canada
| | - Maryam Mahjoubin-Tehran
- grid.411583.a0000 0001 2198 6209Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran ,grid.411583.a0000 0001 2198 6209Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ahmad Movahedpour
- grid.412571.40000 0000 8819 4698Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Student research committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Korosh Morshedi
- grid.444768.d0000 0004 0612 1049School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Amirhossein Sheida
- grid.444768.d0000 0004 0612 1049School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Pouya Taghavi
- grid.444768.d0000 0004 0612 1049School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Mirzaei
- grid.444768.d0000 0004 0612 1049Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Michael R. Hamblin
- grid.38142.3c000000041936754XWellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA ,grid.412988.e0000 0001 0109 131XLaser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, Johannesburg, 2028 South Africa
| |
Collapse
|
18
|
Chu LY, Peng YH, Yang T, Fang WK, Hong CQ, Huang LS, Xu LY, Li EM, Xu YW, Xie JJ. Circulating Levels of L1-cell Adhesion Molecule as a Serum Biomarker for Early Detection of Gastric Cancer and Esophagogastric Junction Adenocarcinoma. J Cancer 2020; 11:5395-5402. [PMID: 32742486 PMCID: PMC7391208 DOI: 10.7150/jca.41100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 04/21/2020] [Indexed: 02/05/2023] Open
Abstract
Background: Low serum L1 cell adhesion molecule (L1CAM) has been found in several malignant tumors. Here, we aimed to evaluate the diagnostic potential for serum L1CAM in patients with gastric cancers (GC) and esophagogastric junction adenocarcinoma (EJA). Methods: Enzyme-linked immunosorbent assay (ELISA) was carried out to detect L1CAM level in sera of 148 GC patients, 59 EJA patients and 148 healthy controls. Receiver operating characteristics (ROC) was employed to evaluate diagnostic accuracy. Results: The concentrations of serum L1CAM were significantly lower in GC and EJA than those in healthy controls (P<0.001). Detection of L1CAM provided a sensitivity of 83.1%, a specificity of 62.2%, and an area under the curve (AUC) of 0.769 (95% CI: 0.715-0.823) in diagnosing GC, and a sensitivity of 66.1%, a specificity of 62.2%, and an AUC of 0.672 (95% CI: 0.590-0.755) in diagnosing EJA. Similar results were observed in the diagnosis of early-stage GC (0.681 (95%CI: 0.596-0.766)) and early-stage EJA (0.674 (95%CI: 0.528-0.820)). Analysis of clinical data showed that the levels of L1CAM were significantly associated with lymph node metastasis in GC (P<0.05). Conclusions: Our study showed that serum L1CAM might be a diagnostic biomarker for GC and EJA.
Collapse
Affiliation(s)
- Ling-Yu Chu
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
| | - Yu-Hui Peng
- Department of Clinical Laboratory Medicine, the Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Tian Yang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Wang-Kai Fang
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
| | - Chao-Qun Hong
- Department of Oncological Laboratory Research, the Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Li-Sheng Huang
- Department of Radiation Oncology, the Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Li-Yan Xu
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, China
| | - En-Min Li
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
| | - Yi-Wei Xu
- Department of Clinical Laboratory Medicine, the Cancer Hospital of Shantou University Medical College, Shantou, China
| | - Jian-Jun Xie
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
| |
Collapse
|
19
|
LIN28B regulates transcription and potentiates MYCN-induced neuroblastoma through binding to ZNF143 at target gene promotors. Proc Natl Acad Sci U S A 2020; 117:16516-16526. [PMID: 32601179 PMCID: PMC7368283 DOI: 10.1073/pnas.1922692117] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
LIN28B is well known as a RNA-binding protein and a suppressor of microRNA biogenesis by selectively blocking the processing of let-7 precursors. However, little is known about let-7–independent roles of LIN28B. Here, we show that LIN28B is recruited to active promoters by binding to the zinc-finger transcription factor ZNF143. LIN28B acts as a cofactor to upregulate expression of a subset of downstream target genes that are essential for neuroblastoma cell survival and migration. Our paper reveals an unexpected role of LIN28B in transcriptional regulation that is independent of let-7 during neuroblastoma pathogenesis. LIN28B is highly expressed in neuroblastoma and promotes tumorigenesis, at least, in part, through inhibition of let-7 microRNA biogenesis. Here, we report that overexpression of either wild-type (WT) LIN28B or a LIN28B mutant that is unable to inhibit let-7 processing increases the penetrance of MYCN-induced neuroblastoma, potentiates the invasion and migration of transformed sympathetic neuroblasts, and drives distant metastases in vivo. Genome-wide chromatin immunoprecipitation coupled with massively parallel DNA sequencing (ChIP-seq) and coimmunoprecipitation experiments show that LIN28B binds active gene promoters in neuroblastoma cells through protein–protein interaction with the sequence-specific zinc-finger transcription factor ZNF143 and activates the expression of downstream targets, including transcription factors forming the adrenergic core regulatory circuitry that controls the malignant cell state in neuroblastoma as well as GSK3B and L1CAM that are involved in neuronal cell adhesion and migration. These findings reveal an unexpected let-7–independent function of LIN28B in transcriptional regulation during neuroblastoma pathogenesis.
Collapse
|
20
|
Karstens KF, Bellon E, Polonski A, Wolters-Eisfeld G, Melling N, Reeh M, Izbicki JR, Tachezy M. Expression and serum levels of the neural cell adhesion molecule L1-like protein (CHL1) in gastrointestinal stroma tumors (GIST) and its prognostic power. Oncotarget 2020; 11:1131-1140. [PMID: 32284790 PMCID: PMC7138165 DOI: 10.18632/oncotarget.27525] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/03/2020] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Diagnosis of gastrointestinal stroma tumors (GIST) is based on the histological evaluation of tissue specimens. Reliable systemic biomarkers are lacking. We investigated the local expression of the neural cell adhesion molecule L1-like protein (CHL1) in GIST and determined whether soluble CHL1 proteoforms could serve as systemic biomarkers. MATERIAL AND METHODS Expression of CHL1 was analyzed in primary tumor specimens and metastases. 58 GIST specimens were immunohistochemically stained for CHL1 on a tissue microarray (TMA). Systemic CHL1 levels were measured in sera derived from 102 GIST patients and 91 healthy controls by ELISA. Results were statistically correlated with clinicopathological parameters. RESULTS CHL1 expression was detected in GIST specimens. Reduced tissue expression was significantly associated with advanced UICC stages (p = 0.036) and unfavorable tumor localization (p = 0.001). CHL1 serum levels are significantly elevated in GIST patients (p < 0.010). Elevated CHL1 levels were significantly associated with larger tumors (p = 0.023), advanced UICC stage (p = 0.021), and an increased Fletcher score (p = 0.041). Moreover, patients with a higher CHL1 serum levels displayed a significantly shortened recurrence free survival independent of other clinicopathological variables. CONCLUSION Local CHL1 expression and serum CHL1 levels show a reverse prognostic behavior, highlighting the relevance of proteolytic shedding of the molecule. The results of the study indicate a potential role of serum CHL1 as a diagnostic and prognostic marker in GIST.
Collapse
Affiliation(s)
- Karl-Frederick Karstens
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendor, Hamburg, Germany
| | - Eugen Bellon
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendor, Hamburg, Germany
| | - Adam Polonski
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendor, Hamburg, Germany
| | - Gerrit Wolters-Eisfeld
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendor, Hamburg, Germany
| | - Nathaniel Melling
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendor, Hamburg, Germany
| | - Matthias Reeh
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendor, Hamburg, Germany
| | - Jakob R Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendor, Hamburg, Germany
| | - Michael Tachezy
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg Eppendor, Hamburg, Germany
| |
Collapse
|
21
|
Hötzel J, Melling N, Müller J, Polonski A, Wolters-Eisfeld G, Izbicki JR, Karstens KF, Tachezy M. Protein expression of close homologue of L1 (CHL1) is a marker for overall survival in non-small cell lung cancer (NSCLC). J Cancer Res Clin Oncol 2019; 145:2285-2292. [PMID: 31372722 DOI: 10.1007/s00432-019-02989-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 07/24/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND The cell adhesion molecule close homologue of L1 (CHL1) is a potential tumour suppressor and was recently detected in non-small cell lung cancer (NSCLC) specimens. The expression pattern, prognostic, and functional role of CHL1 in NSCLCs is unknown. METHODS We evaluated the protein expression of CHL1 by immunohistochemistry in 2161 NSCLC patients based on a tissue microarray. The results were correlated with clinical, histopathological, and patient survival data (Chi square test, t test, and log-rank test, respectively). A multivariate analysis (Cox regression) was performed to validate its impact on patients' survival. RESULTS CHL1 was expressed in NSCLC patients and was significantly overexpressed in lung adenocarcinomas and squamous cell carcinomas compared to neuroendocrine and large cell carcinomas of the lung (p < 0.001). CHL1 expression was associated with the T stage in adenocarcinomas (p = 0.011) and with metastatic lymph node status and UICC stage in squamous cell carcinomas (p = 0.034 and p = 0.035, respectively). Increased CHL1 expression was associated with improved survival in univariate (p = 0.031) and multivariate analyses (odds ratio 0.797, 95% confidence interval 0.677-0.939, p = 0.007). CONCLUSION The prognostic significance of CHL1 makes it a potential prognostic and therapeutic target and underlines its role as a tumour suppressor. Further validation studies and functional analyses are needed to investigate its potential role in tumourigenesis and dissemination.
