1
|
Huang Y, Cheng N, Zhi Y, Qiao D, Wang Y, Ma M, Ge C, Tao W, Liu W. Phillyrin from Forsythia suspensa suppresses the proliferation, angiogenesis, and metastasis of colorectal cancer via targeting CD147. JOURNAL OF ETHNOPHARMACOLOGY 2025; 347:119759. [PMID: 40210179 DOI: 10.1016/j.jep.2025.119759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/26/2025] [Accepted: 04/05/2025] [Indexed: 04/12/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Forsythia suspensa (Thunb.) Vahl, a traditional Chinese herbal medicine, is widely used in clinical practice. Phillyrin (PHN), a major bioactive component of Forsythia suspensa, exhibits significant anti-inflammatory, neuroprotective, and antibacterial properties, offering potential for colorectal cancer (CRC) prevention and treatment. AIM OF THE STUDY This study aimed to clarify the effects of PHN on CRC progression, focusing on epithelial-mesenchymal transition (EMT) and angiogenesis, to elucidate the underlying molecular mechanisms involving CD147. MATERIALS AND METHODS In vitro, cell viability and colony formation were conducted to detect the inhibition of PHN on CRC cells. Wound healing and Transwell assays were used to detect the migration and invasion. PCR, Western blot and ELISA were performed to clarify the relevant molecular levels. Overexpression plasmids were constructed to regulate the target molecule for mechanism research. In vivo, subcutaneous xenograft and lung metastasis models evaluated PHN's anti-cancer effects including histological and immunohistochemical (IHC) analysis. RESULTS PHN inhibited CRC cell proliferation, migration, and invasion in vitro, downregulating CD147 expression, while CD147 overexpression reversed the effects of PHN. In vivo, PHN significantly suppressed tumor growth and lung metastasis, reducing VEGFA, N-Cadherin, Snail1, and MMP-9 expression, and increasing E-Cadherin levels. CONCLUSION These findings indicated that PHN suppressed the proliferation and metastasis of CRC via regulating CD147-mediated EMT and angiogenesis. PHN may be a promising therapeutic agent for CRC treatment in clinic, and CD147 may be a potential target for drug development.
Collapse
Affiliation(s)
- Yuzhen Huang
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Nan Cheng
- Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China
| | - Yingru Zhi
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Dan Qiao
- Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China
| | - Yan Wang
- Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China
| | - Mengqing Ma
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Chun Ge
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Weiwei Tao
- Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China.
| | - Wanli Liu
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
2
|
Xiaoxia F, Rui L, Meiru C, Lu Y, Ying J. CD147 regulates the Rap1 signaling pathway to promote proliferation, migration, and invasion, and inhibit apoptosis in colorectal cancer cells. Sci Rep 2025; 15:13647. [PMID: 40254691 PMCID: PMC12009992 DOI: 10.1038/s41598-025-98266-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 04/10/2025] [Indexed: 04/22/2025] Open
Abstract
The malignant progression of colorectal cancer (CRC) is intimately associated with the abnormal regulation of transmembrane glycoprotein CD147. However, the molecular mechanism via the Rap1/Rap1GAP signaling axis has not been elucidated. This study, through integrated bioinformatics analysis, discovered that the expression of CD147 in CRC tissues was significantly higher than that in adjacent tissues, and patients with high expression had a shorter overall survival. Immunohistochemistry and Western blot confirmed that the expression level of CD147 protein in CRC tissues was higher than that in adjacent tissues. Moreover, qRT-PCR verified a positive correlation between the expressions of CD147 and Rap1. Immunofluorescence clearly indicated that CD147 was specifically enriched in the cell membrane and cytoplasm of SW620 cells. The knockdown of CD147 mediated by shRNA could inhibit the proliferation of HCT116/SW620 cells, induce apoptosis, and weaken the migration and invasion capabilities. The mechanism involved the downregulation of c-Myc, Bcl-2 and the upregulation of Bax, E-cadherin. The mechanistic study found that the knockdown of CD147 increased the expression of Rap1GAP and inhibited Rap1 activity. Overexpression of Rap1 could reverse the inhibitory effects of CD147 knockdown on proliferation, apoptosis, and EMT phenotypes. This study revealed that CD147 upregulated Rap1 expression while inhibiting Rap1GAP, thereby maintaining Rap1 activity and driving the malignant progression of CRC through the c-Myc/Bcl-2/Bax axis and EMT program, providing experimental evidence for precise treatment targeting the CD147-Rap1 signaling axis.
Collapse
Affiliation(s)
- Fu Xiaoxia
- Department of Pathology, Xinzhou Hospital, Shanxi Medical University, Xinzhou, 034000, China
| | - Li Rui
- Clinical Discipline Building Center, Shanxi Medical University, Taiyuan, 030001, China
| | - Chen Meiru
- Department of Pathology, Xinzhou Hospital, Shanxi Medical University, Xinzhou, 034000, China
| | - Yuan Lu
- Department of Pathology, Xinzhou Hospital, Shanxi Medical University, Xinzhou, 034000, China
| | - Jin Ying
- Department of Pathology, Xinzhou Hospital, Shanxi Medical University, Xinzhou, 034000, China.
| |
Collapse
|
3
|
Tummers FHMP, de Koning R, Bazelmans MK, Jansen FW, Blikkendaal MD, van Vlierberghe RLP, Vahrmeijer AL, Hazelbag HM, Kuppen PJK. Immunohistochemical Evaluation of Potential Biomarkers for Targeted Intraoperative Fluorescence Imaging in Endometriosis: Towards Optimizing Surgical Treatment. Reprod Sci 2024; 31:3705-3718. [PMID: 39373851 PMCID: PMC11611954 DOI: 10.1007/s43032-024-01715-4] [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: 06/18/2024] [Accepted: 09/20/2024] [Indexed: 10/08/2024]
Abstract
Surgical intervention for endometriosis is an important treatment modality, yet incomplete resection resulting from poor visibility of affected tissue and consequently recurrence of disease remains a prevalent challenge. Intra-operative visualization of endometriosis, enabling fluorescence-guided surgery (FGS), could help to optimize surgical treatment. A biomarker, upregulated in endometriosis compared to adjacent tissue, is required to use as a target for FGS. Immunohistochemistry was used to evaluate protein expression of a selection of previously identified potential biomarkers. Ten biomarkers were stained in a large cohort of 84 tissues, both deep and peritoneal endometriosis and tissue without endometriosis, all from patients with confirmed endometriosis. MMP11 and VCAN showed the largest upregulation in endometriosis compared to adjacent tissue and showed a membranous or extracellular staining pattern. MMP11 is a promising target for glandular and stromal visualization, VCAN for stromal visualization only. For both biomarkers, upregulation was high in both peritoneal and deep endometriosis and for patients with and without hormonal medication. Other stained biomarkers showed non-beneficial characteristics based on staining pattern or upregulation. Analysis of all endometriosis samples showed that combined glandular and stromal targeting is expected to result in optimal visualization of endometriosis. Further research is needed to determine whether targeting one biomarker is sufficient for this goal, or if dual targeting is necessary. Development of clinical tracers for VCAN and MMP11 is necessary.
Collapse
Affiliation(s)
| | - Rozemarijn de Koning
- Department of Gynecology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maria K Bazelmans
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Frank Willem Jansen
- Department of Gynecology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Biomechanical Engineering, Delft University of Technology, Delft, The Netherlands
| | - Mathijs D Blikkendaal
- Endometriose in Balans, Haaglanden Medical Center, The Hague, The Netherlands
- Nederlandse Endometriose Kliniek, Reinier de Graaf Hospital, Delft, The Netherlands
| | | | | | | | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
4
|
Lee JW, Ahn H, Yoo ID, Hong SP, Baek MJ, Kang DH, Lee SM. Relationship of FDG PET/CT imaging features with tumor immune microenvironment and prognosis in colorectal cancer: a retrospective study. Cancer Imaging 2024; 24:53. [PMID: 38627864 PMCID: PMC11020988 DOI: 10.1186/s40644-024-00698-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Imaging features of colorectal cancers on 2-deoxy-2-[18F]fluoro-d-glucose (FDG) positron emission tomography/computed tomography (PET/CT) have been considered to be affected by tumor characteristics and tumor immune microenvironment. However, the relationship between PET/CT imaging features and immune reactions in tumor tissue has not yet been fully evaluated. This study investigated the association of FDG PET/CT imaging features in the tumor, bone marrow, and spleen with immunohistochemical results of cancer tissue and recurrence-free survival (RFS) in patients with colorectal cancer. METHODS A total of 119 patients with colorectal cancer who underwent FDG PET/CT for staging work-up and received curative surgical resection were retrospectively enrolled. From PET/CT images, 10 first-order imaging features of primary tumors, including intensity of FDG uptake, volumetric metabolic parameters, and metabolic heterogeneity parameters, as well as FDG uptake in the bone marrow and spleen were measured. The degrees of CD4+, CD8+, and CD163 + cell infiltration and interleukin-6 (IL-6) and matrix metalloproteinase-11 (MMP-11) expression were graded through immunohistochemical analysis of surgical specimens. The relationship between FDG PET/CT imaging features and immunohistochemical results was assessed, and prognostic significance of PET/CT imaging features in predicting RFS was evaluated. RESULTS Correlation analysis with immunohistochemistry findings showed that the degrees of CD4 + and CD163 + cell infiltration and IL-6 and MMP-11 expression were correlated with cancer imaging features on PET/CT. Patients with enhanced inflammatory response in cancer tissue demonstrated increased FDG uptake, volumetric metabolic parameters, and metabolic heterogeneity. FDG uptake in the bone marrow and spleen was positively correlated with the degree of CD163 + cell infiltration and IL-6 expression, respectively. In multivariate survival analysis, the coefficient of variation of FDG uptake in the tumor (p = 0.019; hazard ratio, 0.484 for 0.10 increase) and spleen-to-liver uptake ratio (p = 0.020; hazard ratio, 24.901 for 1.0 increase) were significant independent predictors of RFS. CONCLUSIONS The metabolic heterogeneity of tumors and FDG uptake in the spleen were correlated with tumor immune microenvironment and showed prognostic significance in predicting RFS in patients with colorectal cancer.
