1
|
Qin L, Zeng X, Qiu X, Chen X, Liu S. The role of N6-methyladenosine modification in tumor angiogenesis. Front Oncol 2024; 14:1467850. [PMID: 39691597 PMCID: PMC11649548 DOI: 10.3389/fonc.2024.1467850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 11/11/2024] [Indexed: 12/19/2024] Open
Abstract
Tumor angiogenesis is a characteristics of malignant cancer progression that facilitates cancer cell growth, diffusion and metastasis, and has an indispensable role in cancer development. N6-methyladenosine (m6A) is among the most prevalent internal modifications in eukaryotic RNAs, and has considerable influence on RNA metabolism, including its transcription, splicing, localization, translation, recognition, and degradation. The m6A modification is generated by m6A methyltransferases ("writers"), removed by m6A demethylases ("erasers"), and recognized by m6A-binding proteins ("readers"). There is accumulating evidence that abnormal m6A modification is involved in the pathogenesis of multiple diseases, including cancers, and promotes cancer occurrence, development, and progression through its considerable impact on oncoprotein expression. Furthermore, increasing studies have demonstrated that m6A modification can influence angiogenesis in cancers through multiple pathways to regulate malignant processes. In this review, we elaborate the role of m6A modification in tumor angiogenesis-related molecules and pathways in detail, providing insights into the interactions between m6A and tumor angiogenesis. Moreover, we describe how targeting m6A modification in combination with anti-angiogenesis drugs is expected to be a promising anti-tumor treatment strategy, with potential value for addressing the challenge of drug resistance.
Collapse
Affiliation(s)
| | | | | | | | - Shiquan Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Guangxi Medical
University, Nanning, Guangxi, China
| |
Collapse
|
2
|
Hu B, Pei J, Wan C, Liu S, Xu Z, Zou Y, Li Z, Tang Z. Mechanisms of Postischemic Stroke Angiogenesis: A Multifaceted Approach. J Inflamm Res 2024; 17:4625-4646. [PMID: 39045531 PMCID: PMC11264385 DOI: 10.2147/jir.s461427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024] Open
Abstract
Ischemic stroke constitutes a significant global health care challenge, and a comprehensive understanding of its recovery mechanisms is imperative for the development of innovative therapeutic strategies. Angiogenesis, a pivotal element of ischemic tissue repair, facilitates the restoration of blood flow to damaged regions, thereby promoting neuronal regeneration and functional recovery. Nevertheless, the mechanisms underlying postischemic stroke angiogenesis remain incompletely elucidated. This review meticulously examines the constituents of the neurovascular unit, ion channels, molecular mediators, and signaling pathways implicated in angiogenesis following stroke. Furthermore, it delves into prospective therapeutic strategies informed by these factors. Our objective is to provide detailed and exhaustive information on the intricate mechanisms governing postischemic stroke angiogenesis, thus providing a robust scientific foundation for the advancement of novel neurorepair therapies.
Collapse
Affiliation(s)
- Bin Hu
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Jingchun Pei
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Cheng Wan
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
- Department of Medical Imaging, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Shuangshuang Liu
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Zhe Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, People’s Republic of China
- School of Basic Medical Sciences, Qujing Medical College, Qujing, People’s Republic of China
| | - Yongwei Zou
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Zhigao Li
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Zhiwei Tang
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| |
Collapse
|
3
|
Wang L, Zhang L, Dunmall LC, Wang YY, Fan Z, Cheng Z, Wang Y. The dilemmas and possible solutions for CAR-T cell therapy application in solid tumors. Cancer Lett 2024; 591:216871. [PMID: 38604310 DOI: 10.1016/j.canlet.2024.216871] [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: 10/27/2023] [Revised: 03/26/2024] [Accepted: 04/06/2024] [Indexed: 04/13/2024]
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy, as an adoptive immunotherapy, is playing an increasingly important role in the treatment of malignant tumors. CAR-T cells are referred to as "living drugs" as they not only target tumor cells directly, but also induce long-term immune memory that has the potential to provide long-lasting protection. CD19.CAR-T cells have achieved complete response rates of over 90 % for acute lymphoblastic leukemia and over 60 % for non-Hodgkin's lymphoma. However, the response rate of CAR-T cells in the treatment of solid tumors remains extremely low and the side effects potentially severe. In this review, we discuss the limitations that the solid tumor microenvironment poses for CAR-T application and the solutions that are being developed to address these limitations, in the hope that in the near future, CAR-T cell therapy for solid tumors can attain the same success rates as are now being seen clinically for hematological malignancies.
