1
|
Shi Q, He J, Chen G, Xu J, Zeng Z, Zhao X, Zhao B, Gao X, Ye Z, Xiao M, Li H. The chemical composition of Diwu YangGan capsule and its potential inhibitory roles on hepatocellular carcinoma by microarray-based transcriptomics. J Tradit Complement Med 2024; 14:381-390. [PMID: 39035694 PMCID: PMC11259662 DOI: 10.1016/j.jtcme.2023.12.002] [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: 07/18/2023] [Revised: 09/28/2023] [Accepted: 12/24/2023] [Indexed: 07/23/2024] Open
Abstract
The Traditional Chinese Medicine compound preparation known as Diwu Yanggan capsule (DWYG) can effectively hinder the onset and progression of hepatocellular carcinoma (HCC), which is recognized worldwide as a significant contributor to fatalities associated with cancer. Nevertheless, the precise mechanisms implicated have remained ambiguous. In present study, the model of HCC was set up by the 2-acetylaminofluorene (2-AAF)/partial hepatectomy (PH) in rats. To confirm the differentially expressed genes (DEGs) identified in the microarray analysis, real-time quantitative reverse transcription PCR (qRT-PCR) was conducted. In the meantime, the liquid chromatography-quadrupole time of flight mass spectrometry (LC-QTOF-MS/MS) was employed to characterize the component profile of DWYG. Consequently, the DWYG treatment exhibited the ability to reverse 51 variation genes induced by 2-AAF/PH. Additionally, there was an overlap of 54 variation genes between the normal and model groups. Upon conducting RT-qPCR analysis, it was observed that the expression levels of all genes were increased by 2-AAF/PH and subsequently reversed after DWYG treatment. Notably, the fold change of expression levels for all genes was below 0.5, with 3 genes falling below 0.25. Moreover, an investigation was conducted to determine the signaling pathway that was activated/inhibited in the HCC group and subsequently reversed in the DWYG group. Moreover, the component profile of DWYG encompassed a comprehensive compilation of 206 compounds that were identified or characterized. The findings of this study elucidated the potential alleviative mechanisms of DWYG in the context of HCC, thereby holding significant implications for its future clinical utilization and widespread adoption.
Collapse
Affiliation(s)
- Qingxin Shi
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Jiangcheng He
- Wuhan Integrated Traditional Chinese and Western Medicine Orthopedic Hospital, Affiliated Hospital of Wuhan Sports University, Wuhan, 430079, China
| | - Guangya Chen
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Jinlin Xu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Zhaoxiang Zeng
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xueyan Zhao
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Binbin Zhao
- School of Basic Medicine, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Xiang Gao
- Institute of Liver Diseases, Hubei Key Laboratory of the Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, 430074, China
| | - Zhihua Ye
- Institute of Liver Diseases, Hubei Key Laboratory of the Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, 430074, China
| | - Mingzhong Xiao
- Institute of Liver Diseases, Hubei Key Laboratory of the Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, 430074, China
| | - Hanmin Li
- Institute of Liver Diseases, Hubei Key Laboratory of the Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430074, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, 430074, China
| |
Collapse
|
2
|
Seufferlein T, Lausser L, Stein A, Arnold D, Prager G, Kasper-Virchow S, Niedermeier M, Müller L, Kubicka S, König A, Büchner-Steudel P, Wille K, Berger AW, Kestler AMR, Kraus JM, Werle SD, Perkhofer L, Ettrich TJ, Kestler HA. Prediction of resistance to bevacizumab plus FOLFOX in metastatic colorectal cancer-Results of the prospective multicenter PERMAD trial. PLoS One 2024; 19:e0304324. [PMID: 38875244 PMCID: PMC11178165 DOI: 10.1371/journal.pone.0304324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Accepted: 05/08/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND Anti-vascular endothelial growth factor (VEGF) monoclonal antibodies (mAbs) are widely used for tumor treatment, including metastatic colorectal cancer (mCRC). So far, there are no biomarkers that reliably predict resistance to anti-VEGF mAbs like bevacizumab. A biomarker-guided strategy for early and accurate assessment of resistance could avoid the use of non-effective treatment and improve patient outcomes. We hypothesized that repeated analysis of multiple cytokines and angiogenic growth factors (CAFs) before and during treatment using machine learning could provide an accurate and earlier, i.e., 100 days before conventional radiologic staging, prediction of resistance to first-line mCRC treatment with FOLFOX plus bevacizumab. PATIENTS AND METHODS 15 German and Austrian centers prospectively recruited 50 mCRC patients receiving FOLFOX plus bevacizumab as first-line treatment. Plasma samples were collected every two weeks until radiologic progression (RECIST 1.1) as determined by CT scans performed every 2 months. 102 pre-selected CAFs were centrally analyzed using a cytokine multiplex assay (Luminex, Myriad RBM). RESULTS Using random forests, we developed a predictive machine learning model that discriminated between the situations of "no progress within 100 days before radiological progress" and "progress within 100 days before radiological progress". We could further identify a combination of ten out of the 102 CAF markers, which fulfilled this task with 78.2% accuracy, 71.8% sensitivity, and 82.5% specificity. CONCLUSIONS We identified a CAF marker combination that indicates treatment resistance to FOLFOX plus bevacizumab in patients with mCRC within 100 days prior to radiologic progress.