Collapse
Affiliation(s)
- Jenny Hötzel
- Department of General, Visceral and Thoracic Surgery, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Nathaniel Melling
- Department of General, Visceral and Thoracic Surgery, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Julia Müller
- Institute of Pathology, Municipal Hospital Lüneburg, Lüneburg, Germany
| | - Adam Polonski
- Department of General, Visceral and Thoracic Surgery, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Gerrit Wolters-Eisfeld
- Department of General, Visceral and Thoracic Surgery, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Jakob R Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| | - Karl-F Karstens
- Department of General, Visceral and Thoracic Surgery, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany.
| | - Michael Tachezy
- Department of General, Visceral and Thoracic Surgery, University Medical Centre Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Germany
| |
Collapse
|
22
|
Exosomal L1CAM Stimulates Glioblastoma Cell Motility, Proliferation, and Invasiveness. Int J Mol Sci 2019; 20:ijms20163982. [PMID: 31426278 PMCID: PMC6720723 DOI: 10.3390/ijms20163982] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 08/10/2019] [Accepted: 08/12/2019] [Indexed: 12/21/2022] Open
Abstract
Immunoglobulin superfamily protein L1CAM (L1, CD171) normally facilitates neuronal migration, differentiation, and axon guidance during development. Many types of cancers, including glioblastoma (GBM), also abnormally express L1, and this has been associated with poor prognosis due to increased cell proliferation, invasiveness, or metastasis. We showed previously that the soluble L1 ectodomain, which is proteolyzed from the transmembrane form, can stimulate proliferation and motility of GBM cells in vitro by acting through integrins and fibroblast growth factor receptors (FGFRs). Minute L1-decorated exosomal vesicles also are released by GBM cells and potentially could stimulate cell motility, proliferation, and invasiveness, but this needed to be demonstrated. In the present study, we aimed to determine if minute L1-decorated extracellular vesicles (exosomes) were capable of stimulating GBM cell motility, proliferation, and invasiveness. L1-decorated exosomes were isolated from the conditioned media of the human T98G GBM cell line and were evaluated for their effects on the behavior of glioma cell lines and primary tumor cells. L1-decorated exosomes significantly increased cell velocity in the three human glioma cells tested (T98G/shL1, U-118 MG, and primary GBM cells) in a highly quantitative SuperScratch assay compared to L1-reduced exosomes from L1-attenuated T98G/shL1 cells. They also caused a marked increase in cell proliferation as determined by DNA cell cycle analysis and cell counting. In addition, L1-decorated exosomes facilitated initial GBM cell invasion when mixed with non-invasive T98G/shL1 cells in our chick embryo brain tumor model, whereas mixing with L1-reduced exosomes did not. Chemical inhibitors against focal adhesion kinase (FAK) and fibroblast growth factor receptor (FGFR) decreased L1-mediated motility and proliferation to varying degrees. These novel data show that L1-decoratred exosomes stimulate motility, proliferation and invasion to influence GBM cell behavior, which adds to the complexity of how L1 stimulates cancer cells through not only soluble ectodomain but also through exosomes.
Collapse
|
23
|
Chen J, Jiang C, Fu L, Zhu CL, Xiang YQ, Jiang LX, Chen Q, Liu WM, Chen JN, Zhang LY, Liu M, Chen C, Tang H, Wang B, Tsao SW, Kwong DLW, Guan XY. CHL1 suppresses tumor growth and metastasis in nasopharyngeal carcinoma by repressing PI3K/AKT signaling pathway via interaction with Integrin β1 and Merlin. Int J Biol Sci 2019; 15:1802-1815. [PMID: 31523184 PMCID: PMC6743306 DOI: 10.7150/ijbs.34785] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 04/19/2019] [Indexed: 01/24/2023] Open
Abstract
Deletion of Chromosome 3p is one of the most frequently detected genetic alterations in nasopharyngeal carcinoma (NPC). We reported the role of a novel 3p26.3 tumor suppressor gene (TSG) CHL1 in NPC. Down-regulation of CHL1 was detected in 4/6 of NPC cell lines and 71/95 (74.7%) in clinical tissues. Ectopic expressions of CHL1 in NPC cells significantly inhibit colony formation and cell motility in functional study. By up-regulating epithelial markers and down-regulating mesenchymal markers CHL1 could induce mesenchymal-epithelial transition (MET), a key step in preventing tumor invasion and metastasis. CHL1 could also cause the inactivation of RhoA/Rac1/Cdc42 signaling pathway and inhibit the formation of stress fiber, lamellipodia, and filopodia. CHL1 could co-localize with adhesion molecule Integrin-β1, the expression of CHL1 was positively correlated with Integrin-β1 and another known tumor suppressor gene (TSG) Merlin. Down-regulation of Integrin-β1 or Merlin was significantly correlated with the poor survival rate of NPC patients. Further mechanistic studies showed that CHL1 could directly interact with integrin-β1 and link to Merlin, leading to the inactivation of integrin β1-AKT pathway. In conclusion, CHL1 is a vital tumor suppressor in the carcinogenesis of NPC.
Collapse
Affiliation(s)
- Juan Chen
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China;,Department of Clinical Oncology, The Seventh Affiliated Hospital, Sun Yat-sen University
| | - Chen Jiang
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Li Fu
- Guangdong Key Laboratory for Genome Stability & Disease Prevention, Department of Pharmacology and Shenzhen University International Cancer Research Centre, Shenzhen University school of Medicine, Shenzhen, China
| | - Cai-Lei Zhu
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yan-Qun Xiang
- Department of Nasopharyngeal, Sun Yat-Sen Cancer Center, Guangzhou, China
| | - Ling-Xi Jiang
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Qian Chen
- Departments of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Wai Man Liu
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jin-Na Chen
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Li-Yi Zhang
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Ming Liu
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chao Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Science and Technology of Huazhong University, Wuhan, China
| | - Hong Tang
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Bo Wang
- Department of Clinical Oncology, The Seventh Affiliated Hospital, Sun Yat-sen University
| | - Sai Wah Tsao
- Departments of Anatomy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Dora Lai-Wan Kwong
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xin-Yuan Guan
- Departments of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China;,State Key Laboratory of Oncology in Southern China, Sun Yat-Sen University Cancer Center, Guangzhou, China;,✉ Corresponding author: Xin-Yuan Guan, Department of Clinical Oncology, The University of Hong Kong, Room L10-56, Laboratory Block, 21 Sassoon Road, Pokfulam, Hong Kong, Tel: 852-3917-9782, E-Mail: ; or Dora Lai-Wan Kwong, Department of Clinical Oncology, University of Hong Kong, 1/F, Professorial Block, Queen Mary Hospital, Hong Kong, Tel: 852-28554521, E-mail:
| |
Collapse
|
24
|
Manufacture of Chimeric Antigen Receptor T Cells from Mobilized Cyropreserved Peripheral Blood Stem Cell Units Depends on Monocyte Depletion. Biol Blood Marrow Transplant 2019; 25:223-232. [DOI: 10.1016/j.bbmt.2018.10.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 10/02/2018] [Indexed: 11/30/2022]
|
25
|
Role of Immunohistochemistry in the Identification of Supratentorial C11ORF95-RELA Fused Ependymoma in Routine Neuropathology. Am J Surg Pathol 2019; 43:56-63. [DOI: 10.1097/pas.0000000000000979] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
26
|
Li W, Xia S, Aronova A, Min IM, Verma A, Scognamiglio T, Gray KD, Ullmann TM, Liang H, Moore MD, Elemento O, Zarnegar R, Fahey TJ. CHL1 expression differentiates Hürthle cell carcinoma from benign Hürthle cell nodules. J Surg Oncol 2018; 118:1042-1049. [PMID: 30311656 DOI: 10.1002/jso.25214] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/12/2018] [Accepted: 07/30/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND OBJECTIVES Hürthle cell carcinoma (HCC) is an unusual and relatively rare type of differentiated thyroid cancer. Currently, cytologic analysis of fine-needle aspiration biopsy is limited in distinguishing benign Hürthle cell neoplasms from malignant ones. The aim of this study was to determine whether differences in the expression of specific genes could differentiate HCC from benign Hürthle cell nodules by evaluating differential gene expression in Hürthle cell disease. METHODS Eighteen benign Hürthle cell nodules and seven HCC samples were analyzed by whole-transcriptome sequencing. Bioinformatics analysis was carried out to identify candidate differentially expressed genes. Expression of these candidate genes was re-examined by quantitative real-time polymerase chain reaction (qRT-PCR). Protein expression was quantified by immunohistochemistry. RESULTS Close homolog of L1 (CHL1) was identified as overexpressed in HCC. CHL1 was found to have greater than 15-fold higher expression in fragments per kilobase million in HCC compared with benign Hurthle cell tumors. This was confirmed by qRT-PCR. Moreover, the immunoreactivity score of the CHL1 protein was significantly higher in HCC compared with benign Hürthle cell nodules. CONCLUSIONS CHL1 expression may represent a novel and useful prognostic biomarker to distinguish HCC from benign Hürthle cell disease.