Collapse
Affiliation(s)
- Jeong Won Lee
- Department of Nuclear Medicine, Soonchunhyang University Cheonan Hospital, 31 Suncheonhyang 6-gil, Dongnam- gu, 31151, Cheonan, Korea
| | - Hyein Ahn
- Department of Pathology, CHA Gangnam Medical Center, CHA University School of Medicine, 569 Nonhyon-ro, Gangnam-gu, 06135, Seoul, Korea
| | - Ik Dong Yoo
- Department of Nuclear Medicine, Soonchunhyang University Cheonan Hospital, 31 Suncheonhyang 6-gil, Dongnam- gu, 31151, Cheonan, Korea
| | - Sun-Pyo Hong
- Department of Nuclear Medicine, Soonchunhyang University Cheonan Hospital, 31 Suncheonhyang 6-gil, Dongnam- gu, 31151, Cheonan, Korea
| | - Moo-Jun Baek
- Department of Surgery, College of Medicine, Soonchunhyang University Cheonan Hospital, 31 Suncheonhyang 6- gil, Dongnam-gu, 31151, Cheonan, Korea
| | - Dong Hyun Kang
- Department of Colorectal surgery, College of Medicine, Soonchunhyang University Cheonan Hospital, 31 Suncheonhyang 6-gil, Dongnam-gu, 31151, Cheonan, Korea
| | - Sang Mi Lee
- Department of Nuclear Medicine, Soonchunhyang University Cheonan Hospital, 31 Suncheonhyang 6-gil, Dongnam- gu, 31151, Cheonan, Korea.
| |
Collapse
|
5
|
Sallinger K, Gruber M, Müller CT, Bonstingl L, Pritz E, Pankratz K, Gerger A, Smolle MA, Aigelsreiter A, Surova O, Svedlund J, Nilsson M, Kroneis T, El-Heliebi A. Spatial tumour gene signature discriminates neoplastic from non-neoplastic compartments in colon cancer: unravelling predictive biomarkers for relapse. J Transl Med 2023; 21:528. [PMID: 37543577 PMCID: PMC10403907 DOI: 10.1186/s12967-023-04384-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 07/22/2023] [Indexed: 08/07/2023] Open
Abstract
BACKGROUND Opting for or against the administration of adjuvant chemotherapy in therapeutic management of stage II colon cancer remains challenging. Several studies report few survival benefits for patients treated with adjuvant therapy and additionally revealing potential side effects of overtreatment, including unnecessary exposure to chemotherapy-induced toxicities and reduced quality of life. Predictive biomarkers are urgently needed. We, therefore, hypothesise that the spatial tissue composition of relapsed and non-relapsed colon cancer stage II patients reveals relevant biomarkers. METHODS The spatial tissue composition of stage II colon cancer patients was examined by a novel spatial transcriptomics technology with sub-cellular resolution, namely in situ sequencing. A panel of 176 genes investigating specific cancer-associated processes such as apoptosis, proliferation, angiogenesis, stemness, oxidative stress, hypoxia, invasion and components of the tumour microenvironment was designed to examine differentially expressed genes in tissue of relapsed versus non-relapsed patients. Therefore, FFPE slides of 10 colon cancer stage II patients either classified as relapsed (5 patients) or non-relapsed (5 patients) were in situ sequenced and computationally analysed. RESULTS We identified a tumour gene signature that enables the subclassification of tissue into neoplastic and non-neoplastic compartments based on spatial expression patterns obtained through in situ sequencing. We developed a computational tool called Genes-To-Count (GTC), which automates the quantification of in situ signals, accurately mapping their position onto the spatial tissue map and automatically identifies neoplastic and non-neoplastic tissue compartments. The GTC tool was used to quantify gene expression of biological processes upregulated within the neoplastic tissue in comparison to non-neoplastic tissue and within relapsed versus non-relapsed stage II colon patients. Three differentially expressed genes (FGFR2, MMP11 and OTOP2) in the neoplastic tissue compartments of relapsed patients in comparison to non-relapsed patients were identified predicting recurrence in stage II colon cancer. CONCLUSIONS In depth spatial in situ sequencing showed potential to provide a deeper understanding of the underlying mechanisms involved in the recurrence of disease and revealed novel potential predictive biomarkers for disease relapse in colon cancer stage II patients. Our open-access GTC-tool allowed us to accurately capture the tumour compartment and quantify spatial gene expression in colon cancer tissue.
Collapse
Affiliation(s)
- Katja Sallinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Centre, Medical University of Graz, Graz, Austria
- Center for Biomarker Research in Medicine (CBmed), Graz, Austria
| | - Michael Gruber
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Centre, Medical University of Graz, Graz, Austria
| | - Christin-Therese Müller
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Centre, Medical University of Graz, Graz, Austria
| | - Lilli Bonstingl
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Centre, Medical University of Graz, Graz, Austria
- Center for Biomarker Research in Medicine (CBmed), Graz, Austria
| | - Elisabeth Pritz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Centre, Medical University of Graz, Graz, Austria
| | - Karin Pankratz
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Centre, Medical University of Graz, Graz, Austria
| | - Armin Gerger
- Division of Oncology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Maria Anna Smolle
- Department of Orthopaedics and Trauma, Medical University of Graz, Graz, Austria
| | - Ariane Aigelsreiter
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Olga Surova
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, 17165, Solna, Sweden
| | - Jessica Svedlund
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, 17165, Solna, Sweden
- 10x Genomics, Life City, Solnavägen 3H, 113 63, Stockholm, Sweden
| | - Mats Nilsson
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, 17165, Solna, Sweden
| | - Thomas Kroneis
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Centre, Medical University of Graz, Graz, Austria
- Center for Biomarker Research in Medicine (CBmed), Graz, Austria
| | - Amin El-Heliebi
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Centre, Medical University of Graz, Graz, Austria.
- Center for Biomarker Research in Medicine (CBmed), Graz, Austria.
- Biotechmed, Graz, Austria.
| |
Collapse
|
6
|
He L, Kang Q, Chan KI, Zhang Y, Zhong Z, Tan W. The immunomodulatory role of matrix metalloproteinases in colitis-associated cancer. Front Immunol 2023; 13:1093990. [PMID: 36776395 PMCID: PMC9910179 DOI: 10.3389/fimmu.2022.1093990] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/07/2022] [Indexed: 01/22/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are an important class of enzymes in the body that function through the extracellular matrix (ECM). They are involved in diverse pathophysiological processes, such as tumor invasion and metastasis, cardiovascular diseases, arthritis, periodontal disease, osteogenesis imperfecta, and diseases of the central nervous system. MMPs participate in the occurrence and development of numerous cancers and are closely related to immunity. In the present study, we review the immunomodulatory role of MMPs in colitis-associated cancer (CAC) and discuss relevant clinical applications. We analyze more than 300 pharmacological studies retrieved from PubMed and the Web of Science, related to MMPs, cancer, colitis, CAC, and immunomodulation. Key MMPs that interfere with pathological processes in CAC such as MMP-2, MMP-3, MMP-7, MMP-9, MMP-10, MMP-12, and MMP-13, as well as their corresponding mechanisms are elaborated. MMPs are involved in cell proliferation, cell differentiation, angiogenesis, ECM remodeling, and the inflammatory response in CAC. They also affect the immune system by modulating differentiation and immune activity of immune cells, recruitment of macrophages, and recruitment of neutrophils. Herein we describe the immunomodulatory role of MMPs in CAC to facilitate treatment of this special type of colon cancer, which is preceded by detectable inflammatory bowel disease in clinical populations.
