1
|
Liu Y, Xue R. Pancreatic stellate cell: Update on molecular investigations and clinical translation in pancreatic cancer. Int J Cancer 2025; 156:1672-1685. [PMID: 39825771 DOI: 10.1002/ijc.35326] [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: 06/06/2024] [Revised: 12/11/2024] [Accepted: 01/02/2025] [Indexed: 01/20/2025]
Abstract
Pancreatic cancer is a particularly aggressive tumor, distinguished by the presence of a prominent collagenous stroma and desmoplasia that envelops the tumor cells. Pancreatic stellate cell (PSC) contributes to the formation of a dense fibrotic stroma and has been demonstrated to facilitate tumor progression. As the significance of PSCs is increasingly revealed, more explorations are focused on the complex molecular mechanisms and tumor-stromal crosstalk in order to guide potential therapeutic approaches through deactivating or reprogramming PSCs. Nevertheless, significant challenges persist in translating preclinical discoveries into clinical applications. In this review, we expect to offer a comprehensive overview of the latest molecular advancements in PSCs, along with new insights into the clinical therapeutic strategies targeting PSCs.
Collapse
Affiliation(s)
- Yawei Liu
- School of Basic Medicine Sciences, Capital Medical University, Beijing, China
- Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Ran Xue
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Center, Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|
2
|
Tang W, Li J, Zhou Y, Li J, Ma Z, Li X, Wang H, Xiong M, Chen X, Li X, Chen W, Ma H, Ye X. Palmatine attenuates MYH9 mediated nuclear localization of AURKA to induce G2/M phase arrest in colorectal cancer cells. Int Immunopharmacol 2024; 143:113615. [PMID: 39536490 DOI: 10.1016/j.intimp.2024.113615] [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: 08/27/2024] [Revised: 10/26/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
The mitotic kinase Aurora kinase A (AURKA), which plays a crucial role in cell cycle progression, represents a promising target for the treatment of colorectal cancer (CRC). Here, we found that AURKA is a target of a CRC suppressor, the Palmatine (PAL). However, the underlying mechanism remains elusive. This work aims to investigate the underlying mechanism how PAL suppresses CRC through AURKA. It was confirmed that AURKA played an important role in the development of CRC tumors through an Azoxymethane/Dextran sulfate sodium salt induced mice model and tissue microarrays of CRC-patients. Overexpression of AURKA was able to partially reverse the inhibitory effect of PAL on CRC cells, showing that PAL significantly inhibited the malignant phenotype and induced the G2/M phase arrest of CRC cells by down-regulating AURKA. Functional studies indicated that PAL attenuated the stability of AURKA protein and reduced its nuclear level, resulting in reduction of key proteins in the G2/M phase. Importantly, Co-IP and WB experiments suggested that Myosin heavy chain 9 (MYH9) interacted with AURKA and had an impact on its nuclear localization. PAL can decrease nuclear AURKA by reducing the interaction of AURKA and MYH9. Taken together, our study revealed that MYH9 as an auxiliary protein for the nuclear localization of AURKA and elucidated the mechanism that PAL reduced nuclear AURKA through inhibiting the interaction of AURKA and MYH9 to induce G2/M phase arrest in CRC cells. Therefore, this study may provide a theoretical basis of PAL for the treatment of CRC.
Collapse
Affiliation(s)
- Wanyu Tang
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| | - Jingwei Li
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| | - Yuan Zhou
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| | - Juan Li
- School of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China.
| | - Zhengcai Ma
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| | - Xiaoduo Li
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| | - Hongmei Wang
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| | - Mengyuan Xiong
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| | - Xiantao Chen
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| | - Xuegang Li
- School of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China.
| | - Wanqun Chen
- Department of Gastroenterology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400000, China.
| | - Hang Ma
- School of Pharmaceutical Sciences and Chinese Medicine, Southwest University, Chongqing 400715, China.
| | - Xiaoli Ye
- Engineering Research Center of Coptis Development & Utilization (Ministry of Education), School of Life Sciences, Southwest University, Chongqing 400715, China.
| |
Collapse
|
3
|
Koochaki R, Amini E, Zarehossini S, Zareh D, Haftcheshmeh SM, Jha SK, Kesharwani P, Shakeri A, Sahebkar A. Alkaloids in Cancer therapy: Targeting the tumor microenvironment and metastasis signaling pathways. Fitoterapia 2024; 179:106222. [PMID: 39343104 DOI: 10.1016/j.fitote.2024.106222] [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: 02/07/2024] [Revised: 09/17/2024] [Accepted: 09/21/2024] [Indexed: 10/01/2024]
Abstract
The use of phytomedicine in cancer therapy is a growing field of research that takes use of the medicinal properties of plant-derived compounds. Under the domain of cancer therapy and management, alkaloids, a prominent group of natural compounds, have showed significant potential. Alkaloids often affect a wide range of essential cellular mechanisms involved in cancer progression. These multi-targeting capabilities, can give significant advantages to alkaloids in overcoming resistance mechanisms. For example, berberine, an alkaloid found in Berberis species, is widely reported to induce apoptosis by activating caspases and regulating apoptotic pathways. Notably, alkaloids like as quinine have showed promise in inhibiting the formation of new blood vessels required for tumor growth. In addition, alkaloids have shown anti-proliferative and anticancer properties mostly via modulating key signaling pathways involved in metastasis, including those regulating epithelial-mesenchymal transition. This work provides a comprehensive overview of naturally occurring alkaloids that exhibit anticancer properties, with a specific emphasis on their underlying molecular mechanisms of action. Furthermore, many methods to modify previously reported difficult physicochemical properties using nanocarriers in order to enhance its systemic bioavailability have been discussed as well. This study also includes information on newly discovered alkaloids that are now being studied in clinical trials for their potential use in cancer treatment. Further, we have also briefly mentioned on the application of high-throughput screening and molecular dynamics simulation for acceleration on the identification of potent alkaloids based compounds to target and treat cancer.
Collapse
Affiliation(s)
- Raoufeh Koochaki
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Elaheh Amini
- Department of Animal Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Sara Zarehossini
- Department of Cell & Molecular Biology (genetic), Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Danial Zareh
- Department of Cell & Molecular Biology (genetic), Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | | | - Saurav Kumar Jha
- Department of Biological Sciences and Bioengineering (BSBE), Indian Institute of Technology, Kanpur 208016, Uttar Pradesh, India
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Abolfazl Shakeri
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran..
| |
Collapse
|
4
|
Pei X, Luo Y, Zeng H, Jamil M, Liu X, Jiang B. Identification and validation of key genes in gastric cancer: insights from in silico analysis, clinical samples, and functional assays. Aging (Albany NY) 2024; 16:10615-10635. [PMID: 38913913 PMCID: PMC11236316 DOI: 10.18632/aging.205965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 05/16/2024] [Indexed: 06/26/2024]
Abstract
INTRODUCTION The underlying mechanisms of gastric cancer (GC) remain unknown. Therefore, in this study, we employed a comprehensive approach, combining computational and experimental methods, to identify potential key genes and unveil the underlying pathogenesis and prognosis of GC. METHODS Gene expression profiles from GEO databases (GSE118916, GSE79973, and GSE29272) were analyzed to identify DEGs between GC and normal tissues. A PPI network was constructed using STRING and Cytoscape, followed by hub gene identification with CytoHubba. Investigations included expression and promoter methylation analysis, survival modeling, mutational and miRNA analysis, gene enrichment, drug prediction, and in vitro assays for cellular behaviors. RESULTS A total of 83 DEGs were identified in the three datasets, comprising 41 up-regulated genes and 42 down-regulated genes. Utilizing the degree and MCC methods, we identified four hub genes that were hypomethylated and up-regulated: COL1A1, COL1A2, COL3A1, and FN1. Subsequent validation of their expression and promoter methylation on clinical GC samples through targeted bisulfite sequencing and RT-qPCR analysis further confirmed the hypomethylation and overexpression of these genes in local GC patients. Furthermore, it was observed that these hub genes regulate tumor proliferation and metastasis in in vivo and exhibited mutations in GC patients. CONCLUSION We found four potential diagnostic and prognostic biomarkers, including COL1A1, COL1A2, COL3A1, and FN1 that may be involved in the occurrence and progression of GC.
Collapse
Affiliation(s)
- Xiaofeng Pei
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Yuanling Luo
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Huanwen Zeng
- Department of Oncology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| | - Muhammad Jamil
- PARC Arid Zone Research Center, Dera Ismail Khan 29050, Pakistan
| | - Xiaodong Liu
- Department of Pharmacy, The 922 Hospital of Joint Logistics Support Force, PLA, Hengyang 421002, China
| | - Bo Jiang
- Department of Emergency, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, China
| |
Collapse
|
5
|
Li W, Jiao R, Luo S, Liu Z, Song J, Chen Z. Mechanism of action of Coptidis Rhizome in treating periodontitis based on network pharmacology and in vitro validation. BMC Oral Health 2024; 24:530. [PMID: 38704553 PMCID: PMC11069132 DOI: 10.1186/s12903-024-04311-9] [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: 03/01/2024] [Accepted: 04/29/2024] [Indexed: 05/06/2024] Open
Abstract
OBJECTIVE Explore the therapeutic mechanism of Coptidis Rhizome (CR) in periodontitis using network pharmacology, and validate it through molecular docking and in vitro experiments. METHODS Screened potential active components and target genes of CR from TCMSP and Swiss databases. Identified periodontitis-related target genes using GeneCards. Found common target genes using Venny. Conducted GO and KEGG pathway analysis. Performed molecular docking and in vitro experiments using Berberine, the main active component of CR, on lymphocytes from healthy and periodontitis patients. Assessed effects on inflammatory factors using CCK-8, flow cytometry, and ELISA. RESULTS Fourteen active components and 291 targets of CR were identified. 30 intersecting target genes with periodontitis were found. GO and KEGG analysis revealed oxidative stress response and IL-17 signaling pathway as key mechanisms. Molecular docking showed strong binding of Berberine with ALOX5, AKT1, NOS2, and TNF. In vitro experiments have demonstrated the ability of berberine to inhibit the expression of Th17 + and other immune related cells in LPS stimulated lymphocytes, and reduce the secretion of IL-6, IL-8, and IL-17. CONCLUSION CR treats periodontitis through a multi-component, multi-target, and multi-pathway approach. Berberine, its key component, acts through the IL-17 signaling pathway to exert anti-inflammatory effects.
Collapse
Affiliation(s)
- Wei Li
- Guiyang Hospital of Stomatology, Guiyang, Guizhou, 550005, China
| | - Ruofeng Jiao
- Guiyang Hospital of Stomatology, Guiyang, Guizhou, 550005, China
- Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Shiyi Luo
- Guiyang Hospital of Stomatology, Guiyang, Guizhou, 550005, China
- Medical College of Guizhou University, Guiyang, Guizhou, 550025, China
| | - Zefei Liu
- Guiyang Hospital of Stomatology, Guiyang, Guizhou, 550005, China
- Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Jukun Song
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Guizhou Medical University, Guiyang, 550001, China.
| | - Zhu Chen
- Guiyang Hospital of Stomatology, Guiyang, Guizhou, 550005, China.
- Zunyi Medical University, Zunyi, Guizhou, 563000, China.