Collapse
Affiliation(s)
- Lihong Wang
- Department of Oncology, Air Force Medical Center, PLA, Beijing, China; National Centre for International Research in Cell and Gene Therapy, Sino British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Lufang Zhang
- National Centre for International Research in Cell and Gene Therapy, Sino British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Louisa Chard Dunmall
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Yang Yang Wang
- Department of General Pediatrics, Newham General Hospital, E13 8SL, London, United Kingdom
| | - Zaiwen Fan
- Department of Oncology, Air Force Medical Center, PLA, Beijing, China
| | - Zhenguo Cheng
- National Centre for International Research in Cell and Gene Therapy, Sino British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yaohe Wang
- National Centre for International Research in Cell and Gene Therapy, Sino British Research Centre for Molecular Oncology, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China; Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.
| |
Collapse
|
4
|
Mao C, Wang Y, Xu Z, Wang X, Fang B, Chen H. Luteolin-Zn Complex Inhibits Invasion and Migration of M2-Like TAMs via the Downregulation of AMPK/mTOR and PI3K/Akt/mTOR Signaling Pathway Under Hypoxia. Nat Prod Commun 2023. [DOI: 10.1177/1934578x231167996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2023] Open
Abstract
Purpose: The high mortality rate of malignant tumors is often attributable to the loss of surgical opportunities due to late diagnosis when invasion and metastasis have significantly affected the patient. A hypoxic microenvironment can promote the progression of malignant tumors. This study explored the invasion resistance and migration ability of luteolin-Zn complexes. Methods: We created a low-oxygen environment using a 3-atmosphere incubator. The appropriate drug concentration was determined using the CCK8 experiment. We determined its role in cell invasion and migration through scratch and transwell experiments. Western blotting, polymerase chain reaction, and cellular immunity experiments were used to study the mechanism and its impact on the secretion of invasion and migration factors. Results: Our results indicated that the luteolin-Zn complex significantly reduced MMP2, MMP9, N-Ca, and HIF-1ɑ expression. It also upregulated TIMP1 and E-Ca expression. Moreover, its capabilities may be achieved by regulating the AMPK/mTOR and PI3K/Akt/mTOR signaling pathways. Conclusions: The luteolin-Zn complex was highly resistant to the invasion and migration of M2-like tumor-related macrophages. This may exert a unique influence on mTOR by integrating various signals. This study suggests that the luteolin-Zn complex has a strong anticancer effect under hypoxic conditions.
Collapse
Affiliation(s)
- Chenyang Mao
- Department of Hepatobiliary Surgery, The First People's Hospital of Wenling, Wenling, China
| | - Yongling Wang
- Department of Ultrasonography, Taizhou Hospital of Zhejiang Province, Linhai, China
| | - Zhenhua Xu
- Sanya Rehabilitation Center of Joint Support Forces, Hainan, China
| | | | - Binbo Fang
- Department of Medicine, Taizhou University, Jiaojiang, China
| | - Haihua Chen
- Department of Hepatopancreatobiliary Surgery, Taizhou Hospital of Zhejiang Province, Linhai, China
| |
Collapse
|
5
|
Li YT, Yuan WZ, Jin WL. Vagus innervation in the gastrointestinal tumor: Current understanding and challenges. Biochim Biophys Acta Rev Cancer 2023; 1878:188884. [PMID: 36990250 DOI: 10.1016/j.bbcan.2023.188884] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 02/17/2023] [Accepted: 02/28/2023] [Indexed: 03/30/2023]
Abstract
The vagus nerve (VN) is the main parasympathetic nerve of the autonomic nervous system. It is widely distributed in the gastrointestinal tract and maintains gastrointestinal homeostasis with the sympathetic nerve under physiological conditions. The VN communicates with various components of the tumor microenvironment to positively and dynamically affect the progression of gastrointestinal tumors (GITs). The intervention in vagus innervation delays GIT progression. Developments in adeno-associated virus vectors, nanotechnology, and in vivo neurobiological techniques have enabled the creation of precisely regulated "tumor neurotherapies". Furthermore, the combination of neurobiological techniques and single cell sequencing may reveal more insights into VN and GIT. The present review aimed to summarize the mechanisms of communication between the VN and the gastrointestinal TME and to explore the potential and challenges of VN-based tumor neurotherapy in GITs.