Collapse
Affiliation(s)
- Luying He
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Qianming Kang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Ka Iong Chan
- Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, Macao SAR, China
| | - Yang Zhang
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Zhangfeng Zhong
- Macao Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, Macao SAR, China,*Correspondence: Zhangfeng Zhong, ; Wen Tan,
| | - Wen Tan
- School of Pharmacy, Lanzhou University, Lanzhou, China,*Correspondence: Zhangfeng Zhong, ; Wen Tan,
| |
Collapse
|
7
|
Martin‐Morales L, Manzano S, Rodrigo‐Faus M, Vicente‐Barrueco A, Lorca V, Núñez‐Moreno G, Bragado P, Porras A, Caldes T, Garre P, Gutierrez‐Uzquiza A. Germline gain-of-function MMP11 variant results in an aggressive form of colorectal cancer. Int J Cancer 2023; 152:283-297. [PMID: 36093604 PMCID: PMC9827992 DOI: 10.1002/ijc.34289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 06/24/2022] [Accepted: 07/06/2022] [Indexed: 01/12/2023]
Abstract
Matrix metalloproteinase-11 (MMP11) is an enzyme with proteolytic activity against matrix and nonmatrix proteins. Although most MMPs are secreted as inactive proenzymes and are later activated extracellularly, MMP11 is activated intracellularly by furin within the constitutive secretory pathway. It is a key factor in physiological tissue remodeling and its alteration may play an important role in the progression of epithelial malignancies and other diseases. TCGA colon and colorectal adenocarcinoma data showed that upregulation of MMP11 expression correlates with tumorigenesis and malignancy. Here, we provide evidence that a germline variant in the MMP11 gene (NM_005940: c.232C>T; p.(Pro78Ser)), identified by whole exome sequencing, can increase the tumorigenic properties of colorectal cancer (CRC) cells. P78S is located in the prodomain region, which is responsible for blocking MMP11's protease activity. This variant was detected in the proband and all the cancer-affected family members analyzed, while it was not detected in healthy relatives. In silico analyses predict that P78S could have an impact on the activation of the enzyme. Furthermore, our in vitro analyses show that the expression of P78S in HCT116 cells increases tumor cell invasion and proliferation. In summary, our results show that this variant could modify the structure of the MMP11 prodomain, producing a premature or uncontrolled activation of the enzyme that may contribute to an early CRC onset in these patients. The study of this gene in other CRC cases will provide further information about its role in CRC development, which might improve patient treatment in the future.
Collapse
Affiliation(s)
- Lorena Martin‐Morales
- Molecular Oncology LaboratoryHospital Clínico San CarlosMadridSpain,Health Research Institute of the Hospital Clínico San Carlos (IdISSC)MadridSpain,Laboratory of Cancer Stemness, GIGA‐InstituteUniversity of LiegeLiegeBelgium
| | - Sara Manzano
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC)MadridSpain,Department of Biochemistry and Molecular Biology, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain,Biodonostia Health Research InstituteSan Sebastian/DonostiaSpain
| | - Maria Rodrigo‐Faus
- Department of Biochemistry and Molecular Biology, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
| | - Adrian Vicente‐Barrueco
- Department of Biochemistry and Molecular Biology, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain,Center for Cooperative Research in Biosciences (CIC bioGUNE)Basque Research and Technology Alliance (BRTA), Bizkaia Technology ParkDerioSpain
| | - Victor Lorca
- Molecular Oncology LaboratoryHospital Clínico San CarlosMadridSpain,Health Research Institute of the Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - Gonzalo Núñez‐Moreno
- Department of Genetics, Health Research Institute‐Fundación Jiménez Díaz University HospitalUniversidad Autónoma de Madrid (IIS‐FJD, UAM)MadridSpain,Bioinformatics Unit, Health Research Institute‐Fundación Jiménez Díaz University HospitalUniversidad Autónoma de Madrid (IIS‐FJD, UAM)MadridSpain
| | - Paloma Bragado
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC)MadridSpain,Department of Biochemistry and Molecular Biology, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
| | - Almudena Porras
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC)MadridSpain,Department of Biochemistry and Molecular Biology, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
| | - Trinidad Caldes
- Molecular Oncology LaboratoryHospital Clínico San CarlosMadridSpain,Health Research Institute of the Hospital Clínico San Carlos (IdISSC)MadridSpain
| | - Pilar Garre
- Molecular Oncology LaboratoryHospital Clínico San CarlosMadridSpain,Health Research Institute of the Hospital Clínico San Carlos (IdISSC)MadridSpain,Clinical Analysis Service, Molecular Diagnostic UnitIML, Hospital Clínico San CarlosMadridSpain
| | - Alvaro Gutierrez‐Uzquiza
- Health Research Institute of the Hospital Clínico San Carlos (IdISSC)MadridSpain,Department of Biochemistry and Molecular Biology, Facultad de FarmaciaUniversidad Complutense de MadridMadridSpain
| |
Collapse
|
8
|
Li YR, Meng K, Yang G, Liu BH, Li CQ, Zhang JY, Zhang XM. Diagnostic genes and immune infiltration analysis of colorectal cancer determined by LASSO and SVM machine learning methods: a bioinformatics analysis. J Gastrointest Oncol 2022; 13:1188-1203. [PMID: 35837194 PMCID: PMC9274036 DOI: 10.21037/jgo-22-536] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 06/16/2022] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND Genetic factors account for approximately 35% of colorectal cancer risk. The specificity and sensitivity of previous diagnostic biomarkers for colorectal cancer could not meet the need of clinical application. The expanding scale and inherent complexity of biological data have encouraged a growing use of machine learning to build informative and predictive models of the underlying biological processes. The aim of this study is to identify diagnostic genes of colorectal cancer by using machine learning methods. METHODS The GSE41328 and GSE106582 data sets were downloaded from the Gene Expression Omnibus (GEO) database. The gene expression differences between colon cancer and normal tissues were analyzed. The key colorectal cancer genes were screened and validated by Least Absolute Shrinkage and Selection Operator (LASSO) and Support Vector Machine (SVM) regression. Immune cell infiltration and the correlation with the key genes in patients with colon cancer were further analyzed by CIBERSORT. RESULTS Eleven key genes were identified as biomarkers for colon cancer, namely ASCL2, BEST4, CFD, DPEPCFD, FOXQ1, TRIB3, KLF4, MMP7, MMP11, PYY, and PDK4. The mean area under the receiver operating characteristic (ROC) curve (AUC) of all 11 genes for colon cancer diagnosis were 0.94 with a range of 0.91-0.97. In the validation set, the expression of the 11 key genes was significantly different between colon cancer and normal subjects (P<0.05) and the mean AUCs were 0.82 with a range of 0.70-0.88. Immune cell infiltration analyses demonstrated that the relative quantity of plasma cells, T cells, B cells, NK cells, MO, M1, Dendritic cells resting, Mast cells resting, Mast cells activated, and Neutrophils in the tumor group were significantly different to the normal group. CONCLUSIONS ASCL2, BEST4, CFD, DPEPCFD, FOXQ1, TRIB3, KLF4, MMP7, MMP11, PYY, and PDK4 were identified as the key genes for colon cancer diagnosis. These genes are expected to become novel diagnostic markers and targets of new pharmacotherapies for colorectal cancer.
Collapse
Affiliation(s)
- Yan-Rong Li
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Ke Meng
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Guang Yang
- Department of Laboratory, The Red Cross (SEN GONG GENERAL) Hospital of Heilongjiang, Heilongjiang, China
| | - Bao-Hai Liu
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Chu-Qiao Li
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Jia-Yuan Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Xiao-Mei Zhang
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
9
|
Immunoreactivity of humanized single-chain variable fragment against its functional epitope on domain 1 of CD147. Sci Rep 2022; 12:6719. [PMID: 35468972 PMCID: PMC9038914 DOI: 10.1038/s41598-022-10657-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 04/11/2022] [Indexed: 11/08/2022] Open
Abstract
Domain 1 of CD147 participates in matrix metalloproteinase (MMP) production and is a candidate for targeted therapy to prevent cancer invasion and metastasis. A functional mouse anti-CD147 monoclonal antibody, M6-1B9, was found to recognize domain 1 of CD147, and its respective mouse single-chain variable fragment (ScFvM61B9) was subsequently generated. The EDLGS epitope candidate for M6-1B9 was identified using the phage display peptide technique in this study. For future clinical applications, humanized ScFv specific to domain 1 of CD147 (HuScFvM61B9) was partially adopted from the hypervariable sequences of parental mouse ScFvM61B9 and grafted onto suitable human immunoglobulin frameworks. Molecular modelling and simulation were performed in silico to generate the conformational structure of HuScFvM61B9. These results elucidated the amino acid residues that contributed to the interactions between CDRs and the epitope motif. The expressed HuScFvM61B9 specifically interacted with CD147 at the same epitope as the original mAb, M6-1B9, and retained immunoreactivity against CD147 in SupT1 cells. The reactivity of HuScFvM61B9 was confirmed using CD147 knockout Jurkat cells. In addition, the inhibitory effect of HuScFvM61B9 on OKT3-induced T-cell proliferation as M6-1B9 mAb was preserved. As domain 1 is responsible for cancer invasion and metastasis, HuScFvM61B9 would be a candidate for cancer targeted therapy in the future.