- Medical College of Guizhou University, Guiyang, Guizhou, 550025, China.
| |
Collapse
|
6
|
Dong Md S, Xu Md P, Yang Md P, Jiao Md J, Cheng Md PhD CS, Chen Md PhD L. "Huanglianjiedu Decoction" Against Pancreatic Adenocarcinoma Proliferation of by Downregulating the PI3K/AKT/mTOR and MAPK/ERK1/2 Signaling Pathways. J Evid Based Integr Med 2024; 29:2515690X241291381. [PMID: 39410848 PMCID: PMC11489918 DOI: 10.1177/2515690x241291381] [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] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Huanglianjiedu decoction (HLJDD) is a classical Traditional Chinese Medicine (TCM) prescription with thousand years of clinical use against various malignancies, including pancreatic adenocarcinoma (PAAD). However, its potential bioactive component and molecular mechanism remains unclear. AIMS This study is to inspect the HLJDD mechanisms of action against PAAD via integrated computational and pharmacochemistry strategy, in vivo and in vitro experiments to validate associated targets and pathways. METHODS A PAAD xenograft model was established by subcutaneous injecting Panc02 cells into C57BL/6 mice. Ultra-high performance liquid chromatography with tandem mass spectrometry (UHPLC-MS/MS) was engaged to determine constituents of HLJDD and assessed for pharmacokinetic scheme using the TCM Systems Pharmacology Platform (TCM-SP). Differentially expressed genes (DEGs) of PAAD was retrieved from the transcriptome dataset GSE43795, followed by recognizing overlapping targets the oncogenes and target genes of PAAD and HLJDD, respectively. Putative signaling pathways of HLJDD in treating PAAD were enriched using KEGG and GO analyses. The anti-PAAD effects of HLJDD was assessed in vivo and in vitro, besides, the potential mechanism was validated using immunoblotting and immunohistochemical assays. RESULTS HLJDD significantly suppressed the growth of transplanted PAAD tumors, constrained PAAD progression, and induced apoptosis and S-phase arrest. Seventy-five active components meeting the drug-likeness criteria and 278 target genes of HLJDD were identified. KEGG analysis indicated that the top three enriched pathways were cancer, AGE-RAGE signaling, and IL-17 signaling pathways. Disease enrichment analysis highlighted immune, pharmacological, and cancer-related diseases as the top three categories. A total of 47 potential target genes were identified. Immunoblotting revealed that HLJDD inhibited PI3K and MAPK-related signaling pathways, while immunohistochemical staining confirmed that HLJDD suppressed the expression of phosphorylated MAPK and ERK1/2. CONCLUSION HLJDD inhibited PAAD in vitro and in vivo via the modulation of multiple mechanisms, including regulation of PI3K/AKT/mTOR and MAPK/ERK1/2 signaling pathways.
Collapse
Affiliation(s)
- Shu Dong Md
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Panling Xu Md
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Chinese Integrative Medicine Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Peiwen Yang Md
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Juying Jiao Md
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chien-Shan Cheng Md PhD
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Traditional Chinese Medicine, Shanghai Jiao Tong University School of Medicine Affiliated Ruijin Hospital, Shanghai, China
| | - Lianyu Chen Md PhD
- Department of Integrative Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Kakali B. Natural Compounds as Protease Inhibitors in Therapeutic Focus on Cancer Therapy. Anticancer Agents Med Chem 2024; 24:1167-1181. [PMID: 38988167 DOI: 10.2174/0118715206303964240708095110] [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: 02/26/2024] [Revised: 06/12/2024] [Accepted: 06/12/2024] [Indexed: 07/12/2024]
Abstract
Proteases are implicated in every hallmark of cancer and have complicated functions. For cancer cells to survive and thrive, the process of controlling intracellular proteins to keep the balance of the cell proteome is essential. Numerous natural compounds have been used as ligands/ small molecules to target various proteases that are found in the lysosomes, mitochondria, cytoplasm, and extracellular matrix, as possible anticancer therapeutics. Promising protease modulators have been developed for new drug discovery technology through recent breakthroughs in structural and chemical biology. The protein structure, function of significant tumor-related proteases, and their natural compound inhibitors have been briefly included in this study. This review highlights the most current frontiers and future perspectives for novel therapeutic approaches associated with the list of anticancer natural compounds targeting protease and the mode and mechanism of proteinase-mediated molecular pathways in cancer.
Collapse
Affiliation(s)
- Bhadra Kakali
- Department of Zoology, University of Kalyani, Kalyani, 741235, India
| |
Collapse
|
8
|
Xu Q, Zheng J, Su Z, Chen B, Gu S. COL10A1 promotes tumorigenesis by modulating CD276 in pancreatic adenocarcinoma. BMC Gastroenterol 2023; 23:397. [PMID: 37974070 PMCID: PMC10652574 DOI: 10.1186/s12876-023-03045-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Pancreatic adenocarcinoma (PAAD) is a lethal malignant tumour. Further study is needed to determine the molecular mechanism and identify novel biomarkers of PAAD. METHODS Gene expression data from the GSE62165 microarray were analysed with the online software Morpheus to identify differentially expressed genes (DEGs). The STRING database was used to generate a protein‒protein interaction (PPI) network for these DEGs. Hub genes were identified with Cytoscape. COL10A1 expression in PAAD was analysed via the GEPIA database. COL10A1 expression in pancreatic cancer cell lines was measured by using qRT‒PCR. The LinkedOmics database was utilized to perform survival analysis of pancreatic adenocarcinoma patients grouped based on COL10A1 expression level. CCK-8, wound healing, and Transwell assays were used to study the role of COL10A1 in pancreatic cancer cell viability, migration, and invasion. Differentially expressed genes that were related to COL10A1 in PAAD were analysed via the LinkedOmics portal. After COL10A1 was knocked down, CD276 expression was assessed by western blotting. RESULTS COL10A1 was identified as one of the hub genes in PAAD by bioinformatics analysis of the GSE62165 microarray with Morpheus, the STRING database and Cytoscape. GEPIA revealed elevated expression of COL10A1 in PAAD samples vs. normal samples. COL10A1 expression was also increased in pancreatic cancer cells vs. control cells. Survival analysis of PAAD patients via LinkedOmics revealed that high expression of COL10A1 was associated with a poorer prognosis. Knockdown of COL10A1 inhibited the proliferation, migration, and invasion of cells in functional assays. Furthermore, mechanistic studies indicated that CD276 was a target of COL10A1 and that knockdown of COL10A1 decreased CD276 expression. Overexpression of CD276 in cells reversed COL10A1 knockdown-induced repression of proliferation and migration. CONCLUSIONS Our research suggests that COL10A1 promotes pancreatic adenocarcinoma tumorigenesis by regulating CD276. This study provides new insight into biomarkers and possible targets for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Qiaodong Xu
- Department of Hepatobiliary surgery, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515041, China
| | - Jieting Zheng
- Department of pharmacy, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515041, China
| | - Zegeng Su
- Department of anesthesiology, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515041, China
| | - Binlie Chen
- Department of Hepatobiliary surgery, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515041, China
| | - Songgang Gu
- Department of Hepatobiliary surgery, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515041, China.
| |
Collapse
|
9
|
Meng T, Liu J, Chang H, Qie R. Reverse predictive analysis of Rhizoma Pinelliae and Rhizoma Coptidis on differential miRNA target genes in lung adenocarcinoma. Medicine (Baltimore) 2023; 102:e32999. [PMID: 36800601 PMCID: PMC9936040 DOI: 10.1097/md.0000000000032999] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/19/2023] Open
Abstract
To use bioinformatics and network analysis to reveal the mechanism of "Rhizoma Pinelliae-Rhizoma Coptidis" herb pair in the treatment of lung adenocarcinoma. The target and pathway of "Rhizoma Pinelliae-Rhizoma Coptidis" herb pair in the treatment of lung adenocarcinoma were explored by online databases and network analysis tools, and the potential biomarkers of "Rhizoma Pinelliae-Rhizoma Coptidis" herb pair in the treatment of lung adenocarcinoma were predicted in reverse. A total of 59 traditional Chinese medicine compounds and 510 drug targets were screened in this study. A total of 25 micro-RNAs and 15,323 disease targets were obtained through GEO2R software analysis. In the end, 294 therapeutic targets and 47 core targets were obtained. A total of 186 gene ontology enrichment assays were obtained, and core therapeutic targets play multiple roles in biological processes, molecular functions, and cellular composition. Kyoto encyclopedia of genes and genomes pathway enrichment analysis showed that the core targets were mainly enriched in cancer-related pathways, immune-related pathways, endocrine-related pathways, etc, among which the non-small cell lung cancer pathway was the most significant core pathway. Molecular docking shows that the compound and the target have good binding ability. "Rhizoma Pinelliae-Rhizoma Coptidis" herb pair plays a mechanism of action in the treatment of lung adenocarcinoma through multiple targets and pathways. miR-5703, miR-3125, miR-652-5P, and miR-513c-5p may be new biomarkers for the treatment of lung adenocarcinoma.
Collapse
Affiliation(s)
- Tianwei Meng
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Jiawen Liu
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Hong Chang
- Department of Pharmacy, Baotou Medical College, Baotou, Inner Mongolia, China
- * Correspondence: Hong Chang, Department of Pharmacy, Baotou Medical College, Baotou, Inner Mongolia, China (e-mail: )
| | - Rui Qie
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| |
Collapse
|
10
|
Nishimura T, Takadate T, Maeda S, Suzuki T, Minowa T, Fukuda T, Bando Y, Unno M. Disease-related protein co-expression networks are associated with the prognosis of resectable node-positive pancreatic ductal adenocarcinoma. Sci Rep 2022; 12:14709. [PMID: 36038612 PMCID: PMC9424258 DOI: 10.1038/s41598-022-19182-9] [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: 04/09/2022] [Accepted: 08/25/2022] [Indexed: 12/05/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a multifactorial disease, the molecular profile of which remains unclear. This study aimed at unveiling the disease-related protein networks associated with different outcomes of resectable, node-positive PDAC cases. We assessed laser-microdissected cancerous cells from PDAC tissues of a poor outcome group (POG; n = 4) and a better outcome group (BOG; n = 4). Noncancerous pancreatic duct tissues (n = 5) were used as the reference. We identified four representative network modules by applying a weighted network correlation analysis to the obtained quantitative PDAC proteome datasets. Two network modules that were significant for POG were associated with the heat shock response to hypoxia-related stress; in the latter, a large involvement of the non-canonical Hedgehog pathway (regulated by GLI1), the internal ribosome entry site-mediated cap-independent translation, the inositol requiring enzyme 1-alpha (IRE1α)/X-box binding protein 1 pathway of the unfolding protein response (UPR), and the aerobic glycolysis was observed. By contrast, the BOG characteristic module was involved in the inactivation of the UPR pathway via the synoviolin 1-dependent proteasomal degradation of IRE1α, the activation of SOX2, and the loss of PALB2 (partner and localizer of BRCA2) function, all potentially suppressing malignant tumor development. Our findings might facilitate future therapeutic strategies for PDAC.