Collapse
Affiliation(s)
- Wei Li
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Shujun Xia
- Ultrasound Department, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Anna Aronova
- Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical College, New York, New York
| | - Irene M Min
- Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical College, New York, New York
| | - Akanksha Verma
- Department of Physiology and Biophysics, Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York
| | - Theresa Scognamiglio
- Department of Pathology, New York Presbyterian Hospital-Weill Cornell Medical College, New York, New York
| | - Katherine D Gray
- Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical College, New York, New York
| | - Timothy M Ullmann
- Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical College, New York, New York
| | - Heng Liang
- Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical College, New York, New York
| | - Maureen D Moore
- Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical College, New York, New York
| | - Olivier Elemento
- Department of Physiology and Biophysics, Institute for Precision Medicine, Institute for Computational Biomedicine, Weill Cornell Medical College, New York, New York
| | - Rasa Zarnegar
- Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical College, New York, New York
| | - Thomas J Fahey
- Department of Surgery, New York Presbyterian Hospital-Weill Cornell Medical College, New York, New York
| |
Collapse
|
27
|
Wu JD, Hong CQ, Huang WH, Wei XL, Zhang F, Zhuang YX, Zhang YQ, Zhang GJ. L1 Cell Adhesion Molecule and Its Soluble Form sL1 Exhibit Poor Prognosis in Primary Breast Cancer Patients. Clin Breast Cancer 2018; 18:e851-e861. [PMID: 29510897 DOI: 10.1016/j.clbc.2017.12.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Revised: 11/28/2017] [Accepted: 12/20/2017] [Indexed: 02/05/2023]
Abstract
INTRODUCTION The L1 cell adhesion molecule (L1-CAM) and its soluble form sL1 play a prominent role in invasion and metastasis in several cancers. However, its association with breast cancer is still unclear. PATIENTS AND METHODS We analyzed L1-CAM expression and serum sL1 levels in cancer and para-carcinoma tissues from 162 consecutive patients with primary invasive breast cancer (PBC) using immunohistochemistry and an enzyme-linked immunosorbent assay, respectively. The serum sL1 levels were also examined in 38 patients with benign breast disease and 36 healthy controls. RESULTS L1-CAM was expressed more frequently in cancer tissues than in para-carcinoma tissues (24.1% vs. 5.6%; P < .001), and the mean sL1 levels were significantly greater in PBC than in those with benign breast disease and healthy controls (P = .027). Both L1-CAM+ expression and higher mean sL1 levels correlated significantly with larger tumor size, lymph node involvement, higher histologic grade, advanced TNM stage, and shorter disease-free survival for PBC patients. Moreover, higher mean sL1 levels were also significantly associated with estrogen receptor-α-negative expression, human epidermal growth factor receptor 2-positive (HER2+) expression, HER2-enriched and triple-negative molecular subtypes, and L1-CAM+ expression (P < .05). On multivariate analysis, larger tumor size, nodal involvement, HER2+, and higher sL1 levels (≥ 0.7 ng/mL) were independent factors associated with L1-CAM+ expression (P < .05). No association was found between L1-CAM expression or sL1 level with age, gender, histologic type, or expression of progesterone receptor, Ki-67, p53, or vascular endothelial growth factor C (P > .05). CONCLUSION These results indicate that L1-CAM and sL1 are elevated in PBC and both might affect the prognosis of PBC patients. In addition, sL1 might be a useful marker for screening and diagnosis.
Collapse
Affiliation(s)
- Jun-Dong Wu
- The Breast Center, Central Laboratory, Cancer Hospital of Shantou University Medical College, Guangdong, China
| | - Chao-Qun Hong
- Changjiang Scholar's Laboratory, Cancer Hospital of Shantou University Medical College, Guangdong, China
| | - Wen-He Huang
- The Breast Center, Central Laboratory, Cancer Hospital of Shantou University Medical College, Guangdong, China
| | - Xiao-Long Wei
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Guangdong, China
| | - Fan Zhang
- Changjiang Scholar's Laboratory, Cancer Hospital of Shantou University Medical College, Guangdong, China
| | - Yi-Xuan Zhuang
- Changjiang Scholar's Laboratory, Cancer Hospital of Shantou University Medical College, Guangdong, China
| | - Yong-Qu Zhang
- The Breast Center, Central Laboratory, Cancer Hospital of Shantou University Medical College, Guangdong, China
| | - Guo-Jun Zhang
- Changjiang Scholar's Laboratory, Shantou University Medical College, Guangdong, China.
| |
Collapse
|
28
|
Kommoss FK, Karnezis AN, Kommoss F, Talhouk A, Taran FA, Staebler A, Gilks CB, Huntsman DG, Krämer B, Brucker SY, McAlpine JN, Kommoss S. L1CAM further stratifies endometrial carcinoma patients with no specific molecular risk profile. Br J Cancer 2018; 119:480-486. [PMID: 30050154 PMCID: PMC6134076 DOI: 10.1038/s41416-018-0187-6] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/18/2018] [Accepted: 06/25/2018] [Indexed: 12/27/2022] Open
Abstract
Background The newly developed Proactive Molecular Risk Classifier for Endometrial Cancer (ProMisE) has consistently been shown to be prognostically significant in endometrial carcinomas (EC). Recently, we and others have demonstrated L1 cell-adhesion molecule (L1CAM) to be a significant indicator of high-risk disease in EC. In the current study, it was our aim to determine the prognostic significance of aberrant L1CAM expression in ProMisE subgroups in a large, single centre, population-based EC cohort. Methods ProMisE (POLE; MMR-D; p53 wt/NSMP; p53 abn) classification results from a cohort of 452 EC were available for analysis. L1CAM expression was studied by immunohistochemistry on whole slides. Correlations between clinicopathological data and survival were calculated. Results Expression of L1CAM was most frequent in p53 abnormal tumours (80%). L1CAM status was predictive of worse outcome among tumours with no specific molecular profile (p53 wt/NSMP) (p < 0.0001). Among p53 wt/NSMP EC, L1CAM remained a significant prognosticator for disease-specific survival after multivariate analysis (p = 0.035). Conclusion L1CAM status was able to significantly stratify risk among tumours of the large p53 wt/NSMP ProMisE subgroup of EC. Furthermore, our study confirms a highly significant correlation between mutation-type p53 immunostaining and abnormal L1CAM expression in EC.
Collapse
Affiliation(s)
- Felix Kf Kommoss
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Anthony N Karnezis
- Department of Pathology and Laboratory Medicine, University of British Columbia and British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Friedrich Kommoss
- Institute of Pathology, Im Medizin Campus Bodensee, Friedrichshafen, Germany
| | - Aline Talhouk
- Department of Pathology and Laboratory Medicine, University of British Columbia and British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Florin-Andrei Taran
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany
| | - Annette Staebler
- Institute of Pathology, University Hospital Tübingen, Tübingen, Germany
| | - C Blake Gilks
- Department of Pathology and Laboratory Medicine, University of British Columbia and Vancouver General Hospital, Vancouver, BC, Canada
| | - David G Huntsman
- Department of Pathology and Laboratory Medicine, University of British Columbia and British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Bernhard Krämer
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany
| | - Sara Y Brucker
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany
| | - Jessica N McAlpine
- University of British Columbia, Department of Gynecology and Obstetrics, Division of Gynecologic Oncology, Vancouver, BC, Canada
| | - Stefan Kommoss
- Department of Women's Health, Tübingen University Hospital, Tübingen, Germany.
| |
Collapse
|
29
|
Notaro S, Reimer D, Duggan-Peer M, Fiegl H, Wiedermair A, Rössler J, Altevogt P, Marth C, Zeimet AG. Evaluating L1CAM expression in human endometrial cancer using qRT-PCR. Oncotarget 2018; 7:40221-40232. [PMID: 27233077 PMCID: PMC5130004 DOI: 10.18632/oncotarget.9574] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 05/08/2016] [Indexed: 12/22/2022] Open
Abstract
Background Management of endometrial carcinoma (EC) still needs improvement of risk assessment. Recently, L1CAM immunohistochemical (IHC) evaluation showed a unique value to predict the outcome of early EC. However IHC results are often conflicting for lack of inter-laboratory standardisation. Methods Here, as a proof of concept and to increase reproducibility we assayed eighty-two EC and 26 normal endometrium samples for L1CAM expression (L1CAMEXP) via qRT-PCR. The IHC evaluation was performed in 50 cancer samples. Moreover, we aimed to substantiate the in-vitro findings of L1CAM regulation through its promoter methylation (L1CAMMET), miR-34a expression and miR-34a promoter methylation. DNA methylation was assessed with MethyLight PCR technique. Results High overall concordant results between IHC and RT-PCR evaluations were found. L1CAMEXP was detected in 11% of cancer specimens. These positive cancers exhibited a worse DFS (p=0.032) and OS (p=0.016) in a multivariate COX-regression model. L1CAMEXP predicted distant failure (p=0.007) and L1CAMMET predicted risk-reduction of lymph-node involvement (p=0.005). Inverse correlations between L1CAMEXP and L1CAMMET (p=0.004) and between L1CAMEXP and miR-34a expression (p=0.002) were found. Conclusions In conclusion qRT-PCR analysis is a reliable approach to evaluate L1CAM status in EC and L1CAMEXP was highly predictive for distant failure and poor outcome, confirming the large IHC-based studies. Interestingly, L1CAMMET was able to assess the risk of pelvic lymph-node involvement. Especially the latter finding has to be confirmed in larger prospective series.
Collapse
Affiliation(s)
- Sara Notaro
- Department of Gynecology and Obstetrics, Medical University of Innsbruck, Innsbruck, Austria.,Department of Gynecology and Obstetrics, University of Brescia, Brescia, Italy
| | - Daniel Reimer
- Department of Gynecology and Obstetrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Michaela Duggan-Peer
- Department of Gynecology and Obstetrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Heidi Fiegl
- Department of Gynecology and Obstetrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Annamarie Wiedermair
- Department of Gynecology and Obstetrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Julia Rössler
- Department of Gynecology and Obstetrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Peter Altevogt
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany
| | - Christian Marth
- Department of Gynecology and Obstetrics, Medical University of Innsbruck, Innsbruck, Austria
| | - Alain Gustave Zeimet
- Department of Gynecology and Obstetrics, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
30
|
Zaatiti H, Abdallah J, Nasr Z, Khazen G, Sandler A, Abou-Antoun TJ. Tumorigenic proteins upregulated in the MYCN-amplified IMR-32 human neuroblastoma cells promote proliferation and migration. Int J Oncol 2018; 52:787-803. [PMID: 29328367 PMCID: PMC5807036 DOI: 10.3892/ijo.2018.4236] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/05/2017] [Indexed: 12/22/2022] Open
Abstract
Childhood neuroblastoma is one of the most common types of extra-cranial cancer affecting children with a clinical spectrum ranging from spontaneous regression to malignant and fatal progression. In order to improve the clinical outcomes of children with high-risk neuroblastoma, it is crucial to understand the tumorigenic mechanisms that govern its malignant behaviors. MYCN proto-oncogene, bHLH transcription factor (MYCN) amplification has been implicated in the malignant, treatment-evasive nature of aggressive, high-risk neuroblastoma. In this study, we used a SILAC approach to compare the proteomic signatures of MYCN-amplified IMR-32 and non-MYCN-amplified SK-N-SH human neuroblastoma cells. Tumorigenic proteins, including fatty-acid binding protein 5 (FABP5), L1-cell adhesion molecule (L1-CAM), baculoviral IAP repeat containing 5 [BIRC5 (survivin)] and high mobility group protein A1 (HMGA1) were found to be significantly upregulated in the IMR-32 compared to the SK-N-SH cells and mapped to highly tumorigenic pathways including, MYC, MYCN, microtubule associated protein Tau (MAPT), E2F transcription factor 1 (E2F1), sterol regulatory element binding transcription factor 1 or 2 (SREBF1/2), hypoxia-inducible factor 1α (HIF-1α), Sp1 transcription factor (SP1) and amyloid precursor protein (APP). The transcriptional knockdown (KD) of MYCN, HMGA1, FABP5 and L1-CAM significantly abrogated the proliferation of the IMR-32 cells at 48 h post transfection. The early apoptotic rates were significantly higher in the IMR-32 cells in which FABP5 and MYCN were knocked down, whereas cellular migration was significantly abrogated with FABP5 and HMGA1 KD compared to the controls. Of note, L1-CAM, HMGA1 and FABP5 KD concomitantly downregulated MYCN protein expression and MYCN KD concomitantly downregulated L1-CAM, HMGA1 and FABP5 protein expression, while survivin protein expression was significantly downregulated by MYCN, HMGA1 and FABP5 KD. In addition, combined L1-CAM and FABP5 KD led to the concomitant downregulation of HMGA1 protein expression. On the whole, our data indicate that this inter-play between MYCN and the highly tumorigenic proteins which are upregulated in the malignant IMR-32 cells may be fueling their aggressive behavior, thereby signifying the importance of combination, multi-modality targeted therapy to eradicate this deadly childhood cancer.