Collapse
|
10
|
Huang X, Lan Y, Li E, Li J, Deng Q, Deng X. Diagnostic values of MMP-7, MMP-9, MMP-11, TIMP-1, TIMP-2, CEA, and CA19-9 in patients with colorectal cancer. J Int Med Res 2021; 49:3000605211012570. [PMID: 33942633 PMCID: PMC8144491 DOI: 10.1177/03000605211012570] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/30/2021] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE Colorectal cancer (CRC) is one of the most common and lethal malignancies. The identification of precise and noninvasive biomarkers is urgently needed to aid the early diagnosis and clinical management of CRC. METHODS A total of 112 patients with CRC and 115 healthy control subjects were included in this study. Serum levels of matrix metalloproteinase (MMP)-7, MMP-9, MMP-11, tissue inhibitor of metalloproteinase (TIMP)-1, and TIMP-2 were analyzed by enzyme-linked immunosorbent assay, and carcinoembryonic antigen (CEA) and carbohydrate antigen (CA)19-9 levels were measured using an automatic immunoassay analyzer. RESULTS MMP-7, MMP-9, MMP-11, TIMP-1, TIMP-2, CEA, and CA19-9 levels were all significantly higher in CRC patients compared with healthy controls. MMP-7, TIMP-1, and CEA levels were also closely related to clinicopathologic features in patients with CRC. The combination of serum CEA, MMP-7, and TIMP-1 significantly improved the diagnostic value compared with any single marker (area under the curve 0.858-0.890). Furthermore, a combined detection model including MMP-7, TIMP-1, and CEA improved both the specificity and sensitivity for detecting CRC. CONCLUSIONS The results showed that combined detection of CEA, MMP-7, and TIMP-1 in serum could provide a specific and sensitive biomarker for the diagnosis of CRC.
Collapse
Affiliation(s)
- Xiwen Huang
- Department of Oncology, Cancer Center, Meizhou People’s Hospital
(Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital
Affiliated to Sun Yat-sen University, Meizhou, China
| | - Yongquan Lan
- Department of Oncology, Cancer Center, Meizhou People’s Hospital
(Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital
Affiliated to Sun Yat-sen University, Meizhou, China
| | - En Li
- Department of Oncology, Cancer Center, Meizhou People’s Hospital
(Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital
Affiliated to Sun Yat-sen University, Meizhou, China
| | - Jiaquan Li
- Department of Oncology, Cancer Center, Meizhou People’s Hospital
(Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital
Affiliated to Sun Yat-sen University, Meizhou, China
| | - Qiaoting Deng
- Research and Experimental Center, Meizhou People’s Hospital
(Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital
Affiliated to Sun Yat-sen University, Meizhou, China
- Guangdong Provincial Key Laboratory of Precision Medicine and
Clinical Translational Research of Hakka Population, Meizhou, China
| | - Xunwei Deng
- Research and Experimental Center, Meizhou People’s Hospital
(Huangtang Hospital), Meizhou Academy of Medical Sciences, Meizhou Hospital
Affiliated to Sun Yat-sen University, Meizhou, China
- Guangdong Provincial Key Laboratory of Precision Medicine and
Clinical Translational Research of Hakka Population, Meizhou, China
| |
Collapse
|
11
|
Fonseca AS, Ramão A, Bürger MC, de Souza JES, Zanette DL, de Molfetta GA, de Araújo LF, de Barros E Lima Bueno R, Aguiar GM, Plaça JR, Alves CDP, Dos Santos ARD, Vidal DO, Silva GEB, Panepucci RA, Peria FM, Feres O, da Rocha JJR, Zago MA, Silva WA. ETV4 plays a role on the primary events during the adenoma-adenocarcinoma progression in colorectal cancer. BMC Cancer 2021; 21:207. [PMID: 33648461 PMCID: PMC7919324 DOI: 10.1186/s12885-021-07857-x] [Citation(s) in RCA: 9] [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: 10/19/2020] [Accepted: 01/31/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common cancers worldwide; it is the fourth leading cause of death in the world and the third in Brazil. Mutations in the APC, DCC, KRAS and TP53 genes have been associated with the progression of sporadic CRC, occurring at defined pathological stages of the tumor progression and consequently modulating several genes in the corresponding signaling pathways. Therefore, the identification of gene signatures that occur at each stage during the CRC progression is critical and can present an impact on the diagnosis and prognosis of the patient. In this study, our main goal was to determine these signatures, by evaluating the gene expression of paired colorectal adenoma and adenocarcinoma samples to identify novel genetic markers in association to the adenoma-adenocarcinoma stage transition. METHODS Ten paired adenoma and adenocarcinoma colorectal samples were subjected to microarray gene expression analysis. In addition, mutations in APC, KRAS and TP53 genes were investigated by DNA sequencing in paired samples of adenoma, adenocarcinoma, normal tissue, and peripheral blood from ten patients. RESULTS Gene expression analysis revealed a signature of 689 differentially expressed genes (DEG) (fold-change> 2, p< 0.05), between the adenoma and adenocarcinoma paired samples analyzed. Gene pathway analysis using the 689 DEG identified important cancer pathways such as remodeling of the extracellular matrix and epithelial-mesenchymal transition. Among these DEG, the ETV4 stood out as one of the most expressed in the adenocarcinoma samples, further confirmed in the adenocarcinoma set of samples from the TCGA database. Subsequent in vitro siRNA assays against ETV4 resulted in the decrease of cell proliferation, colony formation and cell migration in the HT29 and SW480 colorectal cell lines. DNA sequencing analysis revealed KRAS and TP53 gene pathogenic mutations, exclusively in the adenocarcinomas samples. CONCLUSION Our study identified a set of genes with high potential to be used as biomarkers in CRC, with a special emphasis on the ETV4 gene, which demonstrated involvement in proliferation and migration.
Collapse
Affiliation(s)
- Aline Simoneti Fonseca
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av Bandeirantes, 3900, CEP: 14049-900, Monte Alegre, Ribeirão Preto, SP, Brazil.
- Center for Cell Based Therapy and National Institute of Science and Technology in Stem Cell and Cell Therapy, Ribeirão Preto, SP, Brazil.
- Center for Integrative Systems Biology - CISBi, NAP/USP, Ribeirão Preto, SP, Brazil.
- Research Institute Pelé Pequeno Príncipe, Av Silva Jardim, 1632, CEP: 80250-060, Água Verde, Curitiba, PR, Brazil.
| | - Anelisa Ramão
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av Bandeirantes, 3900, CEP: 14049-900, Monte Alegre, Ribeirão Preto, SP, Brazil
- Center for Cell Based Therapy and National Institute of Science and Technology in Stem Cell and Cell Therapy, Ribeirão Preto, SP, Brazil
| | - Matheus Carvalho Bürger
- Center for Cell Based Therapy and National Institute of Science and Technology in Stem Cell and Cell Therapy, Ribeirão Preto, SP, Brazil
| | - Jorge Estefano Santana de Souza
- Center for Cell Based Therapy and National Institute of Science and Technology in Stem Cell and Cell Therapy, Ribeirão Preto, SP, Brazil
| | - Dalila Lucíola Zanette
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av Bandeirantes, 3900, CEP: 14049-900, Monte Alegre, Ribeirão Preto, SP, Brazil
- Center for Cell Based Therapy and National Institute of Science and Technology in Stem Cell and Cell Therapy, Ribeirão Preto, SP, Brazil
- Center for Integrative Systems Biology - CISBi, NAP/USP, Ribeirão Preto, SP, Brazil
- Laboratory of Applied Science and Technology in Health (LASTH), Instituto Carlos Chagas, Fundação Oswaldo Cruz, Curitiba, PR, Brazil
| | - Greice Andreotti de Molfetta
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av Bandeirantes, 3900, CEP: 14049-900, Monte Alegre, Ribeirão Preto, SP, Brazil
- Center for Cell Based Therapy and National Institute of Science and Technology in Stem Cell and Cell Therapy, Ribeirão Preto, SP, Brazil
- Center for Integrative Systems Biology - CISBi, NAP/USP, Ribeirão Preto, SP, Brazil
| | - Luiza Ferreira de Araújo
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av Bandeirantes, 3900, CEP: 14049-900, Monte Alegre, Ribeirão Preto, SP, Brazil
- Center for Cell Based Therapy and National Institute of Science and Technology in Stem Cell and Cell Therapy, Ribeirão Preto, SP, Brazil
- Center for Integrative Systems Biology - CISBi, NAP/USP, Ribeirão Preto, SP, Brazil
| | - Rafaela de Barros E Lima Bueno
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av Bandeirantes, 3900, CEP: 14049-900, Monte Alegre, Ribeirão Preto, SP, Brazil
- Center for Cell Based Therapy and National Institute of Science and Technology in Stem Cell and Cell Therapy, Ribeirão Preto, SP, Brazil
| | - Graziela Moura Aguiar
- Center for Cell Based Therapy and National Institute of Science and Technology in Stem Cell and Cell Therapy, Ribeirão Preto, SP, Brazil
| | - Jessica Rodrigues Plaça
- Center for Cell Based Therapy and National Institute of Science and Technology in Stem Cell and Cell Therapy, Ribeirão Preto, SP, Brazil
| | - Cleidson de Pádua Alves
- Center for Cell Based Therapy and National Institute of Science and Technology in Stem Cell and Cell Therapy, Ribeirão Preto, SP, Brazil
| | - Anemari Ramos Dinarte Dos Santos
- Center for Cell Based Therapy and National Institute of Science and Technology in Stem Cell and Cell Therapy, Ribeirão Preto, SP, Brazil
| | - Daniel Onofre Vidal
- Center for Cell Based Therapy and National Institute of Science and Technology in Stem Cell and Cell Therapy, Ribeirão Preto, SP, Brazil
| | - Gyl Eanes Barros Silva
- Laboratory of Immunofluorescence and Electron Microscopy (LIME), Presidente Dutra University Hospital (HUUFMA), São Luís, MA, Brazil
| | - Rodrigo Alexandre Panepucci
- Center for Cell Based Therapy and National Institute of Science and Technology in Stem Cell and Cell Therapy, Ribeirão Preto, SP, Brazil
| | - Fernanda Maris Peria
- Departament of Medical Clinic, Medical School of Ribeirão Preto, University of São Paulo, USP, Ribeirão Preto, SP, Brazil
| | - Omar Feres
- Department of Surgery and Anatomy, School of Medicine of Ribeirão Preto, University of São Paulo, Sao Paulo, Brazil
| | | | - Marco Antonio Zago
- Center for Cell Based Therapy and National Institute of Science and Technology in Stem Cell and Cell Therapy, Ribeirão Preto, SP, Brazil
| | - Wilson Araújo Silva
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Av Bandeirantes, 3900, CEP: 14049-900, Monte Alegre, Ribeirão Preto, SP, Brazil.