Collapse
Affiliation(s)
- Toshihide Nishimura
- Department of Translational Medicine Informatics, St. Marianna University School of Medicine, Kawasaki, Kanagawa, 216-8511, Japan. .,Biosys Technologies, Inc., Tokyo, Tokyo, 153-8904, Japan.
| | - Tatsuyuki Takadate
- Department of Surgery, National Hospital Organization Sendai Medical Center, Sendai, Miyagi, 983-8520, Japan
| | - Shimpei Maeda
- Department of Surgery, Saitama Medical Center, Jichi Medical University, Shimotsuke, Tochigi, 329-0498, Japan
| | - Takashi Suzuki
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8574, Japan
| | - Takashi Minowa
- Nanotechnology Innovation Station, National Institute for Materials Science, Tsukuba, Ibaraki, Japan
| | - Tetsuya Fukuda
- Biosys Technologies, Inc., Tokyo, Tokyo, 153-8904, Japan
| | - Yasuhiko Bando
- Biosys Technologies, Inc., Tokyo, Tokyo, 153-8904, Japan
| | - Michiaki Unno
- Department of Surgery, National Hospital Organization Sendai Medical Center, Sendai, Miyagi, 983-8520, Japan.,Department of Surgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980-8574, Japan
| |
Collapse
|
11
|
Lin GS, Zhao MM, Fu QC, Zhao SY, Ba TT, Yu HX. Palmatine attenuates hepatocyte injury by promoting autophagy via the AMPK/mTOR pathway after alcoholic liver disease. Drug Dev Res 2022; 83:1613-1622. [PMID: 35976121 DOI: 10.1002/ddr.21981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/23/2022] [Accepted: 07/16/2022] [Indexed: 11/08/2022]
Abstract
Alcoholic liver disease is one of the diseases with the highest fatality rate worldwide. The cellular process of autophagy which recycles damaged organelles to maintain protein and organelle homeostasis is found to positively influence survival during hepatic insufficiency, although the mechanism is poorly understood. Palmatine (PLT) has a variety of biological functions, such as broad-spectrum antibacterial action, neuroprotective, antioxidant stress, and antiviral and anti-inflammatory activities. However, it is not known whether PLT has a protective effect against alcoholic liver injury. Here, we investigated the protective effect of PLT in a cellular model of alcohol-induced acute liver injury and further explored its mechanism of action. In this study, we show for the first time that PLT attenuates alcohol-induced hepatocyte injury by promoting autophagy to play an essential protective role. As PLT treatment induced a brief increase in LC3-II conversion and p62 degradation, it also upregulated the expression of ATG5 and ATG7. The expression levels of the proapoptotic proteins Bax, Caspase 3, and Caspase 9 significantly decreased, while the antiapoptotic protein levels of Bcl-2 upregulated after treatment with PLT. However, in presence of the autophagy inhibitor, 3-methyladenine, the effect of PLT in inhibiting ethanol-induced hepatocyte injury reversed significantly. Mechanistically, the protective effects of PLT may be mediated by promoting the activation of the AMP-activated protein kinase/mammalian target of rapamycin signaling pathway. Therefore, we believe that the development of alcoholic liver injuries may be controlled by PLT by inhibiting hepatocyte apoptosis through the autophagy pathway. The study lays a solid theoretical and practical basis for future animal models and clinical studies of PLT.
Collapse
Affiliation(s)
- Guo-Shuai Lin
- Department of Infectious Disease, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Mao-Mao Zhao
- Department of Infectious Disease, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Qi-Chao Fu
- Department of Gastroenterology, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Shu-Yi Zhao
- Department of Oncology, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong, China
| | - Tao-Tao Ba
- Department of Infectious Disease, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Hong-Xia Yu
- Department of Infectious Disease, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| |
Collapse
|
12
|
Wang W, Gu W, He C, Zhang T, Shen Y, Pu Y. Bioactive components of Banxia Xiexin Decoction for the treatment of gastrointestinal diseases based on flavor-oriented analysis. JOURNAL OF ETHNOPHARMACOLOGY 2022; 291:115085. [PMID: 35150814 DOI: 10.1016/j.jep.2022.115085] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/23/2022] [Accepted: 02/03/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Banxia Xiexin Decoction (BXD) was first recorded in a Chinese medical classic, Treatise on Febrile Diseases and Miscellaneous Diseases, which was written in the Eastern Han dynasty of China. This ancient prescription consists of seven kinds of Chinese herbal medicine, namely, Pinellia ternata, Rhizoma Coptidis, Radix scutellariae, Rhizoma Zingiberis, Ginseng, Jujube, and Radix Glycyrrhizaepreparata. In clinic practice, its original application in China mainly has focused on the treatment of chronic gastritis for several hundred years. BXD is also effective in treating other gastrointestinal diseases (GIDs) in modern medical application. Despite available literature support and clinical experience, the treatment mechanisms or their relationships with the bioactive compounds in BXD responsible for its pharmacological actions, still need further explorations in more diversified channels. According to the analysis based on the five-flavor theory of TCM, BXD is traditionally viewed as the most representative prescription for pungent-dispersion, bitter-purgation and sweet-tonification. Consequently, based on the flavor-oriented analysis, the compositive herbs in BXD can be divided into three flavor groups, namely, the pungent, bitter, and sweet groups, each of which has specific active ingredients that are possibly relevant to GID treatment. AIM OF THE REVIEW This paper summarized recent literatures on BXD and its bioactive components used in GID treatment, and provided the pharmacological or chemical basis for the further exploration of the ancient prescription and the relative components. METHOD ology: Relevant literature was collected from various electronic databases such as Pubmed, Web of Science, and China National Knowledge Infrastructure (CNKI). Citations were based on peer-reviewed articles published in English or Chinese during the last decade. RESULTS Multiple components were found in the pungent, bitter, and sweet groups in BXD. The corresponding bioactive components include gingerol, shogaol, stigmasterol, and β-sitosterol in the pungent group; berberine, palmatine, coptisine, baicalein, and baicalin in the bitter group; and ginsenosides, polysaccharides, liquiritin, and glycyrrhetinic acid in the sweet group. These components have been found directly or indirectly responsible for the remarkable effects of BXD on GID. CONCLUSION This review provided some valuable reference to further clarify BXD treatment for GID and their possible material basis, based on the perspective of the flavor-oriented analysis.
Collapse
Affiliation(s)
- Weiwei Wang
- Experiment Center of Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Weiliang Gu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chao He
- Experiment Center of Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yao Shen
- Shanghai Center of Biomedicine Development, Shanghai, 201203, China.
| | - Yiqiong Pu
- Experiment Center of Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
13
|
Management of Pancreatic Cancer and Its Microenvironment: Potential Impact of Nano-Targeting. Cancers (Basel) 2022; 14:cancers14122879. [PMID: 35740545 PMCID: PMC9221065 DOI: 10.3390/cancers14122879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary The poor prognosis and survival rates associated with pancreatic cancer show that there is a clear unmet need for better disease management. The heterogeneity of the tumor and its microenvironment, including stroma and fibrosis, creates a challenge for current therapy. The pathogenesis of pancreatic cancer is mediated by several factors, such as severed communication between pancreatic stellate cells and stroma and the consequences of hypoxia-inducible factors that aid in the survival of the pancreatic tumor. Given the multiple limitations of molecular targeting, multiple functional nano-targeting offers a breakthrough in pancreatic cancer treatment through its ability to overcome the physical challenges posed by the tumor microenvironment, amongst many others. Abstract Pancreatic ductal adenocarcinoma (PDAC) is rare and difficult to treat, making it a complicated diagnosis for every patient. These patients have a low survival rate along with a poor quality of life under current pancreatic cancer therapies that adversely affect healthy cells due to the lack of precise drug targeting. Additionally, chemoresistance and radioresistance are other key challenges in PDAC, which might be due in part to the lack of tumor-targeted delivery of sufficient levels of different chemotherapies because of their low therapeutic index. Thus, instead of leaving a trail of off-target damage when killing these cancer cells, it is best to find a way that targets them directly. More seriously, metastatic relapse often occurs after surgery, and therefore, achieving improved outcomes in the management of PDAC in the absence of strategies preventing metastasis is likely to be impossible. Nano-targeting of the tumor and its microenvironment has shown promise for treating various cancers, which might be a promising approach for PDAC. This review updates the advancements in treatment modalities for pancreatic cancer and highlights future directions that warrant further investigation to increase pancreatic patients’ overall survival.
Collapse
|
14
|
Wang C, Wang Y, Fu Z, Huang W, Yu Z, Wang J, Zheng K, Zhang S, Li S, Chen J. MiR-29b-3p Inhibits Migration and Invasion of Papillary Thyroid Carcinoma by Downregulating COL1A1 and COL5A1. Front Oncol 2022; 12:837581. [PMID: 35530352 PMCID: PMC9075584 DOI: 10.3389/fonc.2022.837581] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/16/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction MicroRNAs (miRNAs) are small noncoding RNA molecules that regulate genetic expression and are also vital for tumor initiation and development. MiR-29b-3p was found to be involved in regulating various biological processes of tumors, including tumor cell proliferation, metastasis, and apoptosis inhibition; however, the biofunction and molecule-level mechanisms of miR-29b-3p inpapillary thyroid carcinoma (PTC) remain unclear. Methods The expression of miR-29b-3p in PTC samples was tested via qRT-PCR. Cellular proliferation was analyzed by CCK-8 and EdU assays, and cellular migratory and invasive abilities were assessed utilizing wound-healing and Transwell assays. In addition, protein expressions of COL1A1, COL5A1, E-cadherin, N-cadherin, Snail, and Vimentin were identified via Western blot (WB) assay. Bioinformatics, qRT-PCR, WB, and dual luciferase reporter assays were completed to identify whether miR-29b-3p targeted COL1A1 and COL5A1. In addition, our team explored the treatment effects of miR-29b-3p on a murine heterograft model. Results Our findings revealed that miR-29b-3p proved much more regulated downward in PTC tissue specimens than in adjacent non-cancerous tissues. Meanwhile, decreased expression of miR-29b-3p was strongly related to the TNM stage of PTC patients (p<0.001), while overexpression of miR-29b-3p in PTC cells suppressed cellular migration, invasion, proliferation, and EMT. Conversely, silencing miR-29b-3p yielded the opposite effect. COL1A1 and COL5A1 were affirmed as the target of miR-29b-3p. Additionally, the COL1A1 and COL5A1 were highly expressed in PTC tumor samples than in contrast to neighboring healthy samples. Functional assays revealed that overexpression of COL1A1 or COL5A1 reversed the suppressive role of miR-29b-3p in migration, invasion, and EMT of PTC cells. Finally, miR-29b-3p agomir treatment dramatically inhibited Xenograft tumor growth in the animal model. Conclusions These findings document that miR-29b-3p inhibited PTC cells invasion and metastasis by targeting COL1A1 and COL5A1; this study also sparks new ideas for risk assessment and miRNA replacement therapy in PTC.
Collapse
Affiliation(s)
- Congjun Wang
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Clinical Research Lab, Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Ye Wang
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Clinical Research Lab, Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Zhao Fu
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Clinical Research Lab, Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Weijia Huang
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Clinical Research Lab, Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Zhu Yu
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Clinical Research Lab, Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Jiancheng Wang
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Clinical Research Lab, Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Kaitian Zheng
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Clinical Research Lab, Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Siwen Zhang
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Clinical Research Lab, Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
| | - Shen Li
- Department of Gastrointestinal, Hernia and Enterofistula Surgery, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Junqiang Chen
- Department of Gastrointestinal Gland Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Clinical Research Lab, Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, Nanning, China
- *Correspondence: Junqiang Chen,
| |
Collapse
|
15
|
Mechanism of Synsepalum dulcificum Daniell. Inhibiting Lung Adenocarcinoma. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:5242179. [PMID: 35190747 PMCID: PMC8858071 DOI: 10.1155/2022/5242179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/12/2022] [Indexed: 12/24/2022]
Abstract
Objective: Synsepalum dulcificum Daniell. (SD) is a natural plant fruit and is famous for containing miraculin. It has been reported that SD can be used as an adjuvant treatment to correct patients' loss of taste during the antitumor process, but the effect of SD itself as an antitumor is not clear. In this study, we investigated the mechanism of action of SD on lung adenocarcinoma using network pharmacology. Materials and Methods. The components of SD were identified by liquid chromatography-mass spectrometry, and then the compounds that affect tumor immunity of SD were screened and the related targets were predicted by TCMIO database. At the same time, the results were associated with lung adenocarcinoma targets included in the MalaCards and CTD databases, so as to construct a compound-target action network diagram and explore the mechanism of SD in the treatment of lung adenocarcinoma. In in vitro experiments, cell viability was determined and western blotting was used to detect the related expression of action targets to determine the therapeutic effect of SD. Results. In this experiment, 335 chemical components were identified in SD, and 107 components were related to tumor immunity. After screening by ADME, it was found that 11 compounds might be inhaled into the human body and affect the growth of lung adenocarcinoma. In vitro experiments showed that SD could inhibit the growth of lung adenocarcinoma A549 cells. SD could reduce the expression of PCNA (P < 0.05) and significantly increase the expression of Caspase-3 (P < 0.05). The results of further experiments showed that SD could significantly reduce the phosphorylation of EGFR (P < 0.05), and SD could also effectively inhibit the expression of JAK and STAT3 phosphorylation (P < 0.01) and inhibit the expression of PI3K and AKT phosphorylation (P < 0.01). Conclusion. SD can inhibit the growth of lung adenocarcinoma A549 cells and the potential mechanism was found to be the inhibition of EGFR/JAK/STAT3 and EGFR/PI3K/AKT signaling pathway, and the substance basis for SD to exert antitumor effect may be catechin, taxifolin, betaine, epigallocatechin gallate, erucamide, guanosine, kaempferol, lanosterol, morin, oleanolic acid, and quercetin.