Collapse
Affiliation(s)
- Hayat Zaatiti
- Department of Biology, Faculty of Sciences, University of Balamand, El-Koura, Lebanon
| | - Jad Abdallah
- Department of Pharmaceutical Sciences, School of Pharmacy, Lebanese American University, Byblos 1102-2801, Lebanon
| | - Zeina Nasr
- Department of Biology, Faculty of Sciences, University of Balamand, El-Koura, Lebanon
| | - George Khazen
- School of Arts and Sciences, Lebanese American University, Byblos 1102-2801, Lebanon
| | - Anthony Sandler
- Sheikh Zayed Institute for Pediatric Surgical Innovation, Joseph E. Robert Jr. Center for Surgical Care, Children's National Medical Center, Washington, DC 20010, USA
| | - Tamara J Abou-Antoun
- Department of Pharmaceutical Sciences, School of Pharmacy, Lebanese American University, Byblos 1102-2801, Lebanon
| |
Collapse
|
31
|
Chen SC, Chen FW, Hsu YL, Kuo PL. Systematic Analysis of Transcriptomic Profile of Renal Cell Carcinoma under Long-Term Hypoxia Using Next-Generation Sequencing and Bioinformatics. Int J Mol Sci 2017; 18:ijms18122657. [PMID: 29215599 PMCID: PMC5751259 DOI: 10.3390/ijms18122657] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 11/25/2017] [Accepted: 12/04/2017] [Indexed: 01/28/2023] Open
Abstract
Patients with clear cell renal cell carcinoma (ccRCC) are often diagnosed with both von Hippel-Lindau (VHL) mutations and the constitutive activation of hypoxia-inducible factor-dependent signaling. In this study, we investigated the effects of long-term hypoxia in 786-O, a VHL-defective renal cell carcinoma cell line, to identify potential genes and microRNAs associated with tumor malignancy. The transcriptomic profiles of 786-O under normoxia, short-term hypoxia and long-term hypoxia were analyzed using next-generation sequencing. The results showed that long-term hypoxia promoted the ability of colony formation and transwell migration compared to normoxia. In addition, the differentially expressed genes induced by long-term hypoxia were involved in various biological processes including cell proliferation, the tumor necrosis factor signaling pathway, basal cell carcinoma and cancer pathways. The upregulated (L1CAM and FBN1) and downregulated (AUTS2, MAPT, AGT and USH1C) genes in 786-O under long-term hypoxia were also observed in clinical ccRCC samples along with malignant grade. The expressions of these genes were significantly correlated with survival outcomes in patients with renal cancer. We also found that long-term hypoxia in 786-O resulted in decreased expressions of hsa-mir-100 and hsa-mir-378 and this effect was also observed in samples of metastatic ccRCC compared to samples of non-metastatic ccRCC. These findings may provide a new direction for the study of potential molecular mechanisms associated with the progression of ccRCC.
Collapse
Affiliation(s)
- Szu-Chia Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Internal Medicine, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung Medical University, Kaohsiung 812, Taiwan.
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Feng-Wei Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Ya-Ling Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Po-Lin Kuo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Institute of Medical Science and Technology, National Sun Yat-Sen University, Kaohsiung 804, Taiwan.
| |
Collapse
|
32
|
Fadare O, Roma AA, Desouki MM, Gwin K, Hanley KZ, Jarboe EA, Liang SX, Quick CM, Zheng W, Hecht JL, Parkash V, Wang XJ. The significance of L1CAM expression in clear cell carcinoma of the endometrium. Histopathology 2017; 72:532-538. [PMID: 28941294 DOI: 10.1111/his.13405] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Oluwole Fadare
- Department of Pathology, University of California, San Diego, CA, USA
| | - Andres A Roma
- Department of Pathology, University of California, San Diego, CA, USA
| | - Mohamed M Desouki
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Katja Gwin
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Krisztina Z Hanley
- Department of Pathology and Laboratory Medicine, Emory University Hospital, Atlanta, GA, USA
| | - Elke A Jarboe
- Department of Pathology, ARUP Laboratories, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Sharon X Liang
- Department of Pathology and Laboratory Medicine, Hofstra-Northwell School of Medicine, Lake Success, NY, USA
| | - Charles M Quick
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Wenxin Zheng
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jonathan L Hecht
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Vinita Parkash
- Department of Pathology, Yale University School of Medicine, New Haven, USA
| | - Xuan J Wang
- Department of Pathology and Laboratory Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
33
|
Tangen IL, Kopperud RK, Visser NC, Staff AC, Tingulstad S, Marcickiewicz J, Amant F, Bjørge L, Pijnenborg JM, Salvesen HB, Werner HM, Trovik J, Krakstad C. Expression of L1CAM in curettage or high L1CAM level in preoperative blood samples predicts lymph node metastases and poor outcome in endometrial cancer patients. Br J Cancer 2017; 117:840-847. [PMID: 28751757 PMCID: PMC5589986 DOI: 10.1038/bjc.2017.235] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 06/23/2017] [Accepted: 06/28/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Several studies have identified L1 cell adhesion molecule (L1CAM) as a strong prognostic marker in endometrial cancer. To further underline the clinical usefulness of this biomarker, we investigated L1CAM as a predictive marker for lymph node metastases and its prognostic impact in curettage specimens and preoperative plasma samples. In addition, we aimed to validate the prognostic value of L1CAM in hysterectomy specimen. METHODS Immunohistochemical staining of L1CAM was performed for 795 hysterectomy and 1134 curettage specimen from endometrial cancer patients. The L1CAM level in preoperative blood samples from 372 patients was determined using ELISA. RESULTS Expression of L1CAM in curettage specimen was significantly correlated to L1CAM level in corresponding hysterectomy specimen (P<0.001). Both in curettage and preoperative plasma samples L1CAM upregulation was significantly associated with features of aggressive disease and poor outcome (P<0.001). The L1CAM was an independent predictor of lymph node metastases, after correction for curettage histology, both in curettage specimen (P=0.002) and plasma samples (P=0.048). In the hysterectomy samples L1CAM was significantly associated with poor outcome (P<0.001). CONCLUSIONS We demonstrate that preoperative evaluation of L1CAM levels, both in curettage or plasma samples, predicts lymph node metastases and adds valuable information on patient prognosis.
Collapse
Affiliation(s)
- Ingvild L Tangen
- Department of Gynaecology and Obstetrics, Haukeland University Hospital, 5053 Bergen, Norway.,Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Reidun K Kopperud
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Nicole Cm Visser
- Department of Pathology, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Anne C Staff
- Department of Gynaecology, Oslo University Hospital, 0424 Oslo, Norway.,Faculty of Medicine, University of Oslo, 0424 Oslo, Norway
| | - Solveig Tingulstad
- Department of Gynaecology, St. Olav's Hospital, 7006 Trondheim, Norway.,Department of Laboratory Medicine, Children's and Women's Health (LBK), Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
| | - Janusz Marcickiewicz
- Department of Obstetrics and Gynaecology, Halland's Hospital Varberg, 43281 Varberg, Sweden
| | - Frédéric Amant
- Department of Oncology and Gynaecologic Oncology, Leuven Cancer Institute, 3000 Leuven, Belgium.,Center for Gynaecologic Oncology Amsterdam (CGOA), Netherlands Cancer Institute, 1006 BE Amsterdam, The Netherlands
| | - Line Bjørge
- Department of Gynaecology and Obstetrics, Haukeland University Hospital, 5053 Bergen, Norway.,Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Johanna Ma Pijnenborg
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Helga B Salvesen
- Department of Gynaecology and Obstetrics, Haukeland University Hospital, 5053 Bergen, Norway.,Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Henrica Mj Werner
- Department of Gynaecology and Obstetrics, Haukeland University Hospital, 5053 Bergen, Norway.,Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Jone Trovik
- Department of Gynaecology and Obstetrics, Haukeland University Hospital, 5053 Bergen, Norway.,Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| | - Camilla Krakstad
- Department of Gynaecology and Obstetrics, Haukeland University Hospital, 5053 Bergen, Norway.,Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, 5020 Bergen, Norway
| |
Collapse
|
34
|
Xu YW, Peng YH, Ran LQ, Zhai TT, Guo HP, Qiu SQ, Chen HL, Wu ZY, Li EM, Xie JJ. Circulating levels of autoantibodies against L1-cell adhesion molecule as a potential diagnostic biomarker in esophageal squamous cell carcinoma. Clin Transl Oncol 2017; 19:898-906. [PMID: 28181176 DOI: 10.1007/s12094-017-1623-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/28/2017] [Indexed: 02/05/2023]
Abstract
BACKGROUND Esophageal squamous cell carcinoma (ESCC) is a common malignant disease worldwide, especially in China. We aimed to determine the level of autoantibodies against L1CAM in patients with ESCC. METHODS Levels of circulating autoantibodies against L1CAM antigens were determined by an enzyme-linked immunosorbent assay in cohort 1 (191 patients with ESCC and 94 normal controls) and validated in cohort 2 (47 patients with ESCC and 47 normal controls). Receiver-operating characteristics were employed to calculate diagnostic accuracy. Cumulative survival time was calculated by the Kaplan-Meier method and analyzed by the log-rank test. RESULTS In cohorts 1 and 2, levels of autoantibodies against L1CAM were all significantly higher in sera of patients with ESCC compared to normal controls (P < 0.05). Detection of autoantibodies against L1CAM provided a sensitivity of 26.2%, a specificity of 90.4%, and an area under the curve (AUC) of 0.603 (95% CI 0.535-0.672) in diagnosing ESCC in cohort 1, and a sensitivity of 27.7%, a specificity of 91.5%, and an AUC of 0.628 (95% CI 0.516-0.741). Similar results were observed in the diagnosis of early stage ESCC (25.2% sensitivity, 90.4% specificity, and an AUC of 0.611 (95% CI 0.533-0.689) in cohort 1, and 33.3% sensitivity, 91.5% specificity, and an AUC of 0.636 (95% CI 0.439-0.832) in cohort 2). Moreover, positive rates of autoantibodies against L1CAM had no statistical correlation with clinical outcome of ESCC (P > 0.05). CONCLUSIONS Our results suggest that circulating autoantibodies against L1CAM is a potential biomarker for the early detection of ESCC.