- Center for Cell Based Therapy and National Institute of Science and Technology in Stem Cell and Cell Therapy, Ribeirão Preto, SP, Brazil.
- Center for Integrative Systems Biology - CISBi, NAP/USP, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
12
|
Akhtar MF, Saleem A, Rasul A, Faran Ashraf Baig MM, Bin-Jumah M, Abdel Daim MM. Anticancer natural medicines: An overview of cell signaling and other targets of anticancer phytochemicals. Eur J Pharmacol 2020; 888:173488. [PMID: 32805253 DOI: 10.1016/j.ejphar.2020.173488] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 07/23/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023]
Abstract
Therapies of cancer are as diverse as multifaceted the cancer is. Anticancer drugs include, but not limited to synthetic, semisynthetic and natural drugs and monoclonal antibodies. A recent decline in new drug development has led to the rebirth of herbal therapeutics in the form of dietary supplements and botanical preparations. Medicinal plants comprise of complex phytochemicals due to vast biosynthetic capacity. A wide array of phytochemicals has been pharmacologically evaluated for their chemo-preventive and chemotherapeutic potential for several decades. These phytochemicals target cancer at diverse sites such as apoptotic pathways, genetic and epigenetic mutations, damage to deoxyribonucleic acid, production of reactive oxygen species, autophagy, invasion and metastasis of cancer cells, and modulation of cell signaling through Janus-activated kinase/Signal transducer and activator of transcription, Notch, mitogen-activated protein kinase/Extracellular signal-regulated kinase, phosphatidylinositol 3-kinase/Protein kinase B/mammalian target of rapamycin, Nuclear factor kappa B, Wingless-related integration site and Transforming growth factor β pathways. This review focuses on the therapeutic targets of anticancer and chemo-preventive phytochemicals and their mode of action.
Collapse
Affiliation(s)
- Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Pakistan.
| | - Ammara Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Azhar Rasul
- Department of Zoology, Government College University Faisalabad, Faisalabad, Pakistan
| | | | - May Bin-Jumah
- Biology Department, College of Science, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mohamed M Abdel Daim
- Department of Zoology, College of Science, King Saud University, 2455, Riyadh, 11451, Saudi Arabia; Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, 41522, Egypt
| |
Collapse
|
13
|
Piskór BM, Przylipiak A, Dąbrowska E, Niczyporuk M, Ławicki S. Matrilysins and Stromelysins in Pathogenesis and Diagnostics of Cancers. Cancer Manag Res 2020; 12:10949-10964. [PMID: 33154674 PMCID: PMC7608139 DOI: 10.2147/cmar.s235776] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 09/18/2020] [Indexed: 12/23/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are endopeptidases which are widely studied in terms of their role in the physiological and pathological processes in the organism. In this article, we consider usefulness of matrilysins and stromelysins in pathogenesis and diagnostic of the most common malignancies in the world, e.g., lung, breast, prostate, and colorectal cancers. In all of the mentioned cancers, matrilysins and stromelysins have a pivotal role in their development and also may have diagnostic utility. Influence to the cancerous process is connected with specific dependencies between these enzymes and components of the extracellular matrix (ECM), non-matrix components like cell surface components. All the information provided below allows to take a closer look at matrilysins and stromelysins and their functions in the cancer development.
Collapse
Affiliation(s)
- Barbara Maria Piskór
- Department of Aesthetic Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Andrzej Przylipiak
- Department of Aesthetic Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Emilia Dąbrowska
- Department of Aesthetic Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Marek Niczyporuk
- Department of Aesthetic Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Sławomir Ławicki
- Department of Population Medicine and Civilization Diseases Prevention, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
14
|
Landras A, Reger de Moura C, Jouenne F, Lebbe C, Menashi S, Mourah S. CD147 Is a Promising Target of Tumor Progression and a Prognostic Biomarker. Cancers (Basel) 2019; 11:cancers11111803. [PMID: 31744072 PMCID: PMC6896083 DOI: 10.3390/cancers11111803] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/13/2019] [Accepted: 11/13/2019] [Indexed: 12/18/2022] Open
Abstract
Microenvironment plays a crucial role in tumor development and progression. Cancer cells modulate the tumor microenvironment, which also contribute to resistance to therapy. Identifying biomarkers involved in tumorigenesis and cancer progression represents a great challenge for cancer diagnosis and therapeutic strategy development. CD147 is a glycoprotein involved in the regulation of the tumor microenvironment and cancer progression by several mechanisms—in particular, by the control of glycolysis and also by its well-known ability to induce proteinases leading to matrix degradation, tumor cell invasion, metastasis and angiogenesis. Accumulating evidence has demonstrated the role of CD147 expression in tumor progression and prognosis, suggesting it as a relevant tumor biomarker for cancer diagnosis and prognosis, as well as validating its potential as a promising therapeutic target in cancers.
Collapse
Affiliation(s)
- Alexandra Landras
- INSERM UMRS 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), University of Paris, 75010 Paris, France; (A.L.); (C.R.d.M.); (F.J.); (C.L.); (S.M.)
| | - Coralie Reger de Moura
- INSERM UMRS 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), University of Paris, 75010 Paris, France; (A.L.); (C.R.d.M.); (F.J.); (C.L.); (S.M.)
- Pharmacogenomics Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Saint Louis Hospital, 75010 Paris, France
| | - Fanelie Jouenne
- INSERM UMRS 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), University of Paris, 75010 Paris, France; (A.L.); (C.R.d.M.); (F.J.); (C.L.); (S.M.)
- Pharmacogenomics Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Saint Louis Hospital, 75010 Paris, France
| | - Celeste Lebbe
- INSERM UMRS 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), University of Paris, 75010 Paris, France; (A.L.); (C.R.d.M.); (F.J.); (C.L.); (S.M.)
- Dermatology Department and Centre d’Investigation Clinique (CIC), Assistance Publique-Hôpitaux de Paris (AP-HP), Saint Louis Hospital, 75010 Paris, France
| | - Suzanne Menashi
- INSERM UMRS 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), University of Paris, 75010 Paris, France; (A.L.); (C.R.d.M.); (F.J.); (C.L.); (S.M.)
- Pharmacogenomics Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Saint Louis Hospital, 75010 Paris, France
| | - Samia Mourah
- INSERM UMRS 976, Team 1, Human Immunology Pathophysiology & Immunotherapy (HIPI), University of Paris, 75010 Paris, France; (A.L.); (C.R.d.M.); (F.J.); (C.L.); (S.M.)