Collapse
|
16
|
Shu Y, He L, Gao M, Xiao F, Yang J, Wang S, Wei H, Zhang F, Wei H. EOGT Correlated With Immune Infiltration: A Candidate Prognostic Biomarker for Hepatocellular Carcinoma. Front Immunol 2022; 12:780509. [PMID: 35069551 PMCID: PMC8766744 DOI: 10.3389/fimmu.2021.780509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 12/09/2021] [Indexed: 11/13/2022] Open
Abstract
Background A preliminary study by our group revealed that the deficiency of EGF domain-specific O-linked N-acetylglucosamine transferase (EOGT) impaired regulatory T-cell differentiation in autoimmune hepatitis. Nevertheless, the prognostic value of EOGT in advanced hepatocellular carcinoma (HCC) and its relationship with immune infiltration remain obscured. Methods Initially, EOGT expression was evaluated by Oncomine, TIMER, GEO, and UALCAN databases. Besides, the prognostic potential of EOGT expression was analyzed using GEPIA, Kaplan-Meier plotter, CPTAC, Cox regression, and nomogram in HCC samples. Furthermore, we investigated the association between EOGT expression and tumor mutation burden, DNA methylation, and immune infiltration in addition to its possible mechanism via cBioPortal, TIMER, GEPIA, ESTIMATE, CIBERSORT, GSEA, STRING, and Cytoscape. Results The expression of EOGT in HCC was significantly higher than that in normal tissues. Additionally, elevated EOGT expression was correlated with advanced tumor staging and linked to poor overall survival and relapse-free survival, serving as a significant unfavorable prognostic indicator in HCC patients. Remarkably, our results revealed that high-EOGT expression subgroups with elevated TP53 or low CTNNB1 mutations have worse clinical outcomes than the others. Regarding immune infiltration, immunofluorescent staining showed that immune cells in HCC were positive for EOGT. Besides, elevated EOGT expression was linked to exhausted T cells and immune suppressor cells in HCC samples. More importantly, the proportion of CD8+ T cells was reduced in HCC samples with a high level of EOGT expression, but EOGT did not exhibit prognostic potential in HCC samples with increased CD8+ T cells. Conclusions EOGT may hold great potential as a novel biomarker to distinguish prognosis and immune profiles of HCC patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hongshan Wei
- Department of Gastroenterology, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Tilaoui M, Ait Mouse H, Zyad A. Update and New Insights on Future Cancer Drug Candidates From Plant-Based Alkaloids. Front Pharmacol 2021; 12:719694. [PMID: 34975465 PMCID: PMC8716855 DOI: 10.3389/fphar.2021.719694] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 11/23/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer is a complex multifactorial disease that results from alterations in many physiological and biochemical functions. Over the last few decades, it has become clear that cancer cells can acquire multidrug resistance to conventional anticancer drugs, resulting in tumor relapse. Thus, there is a continuous need to discover new and effective anticancer drugs. Natural products from plants have served as a primary source of cancer drugs and continue to provide new plant-derived anticancer drugs. The present review describes plant-based alkaloids, which have been reported as active or potentially active in cancer treatment within the past 4 years (2017-2020), both in preclinical research and/or in clinical trials. In addition, recent insights into the possible molecular mechanism of action of alkaloid prodrugs naturally present in plants are also highlighted.
Collapse
Affiliation(s)
- Mounir Tilaoui
- Experimental Oncology and Natural Substances Team, Cellular and Molecular Immuno-pharmacology, Faculty of Sciences and Technology, Sultan Moulay Slimane University, Beni-Mellal, Morocco
| | | | | |
Collapse
|
18
|
Ren T, Xue X, Wang X, Zhou X, Dang S. Bioinformatic and experimental analyses of key biomarkers in pancreatic cancer. Exp Ther Med 2021; 22:1359. [PMID: 34659505 PMCID: PMC8515505 DOI: 10.3892/etm.2021.10794] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 09/15/2020] [Indexed: 12/18/2022] Open
Abstract
The present study aimed to screen the key genes in pancreatic cancer and to explore the pathogenesis of pancreatic cancer. A total of three expression profiling datasets (GSE28735, GSE16515 and GSE15471) associated with pancreatic cancer were retrieved from the public gene chip database. The differentially expressed genes (DEGs) were screened by GEO2R and subjected to Gene Ontology (GO) and signaling pathway enrichment analysis. Furthermore, a protein interaction network was constructed. The GEPIA online database was used to screen for genes that affect the prognosis of pancreatic cancer. Finally, cell functional experiments were performed on the selected key genes. A total of 72 DEGs were identified, including 52 upregulated and 20 downregulated genes. Enrichment analysis revealed roles of the DEGs in endodermal cell differentiation, cell adhesion, extracellular matrix-receptor interaction and PI3K-Akt signaling pathway. In total, 10 key nodal genes were identified, including integrin subunit α 2 (ITGA2), ITGB6 and collagen α 1 chain 1. Through survival analysis, two genes with an impact on the prognosis of pancreatic cancer were identified, namely ITGA2 and ITGB6. Silencing of ITGB6 in a pancreatic cancer cell line significantly suppressed cell proliferation and induced cell cycle arrest at G2/M phase. The identified key genes and signaling pathways may help to deepen the understanding of the molecular mechanisms involved in pancreatic cancer and provide a theoretical basis to develop novel therapies.
Collapse
Affiliation(s)
- Tianyu Ren
- Department of General Surgery, The Affiliated Hospital, Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Xiaofei Xue
- Department of General Surgery, Pucheng Hospital, Weinan, Shaanxi 715500, P.R. China
| | - Xiaogang Wang
- Department of General Surgery, Pucheng Hospital, Weinan, Shaanxi 715500, P.R. China
| | - Xingtong Zhou
- Department of General Surgery, The Affiliated Hospital, Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China
| | - Shengchun Dang
- Department of General Surgery, The Affiliated Hospital, Jiangsu University, Zhenjiang, Jiangsu 212001, P.R. China.,Department of General Surgery, Pucheng Hospital, Weinan, Shaanxi 715500, P.R. China
| |
Collapse
|
19
|
Grabarska A, Wróblewska-Łuczka P, Kukula-Koch W, Łuszczki JJ, Kalpoutzakis E, Adamczuk G, Skaltsounis AL, Stepulak A. Palmatine, a Bioactive Protoberberine Alkaloid Isolated from Berberis cretica, Inhibits the Growth of Human Estrogen Receptor-Positive Breast Cancer Cells and Acts Synergistically and Additively with Doxorubicin. Molecules 2021; 26:molecules26206253. [PMID: 34684834 PMCID: PMC8538708 DOI: 10.3390/molecules26206253] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/01/2021] [Accepted: 10/08/2021] [Indexed: 12/24/2022] Open
Abstract
Palmatine (PLT) is a natural isoquinoline alkaloid that belongs to the class of protoberberines and exhibits a wide spectrum of pharmacological and biological properties, including anti-cancer activity. The aim of our study was to isolate PLT from the roots of Berberis cretica and investigate its cytotoxic and anti-proliferative effects in vitro alone and in combination with doxorubicine (DOX) using human ER+/HER2− breast cancer cell lines. The alkaloid was purified by column chromatography filled with silica gel NP and Sephadex LH-20 resin developed in the mixture of methanol: water (50:50 v/v) that provided high-purity alkaloid for bioactivity studies. The purity of the alkaloid was confirmed by high resolution mass measurement and MS/MS fragmentation analysis in the HPLC-ESI-QTOF-MS/MS-based analysis. It was found that PLT treatment inhibited the viability and proliferation of breast cancer cells in a dose-dependent manner as demonstrated by MTT and BrdU assays. PLT showed a quite similar growth inhibition on breast cancer cells with IC50 values ranging from 5.126 to 5.805 µg/mL. In contrast, growth of normal human breast epithelial cells was not affected by PLT. The growth inhibitory activity of PLT was related to the induction of apoptosis, as determined by Annexin V/PI staining. Moreover, PLT sensitized breast cancer cells to DOX. Isobolographic analysis revealed synergistic and additive interactions between studied agents. Our studies suggest that PLT can be a potential candidate agent for preventing and treating breast cancer.
Collapse
Affiliation(s)
- Aneta Grabarska
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1, 20-093 Lublin, Poland;
- Correspondence: ; Tel.: +48-81448-6350
| | - Paula Wróblewska-Łuczka
- Department of Pathophysiology, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland; (P.W.-Ł.); (J.J.Ł.)
| | - Wirginia Kukula-Koch
- Department of Pharmacognosy with Medicinal Plants Garden, Medical University of Lublin, Chodzki 1, 20-093 Lublin, Poland;
| | - Jarogniew J. Łuszczki
- Department of Pathophysiology, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland; (P.W.-Ł.); (J.J.Ł.)
| | - Eleftherios Kalpoutzakis
- Laboratory of Pharmacognosy and Natural Products Chemistry, School of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupoli Zografou, 15771 Athens, Greece; (E.K.); (A.L.S.)
| | - Grzegorz Adamczuk
- Independent Medical Biology Unit, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland;
| | - Alexios Leandros Skaltsounis
- Laboratory of Pharmacognosy and Natural Products Chemistry, School of Pharmacy, National and Kapodistrian University of Athens, Panepistimioupoli Zografou, 15771 Athens, Greece; (E.K.); (A.L.S.)
| | - Andrzej Stepulak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Chodzki 1, 20-093 Lublin, Poland;
| |
Collapse
|
20
|
Network Pharmacology-Based Analysis of the Effects of Corydalis decumbens (Thunb.) Pers. in Non-Small Cell Lung Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:4341517. [PMID: 34497656 PMCID: PMC8421182 DOI: 10.1155/2021/4341517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/17/2021] [Accepted: 07/06/2021] [Indexed: 11/17/2022]
Abstract
Non-small cell lung cancer (NSCLC) is one of the most malignant tumors worldwide. The main treatment for NSCLC is based on Western medicine; however, the overall effect is unsatisfactory. This study aimed to investigate the potential therapeutic targets and pharmacological mechanisms of action of the traditional Chinese medicine Corydalis decumbens (Thunb.) Pers. in NSCLC based on network pharmacology and bioinformatics. The overlapping genes between Corydalis decumbens (Thunb.) Pers. and NSCLCs were screened using Venn analysis. Cytoscape 3.7.1 software was used to analyze the overlapping target protein-protein interaction (PPI) network. Gene ontology and pathway enrichment analysis using the Kyoto Encyclopedia of Genes and Genomics database were performed to exploring biological functions of the overlapping genes. The gene expression profiling interactive analysis dataset was used to analyze the correlation between hub gene expression and disease. This study revealed 38 nodes with 191 edges, which may be therapeutic targets for NSCLC. PPI network analysis showed that the most likely association was between the genes AR and NCOA2, NCOA2, and RXRA and ESR1 and NCOA2. These overlapping genes were mainly enriched in the estrogen signaling pathway, calcium signaling pathway, cholinergic synapse, and PI3K-Akt signaling pathway. ESR2 mRNA levels were significantly downregulated in patients with lung adenocarcinoma (LUAD) getting worse, and KDR levels were lower in lung squamous cell carcinoma (LUSC) than those in normal tissue. PTGS2 expression was correlated with the median survival time of LUAD, and ESR1 expression was correlated with the median survival time of LUSC. The application of network pharmacology revealed the potential mechanism underlying the effects of Corydalis decumbens (Thunb.) Pers. in NSCLC treatment and provided a theoretical basis for further in-depth research in this field.