Collapse
Affiliation(s)
- Thomas Seufferlein
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Ludwig Lausser
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
- Faculty of Computer Science, Technische Hochschule Ingolstadt, Ingolstadt, Germany
| | - Alexander Stein
- Hematology-Oncology Practice Eppendorf, University Cancer Center Hamburg, Hamburg, Germany
| | - Dirk Arnold
- Asklepios Cancer Center Hamburg, AK Altona, Hamburg, Germany
| | - Gerald Prager
- Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Stefan Kasper-Virchow
- Medical Oncology, University Hospital Essen West German Cancer Center, Essen, Germany
| | | | | | - Stefan Kubicka
- Cancer Center Reutlingen, Reutlingen Hospital, Reutlingen, Germany
| | - Alexander König
- Department of Gastroenterology, Gastrointestinal Oncology and Endocrinology, University Medical Center Goettingen, Göttingen, Germany
| | | | - Kai Wille
- Hematology, Oncology, University Hospital Ruhr-University-Bochum, Minden, Germany
| | - Andreas W. Berger
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | | | - Johann M. Kraus
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Silke D. Werle
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| | - Lukas Perkhofer
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Thomas J. Ettrich
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Hans A. Kestler
- Institute of Medical Systems Biology, Ulm University, Ulm, Germany
| |
Collapse
|
3
|
Zhang X, Liu SS, Ma J, Qu W. Secretory leukocyte protease inhibitor (SLPI) in cancer pathophysiology: Mechanisms of action and clinical implications. Pathol Res Pract 2023; 248:154633. [PMID: 37356220 DOI: 10.1016/j.prp.2023.154633] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 06/17/2023] [Accepted: 06/18/2023] [Indexed: 06/27/2023]
Abstract
Cancer is a multifaceted disorder frequently linked to the dysregulation of several biological processes. The SLPI is a multifunctional protein involved in the modulation of immunological response and the inhibition of protease activities. SLPI acts as an inhibitor of proteases, exerts antibacterial properties, and suppresses the transcription of proinflammatory genes through the nuclear factor-kappa B (NF-κB) pathway. The role of this protein as a regulatory agent has been implicated in various types of cancer. Recent research has revealed that SLPI upregulation in cancer cells enhances the metastatic capacity of epithelial malignancies, indicating the deleterious effects of this protein. Furthermore, SLPI interacts intricately with other cancer-promoting factors, including matrix metalloproteinase-2 (MMP-2), MMP-9, the NF-κB and Akt pathways, and the p53-upregulated modulator of apoptosis (PUMA). This review provides an overview of the role of SLPI in cancer pathophysiology, emphasizing its expression in cancer cells and tissues, its potential as a prognostic biomarker, and its therapeutic promise as a target in cancer treatment. The mechanisms of SLPI action in cancer, including its anti-inflammatory effects, regulation of cell proliferation and angiogenesis, and modulation of the tumor microenvironment, have been investigated. The clinical implications of SLPI in cancer have been discussed, including its potential as a diagnostic and prognostic biomarker, its role in chemoresistance, and its therapeutic potential in several types of cancer, such as hepatocellular carcinoma (HCC), colorectal cancer (CRC), pancreatic cancer, head and neck squamous cell carcinoma (HNSCC), ovarian cancer (OvCa), prostate cancer (PC), gastric cancer (GC), breast cancer, and other cancers. In addition, we emphasized the significance of SLPI in cancer, which offers fresh perspectives on potential targets for cancer therapy.