Collapse
|
6
|
Wu Z, Xu B, He Q, Hu Z, Yu Z. Tetrastigma hemsleyanum Ethanolic Extract Inhibited the Growth of Nonsmall Cell Lung Cancer Cells by Suppressing Hypoxia-Inducible Factor-1α-Dependent Glycolysis and Angiogenesis. Nat Prod Commun 2023. [DOI: 10.1177/1934578x221142796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background:The ethanolic extract of Tetrastigma hemsleyanum Diels et Gilg ( T hemsleyanum ethanolic extract [Te-EtOH]) showed positive effects against various tumors. However, there are few studies on the effects of Te-EtOH on nonsmall cell lung cancer (NSCLC). We attempted to examine the inhibiting effect of Te-EtOH on NSCLC cells and to elucidate the relevant mechanisms. Methods: A549 and H1299 cells were pretreated with Te-EtOH at different concentrations. Cell viability was analyzed by Cell Counting Kit-8, flow cytometry, and the 3-dimensional spheroid model; RNA-sequencing was also performed. Moreover, enzyme-linked immunosorbent assay and Western blot tests were performed to determine the metabolic capability, the expressions of energy metabolism-related proteins, and the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/hypoxia-inducible factor-1α (HIF-1α) pathway. Additionally, under hypoxic conditions, the ability of Te-EtOH to inhibit HIF-1α expression and the metabolic capability of NSCLC cells was tested. Results: Te-EtOH considerably repressed cell viability in a dose-dependent manner. RNA-sequencing revealed that Te-EtOH's inhibition of NSCLC cells activity was related to metabolism. In addition, Te-EtOH significantly inhibited glycolysis, and adenosine triphosphate and lactate accumulation in NSCLC cells. Furthermore, we found that Te-EtOH could block PI3K/AKT/HIF-1α pathway activation. Moreover, Te-EtOH significantly inhibited hypoxia-induced expression of HIF-1α, vascular endothelial growth factor, and metabolic capability. Conclusions: Our results suggested that Te-EtOH inhibited the growth of NSCLC cells by suppressing HIF-1α-dependent glycolysis and angiogenesis.