Collapse
|
21
|
Hou L, Lin T, Wang Y, Liu B, Wang M. Collagen type 1 alpha 1 chain is a novel predictive biomarker of poor progression-free survival and chemoresistance in metastatic lung cancer. J Cancer 2021; 12:5723-5731. [PMID: 34475986 PMCID: PMC8408119 DOI: 10.7150/jca.59723] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/19/2021] [Indexed: 01/02/2023] Open
Abstract
Background: Collagen type 1 alpha 1 chain (COL1A1) is an extracellular matrix protein comprising two alpha 1 chains and one alpha 2 chain. Our previous study identified that COL1A1 is the key gene during the development and progression of lung adenocarcinoma by multi-omics analysis. However, the clinical significance of COL1A1 expression in lung cancer samples remains largely unknown. Here, we aimed to evaluate the level of COL1A1 in lung cancer samples and correlate its level with the clinical outcome. Methods:COL1A1 gene expression in lung cancer samples was analyzed using the Oncomine database (www.oncomine.org). A total of 308 lung cancer samples (208 formalin-fixed paraffin-embedded tissues and 100 blood samples) were assessed for protein expression of COL1A1. Immunohistochemistry staining and enzyme-linked immunosorbent assay were used to detect COL1A1 expression in tissues and serum, respectively. Results: We identified an elevation of COL1A1 in mRNA level and gene amplification in lung cancer tissues compared with normal lung tissues. High COL1A1 expression was observed in lung cancer tissues and serum (P < 0.05), it was significantly correlated with the peripheral type tumor, the larger diameter of the tumor, the occurrence of lymph node metastases and distant metastases, a higher TNM stage, and smoking (P < 0.05). High COL1A1 expression was associated with poor progression-free survival (PFS) and chemoresistance in lung cancer patients (P < 0.05). Multivariable Cox-regression analysis showed that COL1A1 expression was an independent prognostic factor (P < 0.05). Furthermore, the area under the receiver operating characteristic (AUC) curve was 0.909 for the combined COL1A1 and carcinoembryonic antigen (CEA) measurement. Conclusion: Our findings revealed that COL1A1 could be used as a novel diagnostic, prognostic, and chemoresistance biomarker of human lung cancer, and these results provide a potential therapeutic strategy for lung cancer patients.
Collapse
Affiliation(s)
- Lingjie Hou
- Department of Respiratory Medical Oncology, Harbin Medical University Cancer Hospital, Heilongjiang, Harbin 150081, China
| | - Tie Lin
- Department of Surgery, The First Affiliated Hospital of Harbin Medical University, Heilongjiang, Harbin 150001, China
| | - Yicun Wang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Jilin 130041, Changchun, China
| | - Bao Liu
- Department of Respiratory Medical Oncology, Harbin Medical University Cancer Hospital, Heilongjiang, Harbin 150081, China
| | - Meng Wang
- Department of Respiratory Medical Oncology, Harbin Medical University Cancer Hospital, Heilongjiang, Harbin 150081, China
| |
Collapse
|
22
|
Yin C, Shen W, Zhang M, Wen L, Huang R, Sun M, Gao Y, Xiong W. Inhibitory Effects of Palmatine on P2X7 Receptor Expression in Trigeminal Ganglion and Facial Pain in Trigeminal Neuralgia Rats. Front Cell Neurosci 2021; 15:672022. [PMID: 34366788 PMCID: PMC8339261 DOI: 10.3389/fncel.2021.672022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 06/01/2021] [Indexed: 11/13/2022] Open
Abstract
Trigeminal Neuralgia (TN) refers to recurrent severe paroxysmal pain in the distribution area of the trigeminal nerve, which seriously affects the quality of life of patients. This research applied the chronic constriction injury of the infraorbital nerve (CCI—ION) approach to induce an animal model of TN in rats. The mechanical pain threshold of each group of rats was determined postoperatively; the expression of P2X7 receptor in trigeminal ganglion (TG) was assessed by qRT-PCR, immunofluorescence and Western blot; and the changes of the proinflammatory cytokines IL-1β and TNF-α in serum of rats were detected by ELISA. The results showed that the administration of palmatine in the TN rats could reduce the mechanical pain threshold, significantly decrease the expression of P2X7 receptor in TG, and lower the serum concentrations of IL-1β and TNF-α, compared to the sham group. In addition, the phosphorylation level of p38 in TG of TN rats was significantly decreased after treatment with palmatine. Likewise, inhibition of P2X7 expression by shRNA treatment could effectively counteract the adversary changes of pain sensitivity, IL-1β and TNF-α production, and p38 phosphorylation in TN rats. Our data suggest that palmatine may alleviate mechanical facial pain in TN rats possibly by reducing the expression of P2X7 receptor in TG of TN rats, which may be attributable to inhibiting p38 phosphorylation and reducing the release of IL-1β and TNF-α.
Collapse
Affiliation(s)
- Cancan Yin
- Affiliated Stomatological Hospital of Nanchang University, Nanchang, China.,Hangzhou Stomatology Hospital, Hangzhou, China
| | - Wenhao Shen
- Affiliated Stomatological Hospital of Nanchang University, Nanchang, China
| | - Mingming Zhang
- Department of Physiology, Basic Medical College, Nanchang University, Nanchang, China
| | - Lequan Wen
- Joint Program of Nanchang University and Queen Mary University of London, Nanchang, China
| | - Ruoyu Huang
- Affiliated Stomatological Hospital of Nanchang University, Nanchang, China
| | - Mengyun Sun
- Department of Physiology, Basic Medical College, Nanchang University, Nanchang, China
| | - Yun Gao
- Department of Physiology, Basic Medical College, Nanchang University, Nanchang, China.,Jiangxi Provincial Key Laboratory of Autonomic Nervous Function and Disease, Nanchang, China
| | - Wei Xiong
- Affiliated Stomatological Hospital of Nanchang University, Nanchang, China.,Jiangxi Provincial Key Laboratory of Oral Biomedicine, Nanchang, China
| |
Collapse
|
23
|
Luo Y, Yin S, Lu J, Zhou S, Shao Y, Bao X, Wang T, Qiu Y, Yu H. Tumor microenvironment: a prospective target of natural alkaloids for cancer treatment. Cancer Cell Int 2021; 21:386. [PMID: 34284780 PMCID: PMC8290600 DOI: 10.1186/s12935-021-02085-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 07/08/2021] [Indexed: 12/17/2022] Open
Abstract
Malignant tumor has become one of the major diseases that seriously endangers human health. Numerous studies have demonstrated that tumor microenvironment (TME) is closely associated with patient prognosis. Tumor growth and progression are strongly dependent on its surrounding tumor microenvironment, because the optimal conditions originated from stromal elements are required for cancer cell proliferation, invasion, metastasis and drug resistance. The tumor microenvironment is an environment rich in immune/inflammatory cells and accompanied by a continuous, gradient of hypoxia and pH. Overcoming immunosuppressive environment and boosting anti-tumor immunity may be the key to the prevention and treatment of cancer. Most traditional Chinese medicine have been proved to have good anti-tumor activity, and they have the advantages of better therapeutic effect and few side effects in the treatment of malignant tumors. An increasing number of studies are giving evidence that alkaloids extracted from traditional Chinese medicine possess a significant anticancer efficiency via regulating a variety of tumor-related genes, pathways and other mechanisms. This paper reviews the anti-tumor effect of alkaloids targeting tumor microenvironment, and further reveals its anti-tumor mechanism through the effects of alkaloids on different components in tumor microenvironment.
Collapse
Affiliation(s)
- Yanming Luo
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shuangshuang Yin
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jia Lu
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shiyue Zhou
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yingying Shao
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xiaomei Bao
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Tao Wang
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yuling Qiu
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China.
| | - Haiyang Yu
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
24
|
Fan C, Lu W, Li K, Zhao C, Wang F, Ding G, Wang J. Identification of immune cell infiltration pattern and related critical genes in metastatic castration-resistant prostate cancer by bioinformatics analysis. Cancer Biomark 2021; 32:363-377. [PMID: 34151837 DOI: 10.3233/cbm-203222] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Metastatic castration-resistant prostate cancer (mCRPC) is the lethal stage of prostate cancer and the main cause of morbidity and mortality, which is also a potential target for immunotherapy. METHOD In this study, using the Approximate Relative Subset of RNA Transcripts (CIBERSORT) online method, we analysed the immune cell abundance ratio of each sample in the mCRPC dataset. The EdgeR (an R package) was used to classify differentially expressed genes (DEGs). Using the Database for annotation, visualisation and interactive exploration (DAVID) online method, we performed functional enrichment analyses. STRING online database and Cytoscape tools have been used to analyse protein-protein interaction (PPI) and classify hub genes. RESULTS The profiles of immune infiltration in mCRPC showed that Macrophages M2, Macrophages M0, T cells CD4 memory resting, T cells CD8 and Plasma cells were the main infiltration cell types in mCRPC samples. Macrophage M0 and T cell CD4 memory resting abundance ratios were correlated with clinical outcomes. We identified 1102 differentially expressed genes (DEGs) associated with the above two immune cells to further explore the underlying mechanisms. Enrichment analysis found that DEGs were substantially enriched in immune response, cell metastasis, and metabolism related categories. We identified 20 hub genes by the protein-protein interaction network analysis. Further analysis showed that three critical hub genes, CCR5, COL1A1 and CXCR3, were significantly associated with prostate cancer prognosis. CONCLUSION Our findings revealed the pattern of immune cell infiltration in mCRPC, and identified the types and genes of immune cells correlated with clinical outcomes. A new theoretical basis for immunotherapy may be given by our results.