Collapse
Affiliation(s)
- Xiaohua Zhang
- Department of Clinical Laboratory, the Second Hospital of Jilin University, Changchun 130000, China
| | - Shan Shan Liu
- Department of General Medicine, the Second Hospital of Jilin University, Changchun 130000, China.
| | - Jingru Ma
- Department of Clinical Laboratory, the Second Hospital of Jilin University, Changchun 130000, China
| | - Wei Qu
- Department of General Medicine, the Second Hospital of Jilin University, Changchun 130000, China
| |
Collapse
|
4
|
Liu T, Zhao M, Peng L, Chen J, Xing P, Gao P, Chen L, Qiao X, Wang Z, Di J, Qu H, Jiang B, Su X. WFDC3 inhibits tumor metastasis by promoting the ERβ-mediated transcriptional repression of TGFBR1 in colorectal cancer. Cell Death Dis 2023; 14:425. [PMID: 37443102 PMCID: PMC10345115 DOI: 10.1038/s41419-023-05956-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 06/20/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023]
Abstract
Estrogen plays a protective role in colorectal cancer (CRC) and primarily functions through estrogen receptor β (ERβ). However, clinical strategies for CRC therapy associated with ERβ are still under investigation. Our discoveries identified WFDC3 as a tumor suppressor that facilitates estrogen-induced inhibition of metastasis through the ERβ/TGFBR1 signaling axis. WFDC3 interacts with ERβ and increases its protein stability by inhibiting its proteasome-dependent degradation. WFDC3 represses TGFBR1 expression through ERβ-mediated transcription. Blocking TGFβ signaling with galunisertib, a drug used in clinical trials that targets TGFBR1, impaired the migration of CRC cells induced by WFDC3 depletion. Moreover, there was clinical significance to WFDC3 in CRC, as CRC patients with high WFDC3 expression in tumor cells had favorable prognoses. Therefore, this work suggests that WFDC3 could be an indicator for therapies targeting the estrogen/ERβ pathway in CRC patients.
Collapse
Affiliation(s)
- Tianqi Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Min Zhao
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore DC, Sunshine Coast, QLD, 4556, Australia
| | - Lin Peng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Jiangbo Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Pu Xing
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Pin Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Lei Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Xiaowen Qiao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Zaozao Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Jiabo Di
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China
| | - Hong Qu
- Center for Bioinformatics, State Key Laboratory of Protein and Plant Gene Research, College of Life Sciences, Peking University, 100871, Beijing, People's Republic of China.
| | - Beihai Jiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China.
| | - Xiangqian Su
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery IV, Peking University Cancer Hospital & Institute, 100142, Beijing, China.