Collapse
Affiliation(s)
- Y-W Xu
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China
| | - Y-H Peng
- Department of Clinical Laboratory Medicine, The Cancer Hospital of Shantou University Medical College, Shantou, China
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China
| | - L-Q Ran
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China
| | - T-T Zhai
- Department of Radiation Oncology, The Cancer Hospital of Shantou University Medical College, Shantou, China
| | - H-P Guo
- Department of Head and Neck Surgery, The Cancer Hospital of Shantou University Medical College, Shantou, China
| | - S-Q Qiu
- The Breast Center, The Cancer Hospital of Shantou University Medical College, Shantou, China
| | - H-L Chen
- Department of Surgical Oncology, Shantou Central Hospital, Shantou, China
| | - Z-Y Wu
- Department of Surgical Oncology, Shantou Central Hospital, Shantou, China
| | - E-M Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China.
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China.
| | - J-J Xie
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, China.
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, China.
| |
Collapse
|
35
|
Kim H, Hwang H, Lee H, Hong HJ. L1 Cell Adhesion Molecule Promotes Migration and Invasion via JNK Activation in Extrahepatic Cholangiocarcinoma Cells with Activating KRAS Mutation. Mol Cells 2017; 40:363-370. [PMID: 28535665 PMCID: PMC5463045 DOI: 10.14348/molcells.2017.2282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 04/10/2017] [Accepted: 04/28/2017] [Indexed: 02/07/2023] Open
Abstract
Extrahepatic cholangiocarcinoma (ECC), a malignant tumor of biliary origin, has a poor prognosis with limited treatment options. The KRAS oncogene is the most commonly mutated gene in ECC and one of the factors that predicts a poor prognosis and low survival rate. L1 cell adhesion molecule (L1CAM) is expressed in ECC cells and acts as an independent poor prognostic factor in predicting patient survival. In this study we investigate the functional significance of L1CAM in ECC cells with activating KRAS mutation. We selected an ECC cell line, EGI-1, with activating KRAS mutation, and then confirmed its expression of L1CAM by RT-PCR, western blot analysis, and flow cytometry. The suppression of L1CAM expression (using a specific lentivirus-delivered shRNA) significantly decreased the migratory and invasive properties of EGI-1 cells, without altering their proliferation or survival. Analyses of signaling effectors in L1CAM-depleted and control EGI-1 cells indicated that L1CAM suppression decreased the levels of both phosphorylated MKK4 and total MKK4, together with c-Jun N-terminal kinase (JNK) phosphorylation. Further, exposure to a JNK inhibitor (SP600125) decreased migration and invasion of EGI-1 cells. These results suggest that L1CAM promotes cellular migration and invasion via the induction of MKK4 expression, leading to JNK activation. Our study is the first to demonstrate a functional role for L1CAM in ECC carrying the activating KRAS mutation. Given that KRAS is the most commonly mutated oncogene in ECC, L1CAM may serve as an attractive therapeutic target for ECC cells with activating KRAS mutation.
Collapse
Affiliation(s)
- Haejung Kim
- Department of Biology, College of National Science, College of Biomedical Science, Kangwon National University, Chuncheon 24341,
Korea
- Institute of Bioscience and Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon 24341,
Korea
| | - Haein Hwang
- Department of Systems Immunology, College of Biomedical Science, Kangwon National University, Chuncheon 24341,
Korea
| | - Hansoo Lee
- Department of Biology, College of National Science, College of Biomedical Science, Kangwon National University, Chuncheon 24341,
Korea
| | - Hyo Jeong Hong
- Institute of Bioscience and Biotechnology, College of Biomedical Science, Kangwon National University, Chuncheon 24341,
Korea
- Department of Systems Immunology, College of Biomedical Science, Kangwon National University, Chuncheon 24341,
Korea
| |
Collapse
|
36
|
Samulin Erdem J, Arnoldussen YJ, Skaug V, Haugen A, Zienolddiny S. Copy number variation, increased gene expression, and molecular mechanisms of neurofascin in lung cancer. Mol Carcinog 2017; 56:2076-2085. [PMID: 28418179 PMCID: PMC6084301 DOI: 10.1002/mc.22664] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 03/31/2017] [Accepted: 04/13/2017] [Indexed: 12/21/2022]
Abstract
Metastasis and cell adhesion are key aspects of cancer progression. Neurofascin (NFASC) is a member of the immunoglobulin superfamily of adhesion molecules and, while studies on NFASC are inadequate, other members have been indicated pivotal roles in cancer progression and metastasis. This study aimed at increasing the knowledge on the involvement of adhesion molecules in lung cancer progression by studying the regulation and role of NFASC in non‐small cell lung cancer (NSCLC). Here, copy number variations in the NFASC gene were analyzed in tumor and non‐tumorous lung tissues of 204 NSCLC patients. Frequent gene amplifications (OR = 4.50, 95%CI: 2.27‐8.92, P ≤ 0.001) and increased expression of NFASC (P = 0.034) were identified in tumors of NSCLC patients. Furthermore, molecular mechanisms of NFASC in lung cancer progression were evaluated by investigating the effects of NFASC silencing on cell proliferation, viability, migration, and invasion using siRNA technology in four NSCLC cell lines. Silencing of NFASC did not affect cell proliferation or viability but rather decreased NSCLC cell migration (P ≤ 0.001) and led to morphological changes, rearrangements in the actin cytoskeleton and changes in F‐actin networks in migrating NSCLC cell lines. This study is the first to report frequent copy number gain and increased expression of NFASC in NSCLC. Moreover, these data suggest that NFASC is a novel regulator of NSCLC cell motility and support a role of NFASC in the regulation of NSCLC progression.
Collapse
Affiliation(s)
- Johanna Samulin Erdem
- Department of Chemical and Biological Work Environment, National Institute of Occupational Health, Oslo, Norway
| | - Yke Jildouw Arnoldussen
- Department of Chemical and Biological Work Environment, National Institute of Occupational Health, Oslo, Norway
| | - Vidar Skaug
- Department of Chemical and Biological Work Environment, National Institute of Occupational Health, Oslo, Norway
| | - Aage Haugen
- Department of Chemical and Biological Work Environment, National Institute of Occupational Health, Oslo, Norway
| | - Shanbeh Zienolddiny
- Department of Chemical and Biological Work Environment, National Institute of Occupational Health, Oslo, Norway
| |
Collapse
|
37
|
C1QBP suppresses cell adhesion and metastasis of renal carcinoma cells. Sci Rep 2017; 7:999. [PMID: 28428626 PMCID: PMC5430506 DOI: 10.1038/s41598-017-01084-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 03/23/2017] [Indexed: 11/09/2022] Open
Abstract
Complement component 1q subcomponent binding protein (C1QBP) is a ubiquitously expressed cellular protein and can be upregulated or activated in a variety of malignant tumors, including those from thyroid, colon and breast, but its role remains unclear in renal cell carcinoma (RCC). In this study, C1QBP knockdown in RCC cell influenced expression of multiple genes associated with cell adhesion, among which L1 cell adhesion molecule (L1CAM) was significantly higher upon a reduction of C1QBP. In turn, cell adhesion and invasion abilities were significantly increased with increased metastasis to lung and liver in vivo. C1QBP may regulate RCC cell adhesion and invasion through influencing the p-GSK3/β-Catenin/L1CAM expression. Over all, our study demonstrated that C1QBP could regulate RCC metastasis by regulating the GSK3/β-Catenin/L1CAM signaling pathway.