Collapse
Affiliation(s)
- Caibin Fan
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Wei Lu
- School of Nursing, Suzhou Vocational Health and Technical College, Suzhou, Jiangsu, China.,Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Kai Li
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chunchun Zhao
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fei Wang
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guanxiong Ding
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianqing Wang
- Department of Urology, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
25
|
Geng Q, Shen Z, Li L, Zhao J. COL1A1 is a prognostic biomarker and correlated with immune infiltrates in lung cancer. PeerJ 2021; 9:e11145. [PMID: 33850663 PMCID: PMC8018245 DOI: 10.7717/peerj.11145] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/03/2021] [Indexed: 12/23/2022] Open
Abstract
Objective Lung cancer (LC) is one of the top ten malignant tumors and the first leading cause of cancer-related death among both men and women worldwide. It is imperative to identify immune-related biomarkers for early LC diagnosis and treatment. Methods Three Gene Expression Omnibus (GEO) datasets were selected to acquire the differentially expressed genes(DEGs) between LC and normal lung samples through GEO2R tools of NCBI. To identify hub genes, the DEGs were performed functional enrichment analysis, the protein-protein interaction (PPI) network construction, and Lasso regression. Then, a nomogram was constructed to predict the prognosis of patients with carcinoma based on hub genes. We further evaluated the influence of COL1A1 on clinical prognosis using GSE3141, GSE31210, and TCGA database. Also, the correlations between COL1A1 and cancer immune infiltrates and the B7-CD28 family was investigated via TIMER and GEPIA. Further analysis of immunohistochemistry shown that the COL1A1 expression level is positively correlated with CD276 expression level. Results By difference analysis, there were 340 DEGs between LC and normal lung samples. Then, we picked out seven hub genes, which were identified as components of the risk signature to divide LC into low and high-risk groups. Among them, the expression of COL1A1 is highly correlated with overall survival(OS) and progression-free survival (PFS) (p < 0.05). Importantly, there is a moderate to strong positive relationships between COL1A1 expression level and infiltration level of CD4+ T cells, Macrophage, Neutrophil, and Dendritic cell, as well as CD276 expression level. Conclusion These findings suggest that COL1A1 is correlated with prognosis and immune infiltrating levels, including CD4+ T cells, Macrophage, Neutrophil, and Dendritic cell, as well as CD276 expression level, indicating COL1A1 can be a potential immunity-related biomarker and therapeutic target in LC.
Collapse
Affiliation(s)
- Qishun Geng
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhibo Shen
- Engineering Laboratory for Digital Telemedicine Service, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lifeng Li
- Engineering Laboratory for Digital Telemedicine Service, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jie Zhao
- Engineering Laboratory for Digital Telemedicine Service, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
26
|
Gao Y, Chen S, Sun J, Su S, Yang D, Xiang L, Meng X. Traditional Chinese medicine may be further explored as candidate drugs for pancreatic cancer: A review. Phytother Res 2020; 35:603-628. [PMID: 32965773 DOI: 10.1002/ptr.6847] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/19/2020] [Accepted: 07/20/2020] [Indexed: 12/15/2022]
Abstract
Pancreatic cancer is a disease with a high mortality rate. Although survival rates for different types of cancers have improved in recent years, the five-year survival rate of pancreatic cancer stands at 8%. Moreover, the current first-line therapy, gemcitabine, results in low remission rates and is associated with drug resistance problems. Alternative treatments for pancreatic cancer such as surgery, chemotherapy and radiation therapy provide marginal remission and survival rates. This calls for the search of more effective drugs or treatments. Traditional Chinese medicine contains numerous bioactive ingredients some of which show activity against pancreatic cancer. In this review, we summarize the mechanisms of five types of traditional Chinese medicine monomers. In so-doing, we provide new potential drug candidates for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Yue Gao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shiyu Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiayi Sun
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Siyu Su
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dong Yang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Xiang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xianli Meng
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
27
|
An Q, Liu T, Wang MY, Yang YJ, Zhang ZD, Lin ZJ, Yang B. circKRT7-miR-29a-3p-COL1A1 Axis Promotes Ovarian Cancer Cell Progression. Onco Targets Ther 2020; 13:8963-8976. [PMID: 32982288 PMCID: PMC7490051 DOI: 10.2147/ott.s259033] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 07/28/2020] [Indexed: 12/12/2022] Open
Abstract
Background Circular RNA (circRNA) has emerged as an important regulator in the progression of human diseases. However, the role of circRNAs in ovarian cancer remains largely unknown. Materials and Methods DNA sequencing and PCR were used to identify the existence and expression of circKRT7. The targeting relationship between circKRT7/miR-29a-3p and miR-29a-3p/COL1A1 was verified by fluorescence reporter assay. In vitro, colony formation, transwell and wound healing assay were used to detect the effects of circKRT7 and miR-29a-3p on the proliferation, migration and invasion ability of ovarian cancer cells. In vivo, xenograft tumor model was performed to validate the role of circKRT7 and miR-29a-3p in tumor growth. Results We found that circKRT7 can promote the proliferation and metastasis of ovarian cancer cells by absorbing miR-29a-3p, which leads to the up-regulation of COL1A1. In vitro, knock-down of circKRT7 can inhibit the migration and invasion of ovarian cancer cells. This effect of circKRT7 is achieved by adsorbing miR-29a-3p and subsequently COL1A1 release. In vivo experiments, the reduction of circKRT7 expression can also slow tumor growth, and this inhibition was partly counteracted after miR-29a-3p repression. Conclusion Overall, circKRT7 promotes EMT-related cell progression by absorbing miR-29a-3p in ovarian cancer. This suggests the crucial role of circular RNA in the malignant evolution in cancer.
Collapse
Affiliation(s)
- Qiang An
- Department of Gynecology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China
| | - Ting Liu
- Department of Gynecology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China
| | - Ming-Yang Wang
- Department of Gynecology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China
| | - Yu-Jia Yang
- Department of Gynecology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China
| | - Zhen-Dong Zhang
- Department of Gynecology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China
| | - Zhen-Jiang Lin
- Department of Gynecology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China
| | - Bing Yang
- Department of Gynecology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563000, People's Republic of China
| |
Collapse
|
28
|
Wang L, Bi R, Li L, Zhou K, Liu H. Functional characteristics of autophagy in pancreatic cancer induced by glutamate metabolism in pancreatic stellate cells. J Int Med Res 2020; 48:300060519865368. [PMID: 31856624 PMCID: PMC7607760 DOI: 10.1177/0300060519865368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/02/2019] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVE To observe the effects of glutaminase (GLS) inhibitors on autophagy and proliferation of pancreatic stellate cells, and to explore their functions in pancreatic cancer. METHODS Pancreatic cancer cells were divided into two groups. Group A was the control untreated group, and group B cells were treated with GLS inhibitors. Western blotting was used to detect the expression of Atg5, Bcl-2, Bax, and Bid proteins. The bromodeoxyuridine assay and scratch test were employed to investigate cell proliferation and invasion, respectively. The expression of E-cadherin, vimentin, cell adhesion molecule 2 (CADM2), and Snail protein was investigated by immunofluorescence. RESULTS The expression of Atg5, Bax, and Bid was higher in group A than in group B, while Bcl-2 expression was lower in group A than in group B. Group A cells demonstrated greater proliferation and invasion than group B cells. The expression of E-cadherin was lower in group A cells than group B cells, while vimentin, CADM2, and Snail were expressed at higher levels in group A than group B cells. CONCLUSION The inhibition of glutamine isozymes reduces autophagy and apoptosis in astrocytes, and inhibits pancreatic cancer cell proliferation and metastasis, while reducing their invasiveness.
Collapse
Affiliation(s)
- Lei Wang
- Department of Gastroenterology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University, Shanghai, China
| | - RongRong Bi
- Department of Pulmonary, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lei Li
- Department of Gastroenterology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Kun Zhou
- Department of Gastroenterology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University, Shanghai, China
| | - HaiLin Liu
- Department of Gastroenterology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
29
|
Shinji S, Nakamura S, Nihashi Y, Umezawa K, Takaya T. Berberine and palmatine inhibit the growth of human rhabdomyosarcoma cells. Biosci Biotechnol Biochem 2020; 84:63-75. [DOI: 10.1080/09168451.2019.1659714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
ABSTRACT
A natural isoquinoline alkaloid, berberine, has been known to exhibit anti-tumor activity in various cancer cells via inducing cell cycle arrest. However, it has not been investigated whether berberine and its analogs inhibit the growth of rhabdomyosarcoma (RMS), which is the most frequent soft tissue tumor in children. The present study examined the anti-tumor effects of berberine and palmatine on expansions of three human embryonal RMS cell lines; ERMS1, KYM1, and RD. Intracellular incorporation of berberine was relatively higher than that of palmatine in every RMS cell line. Berberine significantly inhibited the cell cycle of all RMS cells at G1 phase. On the other hand, palmatine only suppressed the growth of RD cells. Both of berberine and palmatine strongly inhibited the growth of tumorsphere of RD cells in three-dimensional culture. These results indicate that berberine derivatives have the potential of anti-tumor drugs for RMS therapy.
Abbreviations: ARMS: alveolar rhabdomyosarcoma; ERMS: embryonal rhabdomyosarcoma; RMS: rhabdomyosarcoma
Collapse
Affiliation(s)
- Sayaka Shinji
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Shunichi Nakamura
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
| | - Yuma Nihashi
- Department of Science and Technology, Graduate School of Medicine, Science and Technology, Shinshu University, Nagano, Japan
| | - Koji Umezawa
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| | - Tomohide Takaya
- Department of Agriculture, Graduate School of Science and Technology, Shinshu University, Nagano, Japan
- Department of Science and Technology, Graduate School of Medicine, Science and Technology, Shinshu University, Nagano, Japan
- Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, Nagano, Japan
| |
Collapse
|
30
|
Wu W, Yang Z, Long F, Luo L, Deng Q, Wu J, Ouyang S, Tang D. COL1A1 and MZB1 as the hub genes influenced the proliferation, invasion, migration and apoptosis of rectum adenocarcinoma cells by weighted correlation network analysis. Bioorg Chem 2019; 95:103457. [PMID: 31901757 DOI: 10.1016/j.bioorg.2019.103457] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 10/15/2019] [Accepted: 11/19/2019] [Indexed: 02/07/2023]
Abstract
OBJECTIVES The influences of COL1A1 and MZB1 on the proliferation, migration, invasion and apoptosis abilities of rectum adenocarcinoma was aimed to explore in this study. METHODS Gene expression levels in rectum adenocarcinoma and adjacent tissues were analyzed by differential analysis. Weighted gene correlation network analysis (WGCNA) was employed to investigate rectal adenocarcinoma (READ) hub genes. MCODE was performed to screen the modules of protein-protein interaction network in Cytoscape software. Database for Annotation, Visualization and Integrated Discovery (DAVID) online tool is considered to be the most effective tool in gene ontology (GO) enrichment and Kyoto Gene and Genomics Encyclopedia (KEGG) pathway analysis. Survival analysis was performed using READ patient information from TCGA-READ project database. Quantitative Real-time Polymerase Chain Reaction (qRT-PCR) and western blot were employedto examinemRNA and protein expressions of COL1A1 and MZB1 in tumor tissues and cell lines. After transfecting various interference sequences by liposome-mediated transfection, the influence of COL1A1 and MZB1 on the proliferation, apoptosis, migration and invasion of rectal cancer cells were observed by plate clone formation assay, flow cytometry, wound healing assay and transwell assay respectively. Moreover, xenograft tumor growth assay in vivo validated the results. RESULTS Higher expression levels of COL1A1 and lower expression levels of MZB1 were discovered in tumor tissues of patients with colorectal adenocarcinoma. Overexpression of MZB1 and silencing COL1A1 significantly inhibited proliferation, migration and invasion, while cell apoptosis was promoted. Overexpression of MZB1 and silencing COL1A1 inhibited the orthotopic growth of tumor in vivo. CONCLUSION COL1A1 promoted proliferation, migration and invasion but inhibited apoptosis of rectal adenocarcinoma cells while MZB1 was totally on the contrary.