| |
Collapse
|
5
|
Nugteren S, den Uil SH, Delis-van Diemen PM, Simons-Oosterhuis Y, Lindenbergh-Kortleve DJ, van Haaften DH, Stockmann HBAC, Sanders J, Meijer GA, Fijneman RJA, Samsom JN. High expression of secretory leukocyte protease inhibitor (SLPI) in stage III micro-satellite stable colorectal cancer is associated with reduced disease recurrence. Sci Rep 2022; 12:12174. [PMID: 35842496 PMCID: PMC9288430 DOI: 10.1038/s41598-022-16427-5] [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: 03/31/2022] [Accepted: 07/11/2022] [Indexed: 12/02/2022] Open
Abstract
Secretory leukocyte protease inhibitor (SLPI) is a pleiotropic protein produced by healthy intestinal epithelial cells. SLPI regulates NF-κB activation, inhibits neutrophil proteases and has broad antimicrobial activity. Recently, increased SLPI expression was found in various types of carcinomas and was suggested to increase their metastatic potential. Indeed, we demonstrated that SLPI protein expression in colorectal cancer (CRC) liver metastases and matched primary tumors is associated with worse outcome, suggesting that SLPI promotes metastasis in human CRC. However, whether SLPI plays a role in CRC before distant metastases have formed is unclear. Therefore, we examined whether SLPI expression is associated with prognosis in CRC patients with localized disease. Using a cohort of 226 stage II and 160 stage III CRC patients we demonstrate that high SLPI protein expression is associated with reduced disease recurrence in patients with stage III micro-satellite stable tumors treated with adjuvant chemotherapy, independently of established clinical risk factors (hazard rate ratio 0.54, P-value 0.03). SLPI protein expression was not associated with disease-free survival in stage II CRC patients. Our data suggest that the role of SLPI in CRC may be different depending on the stage of disease. In stage III CRC, SLPI expression may be unfavorable for tumors, whereas SLPI expression may be beneficial for tumors once distant metastases have established.
Collapse
Affiliation(s)
- Sandrine Nugteren
- Division Gastroenterology and Nutrition, Laboratory of Pediatrics, Erasmus University Medical Center, Sophia Children's Hospital, Room Ee1567A, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Sjoerd H den Uil
- Department of Surgery, Spaarne Gasthuis, Haarlem, The Netherlands
| | | | - Ytje Simons-Oosterhuis
- Division Gastroenterology and Nutrition, Laboratory of Pediatrics, Erasmus University Medical Center, Sophia Children's Hospital, Room Ee1567A, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Dicky J Lindenbergh-Kortleve
- Division Gastroenterology and Nutrition, Laboratory of Pediatrics, Erasmus University Medical Center, Sophia Children's Hospital, Room Ee1567A, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Daniëlle H van Haaften
- Division Gastroenterology and Nutrition, Laboratory of Pediatrics, Erasmus University Medical Center, Sophia Children's Hospital, Room Ee1567A, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | | | - Joyce Sanders
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Gerrit A Meijer
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Remond J A Fijneman
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Janneke N Samsom
- Division Gastroenterology and Nutrition, Laboratory of Pediatrics, Erasmus University Medical Center, Sophia Children's Hospital, Room Ee1567A, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands.
| |
Collapse
|
6
|
Cannon AS, Holloman BL, Wilson K, Miranda K, Dopkins N, Nagarkatti P, Nagarkatti M. AhR Activation Leads to Attenuation of Murine Autoimmune Hepatitis: Single-Cell RNA-Seq Analysis Reveals Unique Immune Cell Phenotypes and Gene Expression Changes in the Liver. Front Immunol 2022; 13:899609. [PMID: 35720411 PMCID: PMC9204231 DOI: 10.3389/fimmu.2022.899609] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) is a ubiquitously expressed ligand-activated transcription factor. While initially identified as an environmental sensor, this receptor has been shown more recently to regulate a variety of immune functions. AhR ligands vary in structure and source from environmental chemicals such as 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) and indoles found in cruciferous vegetables to endogenous ligands derived from tryptophan metabolism. In the current study, we used TCDD, a high affinity AhR ligand to study the impact of AhR activation in the murine model of autoimmune hepatitis (AIH). Primarily, we used single-cell RNA-sequencing (scRNA-seq) technology to study the nature of changes occurring in the immune cells in the liver at the cellular and molecular level. We found that AhR activation attenuated concanavalin A (ConA)-induced AIH by limiting chemotaxis of pro-inflammatory immune cell subsets, promoting anti-inflammatory cytokine production, and suppressing pro-inflammatory cytokine production. scRNA-seq analysis showed some unusual events upon ConA injection such as increased presence of mature B cells, natural killer (NK) T cells, CD4+ or CD8+ T cells, Kupffer cells, memory CD8+ T cells, and activated T cells while TCDD treatment led to the reversal of most of these events. Additionally, the immune cells showed significant alterations in the gene expression profiles. Specifically, we observed downregulation of inflammation-associated genes including Ptma, Hspe1, and CD52 in TCDD-treated AIH mice as well as alterations in the expression of migratory markers such as CXCR2. Together, the current study characterizes the nature of inflammatory changes occurring in the liver during AIH, and sheds light on how AhR activation during AIH attenuates liver inflammation by inducing phenotypic and genotypic changes in immune cells found in the liver.