Collapse
|
38
|
Kommoss F, Kommoss F, Grevenkamp F, Bunz AK, Taran FA, Fend F, Brucker SY, Wallwiener D, Schönfisch B, Greif K, Lax S, Staebler A, Kommoss S. L1CAM: amending the "low-risk" category in endometrial carcinoma. J Cancer Res Clin Oncol 2017; 143:255-262. [PMID: 27695947 DOI: 10.1007/s00432-016-2276-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 09/20/2016] [Indexed: 11/25/2022]
Abstract
PURPOSE Low- and intermediate-risk endometrial carcinomas have an excellent prognosis. Nonetheless, a small subgroup of such patients will experience unexpected relapse. Recently L1CAM was suggested to be a strong prognosticator in endometrial carcinoma. The focus of our study was on low- and intermediate-risk disease, where no or only limited adjuvant treatment is recommended according to current guidelines. METHODS Endometrial carcinomas of low, intermediate and high-intermediate risk according to published 2016 consensus guidelines were identified. The study was limited to cases with previous central pathology review focusing on histotype, depth of myometrial invasion, presence of lymphovascular space invasion (LVSI) and MELF pattern of invasion. Standard L1CAM immunohistochemistry was performed. Disease-specific uni- and multivariate survival analyses were calculated. RESULTS A total of 344 cases were available for immunohistochemistry (low-risk: n = 250; intermediate-risk: n = 67; high-intermediate-risk: n = 27). L1CAM positivity rates were: 29/344 (8.4 %; all cases), 18/250 (7.2 %; low-risk), 6/67 (9.0 %; intermediate-risk) and 5/27 (18.5 %; high-intermediate-risk). Expression of L1CAM was independent of LVSI and MELF. L1CAM was a significant independent prognosticator for disease-specific survival with a hazard ratio of 5.98 [CI 1.50-22.14, p = 0.012]. Adverse prognostic significance of L1CAM positivity was maintained after low-risk subgroup analysis (5-year disease-specific survival rates 71.8 vs. 100 %, p < 0.0001). All four tumour-related deaths in the subgroup of low-risk disease occurred in patients with L1CAM-positive tumours. CONCLUSION The current definition of "low-risk" in endometrial carcinoma should be amended. "Low-risk carcinomas" should be limited to L1CAM-negative tumours. L1CAM status will play a key role in future algorithms to tailor adjuvant treatment and patient follow-up strategies.
Collapse
Affiliation(s)
- Felix Kommoss
- Department für Frauengesundheit, Universitätsklinikum Tübingen, Calwerstr. 7, 72076, Tübingen, Germany
| | - Friedrich Kommoss
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Friederike Grevenkamp
- Department für Frauengesundheit, Universitätsklinikum Tübingen, Calwerstr. 7, 72076, Tübingen, Germany
| | - Anne-Kathrin Bunz
- Department für Frauengesundheit, Universitätsklinikum Tübingen, Calwerstr. 7, 72076, Tübingen, Germany
| | - Florin-Andrei Taran
- Department für Frauengesundheit, Universitätsklinikum Tübingen, Calwerstr. 7, 72076, Tübingen, Germany
| | - Falko Fend
- Institut für Pathologie, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Sara Y Brucker
- Department für Frauengesundheit, Universitätsklinikum Tübingen, Calwerstr. 7, 72076, Tübingen, Germany
| | - Diethelm Wallwiener
- Department für Frauengesundheit, Universitätsklinikum Tübingen, Calwerstr. 7, 72076, Tübingen, Germany
| | - Birgitt Schönfisch
- Department für Frauengesundheit, Universitätsklinikum Tübingen, Calwerstr. 7, 72076, Tübingen, Germany
| | - Karen Greif
- Institut für Pathologie, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Sigurd Lax
- Institut für Pathologie, LKH Graz West, Graz, Austria
| | - Annette Staebler
- Institut für Pathologie, Universitätsklinikum Tübingen, Tübingen, Germany
| | - Stefan Kommoss
- Department für Frauengesundheit, Universitätsklinikum Tübingen, Calwerstr. 7, 72076, Tübingen, Germany.
| |
Collapse
|
39
|
Wojciechowski M, Głowacka E, Wilczyński M, Pękala-Wojciechowska A, Malinowski A. The sL1CAM in sera of patients with endometrial and ovarian cancers. Arch Gynecol Obstet 2017; 295:225-232. [PMID: 27832351 PMCID: PMC5225192 DOI: 10.1007/s00404-016-4226-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 11/02/2016] [Indexed: 10/25/2022]
Abstract
PURPOSE L1CAM is a cell adhesion molecule suspected to play an important role in carcinogenesis. The objective of the study was to evaluate the level of soluble L1CAM in the sera of patients with endometrial and ovarian carcinomas and verify the feasibility of the sL1CAM as a marker of these carcinomas. METHODS 35 endometrial and 18 ovarian cancer patients were enrolled in the study. 43 patients with benign gynecological conditions constituted a control group. The sL1CAM serum level was measured with ELISA test in each patient and it was referred to the data from the surgical staging of the cancers. RESULTS The sL1CAM serum level was significantly lower in patients with endometrial cancer than in healthy women and slightly lower in the ovarian cancer group than in the control group. In the endometrial cancer group there was no correlation between sL1CAM concentration and cancer histopathology, stage or grade. sL1CAM concentration positively correlated with ovarian cancer stage and (not significantly) with grade. CONCLUSIONS Despite the increasing data about the possible role of L1CAM as a strong prognostic factor of poor outcome in many cancers, we did not find evidence supporting the use of sL1CAM as a marker of endometrial or ovarian cancers.
Collapse
Affiliation(s)
- Michał Wojciechowski
- Department of Surgical, Endoscopic Gynecology and Oncology, Polish Mother's Memorial Hospital-Research Institute, 281/283 Rzgowska St., 93-338, Łódź, Poland.
- Department of Surgical and Endoscopic Gynecology, Medical University of Lodz, Łódź, Poland.
| | - Ewa Głowacka
- Department of Laboratory Diagnostics, Polish Mother's Memorial Hospital-Research Institute, Łódź, Poland
| | - Miłosz Wilczyński
- Department of Surgical, Endoscopic Gynecology and Oncology, Polish Mother's Memorial Hospital-Research Institute, 281/283 Rzgowska St., 93-338, Łódź, Poland
| | | | - Andrzej Malinowski
- Department of Surgical, Endoscopic Gynecology and Oncology, Polish Mother's Memorial Hospital-Research Institute, 281/283 Rzgowska St., 93-338, Łódź, Poland
- Department of Surgical and Endoscopic Gynecology, Medical University of Lodz, Łódź, Poland
| |
Collapse
|
40
|
Pechriggl EJ, Concin N, Blumer MJ, Bitsche M, Zwierzina M, Dudas J, Koziel K, Altevogt P, Zeimet AG, Fritsch H. L1CAM in the Early Enteric and Urogenital System. J Histochem Cytochem 2016; 65:21-32. [PMID: 28026654 DOI: 10.1369/0022155416677241] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
L1 cell adhesion molecule (L1CAM) is a transmembrane molecule belonging to the L1 protein family. It has shown to be a key player in axonal guidance in the course of neuronal development. Furthermore, L1CAM is also crucial for the establishment of the enteric and urogenital organs and is aberrantly expressed in cancer originating in these organs. Carcinogenesis and embryogenesis follow a lot of similar molecular pathways, but unfortunately, comprehensive data on L1CAM expression and localization in human developing organs are lacking so far. In the present study we, therefore, examined the spatiotemporal distribution of L1CAM in the early human fetal period (weeks 8-12 of gestation) by means of immunohistochemistry and in situ hybridization (ISH). In the epithelia of the gastrointestinal organs, L1CAM localization cannot be observed in the examined stages most likely due to their advanced polarization and differentiation. Despite these results, our ISH data indicate weak L1CAM expression, but only in few epithelial cells. The genital tracts, however, are distinctly L1CAM positive throughout the entire fetal period. We, therefore, conclude that in embryogenesis L1CAM is crucial for further differentiation of epithelia.
Collapse
Affiliation(s)
- Elisabeth Judith Pechriggl
- Division of Clinical and Functional Anatomy, Department of Anatomy, Histology and Embryology (EJP, MJB, MB, MZ, HF), Medical University of Innsbruck, Innsbruck, Austria
| | - Nicole Concin
- Department of Gynaecology and Obstetrics (NC, KK, A-GZ), Medical University of Innsbruck, Innsbruck, Austria
| | - Michael J Blumer
- Division of Clinical and Functional Anatomy, Department of Anatomy, Histology and Embryology (EJP, MJB, MB, MZ, HF), Medical University of Innsbruck, Innsbruck, Austria
| | - Mario Bitsche
- Division of Clinical and Functional Anatomy, Department of Anatomy, Histology and Embryology (EJP, MJB, MB, MZ, HF), Medical University of Innsbruck, Innsbruck, Austria
| | - Marit Zwierzina
- Division of Clinical and Functional Anatomy, Department of Anatomy, Histology and Embryology (EJP, MJB, MB, MZ, HF), Medical University of Innsbruck, Innsbruck, Austria
| | - Jozsef Dudas
- Department of Otolaryngology (JD), Medical University of Innsbruck, Innsbruck, Austria
| | - Katarzyna Koziel
- Department of Gynaecology and Obstetrics (NC, KK, A-GZ), Medical University of Innsbruck, Innsbruck, Austria
| | - Peter Altevogt
- Skin Cancer Unit, German Cancer Research Center, Heidelberg, Germany (PA).,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Heidelberg, Germany (PA)
| | - Alain-Gustave Zeimet
- Department of Gynaecology and Obstetrics (NC, KK, A-GZ), Medical University of Innsbruck, Innsbruck, Austria
| | - Helga Fritsch
- Division of Clinical and Functional Anatomy, Department of Anatomy, Histology and Embryology (EJP, MJB, MB, MZ, HF), Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
41
|
Hoja-Łukowicz D, Przybyło M, Duda M, Pocheć E, Bubka M. On the trail of the glycan codes stored in cancer-related cell adhesion proteins. Biochim Biophys Acta Gen Subj 2016; 1861:3237-3257. [PMID: 27565356 DOI: 10.1016/j.bbagen.2016.08.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 07/22/2016] [Accepted: 08/14/2016] [Indexed: 12/14/2022]
Abstract
Changes in the profile of protein glycosylation are a hallmark of ongoing neoplastic transformation. A unique set of tumor-associated carbohydrate antigens expressed on the surface of malignant cells may serve as powerful diagnostic and therapeutic targets. Cell-surface proteins with altered glycosylation affect the growth, proliferation and survival of those cells, and contribute to their acquisition of the ability to migrate and invade. They may also facilitate tumor-induced immunosuppression and the formation of distant metastases. Deciphering the information encoded in these particular glycan portions of glycoconjugates may shed light on the mechanisms of cancer progression and metastasis. A majority of the related review papers have focused on overall changes in the patterns of cell-surface glycans in various cancers, without pinpointing the molecular carriers of these glycan structures. The present review highlights the ways in which particular tumor-associated glycan(s) coupled with a given membrane-bound protein influence neoplastic cell behavior during the development and progression of cancer. We focus on altered glycosylated cell-adhesion molecules belonging to the cadherin, integrin and immunoglobulin-like superfamilies, examined in the context of molecular interactions.