Collapse
Affiliation(s)
- Wenyu Wu
- Department of Oncology, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, Guizhou, China
| | - Zhu Yang
- Party Committee Office, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, Guizhou, China
| | - Fengxi Long
- Graduate School, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, Guizhou, China
| | - Li Luo
- Department of Oncology, Guihang Guiyang Hospital, Guiyang 550009, Guizhou, China
| | - Qian Deng
- Graduate School, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, Guizhou, China
| | - Jinlin Wu
- Graduate School, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, Guizhou, China
| | - Silu Ouyang
- Graduate School, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, Guizhou, China
| | - Dongxin Tang
- Department of Science and Education, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang 550001, Guizhou, China.
| |
Collapse
|
31
|
Wang J, Wang L, Lou GH, Zeng HR, Hu J, Huang QW, Peng W, Yang XB. Coptidis Rhizoma: a comprehensive review of its traditional uses, botany, phytochemistry, pharmacology and toxicology. PHARMACEUTICAL BIOLOGY 2019; 57:193-225. [PMID: 30963783 PMCID: PMC6461078 DOI: 10.1080/13880209.2019.1577466] [Citation(s) in RCA: 242] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 01/04/2019] [Accepted: 01/20/2019] [Indexed: 05/09/2023]
Abstract
CONTEXT Coptidis rhizome (CR), also known as Huanglian in Chinese, is the rhizome of Coptis chinensis Franch., C. deltoidea C.Y. Cheng et Hsiao, or C. teeta Wall (Ranunculaceae). It has been widely used to treat bacillary dysentery, diabetes, pertussis, sore throat, aphtha, and eczema in China. OBJECTIVES The present paper reviews the latest advances of CR, focusing on the botany, phytochemistry, traditional usages, pharmacokinetics, pharmacology and toxicology of CR and its future perspectives. METHODS Studies from 1985 to 2018 were reviewed from books; PhD. and MSc. dissertations; the state and local drug standards; PubMed; CNKI; Scopus; the Web of Science; and Google Scholar using the keywords Coptis, Coptidis Rhizoma, Huanglian, and goldthread. RESULTS Currently, 128 chemical constituents have been isolated and identified from CR. Alkaloids are the characteristic components, together with organic acids, coumarins, phenylpropanoids and quinones. The extracts/compounds isolated from CR cover a wide pharmacological spectrum, including antibacterial, antivirus, antifungal, antidiabetic, anticancer and cardioprotective effects. Berberine is the most important active constituent and the primary toxic component of CR. CONCLUSIONS As an important herbal medicine in Chinese medicine, CR has the potential to treat various diseases. However, further research should be undertaken to investigate the clinical effects, toxic constituents, target organs and pharmacokinetics, and to establish criteria for quality control, for CR and its related medications. In addition, the active constituents, other than alkaloids, in both raw and processed products of CR should be investigated.
Collapse
Affiliation(s)
- Jin Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lin Wang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Guan-Hua Lou
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hai-Rong Zeng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ju Hu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qin-Wan Huang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Peng
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiang-Bo Yang
- Ya'an Xun Kang Pharmaceutical Co., Ltd, Ya'an, China
| |
Collapse
|
32
|
Xu Q, Wu N, Li X, Guo C, Li C, Jiang B, Wang H, Shi D. Inhibition of PTP1B blocks pancreatic cancer progression by targeting the PKM2/AMPK/mTOC1 pathway. Cell Death Dis 2019; 10:874. [PMID: 31745071 PMCID: PMC6864061 DOI: 10.1038/s41419-019-2073-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 08/27/2019] [Accepted: 09/03/2019] [Indexed: 02/08/2023]
Abstract
Pancreatic cancer is a highly malignant cancer and lacks effective therapeutic targets. Protein-tyrosine phosphatase 1B (PTP1B), a validated therapeutic target for diabetes and obesity, also plays a critical positive or negative role in tumorigenesis. However, the role of PTP1B in pancreatic cancer remains elusive. Here, we initially demonstrated that PTP1B was highly expressed in pancreatic tumors, and was positively correlated with distant metastasis and tumor staging, and indicated poor survival. Then, inhibition of PTP1B either by shRNA or by a specific small-molecule inhibitor significantly suppressed pancreatic cancer cell growth, migration and colony formation with cell cycle arrest in vitro and inhibited pancreatic cancer progression in vivo. Mechanism studies revealed that PTP1B targeted the PKM2/AMPK/mTOC1 signaling pathway to regulate cell growth. PTP1B inhibition directly increased PKM2 Tyr-105 phosphorylation to further result in significant activation of AMPK, which decreased mTOC1 activity and led to inhibition of p70S6K. Meanwhile, the decreased phosphorylation of PRAS40 caused by decreased PKM2 activity also helped to inhibit mTOC1. Collectively, these findings support the notion of PTP1B as an oncogene and a promising therapeutic target for PDAC.
Collapse
MESH Headings
- AMP-Activated Protein Kinases/antagonists & inhibitors
- AMP-Activated Protein Kinases/metabolism
- Animals
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Carcinoma, Pancreatic Ductal/therapy
- Carrier Proteins/antagonists & inhibitors
- Carrier Proteins/metabolism
- Cell Line, Tumor
- Disease Progression
- Female
- Humans
- Male
- Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors
- Mechanistic Target of Rapamycin Complex 1/metabolism
- Membrane Proteins/antagonists & inhibitors
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/therapy
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/biosynthesis
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism
- RNA, Small Interfering/administration & dosage
- RNA, Small Interfering/genetics
- Random Allocation
- Signal Transduction/drug effects
- Small Molecule Libraries/pharmacology
- Thyroid Hormones/metabolism
- Xenograft Model Antitumor Assays
- Thyroid Hormone-Binding Proteins
Collapse
Affiliation(s)
- Qi Xu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
- The University of Chinese Academy of Sciences, Beijing, China
| | - Ning Wu
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Xiangqian Li
- State Key Laboratory of Microbial Technology, Shandong University, Jinan, 250100, Shandong, China
- The University of Chinese Academy of Sciences, Beijing, China
| | - Chuanlong Guo
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
- The University of Chinese Academy of Sciences, Beijing, China
| | - Chao Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
- The University of Chinese Academy of Sciences, Beijing, China
| | - Bo Jiang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Huaizhi Wang
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China.
| | - Dayong Shi
- State Key Laboratory of Microbial Technology, Shandong University, Jinan, 250100, Shandong, China.
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
- The University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
33
|
Li C, Shao T, Bao G, Gao Z, Zhang Y, Ding H, Zhang W, Liu F, Guo C. Identification of potential core genes in metastatic renal cell carcinoma using bioinformatics analysis. Am J Transl Res 2019; 11:6812-6825. [PMID: 31814889 PMCID: PMC6895538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 10/25/2019] [Indexed: 06/10/2023]
Abstract
Most cases of mRCC without an early finding are not candidates for curative therapies, which may be one of the reasons for the poor patient prognosis. Therefore, candidate markers to diagnose the disease and treatment with high efficiency are urgently demanded. Three datasets of mRNA microarray have been assessed to discover DEGs between tissues from metastatic RCC and RCC. 111 DEGs in total were identified according to the expression profile result of genes in the database of GEO. Enrichment analyses for GO and KEGG have been conducted to reveal the interacting activities in the DEGs. A network of PPI has been established to reveal the interconnection among the DEGs, and we selected 10 hub genes. Subsequently, the disease-free survival rate and total survival rate analysis for the hub genes have been carried out with the method of Kaplan-Meier curve. RCC patients with CDH11, COL3A1, COL5A1, COL5A2, COL6A3 and COL11A1 alteration showed worse overall survival. Nonetheless, RCC patients with CDH11, COL3A1, COL5A1, COL5A2 and COL11A1 alteration showed worse disease-free survival. In the Jones Renal dataset, mRNA levels of 10 hub genes were associated with metastasis, and the gene expression level in patients with mRCC was higher than that in patients without metastasis. COL5A1, COL6A3 and COL11A1 expression levels were remarkably related to RCC patient survival rate using UALCAN. COL5A1, COL6A3 and COL11A1 were positively correlated with each other in RCC. These genes have been recognized as genes with clinical relevance, revealing that they might have important roles in carcinogenesis or development of mRCC.
Collapse
Affiliation(s)
- Chunsheng Li
- Department of Urology, Affiliated Hospital of Chifeng UniversityChifeng 024000, China
- Urology Research Center, Chifeng UniversityChifeng 024000, China
| | - Ting Shao
- Department of Gynecology, Affiliated Hospital of Chifeng UniversityChifeng 024000, China
| | - Guochang Bao
- Department of Urology, Affiliated Hospital of Chifeng UniversityChifeng 024000, China
- Urology Research Center, Chifeng UniversityChifeng 024000, China
| | - Zhiming Gao
- Department of Urology, Affiliated Hospital of Chifeng UniversityChifeng 024000, China
- Urology Research Center, Chifeng UniversityChifeng 024000, China
| | - Yinglang Zhang
- Department of Urology, Affiliated Hospital of Chifeng UniversityChifeng 024000, China
| | - Honglin Ding
- Department of Urology, Affiliated Hospital of Chifeng UniversityChifeng 024000, China
| | - Wei Zhang
- Department of Urology, Affiliated Hospital of Chifeng UniversityChifeng 024000, China
| | - Fengjun Liu
- Department of Urology, Affiliated Hospital of Chifeng UniversityChifeng 024000, China
| | - Changgang Guo
- Department of Urology, Affiliated Hospital of Chifeng UniversityChifeng 024000, China
- Urology Research Center, Chifeng UniversityChifeng 024000, China
| |
Collapse
|
34
|
Tarabasz D, Kukula-Koch W. Palmatine: A review of pharmacological properties and pharmacokinetics. Phytother Res 2019; 34:33-50. [PMID: 31496018 DOI: 10.1002/ptr.6504] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/18/2019] [Accepted: 08/19/2019] [Indexed: 12/11/2022]
Abstract
The aim of this review is to collect together the results of the numerous studies over the last two decades on the pharmacological properties of palmatine published in scientific databases like Scopus and PubMed, which are scattered across different publications. Palmatine, an isoquinoline alkaloid from the class of protoberberines, is a yellow compound present in the extracts from different representatives of Berberidaceae, Papaveraceae, Ranunculaceae, and Menispermaceae. It has been extensively used in traditional medicine of Asia in the treatment of jaundice, liver-related diseases, hypertension, inflammation, and dysentery. New findings describe its possible applications in the treatment of civilization diseases like central nervous system-related problems. This review intends to let this alkaloid come out from the shade of a more frequently described alkaloid: berberine. The toxicity, pharmacokinetics, and biological activities of this protoberberine alkaloid will be developed in this work.
Collapse
Affiliation(s)
| | - Wirginia Kukula-Koch
- Chair and Department of Pharmacognosy with Medicinal Plants Unit, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
35
|
Wang Y, Xu Q, Shen Z, Wang Y, Dong L, Fu S, Yang L, Qin M, Zhang Y, Guo S. Simultaneous Determination of Eight Active Compounds in Baitouweng Decotion and its Single Herbs. J Chromatogr Sci 2019; 57:502-510. [PMID: 30929002 DOI: 10.1093/chromsci/bmz022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 01/22/2019] [Accepted: 03/05/2019] [Indexed: 11/12/2022]
Abstract
Baitouweng Decotion (BD) is a famous traditional Chinese medicinal prescription, which is composed of Pulsatilla chinensis (bunge) regel, Coptis chinensis franch., Phellodendron chinense and Cortex Fraxini. In this study, a simple and sensitive high performance liquid chromatography coupled with ultraviolet detection method was established for the simultaneous determination of eight marker compounds including Esculin, Fraxin, Esculetin, Fraxetin, Columbamine, Coptisine, Palmatine Chloride and Berberine hydrochloride in BD, the single herbs and their negative controls. The chromatographic separation was performed using an Agilent Eclipse XDB-C18 column with a gradient elution system of acetonitrile and 0.1% phosphoric acid (contained 0.2% triethylamine) solution at a flow rate of 0.8 mL/min. The results demonstrated that the validated method was simple, reliable and successfully applied to evaluate the selected compounds in water extraction (BDW) and ethanol extraction (BDE) of BD, the single herbs and their negative control for quality control. Moreover, the experimental data showed that the contents of the major active components detected in BDE were significantly higher than those in the BDW, while the BDW had several peaks BDE without. The paper also suggested a method to extract Fraxin, Esculin, Fraxetin, Esculetin and Berberine from Baitouweng Decotion more effectively.