Collapse
Affiliation(s)
| | | | | | | | | | - Prakash Nagarkatti
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| |
Collapse
|
7
|
Zhang G, Gao Y, Yu Z, Su H. Upregulated long intergenic non-protein coding RNA 1094 (LINC01094) is linked to poor prognosis and alteration of cell function in colorectal cancer. Bioengineered 2022; 13:8526-8537. [PMID: 35287563 PMCID: PMC9161846 DOI: 10.1080/21655979.2022.2051839] [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] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) showed high cancer-related mortality in recent years partly due to the absence of an effective prognostic predictor. This research intended to evaluate the prognostic value and potential role of long intergenic non-protein coding RNA 1094 (LINC01094) in CRC. In this work, we evaluated the LINC01094 level in 122 CRC patients’ tissues and in human CRC cell lines. We explored the ability of LINC01094 in overall survival and progression-free survival estimate. The effect of LINC01094 dysregulation on the CRC cells was investigated. LINC01094 is highly expressed in CRC tissues and cells than normal ones. This high expression was correlated with absent vascular invasion, positive lymph node metastasis, and advanced TNM stage. With the result of Kaplan-Meier analysis and multivariate Cox’s proportional hazard analysis, LINC01094 was an effective biomarker for CRC overall survival. Downregulation of LINC01094 impeded the malignant biological behavior (proliferation, invasion, and migration) of CRC cells, while overexpression of LINC01094 boosted that maybe by sponging miR-1266-5p. LINC01094 might function as an oncogene in CRC and allowed the discovery of a new biomarker for prognosis and therapy of CRC.
Collapse
Affiliation(s)
- Guangliang Zhang
- Oncology Department, Liaocheng People's Hospital, Liaocheng, 252000, China
| | - Yingjie Gao
- Oncology Department, Liaocheng People's Hospital, Liaocheng, 252000, China
| | - Zhen Yu
- Intervention Therapy Department, Liaocheng People's Hospital, Liaocheng, 252000, China
| | - Hui Su
- Oncology Department, Liaocheng People's Hospital, Liaocheng, 252000, China
| |
Collapse
|
8
|
Nugteren S, Samsom JN. Secretory Leukocyte Protease Inhibitor (SLPI) in mucosal tissues: Protects against inflammation, but promotes cancer. Cytokine Growth Factor Rev 2021; 59:22-35. [PMID: 33602652 DOI: 10.1016/j.cytogfr.2021.01.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 01/24/2021] [Indexed: 12/20/2022]
Abstract
The immune system is continuously challenged with large quantities of exogenous antigens at the barriers between the external environment and internal human tissues. Antimicrobial activity is essential at these sites, though the immune responses must be tightly regulated to prevent tissue destruction by inflammation. Secretory Leukocyte Protease Inhibitor (SLPI) is an evolutionarily conserved, pleiotropic protein expressed at mucosal surfaces, mainly by epithelial cells. SLPI inhibits proteases, exerts antimicrobial activity and inhibits nuclear factor-kappa B (NF-κB)-mediated inflammatory gene transcription. SLPI maintains homeostasis at barrier tissues by preventing tissue destruction and regulating the threshold of inflammatory immune responses, while protecting the host from infection. However, excessive expression of SLPI in cancer cells may have detrimental consequences, as recent studies demonstrate that overexpression of SLPI increases the metastatic potential of epithelial tumors. Here, we review the varied functions of SLPI in the respiratory tract, skin, gastrointestinal tract and genitourinary tract, and then discuss the mechanisms by which SLPI may contribute to cancer.
Collapse
Affiliation(s)
- Sandrine Nugteren
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Janneke N Samsom
- Laboratory of Pediatrics, Division Gastroenterology and Nutrition, Erasmus University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|