Collapse
Affiliation(s)
- Zhiqiang Wu
- Department of Traditional Chinese Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Bin Xu
- Department of Traditional Chinese Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Qin He
- Department of Traditional Chinese Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Zhuyuan Hu
- Department of Traditional Chinese Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| | - Zhiyi Yu
- Department of Traditional Chinese Medicine, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, China
| |
Collapse
|
7
|
Lei ZN, Teng QX, Tian Q, Chen W, Xie Y, Wu K, Zeng Q, Zeng L, Pan Y, Chen ZS, He Y. Signaling pathways and therapeutic interventions in gastric cancer. Signal Transduct Target Ther 2022; 7:358. [PMID: 36209270 PMCID: PMC9547882 DOI: 10.1038/s41392-022-01190-w] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 08/14/2022] [Accepted: 09/07/2022] [Indexed: 11/23/2022] Open
Abstract
Gastric cancer (GC) ranks fifth in global cancer diagnosis and fourth in cancer-related death. Despite tremendous progress in diagnosis and therapeutic strategies and significant improvements in patient survival, the low malignancy stage is relatively asymptomatic and many GC cases are diagnosed at advanced stages, which leads to unsatisfactory prognosis and high recurrence rates. With the recent advances in genome analysis, biomarkers have been identified that have clinical importance for GC diagnosis, treatment, and prognosis. Modern molecular classifications have uncovered the vital roles that signaling pathways, including EGFR/HER2, p53, PI3K, immune checkpoint pathways, and cell adhesion signaling molecules, play in GC tumorigenesis, progression, metastasis, and therapeutic responsiveness. These biomarkers and molecular classifications open the way for more precise diagnoses and treatments for GC patients. Nevertheless, the relative significance, temporal activation, interaction with GC risk factors, and crosstalk between these signaling pathways in GC are not well understood. Here, we review the regulatory roles of signaling pathways in GC potential biomarkers, and therapeutic targets with an emphasis on recent discoveries. Current therapies, including signaling-based and immunotherapies exploited in the past decade, and the development of treatment for GC, particularly the challenges in developing precision medications, are discussed. These advances provide a direction for the integration of clinical, molecular, and genomic profiles to improve GC diagnosis and treatments.
Collapse
Affiliation(s)
- Zi-Ning Lei
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Qin Tian
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China
| | - Wei Chen
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China
| | - Yuhao Xie
- Institute for Biotechnology, St. John's University, Queens, NY, 11439, USA
| | - Kaiming Wu
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China
| | - Qianlin Zeng
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China
| | - Leli Zeng
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China.
| | - Yihang Pan
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China.
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
- Institute for Biotechnology, St. John's University, Queens, NY, 11439, USA.
| | - Yulong He
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, 518107, Shenzhen, Guangdong, China.
| |
Collapse
|
8
|
Wang Z, Liang X, Zhang H, Wang Z, Zhang X, Dai Z, Liu Z, Zhang J, Luo P, Li J, Cheng Q. Identification of a Hypoxia-Angiogenesis lncRNA Signature Participating in Immunosuppression in Gastric Cancer. J Immunol Res 2022; 2022:5209607. [PMID: 36052279 PMCID: PMC9427269 DOI: 10.1155/2022/5209607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 06/06/2022] [Indexed: 01/17/2023] Open
Abstract
Hypoxia and angiogenesis are the leading causes of tumor progression, and their strong correlation has been discovered in many cancers. However, their collective function's prognostic and biological roles were not reported in gastric cancer. Hence, we aimed to investigate the effects of hypoxia and angiogenesis on gastric cancer via sequencing data. This study used weighted gene coexpression network analysis and random forest regression to build a hypoxia-angiogenesis-related model (HARM) via the TCGA-STAD lncRNA data. It estimated the HARM's correlation with clinical features and its accuracy for survival prediction. Sequential functional analyses were conducted to investigate its biological role, and we next sought the immune landscape status and immunological function variation by ESTIMATE score calculation and GSVA, respectively. Seven different algorithms were conducted to assess the immunocyte infiltration, and TIDE score and immune checkpoint levels were compared between the high- and low-HARM groups. As a result, we found that HARM predicted patient survival with high accuracy and was correlated with higher stages of gastric cancer. Various cancer-associated pathways and macrophage-related regulations were upregulated in the high-HRAM group. The high-HARM group harbored higher immune levels, and M2 macrophages and cancer-associated fibroblasts were particularly highly unfiltered. Furthermore, globally upregulated immune checkpoints and higher TIDE scores were observed in the high-HARM group. Finally, we filtered eight drugs with lower IC50 in the high-HARM group as potential drugs for the HARM-targeted therapy. We believe this study opens up novel perspectives into the interaction between hypoxia-angiogenesis and immunosuppression and will provide novel insights for gastric cancer immunotherapy.