- Pharmacogenomics Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Saint Louis Hospital, 75010 Paris, France
- Correspondence: ; Tel.: +33-1-42-49-48-85
| |
Collapse
|
15
|
Eiro N, Cid S, Fernández B, Fraile M, Cernea A, Sánchez R, Andicoechea A, DeAndrés Galiana EJ, González LO, Fernández‐Muñiz Z, Fernández‐Martínez JL, Vizoso FJ. MMP11 expression in intratumoral inflammatory cells in breast cancer. Histopathology 2019; 75:916-930. [DOI: 10.1111/his.13956] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/18/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Noemi Eiro
- Research Unit Fundación Hospital de Jove Gijón Spain
| | - Sandra Cid
- Research Unit Fundación Hospital de Jove Gijón Spain
| | - Berta Fernández
- Department of Surgery Hospital Universitario Central de Asturias Oviedo Spain
| | - Maria Fraile
- Research Unit Fundación Hospital de Jove Gijón Spain
| | - Ana Cernea
- Department of Mathematics Group of Inverse Problems, Optimization and Machine Learning University of Oviedo Oviedo Spain
| | - Rosario Sánchez
- Department of Surgery Fundación Hospital de Jove Gijón Spain
| | | | - Enrique J DeAndrés Galiana
- Department of Mathematics Group of Inverse Problems, Optimization and Machine Learning University of Oviedo Oviedo Spain
- Department of Informatics and Computer Science University of Oviedo Oviedo Spain
| | - Luis O González
- Department of Anatomical Pathology Fundación Hospital de Jove Gijón Spain
| | - Zulima Fernández‐Muñiz
- Department of Mathematics Group of Inverse Problems, Optimization and Machine Learning University of Oviedo Oviedo Spain
| | - Juan L Fernández‐Martínez
- Department of Mathematics Group of Inverse Problems, Optimization and Machine Learning University of Oviedo Oviedo Spain
| | - Francisco J Vizoso
- Research Unit Fundación Hospital de Jove Gijón Spain
- Department of Surgery Fundación Hospital de Jove Gijón Spain
| |
Collapse
|
16
|
Guo F, Li X, Yao G, Zeng G, Yu L. Correlation between 18F-FDG maximum standardized uptake value with CD147 expression in lung adenocarcinomas: a retrospective study. PeerJ 2019; 7:e7635. [PMID: 31565568 PMCID: PMC6741284 DOI: 10.7717/peerj.7635] [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: 01/19/2019] [Accepted: 08/06/2019] [Indexed: 11/20/2022] Open
Abstract
Background The pro-tumoral action of the cluster of differentiation 147 (CD147), which is associated with the chemotherapy resistance of lung adenocarcinoma, is partly due to accelerated tumor cell glycolysis. 18F-fluorodeoxyglucose positron emission tomography (18F-FDG PET) metabolic parameters included maximal standardized uptake value (SUVmax), mean standardized uptake value (SUVmean), metabolic tumor volume (MTV), and total lesion glycolysis (TLG), which are non-invasive markers of the glucose metabolism of tumor cells in vivo. This study aimed to clarify the correlation between PET metabolic parameters and CD147 expression, and to evaluate the prognostic value of CD147 expression in resectable lung adenocarcinoma patients. Methods A total of 89 lung adenocarcinoma chemotherapy-naive patients who underwent 18F-fluorodeoxyglucose positron emission tomography and computerized tomography scan before pulmonary surgery were retrospectively analyzed. The PET metabolic parameters were calculated by 18F-FDG PET imaging, and CD147 expression was analyzed by immunohistochemistry. SUVmax, SUVmean, MTV, and TLG compared for their performance in predicting the expression of CD147 were illustrated with statistical analysis. All patients were then followed-up for survival analysis. Results The SUVmax was significantly correlated with the CD147 expression and was the primary predictor for the CD147 expression of lung adenocarcinoma. A cut-off value of the SUVmax, 9.77 allowed 85.1% sensitivity and 64.3% specificity for predicting the CD147 positive lung adenocarcinoma. CD147 expression was correlated with tumor differentiation and metastasis. Univariate survival analysis showed that CD147 expression was significantly associated with a shorter overall survival (OS) time. Multivariate analysis revealed that CD147 was an independent prognostic factor of lung adenocarcinoma patients. Conclusion The SUVmax of a primary tumor measured with 18F-FDG PET may be a simple and non-invasive marker for predicting CD147 expression in lung adenocarcinoma. CD147 is an independent prognostic factor related to OS of postoperative lung adenocarcinoma patients.
Collapse
Affiliation(s)
- Fei Guo
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xueyan Li
- Department of PET/CT, Harbin Medical University Cancer Hospital, Harbin, China
| | - Guodong Yao
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Guangchun Zeng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Lijuan Yu
- Department of PET/CT, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
17
|
Bozhenko VK, Stanojevic US, Trotsenko ID, Zakharenko MV, Kiseleva YY, Solodkiy VA. [Comparison of matrix proteinase mRNA expression in morphologically normal, neoplastic, and metastatic colon tissue and colon biopsies from healthy donors]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2019; 64:46-52. [PMID: 29460834 DOI: 10.18097/pbmc20186401046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Matrix metalloproteinases (MMPs) responsible for the extracellular matrix remodeling, the activation of various growth factors, and angiogenesis play an important role in the colorectal cancer (CRC) development. In the present work the comparative analysis of MMP-7, -8, -9, and -11 mRNA as well mRNA of the Ki-67 proliferation marker in tissue samples obtained from CRC patients and healthy individuals. Employing the real time PCR method the expression levels of several MMPs (MMP-7, -8, -9, and -11) and cell proliferation marker, Ki-67, were simultaneously measured in 256 tissue samples obtained from 112 patients with CRC: 112 samples of the primary tumor (CRC), 112 samples of the most distant border of morphologically normal colonic mucosa (MNT), 16 samples of liver metastases) and from 16 healthy volunteers who underwent colonoscopy and biopsy. The expression of both MMPs studied and Ki-67 was found to be elevated in CRC primary tumors and liver metastases compared with the normal mucosa. CRC tumor and metastatic cells exhibited similar proliferative activity. The metastases are characterized by the highest cross-correlation of MMPs among tissue types tested. For the first time it was shown that normal mucosa from healthy individuals and CRC patients varied in the MMP-8 expression level. They also had dissimilar MMP correlation patterns thus suggesting that epithelial cells adjusted to CRC tumor differ from mucosal epithelial cells of healthy individuals.
Collapse
Affiliation(s)
- V K Bozhenko
- Russian Scientific Center of Roentgenoradiology, Moscow, Russia
| | - U S Stanojevic
- Russian Scientific Center of Roentgenoradiology, Moscow, Russia
| | - I D Trotsenko
- Russian Scientific Center of Roentgenoradiology, Moscow, Russia; Peoples' Friendship University of Russia, Moscow, Russia
| | - M V Zakharenko
- Russian Scientific Center of Roentgenoradiology, Moscow, Russia
| | - Y Y Kiseleva
- Russian Scientific Center of Roentgenoradiology, Moscow, Russia
| | - V A Solodkiy
- Russian Scientific Center of Roentgenoradiology, Moscow, Russia
| |
Collapse
|
18
|
Kumar D, Vetrivel U, Parameswaran S, Subramanian KK. Structural insights on druggable hotspots in CD147: A bull's eye view. Life Sci 2019; 224:76-87. [DOI: 10.1016/j.lfs.2019.03.044] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/11/2019] [Accepted: 03/19/2019] [Indexed: 12/13/2022]
|
19
|
Nasry WHS, Rodriguez-Lecompte JC, Martin CK. Role of COX-2/PGE2 Mediated Inflammation in Oral Squamous Cell Carcinoma. Cancers (Basel) 2018; 10:cancers10100348. [PMID: 30248985 PMCID: PMC6211032 DOI: 10.3390/cancers10100348] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/16/2018] [Accepted: 09/20/2018] [Indexed: 12/24/2022] Open
Abstract
A significant amount of research indicates that the cyclooxygenase/prostaglandin E2 (PGE2) pathway of inflammation contributes to the development and progression of a variety of cancers, including squamous cell carcinoma of the oral cavity and oropharynx (OSCC). Although there have been promising results from studies examining the utility of anti-inflammatory drugs in the treatment of OSCC, this strategy has been met with only variable success and these drugs are also associated with toxicities that make them inappropriate for some OSCC patients. Improved inflammation-targeting therapies require continued study of the mechanisms linking inflammation and progression of OSCC. In this review, a synopsis of OSCC biology will be provided, and recent insights into inflammation related mechanisms of OSCC pathobiology will be discussed. The roles of prostaglandin E2 and cluster of differentiation factor 147 (CD147) will be presented, and evidence for their interactions in OSCC will be explored. Through continued investigation into the protumourigenic pathways of OSCC, more treatment modalities targeting inflammation-related pathways can be designed with the hope of slowing tumour progression and improving patient prognosis in patients with this aggressive form of cancer.