Collapse
Affiliation(s)
- Yubo Wang
- Department of Thoracic Surgery, Binzhou Medical College Hospital, No. 661 Huanghe 2 Road, Bincheng Area, Binzhou, China
| | - Qianqian Xu
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, No. 169 Huanghe 2 Road, Economic-Technological Development Area, Binzhou, China.,Shandong Lvdu Ante Veterinary Drug Industry Co., Ltd, No. 169 Huanghe 2 Road, Economic-Technological Development Area, Binzhou, China
| | - Zhiqiang Shen
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, No. 169 Huanghe 2 Road, Economic-Technological Development Area, Binzhou, China.,Shandong Lvdu Ante Veterinary Drug Industry Co., Ltd, No. 169 Huanghe 2 Road, Economic-Technological Development Area, Binzhou, China
| | - Yanping Wang
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, No. 169 Huanghe 2 Road, Economic-Technological Development Area, Binzhou, China.,Shandong Lvdu Ante Veterinary Drug Industry Co., Ltd, No. 169 Huanghe 2 Road, Economic-Technological Development Area, Binzhou, China
| | - Lin Dong
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, No. 169 Huanghe 2 Road, Economic-Technological Development Area, Binzhou, China
| | - Shijun Fu
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, No. 169 Huanghe 2 Road, Economic-Technological Development Area, Binzhou, China
| | - Limei Yang
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, No. 169 Huanghe 2 Road, Economic-Technological Development Area, Binzhou, China.,Shandong Lvdu Ante Veterinary Drug Industry Co., Ltd, No. 169 Huanghe 2 Road, Economic-Technological Development Area, Binzhou, China
| | - Mimi Qin
- Department of Pathology, Binzhou Medical College Hospital, No. 661 Huanghe 2 Road, Bincheng Area, Binzhou, China
| | - Ying Zhang
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, No. 169 Huanghe 2 Road, Economic-Technological Development Area, Binzhou, China.,Shandong Lvdu Ante Veterinary Drug Industry Co., Ltd, No. 169 Huanghe 2 Road, Economic-Technological Development Area, Binzhou, China
| | - Shijin Guo
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, No. 169 Huanghe 2 Road, Economic-Technological Development Area, Binzhou, China.,Shandong Lvdu Ante Veterinary Drug Industry Co., Ltd, No. 169 Huanghe 2 Road, Economic-Technological Development Area, Binzhou, China
| |
Collapse
|
36
|
Chen X, Tang WJ, Shi JB, Liu MM, Liu XH. Therapeutic strategies for targeting telomerase in cancer. Med Res Rev 2019; 40:532-585. [PMID: 31361345 DOI: 10.1002/med.21626] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/12/2019] [Accepted: 07/16/2019] [Indexed: 12/13/2022]
Abstract
Telomere and telomerase play important roles in abnormal cell proliferation, metastasis, stem cell maintenance, and immortalization in various cancers. Therefore, designing of drugs targeting telomerase and telomere is of great significance. Over the past two decades, considerable knowledge regarding telomere and telomerase has been accumulated, which provides theoretical support for the design of therapeutic strategies such as telomere elongation. Therefore, the development of telomere-based therapies such as nucleoside analogs, non-nucleoside small molecules, antisense technology, ribozymes, and dominant negative human telomerase reverse transcriptase are being prioritized for eradicating a majority of tumors. While the benefits of telomere-based therapies are obvious, there is a need to address the limitations of various therapeutic strategies to improve the possibility of clinical applications. In this study, current knowledge of telomere and telomerase is discussed, and therapeutic strategies based on recent research are reviewed.
Collapse
Affiliation(s)
- Xing Chen
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, People's Republic of China
| | - Wen-Jian Tang
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, People's Republic of China
| | - Jing Bo Shi
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, People's Republic of China
| | - Ming Ming Liu
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, People's Republic of China
| | - Xin-Hua Liu
- School of Pharmacy, Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei, People's Republic of China
| |
Collapse
|
37
|
Palmatine: A review of its pharmacology, toxicity and pharmacokinetics. Biochimie 2019; 162:176-184. [DOI: 10.1016/j.biochi.2019.04.008] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 04/07/2019] [Indexed: 12/22/2022]
|
38
|
Mock A, Rapp C, Warta R, Abdollahi A, Jäger D, Sakowitz O, Brors B, von Deimling A, Jungk C, Unterberg A, Herold-Mende C. Impact of post-surgical freezing delay on brain tumor metabolomics. Metabolomics 2019; 15:78. [PMID: 31087206 DOI: 10.1007/s11306-019-1541-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 05/04/2019] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Translational cancer research has seen an increasing interest in metabolomic profiling to decipher tumor phenotypes. However, the impact of post-surgical freezing delays on mass spectrometric metabolomic measurements of the cancer tissue remains elusive. OBJECTIVES To evaluate the impact of post-surgical freezing delays on cancer tissue metabolomics and to investigate changes per metabolite and per metabolic pathway. METHODS We performed untargeted metabolomics on three cortically located and bulk-resected glioblastoma tissues that were sequentially frozen as duplicates at up to six different time delays (0-180 min, 34 samples). RESULTS Statistical modelling revealed that 10% of the metabolome (59 of 597 metabolites) changed significantly after a 3 h delay. While carbohydrates and energy metabolites decreased, peptides and lipids increased. After a 2 h delay, these metabolites had changed by as much as 50-100%. We present the first list of metabolites in glioblastoma tissues that are sensitive to post-surgical freezing delays and offer the opportunity to define individualized fold change thresholds for future comparative metabolomic studies. CONCLUSION More researchers should take these pre-analytical factors into consideration when analyzing metabolomic data. We present a strategy for how to work with metabolites that are sensitive to freezing delays.
Collapse
Affiliation(s)
- Andreas Mock
- Division of Experimental Neurosurgery, Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Carmen Rapp
- Division of Experimental Neurosurgery, Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Rolf Warta
- Division of Experimental Neurosurgery, Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Amir Abdollahi
- Molecular and Translational Radiation Oncology, National Center for Tumor Diseases (NCT), Heidelberg University Hospital and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk Jäger
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg University Hospital, Heidelberg, Germany
| | - Oliver Sakowitz
- Division of Experimental Neurosurgery, Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Benedikt Brors
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andreas von Deimling
- Department of Neuropathology & Clinical Cooperation Unit Neuropathology, University Hospital Heidelberg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christine Jungk
- Division of Experimental Neurosurgery, Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas Unterberg
- Division of Experimental Neurosurgery, Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Christel Herold-Mende
- Division of Experimental Neurosurgery, Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
39
|
Johnson-Ajinwo OR, Richardson A, Li WW. Palmatine from Unexplored Rutidea parviflora Showed Cytotoxicity and Induction of Apoptosis in Human Ovarian Cancer Cells. Toxins (Basel) 2019; 11:toxins11040237. [PMID: 31027283 PMCID: PMC6521182 DOI: 10.3390/toxins11040237] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/17/2019] [Accepted: 04/18/2019] [Indexed: 12/25/2022] Open
Abstract
Ovarian cancer ranks amongst the deadliest cancers in the gynaecological category of cancers. This research work aims to evaluate in vitro anti-ovarian cancer activities and identify phytochemical constituents of a rarely explored plant species—Rutidea parviflora DC. The aqueous and organic extracts of the plant were evaluated for cytotoxicity using sulforhodamine B assay in four ovarian cancer cell lines and an immortalized human ovarian epithelial (HOE) cell line. The bioactive compounds were isolated and characterized by gas/liquid chromatography mass spectrometry and nuclear magnetic resonance spectroscopy. Caspase 3/7 activity assay, western blotting and flow cytometry were carried out to assess apoptotic effects of active compounds. The extracts/fractions of R. parviflora showed promising anti-ovarian cancer activities in ovarian cancer cell lines. A principal cytotoxic alkaloid was identified as palmatine whose IC50 was determined as 5.5–7.9 µM. Palmatine was relatively selective towards cancer cells as it was less cytotoxic toward HOE cells, also demonstrating interestingly absence of cross-resistance in cisplatin-resistant A2780 cells. Palmatine further induced apoptosis by increasing caspase 3/7 activity, poly-ADP-ribose polymerase cleavage, and annexin V and propidium iodide staining in OVCAR-4 cancer cells. Our studies warranted further investigation of palmatine and R. parviflora extracts in preclinical models of ovarian cancer.
Collapse
Affiliation(s)
- Okiemute Rosa Johnson-Ajinwo
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent ST4 7QB, UK.
- Faculty of Pharmaceutical Sciences, University of Port Harcourt, Port Harcourt, PMB 5323, Nigeria.
| | - Alan Richardson
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent ST4 7QB, UK.
| | - Wen-Wu Li
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent ST4 7QB, UK.
| |
Collapse
|
40
|
Martínez-García D, Manero-Rupérez N, Quesada R, Korrodi-Gregório L, Soto-Cerrato V. Therapeutic strategies involving survivin inhibition in cancer. Med Res Rev 2018; 39:887-909. [PMID: 30421440 DOI: 10.1002/med.21547] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 09/13/2018] [Accepted: 10/09/2018] [Indexed: 02/06/2023]
Abstract
Survivin is a small protein that belongs to the inhibitor of apoptosis protein family. It is abundantly expressed in tumors compared with adult differentiated tissues, being associated with poor prognosis in many human neoplasms. This apoptotic inhibitor has a relevant role in both the promotion of cancer cell survival and in the inhibition of cell death. Consequently, aberrant survivin expression stimulates tumor progression and confers resistance to several therapeutic strategies in a variety of tumors. In fact, efficient survivin downregulation or inhibition results in spontaneous apoptosis or sensitization to chemotherapy and radiotherapy. Therefore, all these features make survivin an attractive therapeutic target to treat cancer. Currently, there are several survivin inhibitors under clinical evaluation, although more specific and efficient survivin inhibitors are being developed. Moreover, novel combination regimens targeting survivin together with other therapeutic approaches are currently being designed and assessed. In this review, recent progress in the therapeutic options targeting survivin for cancer treatment is analyzed. Direct survivin inhibitors and their current development status are explored. Besides, the major signaling pathways implicated in survivin regulation are described and different therapeutic approaches involving survivin indirect inhibition are evaluated. Finally, promising novel inhibitors under preclinical or clinical evaluation as well as challenges of developing survivin inhibitors as a new therapy for cancer treatment are discussed.
Collapse
Affiliation(s)
- David Martínez-García
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine, University of Barcelona, Barcelona, Spain.,Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Noemí Manero-Rupérez
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine, University of Barcelona, Barcelona, Spain.,Hospital del Mar Medical Research Institute, Barcelona Biomedical Research Park, Barcelona, Spain
| | - Roberto Quesada
- Department of Chemistry, Universidad de Burgos, Burgos, Spain
| | - Luís Korrodi-Gregório
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Vanessa Soto-Cerrato
- Department of Pathology and Experimental Therapeutics, Faculty of Medicine, University of Barcelona, Barcelona, Spain.,Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| |
Collapse
|