Collapse
Affiliation(s)
- Zicheng Wang
- Hepatobiliary Surgery, Hunan Provincial People's Hospital (The First Affiliate Hospital of Hunan Normal University), Changsha, China
| | - Xisong Liang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xun Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510000, China
| | - Jiarong Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
9
|
Zhang M, Jin M, Gao Z, Yu W, Zhang W. High COL10A1 expression potentially contributes to poor outcomes in gastric cancer with the help of LEF1 and Wnt2. J Clin Lab Anal 2022; 36:e24612. [PMID: 35929139 PMCID: PMC9459277 DOI: 10.1002/jcla.24612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/15/2022] [Accepted: 06/13/2022] [Indexed: 11/14/2022] Open
Abstract
Background COL10A1 is a secreted, short‐chain collagen found in several types of cancer. Studies have shown that COL10A1 aberrant expression is considered an oncogenic factor. However, its underlying mechanisms and regulation of gastric cancer remain undefined. Methods The data on the expression of COL10A1, clinicopathological characteristics, and outcome of patients with GC were obtained from The Cancer Genome Atlas. The ALGGEN‐PROMO database defined the related transcription factors. Quantitative real‐time reverse transcription‐polymerase chain reaction and western blotting analysis were used to identify the differential expression levels of COL10A1 and related transcription factors. Results We found that high COL10A1 expression is an independent risk factor for gastric cancer. Upregulation of LEF1 and Wnt2 was also observed in gastric cancer, suggesting a potential correlation between LEF1/COL10A1 regulation in the Wnt2 signaling pathway. Conclusion High COL10A1 expression may contribute to poor outcomes via upregulation of LEF1 and Wnt2 in gastric cancer.
Collapse
Affiliation(s)
- Miaozun Zhang
- Department of Gastrointestinal Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
| | - Ming Jin
- Department of Radiation Oncology, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
| | - Zhiqiang Gao
- Department of Gastroenterology, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
| | - Weiming Yu
- Department of Gastrointestinal Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
| | - Wei Zhang
- Department of Gastroenterology, The HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| |
Collapse
|
10
|
HIF in Gastric Cancer: Regulation and Therapeutic Target. Molecules 2022; 27:molecules27154893. [PMID: 35956843 PMCID: PMC9370240 DOI: 10.3390/molecules27154893] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/25/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
HIF means hypoxia-inducible factor gene family, and it could regulate various biological processes, including tumor development. In 2021, the FDA approved the new drug Welireg for targeting HIF-2a, and it is mainly used to treat von Hippel-Lindau syndrome, which demonstrated its good prospects in tumor therapy. As the fourth deadliest cancer worldwide, gastric cancer endangers the health of people all across the world. Currently, there are various treatment methods for patients with gastric cancer, but the five-year survival rate of patients with advanced gastric cancer is still not high. Therefore, here we reviewed the regulatory role and target role of HIF in gastric cancer, and provided some references for the treatment of gastric cancer.