Collapse
Affiliation(s)
- Walaa Hamed Shaker Nasry
- Department of Pathology and Microbiology, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada.
| | - Juan Carlos Rodriguez-Lecompte
- Department of Pathology and Microbiology, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada.
| | - Chelsea K Martin
- Department of Pathology and Microbiology, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada.
| |
Collapse
|
20
|
Manning S, Danielson KM. The immunomodulatory role of tumor-derived extracellular vesicles in colorectal cancer. Immunol Cell Biol 2018; 96:733-741. [PMID: 29575270 DOI: 10.1111/imcb.12038] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/08/2018] [Accepted: 03/09/2018] [Indexed: 12/15/2022]
Abstract
Colorectal cancer (CRC) is one of the most prevalent cancers worldwide with rising mortality rates predicted in the coming decades. In light of this, there is a continued need for improvement in our understanding of CRC biology and the development of novel treatment options. Tumor-derived extracellular vesicles (tEVs) have emerged as both novel cancer biomarkers and functional mediators of carcinogenesis. tEVs are released by tumor cells in abundance and play an important role in mediating tumor cell-immune cell interactions in the tumor microenvironment. Furthermore, tEVs are released into the circulation in humans where they could also interact with circulating immune cells. This review aims to describe CRC-specific tEVs and what is currently known about their role in immunomodulation. In particular, we discuss the ability of CRC-derived tEVs to affect monocyte differentiation into macrophages and dendritic cells, and their effects on T-cell viability and activity. Finally, the potential for tEVs in the development of immunotherapies will be discussed.
Collapse
Affiliation(s)
- Stephanie Manning
- Department of Surgery and Anaesthesia, University of Otago, Wellington, New Zealand
| | - Kirsty M Danielson
- Department of Surgery and Anaesthesia, University of Otago, Wellington, New Zealand
| |
Collapse
|
21
|
Shang G, Mi Y, Mei Y, Wang G, Wang Y, Li X, Wang Y, Li Y, Zhao G. MicroRNA-192 inhibits the proliferation, migration and invasion of osteosarcoma cells and promotes apoptosis by targeting matrix metalloproteinase-11. Oncol Lett 2018; 15:7265-7272. [PMID: 29731885 DOI: 10.3892/ol.2018.8239] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 01/29/2018] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression during stem cell growth, proliferation and differentiation. miRNAs are also involved in the development and progression of a number of cancer types, including osteosarcoma (OS). miR-192 is significantly downregulated in various tumors, including lung, bladder and rectal cancer. miR-192 expression is associated with the migration and invasion of OS cells. However, the expression of miR-192 and its effects on the development of OS have not been reported. In the present study, the involvement of miR-192 and its molecular mechanisms in the development of OS was investigated. The results indicate that miR-192 expression was significantly downregulated in OS tissues compared with non-tumor tissues (P<0.05). Next, a miR-192 agomir was transfected into the OS cell line MG-63 to upregulate miR-192. The effects of miR-192 overexpression were then investigated by examining cell proliferation, apoptosis, migration and invasion. Matrix metalloproteinase (MMP)-11 belongs to a family of nine or more highly homologous Zn2+-endopeptidases. It was demonstrated that the mRNA and protein expression of MMP-11 were upregulated in OS tissues compared with non-tumor tissues (P<0.05). MMP-11 was predicted by TargetScan and miRanda as a miR-192 target, which was confirmed by western blotting and dual-luciferase assays. Finally, it was demonstrated that the overexpression of miR-192 was able to downregulate MMP-11 expression and reduce proliferation, migration and invasion, and promote apoptosis in OS cells. Together, these data indicate that miR-192 may be a tumor suppressor that inhibits the progression and invasion of OS by targeting MMP-11. Therefore, miR-192 may be useful for the diagnosis and treatment of OS.
Collapse
Affiliation(s)
- Guowei Shang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yang Mi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yingwu Mei
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Guanghui Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yadong Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Xinjie Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yisheng Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yuebai Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Guoqiang Zhao
- Department of Microbiology and Immunology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| |
Collapse
|
22
|
Butvilovskaya VI, Tikhonov AA, Savvateeva EN, Ragimov AA, Salimov EL, Voloshin SA, Sidorov DV, Chernichenko MA, Polyakov AP, Filushin MM, Tsybulskaya MV, Rubina AY. Hydrogel microchip as a tool for studying exosomes in human serum. Mol Biol 2017. [DOI: 10.1134/s0026893317050053] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
23
|
Mohanta S, Siddappa G, Valiyaveedan SG, Dodda Thimmasandra Ramanjanappa R, Das D, Pandian R, Khora SS, Kuriakose MA, Suresh A. Cancer stem cell markers in patterning differentiation and in prognosis of oral squamous cell carcinoma. Tumour Biol 2017. [PMID: 28631562 DOI: 10.1177/1010428317703656] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Differentiation is a major histological parameter determining tumor aggressiveness and prognosis of the patient; cancer stem cells with their slow dividing and undifferentiated nature might be one of the factors determining the same. This study aims to correlate cancer stem cell markers (CD44 and CD147) with tumor differentiation and evaluate their subsequent effect on prognosis. Immunohistochemical analysis in treatment naïve oral cancer patients (n = 53) indicated that the expression of CD147 was associated with poorly differentiated squamous cell carcinoma and moderately differentiated squamous cell carcinoma (p < 0.01). Furthermore, co-expression analysis showed that 45% each of moderately differentiated squamous cell carcinoma and poorly differentiated squamous cell carcinoma patients were CD44high/CD147high as compared to only 10% of patients with well-differentiated squamous cell carcinoma. A three-way analysis indicated that differentiation correlated with recurrence and survival (p < 0.05) in only the patients with CD44high/CD147high cohort. Subsequently, relevance of these cancer stem cell markers in patterning the differentiation characteristics was evaluated in oral squamous cell carcinoma cell lines originating from different grades of oral cancer. Flowcytometry-based analysis indicated an increase in CD44+/CD147+ cells in cell lines of poorly differentiated squamous cell carcinoma (94.35 ± 1.14%, p < 0.001) and moderately differentiated squamous cell carcinoma origin (93.49 ± 0.47%, p < 0.001) as compared to cell line of well-differentiated squamous cell carcinoma origin (23.12% ± 0.49%). Expression profiling indicated higher expression of cancer stem cell and epithelial-mesenchymal transition markers in SCC029B (poorly differentiated squamous cell carcinoma originated; p ≤ 0.001), which was further translated into increased spheroid formation, migration, and invasion (p < 0.001) as compared to cell line of well-differentiated squamous cell carcinoma origin. This study suggests that CD44 and CD147 together improve the prognostic efficacy of tumor differentiation; in vitro results further point out that these markers might be determinant of differentiation characteristics, imparting properties of increased self-renewal, migration, and invasion.
Collapse
Affiliation(s)
- Simple Mohanta
- 1 Integrated Head and Neck Oncology Program, DSRG-5, Mazumdar Shaw Center for Translational Research, Mazumdar Shaw Medical Foundation, Narayana Health City, Bangalore, India.,2 School of Bio Sciences & Technology, Vellore Institute of Technology (VIT) University, Vellore, India.,3 Department of Head and Neck Oncology, Mazumdar Shaw Medical Center, Narayana Hrudayalaya, Bangalore, India
| | - Gangotri Siddappa
- 1 Integrated Head and Neck Oncology Program, DSRG-5, Mazumdar Shaw Center for Translational Research, Mazumdar Shaw Medical Foundation, Narayana Health City, Bangalore, India
| | - Sindhu Govindan Valiyaveedan
- 1 Integrated Head and Neck Oncology Program, DSRG-5, Mazumdar Shaw Center for Translational Research, Mazumdar Shaw Medical Foundation, Narayana Health City, Bangalore, India
| | - Ravindra Dodda Thimmasandra Ramanjanappa
- 1 Integrated Head and Neck Oncology Program, DSRG-5, Mazumdar Shaw Center for Translational Research, Mazumdar Shaw Medical Foundation, Narayana Health City, Bangalore, India.,3 Department of Head and Neck Oncology, Mazumdar Shaw Medical Center, Narayana Hrudayalaya, Bangalore, India
| | - Debashish Das
- 4 Stem Cell Research Lab, GROW Lab, Narayana Nethralaya Foundation, Bangalore, India
| | - Ramanan Pandian
- 5 GROW Lab, Narayana Nethralaya Foundation, Bangalore, India
| | - Samanta Sekhar Khora
- 2 School of Bio Sciences & Technology, Vellore Institute of Technology (VIT) University, Vellore, India
| | - Moni Abraham Kuriakose
- 3 Department of Head and Neck Oncology, Mazumdar Shaw Medical Center, Narayana Hrudayalaya, Bangalore, India.,6 Mazumdar Shaw Medical Centre-Roswell Park Collaboration Program, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Amritha Suresh
- 1 Integrated Head and Neck Oncology Program, DSRG-5, Mazumdar Shaw Center for Translational Research, Mazumdar Shaw Medical Foundation, Narayana Health City, Bangalore, India.,6 Mazumdar Shaw Medical Centre-Roswell Park Collaboration Program, Roswell Park Cancer Institute, Buffalo, NY, USA
| |
Collapse
|
24
|
Ma C, Wang J, Fan L, Guo Y. Inhibition of CD147 expression promotes chemosensitivity in HNSCC cells by deactivating MAPK/ERK signaling pathway. Exp Mol Pathol 2017; 102:59-64. [PMID: 28062212 DOI: 10.1016/j.yexmp.2017.01.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 01/02/2017] [Accepted: 01/02/2017] [Indexed: 10/20/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is one of the most common cancers in the world. CD147, a transmembrane glycoprotein, has been reported to be correlated with cancer progression, metastasis, and chemoresistance in various cancers. In this study, we aimed to investigate the mechanism of CD147 in regulating drug resistance in HNSCC cells. qRT-PCR were used to evaluated the expression of CD147 in 57 HNSCC tumorous tissues and 2 cell lines. Increased expression of CD147 was found in most HNSCC samples, and the expression level of CD147 was correlated with multidrug resistance. CD147 RNA silencing decreased the chemoresistance of HNSCC cells by deactivating MAPK/ERK signaling pathway. Further investigation revealed that either rescue expression of CD147 or treatment of MAPK/ERK activator phorbol 12-myristate 13-acetate (PMA) in CD147 knockdown CRC cell line attenuated the decreased chemoresistance in CD147 knockdown cells. Taken together, our results suggest that CD147 promotes chemoresistance by activating MAPK/ERK signaling pathway in HNSCC.