Collapse
Affiliation(s)
- Dorota Hoja-Łukowicz
- Department of Glycoconjugate Biochemistry, Institute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Krakow, Poland.
| | - Małgorzata Przybyło
- Department of Glycoconjugate Biochemistry, Institute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Krakow, Poland.
| | - Małgorzata Duda
- Department of Endocrinology, Institute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Krakow, Poland.
| | - Ewa Pocheć
- Department of Glycoconjugate Biochemistry, Institute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Krakow, Poland.
| | - Monika Bubka
- Department of Glycoconjugate Biochemistry, Institute of Zoology, Jagiellonian University, 9 Gronostajowa Street, 30-387 Krakow, Poland.
| |
Collapse
|
42
|
Rached J, Nasr Z, Abdallah J, Abou-Antoun T. L1-CAM knock-down radiosensitizes neuroblastoma IMR-32 cells by simultaneously decreasing MycN, but increasing PTEN protein expression. Int J Oncol 2016; 49:1722-30. [PMID: 27432152 DOI: 10.3892/ijo.2016.3625] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 06/28/2016] [Indexed: 11/06/2022] Open
Abstract
Childhood neuroblastoma is one of the most malignant types of cancers leading to a high mortality rate. These cancerous cells can be highly metastatic and malignant giving rise to disease recurrence and poor prognosis. The proto-oncogene myelocytomatosis neuroblastoma (MycN) is known to be amplified in this type of cancer, thus, promoting high malignancy and resistance. The L1 cell adhesion molecule (L1-CAM) cleavage has been found upregulated in many types of malignant cancers. In the present study, we explored the interplay between L1-CAM, MycN and PTEN as well as the role played by PDGFR and VEGFR on tumorigenicity in neuroblastoma cells. We investigated the effect of L1-CAM knock-down (KD) and PDGFR/VEGFR inhibition with sunitinib malate (Sutent®) treatment on subsequent tumorsphere formation and cellular proliferation and migration in the MycN-amplified IMR-32 neuroblastoma cells. We further examined the effect of combined L1-CAM KD with Sutent treatment or radiotherapy on these cellular functions in our cells. Tumorsphere formation is one of the indicators of aggressiveness in malignant cancers, which was significantly inhibited in IMR-32 cells after L1-CAM KD or Sutent treatment, however, no synergistic effect was observed with dual treatments, rather L1-CAM KD alone showed a greater inhibition on tumorsphere formation compared to Sutent treatment alone. In addition, cellular proliferation and migration were significantly inhibited after L1-CAM KD in the IMR-32 cells with no synergistic effect observed on the rate of cell proliferation when combined with Sutent treatment. Again, L1-CAM KD alone exhibited greater inhibitory effect than Sutent treatment on cell proliferation. L1-CAM KD led to the simultaneous downregulation of MycN, but the upregulation of PTEN protein expression. Notably, radiotherapy (2 Gy) of the IMR-32 cells led to significant upregulation of both L1-CAM and MycN, which was abrogated with L1-CAM KD in our cells. In addition, L1-CAM KD radiosensitized the cells as exhibited by the synergistic effect on the reduction in cell proliferation compared to radiotherapy alone. Taken together, our data show the importance of L1-CAM interplay with MycN and PTEN on the MycN amplified neuroblastoma cell radioresistance, proliferation and motility.
Collapse
Affiliation(s)
- Johnny Rached
- Faculty of Sciences, University of Balamand, Koura, Lebanon
| | - Zeina Nasr
- Faculty of Sciences, University of Balamand, Koura, Lebanon
| | - Jad Abdallah
- School of Pharmacy, Pharmaceutical Sciences Department, Lebanese American University, Byblos, Lebanon
| | - Tamara Abou-Antoun
- School of Pharmacy, Pharmaceutical Sciences Department, Lebanese American University, Byblos, Lebanon
| |
Collapse
|
43
|
Abstract
The local extension of cancer cells along nerves is a frequent clinical finding for various tumours. Traditionally, nerve invasion was assumed to occur via the path of least resistance; however, recent animal models and human studies have revealed that cancer cells have an innate ability to actively migrate along axons in a mechanism called neural tracking. The tendency of cancer cells to track along nerves is supported by various cell types in the perineural niche that secrete multiple growth factors and chemokines. We propose that the perineural niche should be considered part of the tumour microenvironment, describe the molecular cues that facilitate neural tracking and suggest methods for its inhibition.
Collapse
Affiliation(s)
- Moran Amit
- Laboratory for Applied Cancer Research, Department of Otolaryngology Head and Neck Surgery, Head and Neck Center, Rambam Healthcare Campus, Clinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, The Technion-Israel Institute of Technology, Haalia Street No. 8, Haifa, Israel
| | - Shorook Na'ara
- Laboratory for Applied Cancer Research, Department of Otolaryngology Head and Neck Surgery, Head and Neck Center, Rambam Healthcare Campus, Clinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, The Technion-Israel Institute of Technology, Haalia Street No. 8, Haifa, Israel
| | - Ziv Gil
- Laboratory for Applied Cancer Research, Department of Otolaryngology Head and Neck Surgery, Head and Neck Center, Rambam Healthcare Campus, Clinical Research Institute at Rambam, Rappaport Institute of Medicine and Research, The Technion-Israel Institute of Technology, Haalia Street No. 8, Haifa, Israel
| |
Collapse
|
44
|
Ferese R, Zampatti S, Griguoli AMP, Fornai F, Giardina E, Barrano G, Albano V, Campopiano R, Scala S, Novelli G, Gambardella S. A New Splicing Mutation in the L1CAM Gene Responsible for X-Linked Hydrocephalus (HSAS). J Mol Neurosci 2016; 59:376-81. [DOI: 10.1007/s12031-016-0754-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 04/08/2016] [Indexed: 01/30/2023]
|
45
|
Abstract
L1 cell adhesion molecule (L1CAM) is the prototype member of the L1-family of closely related neural adhesion molecules. L1CAM is differentially expressed in the normal nervous system as well as pathological tissues and displays a wide range of biological activities. In human malignancies, L1CAM plays a vital role in tumor growth, invasion and metastasis. Recently, increasing evidence has suggested that L1CAM exerts a variety of functions at different steps of tumor progression through a series of signaling pathways. In addition, L1CAM has been identified as a promising target for cancer therapy by using synthetic and natural inhibitors. In this review, we provide an up-to-date overview of the role of L1CAM involved in cancers and the rationale for L1CAM as a novel molecular target for cancer therapy.
Collapse
Affiliation(s)
- Xinzhe Yu
- a Department of Pancreatic Surgery, Huashan Hospital , Fudan University , Shanghai , China
| | - Feng Yang
- a Department of Pancreatic Surgery, Huashan Hospital , Fudan University , Shanghai , China
| | - De-Liang Fu
- a Department of Pancreatic Surgery, Huashan Hospital , Fudan University , Shanghai , China
| | - Chen Jin
- a Department of Pancreatic Surgery, Huashan Hospital , Fudan University , Shanghai , China
| |
Collapse
|
46
|
Cho S, Park I, Kim H, Jeong MS, Lim M, Lee ES, Kim JH, Kim S, Hong HJ. Generation, characterization and preclinical studies of a human anti-L1CAM monoclonal antibody that cross-reacts with rodent L1CAM. MAbs 2016; 8:414-25. [PMID: 26785809 PMCID: PMC5037990 DOI: 10.1080/19420862.2015.1125067] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
L1 cell adhesion molecule (L1CAM) is aberrantly expressed in malignant tumors and plays important roles in tumor progression. Thus, L1CAM could serve as a therapeutic target and anti-L1CAM antibodies may have potential as anticancer agents. However, L1CAM is expressed in neural cells and the druggability of anti-L1AM antibody must be validated at the earliest stages of preclinical study. Here, we generated a human monoclonal antibody that is cross-reactive with mouse L1CAM and evaluated its pharmacokinetic properties and anti-tumor efficacy in rodent models. First, we selected an antibody (Ab4) that binds human and mouse L1CAM from the human naïve Fab library using phage display, then increased its affinity 45-fold through mutation of 3 residues in the complementarity-determining regions (CDRs) to generate Ab4M. Next, the affinity of Ab4M was increased 1.8-fold by yeast display of single-chain variable fragment containing randomly mutated light chain CDR3 to generate Ab417. The affinities (KD) of Ab417 for human and mouse L1CAM were 0.24 nM and 79.16 pM, respectively. Ab417 specifically bound the Ig5 domain of L1CAM and did not exhibit off-target activity, but bound to the peripheral nerves embedded in normal human tissues as expected in immunohistochemical analysis. In a pharmacokinetics study, the mean half-life of Ab417 was 114.49 h when a single dose (10 mg/kg) was intravenously injected into SD rats. Ab417 significantly inhibited tumor growth in a human cholangiocarcinoma xenograft nude mouse model and did not induce any adverse effect in in vivo studies. Thus, Ab417 may have potential as an anticancer agent.