Collapse
|
11
|
Zhu Y, Zhao Y, Cao Z, Chen Z, Pan W. Identification of three immune subtypes characterized by distinct tumor immune microenvironment and therapeutic response in stomach adenocarcinoma. Gene X 2022; 818:146177. [PMID: 35065254 DOI: 10.1016/j.gene.2021.146177] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 11/05/2021] [Accepted: 12/06/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND In primary stomach adenocarcinoma (STAD), the tumor immune microenvironment (TIME) is important for cancer occurrence and progression; however, its clinical significance remains unclear. This study investigated the association between patient survival, TIME, and therapeutic response to STAD. METHODS Gene expression profiles of STAD cases were collected from the Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus. Molecular subtypes were explored with consistent clustering methods according to 119 immune signatures and the infiltrating scores of 22 immune cells using the Multi-Omics Immuno-Oncology Biological Research algorithm. We determined IFNγ scores and immune cytolytic activity (CYT) scores on the basis of corresponding gene signatures via single-sample Gene Set Enrichment Analysis. Comparisons of survival, TIME, 10 immunity-related oncogenic pathways, immune checkpoint expression, and therapeutic response were conducted among the three subtypes. We further applied linear discriminant analysis to construct a characteristic index to classify the subtypes, and the Pearson correlation coefficient for the relationship between the index and immune checkpoint genes. Weighted Correlation Network Analysis (WGCNA) was used to mine the associated modules and specific genes. RESULTS We collected gene expression profiles from 352 STAD cases in the TCGA database, 300 in GSE62254, and 344 in GSE84437. Three STAD subtypes (IS1-IS3) were established according to the TIME signatures. The IS3 subtype had the highest immune score and the best prognosis, as well as markedly increased immune T-cell CYT, Th1/IFNγ scores, and immune checkpoint gene expression, compared to the other two subtypes. It was highly similar to the PD-1 response group in the previous study samples of GSE91061. The established TIME classification index performed well in classifying subtypes and was directly proportional to immune checkpoint-related gene expression levels. WGCNA explored 6 modules and 14 genes, namely DYSF, MAN1C1, HTRA3, EMCN, RFLNB, KANK3, MAGEH1, CD93, PCAT19, FUT11, BMP1, FOSB, DCHS1, and TCF3, which were associated with the established TIME classification index and STAD patient prognosis. CONCLUSION TIME phenotypes of STAD patients could be divided into three different molecular subtypes, which displayed different prognoses, immune features, and therapeutic responses. Our results shed new light on predicting patient outcomes and the discovery of new anti-STAD therapeutic strategies according to the TIME.
Collapse
Affiliation(s)
- Yimiao Zhu
- Department of Clinical Medicine, Medical College of Soochow University, Suzhou 215006, People's Republic of China; Department of Gastroenterology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, People's Republic of China
| | - Yu Zhao
- Department of Endocrinology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, People's Republic of China
| | - Zhongsheng Cao
- Department of Gastroenterology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, People's Republic of China
| | - Zhihao Chen
- Department of Gastroenterology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, People's Republic of China
| | - Wensheng Pan
- Department of Clinical Medicine, Medical College of Soochow University, Suzhou 215006, People's Republic of China; Department of Gastroenterology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, People's Republic of China.
| |
Collapse
|
12
|
Yu B, Dai W, Pang L, Sang Q, Li F, Yu J, Feng H, Li J, Hou J, Yan C, Su L, Zhu Z, Li YY, Liu B. The dynamic alteration of transcriptional regulation by crucial TFs during tumorigenesis of gastric cancer. Mol Med 2022; 28:41. [PMID: 35421923 PMCID: PMC9008954 DOI: 10.1186/s10020-022-00468-7] [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: 12/12/2021] [Accepted: 04/04/2022] [Indexed: 11/26/2022] Open
Abstract
Background The mechanisms of Gastric cancer (GC) initiation and progression are complicated, at least partly owing to the dynamic changes of gene regulation during carcinogenesis. Thus, investigations on the changes in regulatory networks can improve the understanding of cancer development and provide novel insights into the molecular mechanisms of cancer. Methods Differential co-expression analysis (DCEA), differential gene regulation network (GRN) modeling and differential regulation analysis (DRA) were integrated to detect differential transcriptional regulation events between gastric normal mucosa and cancer samples based on GSE54129 dataset. Cytological experiments and IHC staining assays were used to validate the dynamic changes of CREB1 regulated targets in different stages. Results A total of 1955 differentially regulated genes (DRGs) were identified and prioritized in a quantitative way. Among the top 1% DRGs, 14 out of 19 genes have been reported to be GC relevant. The four transcription factors (TFs) among the top 1% DRGs, including CREB1, BPTF, GATA6 and CEBPA, were regarded as crucial TFs relevant to GC progression. The differentially regulated links (DRLs) around the four crucial TFs were then prioritized to generate testable hypotheses on the differential regulation mechanisms of gastric carcinogenesis. To validate the dynamic alterations of gene regulation patterns of crucial TFs during GC progression, we took CREB1 as an example to screen its differentially regulated targets by using cytological and IHC staining assays. Eventually, TCEAL2 and MBNL1 were proved to be differentially regulated by CREB1 during tumorigenesis of gastric cancer. Conclusions By combining differential networking information and molecular cell experiments verification, testable hypotheses on the regulation mechanisms of GC around the core TFs and their top ranked DRLs were generated. Since TCEAL2 and MBNL1 have been reported to be potential therapeutic targets in SCLC and breast cancer respectively, their translation values in GC are worthy of further investigation. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00468-7.