Collapse
Affiliation(s)
- Chao Ma
- Department of oral and maxillofacial surgery, Cangzhou central hospital of Hebei province, Cangzhou 061001, China
| | - Jianqi Wang
- Department of oral and maxillofacial surgery, Cangzhou central hospital of Hebei province, Cangzhou 061001, China
| | - Longkun Fan
- Department of oral and maxillofacial surgery, Cangzhou central hospital of Hebei province, Cangzhou 061001, China
| | - Yanjun Guo
- Department of oral and maxillofacial surgery, Cangzhou central hospital of Hebei province, Cangzhou 061001, China.
| |
Collapse
|
25
|
Yonemori M, Seki N, Yoshino H, Matsushita R, Miyamoto K, Nakagawa M, Enokida H. Dual tumor-suppressors miR-139-5p and miR-139-3p targeting matrix metalloprotease 11 in bladder cancer. Cancer Sci 2016; 107:1233-42. [PMID: 27355528 PMCID: PMC5021030 DOI: 10.1111/cas.13002] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/21/2016] [Accepted: 06/29/2016] [Indexed: 12/14/2022] Open
Abstract
Our recent study of the microRNA (miRNA) expression signature of bladder cancer (BC) by deep‐sequencing revealed that two miRNA, microRNA‐139‐5p/microRNA‐139‐3p were significantly downregulated in BC tissues. The aim of this study was to investigate the functional roles of these miRNA and their modulation of cancer networks in BC cells. Functional assays of BC cells were performed using transfection of mature miRNA or small interfering RNA (siRNA). Genome‐wide gene expression analysis, in silico analysis and dual‐luciferase reporter assays were applied to identify miRNA targets. The associations between the expression of miRNA and its targets and overall survival were estimated by the Kaplan–Meier method. Gain‐of‐function studies showed that miR‐139‐5p and miR‐139‐3p significantly inhibited cell migration and invasion by BC cells. The matrix metalloprotease 11 gene (MMP11) was identified as a direct target of miR‐139‐5p and miR‐139‐3p. Kaplan–Meier survival curves showed that higher expression of MMP11 predicted shorter survival of BC patients (P = 0.029). Downregulated miR‐139‐5p or miR‐139‐3p enhanced BC cell migration and invasion in BC cells. MMP11 was directly regulated by these miRNA and might be a good prognostic marker for survival of BC patients.
Collapse
Affiliation(s)
- Masaya Yonemori
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Naohiko Seki
- Department of Functional Genomics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hirofumi Yoshino
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Ryosuke Matsushita
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Kazutaka Miyamoto
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Masayuki Nakagawa
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Hideki Enokida
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan.
| |
Collapse
|
26
|
Wan X, Pu H, Huang W, Yang S, Zhang Y, Kong Z, Yang Z, Zhao P, Li A, Li T, Li Y. Androgen-induced miR-135a acts as a tumor suppressor through downregulating RBAK and MMP11, and mediates resistance to androgen deprivation therapy. Oncotarget 2016; 7:51284-51300. [PMID: 27323416 PMCID: PMC5239475 DOI: 10.18632/oncotarget.9992] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 05/25/2016] [Indexed: 12/27/2022] Open
Abstract
The main challenge in the treatment of prostate cancer (PCa) is that the majority of patients inevitably develop resistance to androgen deprivation. However, the mechanisms involved in hormone independent behavior of PCa remain unclear. In the present study, we identified androgen-induced miR-135a as a direct target of AR. Functional studies revealed that overexpression of miR-135a could significantly decrease cell proliferation and migration, and induce cell cycle arrest and apoptosis in PCa. We identified RBAK and MMP11 as direct targets of miR-135a in PCa by integrating bioinformatics analysis and experimental assays. Mechanistically, miR-135a repressed PCa migration through downregulating MMP11 and induced PCa cell cycle arrest and apoptosis by suppressing RBAK. Consistently, inverse correlations were also observed between the expression of miR-135a and RBAK or MMP11 in PCa samples. In addition, low miR-135a and high RBAK and MMP11 expression were positively correlated with PCa progression. Also, PI3K/AKT pathway was confirmed to be an upstream regulation signaling of miR-135a in androgen-independent cell lines. Accordingly, we reported a resistance mechanism to androgen deprivation therapy (ADT) mediated by miR-135a which might be downregulated by androgen depletion and/or PI3K/AKT hyperactivation, in castration-resistant prostate cancer (CRPC), thus promoting tumor progression. Taken together, miR-135a may represent a new diagnostic and therapeutic biomarker for castration-resistant PCa.
Collapse
Affiliation(s)
- Xuechao Wan
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Science, Fudan University, Shanghai, 200433, PR China
| | - Honglei Pu
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Science, Fudan University, Shanghai, 200433, PR China
| | - Wenhua Huang
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Science, Fudan University, Shanghai, 200433, PR China
| | - Shu Yang
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Science, Fudan University, Shanghai, 200433, PR China
| | - Yalong Zhang
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Science, Fudan University, Shanghai, 200433, PR China
| | - Zhe Kong
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Science, Fudan University, Shanghai, 200433, PR China
| | - Zhuoran Yang
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Science, Fudan University, Shanghai, 200433, PR China
| | - Peiqing Zhao
- Center of Translational Medicine, Central Hospital of Zibo, Zibo, Shangdong, 255036, PR China
| | - Ao Li
- Center of Translational Medicine, Central Hospital of Zibo, Zibo, Shangdong, 255036, PR China
| | - Tao Li
- Center of Translational Medicine, Central Hospital of Zibo, Zibo, Shangdong, 255036, PR China
| | - Yao Li
- State Key Laboratory of Genetic Engineering, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Science, Fudan University, Shanghai, 200433, PR China
- Key Laboratory of Reproduction Regulation of NPFPC, Fudan University, Shanghai, 200433, PR China
| |
Collapse
|
27
|
Zhang X, Huang S, Guo J, Zhou L, You L, Zhang T, Zhao Y. Insights into the distinct roles of MMP-11 in tumor biology and future therapeutics (Review). Int J Oncol 2016; 48:1783-93. [PMID: 26892540 DOI: 10.3892/ijo.2016.3400] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 01/21/2016] [Indexed: 11/06/2022] Open
Abstract
The biological processes of cancer cells such as tumorigenesis, proliferation, angiogenesis, apoptosis and invasion are greatly influenced by the surrounding microenvironment. The ability of solid malignant tumors to alter the microenvironment represents an important characteristic through which tumor cells are able to acquire specific functions necessary for their malignant biological behaviors. Matrix metalloproteinases (MMPs) are a family of zinc-dependent endopeptidases with the capacity of remodeling extracellular matrix (ECM) by degrading almost all ECM proteins, which plays essential roles during the invasion and metastasis process of solid malignant tumors, including allowing tumor cells to modify the ECM components and release cytokines, ultimately facilitating protease-dependent tumor progression. MMP-11, also named stromelysin-3, is a member of the stromelysin subgroup belonging to MMPs superfamily, which has been detected in cancer cells, stromal cells and adjacent microenvironment. Differently, MMP-11 exerts a dual effect on tumors. On the one hand MMP-11 promotes cancer development by inhibiting apoptosis as well as enhancing migration and invasion of cancer cells, on the other hand MMP-11 plays a negative role against cancer development via suppressing metastasis in animal models. Overexpression of MMP-11 was discovered in sera of cancer patients compared with normal control group as well as in multiple tumor tissue specimens, such as gastric cancer, breast cancer, and pancreatic cancer. At present, some evidence supports that MMP-11 may work as a significant tumor biomarker for early detection of cancer, tumor staging, prognostic analysis, monitoring recurrence during follow-up and also a potential target for immunotherapy against cancer. In view of the importance of MMP-11 in modifying tumor microenvironment and potent antitumoral effects on solid tumors, there is an urgent need for a deeper understanding of how MMP-11 modulates tumor progression, and exploring its potential clinical application.
Collapse
Affiliation(s)
- Xu Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Shuai Huang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Junchao Guo
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Li Zhou
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|