Collapse
Affiliation(s)
- Seulki Cho
- a Department of Functional Genomics , University of Science & Technology , Daejeon , Korea.,b Institute of Bioscience and Biotechnology, Kangwon National University , Chuncheon , Korea
| | - Insoo Park
- c Immunotherapy Research Center, Korea Research Institute of Bioscience & Biotechnology , Daejeon , Korea
| | - Haejung Kim
- b Institute of Bioscience and Biotechnology, Kangwon National University , Chuncheon , Korea
| | - Mun Sik Jeong
- d Department of Systems Immunology , Kangwon National University , Chuncheon , Korea
| | - Mooney Lim
- d Department of Systems Immunology , Kangwon National University , Chuncheon , Korea
| | - Eung Suk Lee
- b Institute of Bioscience and Biotechnology, Kangwon National University , Chuncheon , Korea
| | - Jin Hong Kim
- b Institute of Bioscience and Biotechnology, Kangwon National University , Chuncheon , Korea
| | - Semi Kim
- c Immunotherapy Research Center, Korea Research Institute of Bioscience & Biotechnology , Daejeon , Korea
| | - Hyo Jeong Hong
- b Institute of Bioscience and Biotechnology, Kangwon National University , Chuncheon , Korea.,d Department of Systems Immunology , Kangwon National University , Chuncheon , Korea
| |
Collapse
|
47
|
Altevogt P, Doberstein K, Fogel M. L1CAM in human cancer. Int J Cancer 2015; 138:1565-76. [DOI: 10.1002/ijc.29658] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 06/19/2015] [Indexed: 12/18/2022]
Affiliation(s)
- Peter Altevogt
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany and Department of Dermatology, Venereology and Allergology; University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg; Mannheim Germany
| | - Kai Doberstein
- Ovarian Cancer Research Center, Perelman School of Medicine; University of Pennsylvania; Philadelphia, PA
| | - Mina Fogel
- Central Laboratories; Kaplan Medical Center; Rehovot Israel
| |
Collapse
|
48
|
Sobotič B, Vizovišek M, Vidmar R, Van Damme P, Gocheva V, Joyce JA, Gevaert K, Turk V, Turk B, Fonović M. Proteomic Identification of Cysteine Cathepsin Substrates Shed from the Surface of Cancer Cells. Mol Cell Proteomics 2015; 14:2213-28. [PMID: 26081835 DOI: 10.1074/mcp.m114.044628] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Indexed: 01/08/2023] Open
Abstract
Extracellular cysteine cathepsins are known to drive cancer progression, but besides degradation of extracellular matrix proteins little is known about their physiological substrates and thus the molecular mechanisms they deploy. One of the major mechanisms used by other extracellular proteases to facilitate cancer progression is proteolytic release of the extracellular domains of transmembrane proteins or ectodomain shedding. Here we show using a mass spectrometry-based approach that cathepsins L and S act as sheddases and cleave extracellular domains of CAM adhesion proteins and transmembrane receptors from the surface of cancer cells. In cathepsin S-deficient mouse pancreatic cancers, processing of these cathepsin substrates is highly reduced, pointing to an essential role of cathepsins in extracellular shedding. In addition to influencing cell migration and invasion, shedding of surface proteins by extracellular cathepsins impacts intracellular signaling as demonstrated for regulation of Ras GTPase activity, thereby providing a putative mechanistic link between extracellular cathepsin activity and cancer progression. The MS data is available via ProteomeXchange with identifier PXD002192.
Collapse
Affiliation(s)
- Barbara Sobotič
- From the ‡Department of Biochemistry, Molecular and Structural Biology, Jozef Stefan Institute, Jamova cesta 39, SI-1000 Ljubljana, Slovenia; §International Postgraduate School Jozef Stefan, Jamova 39, SI-1000 Ljubljana, Slovenia
| | - Matej Vizovišek
- From the ‡Department of Biochemistry, Molecular and Structural Biology, Jozef Stefan Institute, Jamova cesta 39, SI-1000 Ljubljana, Slovenia; §International Postgraduate School Jozef Stefan, Jamova 39, SI-1000 Ljubljana, Slovenia
| | - Robert Vidmar
- From the ‡Department of Biochemistry, Molecular and Structural Biology, Jozef Stefan Institute, Jamova cesta 39, SI-1000 Ljubljana, Slovenia; §International Postgraduate School Jozef Stefan, Jamova 39, SI-1000 Ljubljana, Slovenia
| | - Petra Van Damme
- ¶Department of Biochemistry, Ghent University, B-9000 Ghent, Belgium; ‖Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium
| | - Vasilena Gocheva
- **Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Johanna A Joyce
- **Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Kris Gevaert
- ¶Department of Biochemistry, Ghent University, B-9000 Ghent, Belgium; ‖Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium
| | - Vito Turk
- From the ‡Department of Biochemistry, Molecular and Structural Biology, Jozef Stefan Institute, Jamova cesta 39, SI-1000 Ljubljana, Slovenia; §International Postgraduate School Jozef Stefan, Jamova 39, SI-1000 Ljubljana, Slovenia; ‡‡Centre of Excellence for Integrated Approaches in Chemistry and Biology of Proteins, Jamova cesta 39, SI-1000 Ljubljana, Slovenia
| | - Boris Turk
- From the ‡Department of Biochemistry, Molecular and Structural Biology, Jozef Stefan Institute, Jamova cesta 39, SI-1000 Ljubljana, Slovenia; ‡‡Centre of Excellence for Integrated Approaches in Chemistry and Biology of Proteins, Jamova cesta 39, SI-1000 Ljubljana, Slovenia; §§Center of Excellence NIN, Ljubljana, Slovenia; ¶¶Faculty of Chemistry and Chemical Technology, University of Ljubljana, Slovenia
| | - Marko Fonović
- From the ‡Department of Biochemistry, Molecular and Structural Biology, Jozef Stefan Institute, Jamova cesta 39, SI-1000 Ljubljana, Slovenia; ‡‡Centre of Excellence for Integrated Approaches in Chemistry and Biology of Proteins, Jamova cesta 39, SI-1000 Ljubljana, Slovenia;
| |
Collapse
|
49
|
Slug-dependent upregulation of L1CAM is responsible for the increased invasion potential of pancreatic cancer cells following long-term 5-FU treatment. PLoS One 2015; 10:e0123684. [PMID: 25860483 PMCID: PMC4393253 DOI: 10.1371/journal.pone.0123684] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/02/2015] [Indexed: 01/05/2023] Open
Abstract
Background Pancreatic adenocarcinoma is a lethal disease with 5-year survival of less than 5%. 5-fluorouracil (5-FU) is a principal first-line therapy, but treatment only extends survival modestly and is seldom curative. Drug resistance and disease recurrence is typical and there is a pressing need to overcome this. To investigate acquired 5-FU resistance in pancreatic adenocarcinoma, we established chemoresistant monoclonal cell lines from the Panc 03.27 cell line by long-term exposure to increasing doses of 5-FU. Results 5-FU-resistant cell lines exhibited increased expression of markers associated with multidrug resistance explaining their reduced sensitivity to 5-FU. In addition, 5-FU-resistant cell lines showed alterations typical for an epithelial-to-mesenchymal transition (EMT), including upregulation of mesenchymal markers and increased invasiveness. Microarray analysis revealed the L1CAM pathway as one of the most upregulated pathways in the chemoresistant clones, and a significant upregulation of L1CAM was seen on the RNA and protein level. In pancreatic cancer, expression of L1CAM is associated with a chemoresistant and migratory phenotype. Using esiRNA targeting L1CAM, or by blocking the extracellular part of L1CAM with antibodies, we show that the increased invasiveness observed in the chemoresistant cells functionally depends on L1CAM. Using esiRNA targeting β-catenin and/or Slug, we demonstrate that in the chemoresistant cell lines, L1CAM expression depends on Slug rather than β-catenin. Conclusion Our findings establish Slug-induced L1CAM expression as a mediator of a chemoresistant and migratory phenotype in pancreatic adenocarcinoma cells.
Collapse
|
50
|
DU Y, Zhang H, Jiang Z, Huang G, Lu W, Wang H. Expression of L1 protein correlates with cluster of differentiation 24 and integrin β1 expression in gastrointestinal stromal tumors. Oncol Lett 2015; 9:2595-2602. [PMID: 26137113 DOI: 10.3892/ol.2015.3096] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 01/29/2015] [Indexed: 12/11/2022] Open
Abstract
The present study examined 66 cases of gastrointestinal stromal tumors (GISTs), 20 cases of smooth muscle tumors, 20 cases of schwannomas and 20 cases of normal gastric tissues in order to analyze the expression of L1, cluster of differentiation (CD)24 and integrin β1 by immunohistochemical staining. Patients were subjected to follow-up, and survival data were evaluated. L1 expression was detected in 57.6% of GIST cases; this was a significantly higher percentage compared with that found in the smooth muscle tumor cases or the normal control group. CD24 and integrin β1 were also expressed at significantly higher levels in the GIST cases than in the normal control group, although no significant difference was found in the expression levels of these proteins in smooth muscle tumor or schwannoma cases. These higher levels of L1 and integrin β1 expression were associated with an increased risk of invasive GIST, and were significantly positively correlated with Ki-67 expression. CD24 expression was not associated with the risk of GIST invasion or Ki-67 expression. There were positive correlations between L1, CD24 and integrin β1 expression; however, these had no significant association with patient survival. Therefore, L1 alone or in conjunction with CD24 (L1 + CD24), or integrin β1 (L1 + integrin β1) can be considered a valuable indicator for the differential diagnosis of GIST. Furthermore, L1 and integrin β1 can be used alone or in combination to evaluate the biological behavior of GISTs. Future studies are required to evaluate the prognostic value of these markers.
Collapse
Affiliation(s)
- Yue DU
- Department of Public Health, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Haihong Zhang
- Department of Public Health, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Zhongmin Jiang
- Department of Pathology, Tianjin Fifth Central Hospital, Tianjin 300450, P.R. China
| | - Guowei Huang
- Department of Public Health, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Wenli Lu
- Department of Public Health, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Hesheng Wang
- Department of Public Health, Tianjin Medical University, Tianjin 300070, P.R. China
| |
Collapse
|