Collapse
|
13
|
Target Therapy for Hepatocellular Carcinoma: Beyond Receptor Tyrosine Kinase Inhibitors and Immune Checkpoint Inhibitors. BIOLOGY 2022; 11:biology11040585. [PMID: 35453784 PMCID: PMC9027240 DOI: 10.3390/biology11040585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/19/2022] [Accepted: 04/07/2022] [Indexed: 11/24/2022]
Abstract
Simple Summary Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer and its incidence is steadily increasing. The development of HCC is a complex, multi-step process that is accompanied by alterations in multiple signaling cascades. Recent years have seen advancement in understanding molecular signaling pathways that play central roles in hepatocarcinogenesis. Aberrant activation of YAP/TAZ, Hedgehog, or Wnt/β-catenin signaling is frequently found in a subset of HCC patients. Targeting the signaling pathway via small molecule inhibitors could be a promising therapeutic option for the subset of patients. In this review, we will introduce the signaling pathways, discuss their roles in the development of HCC, and propose a therapeutic approach targeting the signaling pathways in the context of HCC. Abstract Hepatocellular carcinoma (HCC) is a major health concern worldwide, and its incidence is increasing steadily. To date, receptor tyrosine kinases (RTKs) are the most favored molecular targets for the treatment of HCC, followed by immune checkpoint regulators such as PD-1, PD-L1, and CTLA-4. With less than desirable clinical outcomes from RTK inhibitors as well as immune checkpoint inhibitors (ICI) so far, novel molecular target therapies have been proposed for HCC. In this review, we will introduce diverse molecular signaling pathways that are aberrantly activated in HCC, focusing on YAP/TAZ, Hedgehog, and Wnt/β-catenin signaling pathways, and discuss potential therapeutic strategies targeting the signaling pathways in HCC.
Collapse
|
14
|
Scalp Electroacupuncture Promotes Angiogenesis after Stroke in Rats by Activation of Wnt/ β-Catenin Signal Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1649605. [PMID: 35321503 PMCID: PMC8938052 DOI: 10.1155/2022/1649605] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 10/31/2021] [Accepted: 02/08/2022] [Indexed: 11/18/2022]
Abstract
Background Scalp acupuncture is a contemporary acupuncture method based on the fundamental theories of traditional acupuncture, which has been widely used in patients with stroke in China. However, the effectiveness is controversial due to lack of solid experimental evidence. Methods In this study, a rat model of cerebral ischemia-reperfusion injury (CIRI) was established by the middle cerebral artery occlusion/recirculation. The efficacy of scalp acupuncture against CIRI was evaluated by the mNSS scores, TTC staining for brain slices, and laser Doppler perfusion imaging. Immunohistochemical staining for angiogenetic factors indicated the vascularization after CIRI, including VEGF, Ang2, and bFGF. Activation of the Wnt/β-catenin signaling pathway and p-GSK3β (ser9)/VEGF pathway in the injured brain tissues was assessed by western blotting and qRT-PCR. Results On the 7, 14, and 21 days after CIRI, scalp acupuncture could reduce the mNSS scores, decrease the cerebral infarction area, and accelerate the recirculation of ischemic brain tissues. VEGF, FLK1, bFGF, and Ang2 were upregulated on both the mRNA and protein levels in the ischemic brain tissues of the AC group, suggesting that the recirculation might result from angiogenesis, which was also confirmed with the IHC staining in the angiogenetic markers of VEGF, Ang2, and bFGF. Moreover, Wnt3a, β-catenin, and cyclin D1 were also upregulated on both the mRNA and protein levels in the ischemic brain tissues of the AC group on day 7, 14, and 21, indicating that the Wnt/β-catenin signaling pathway was activated after the treatment of scalp acupuncture. In contrast, dikkoppf-1 (DKK1) pretreatment, a specific inhibitor for the Wnt/β-catenin signaling pathway, inactivated the Wnt3a/β-catenin signaling pathway and exacerbated the infarct size induced by the cerebral IR injury on day 7. Conclusion Together, our findings demonstrated a mechanism whereby scalp acupuncture led to the activation of Wnt/β-catenin signaling pathway, promoting angiogenetic factor expression and restoring blood perfusion in the ischemic zone.
Collapse
|
15
|
Jiang L, Li Y, He Y, Wei D, Yan L, Wen H. Knockdown of m6A Reader IGF2BP3 Inhibited Hypoxia-Induced Cell Migration and Angiogenesis by Regulating Hypoxia Inducible Factor-1α in Stomach Cancer. Front Oncol 2021; 11:711207. [PMID: 34621671 PMCID: PMC8490730 DOI: 10.3389/fonc.2021.711207] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/02/2021] [Indexed: 01/01/2023] Open
Abstract
Hypoxia is a common feature of solid tumors including stomach cancer (SC) and is closely associated with cancer malignant progression. N6-methyladenosine (m6A), a common modification on RNA, is involved in the regulation of RNA fate and hypoxic responses in cancers. However, the interaction between m6A reader insulin-like growth factor-II mRNA-binding protein 3 (IGF2BP3) and SC hypoxic microenvironment is poorly defined. In the present study, expression levels of IGF2BP3 and hypoxia inducible factor-1α (HIF1A) were examined by bioinformatics analysis and RT-qPCR and western blot assays. Cell migratory ability was assessed through Transwell and wound healing assays. The angiogenic potential was evaluated by VEGF secretion, tube formation, and chick embryo chorioallantoic membrane (CAM) assays. The interaction between IGF2BP3 and HIF1A was explored using bioinformatics analysis and RIP and luciferase reporter assays. The results showed that IGF2BP3 and HIF1A were highly expressed in SC tissues and hypoxia-treated SC cells. IGF2BP3 knockdown inhibited hypoxia-induced cell migration and angiogenesis in SC. IGF2BP3 positively regulated HIF1A expression by directly binding to a specific m6A site in the coding region of HIF1A mRNA in SC cells. HIF1A overexpression abrogated the effects of IGF2BP3 knockdown on hypoxia-induced cell migration and angiogenesis in SC. In conclusion, IGF2BP3 knockdown inhibited hypoxia-induced cell migration and angiogenesis by down-regulating HIF1A in SC.
Collapse
Affiliation(s)
- Libin Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yingxia Li
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yixin He
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dapeng Wei
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lvyin Yan
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongtao Wen
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
16
|
Wen JX, Tong YL, Ma X, Wang RL, Li RS, Song HT, Zhao YL. Therapeutic effects and potential mechanism of dehydroevodiamine on N-methyl-N'-nitro-N-nitrosoguanidine-induced chronic atrophic gastritis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 91:153619. [PMID: 34320422 DOI: 10.1016/j.phymed.2021.153619] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/27/2021] [Accepted: 05/28/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUNDS Dehydroevodiamine (DHE) is a quinazoline alkaloid isolated from a Chinese herbal medicine, named Euodiae Fructus (Wu-Zhu-Yu in Chinese). This study aimed to investigate the therapeutic effects and potential mechanism of DHE on N-methyl-N'-nitro-N-nitrosoguanidine (MNNG)-induced chronic atrophic gastritis (CAG) based on integrated approaches. METHODS Therapeutic effects of DHE on serum biochemical indices and histopathology of gastric tissue in MNNG-induced CAG rats were analyzed. MNNG-induced GES-1 human gastric epithelial cell injury model was established. Cell viability and proliferation was quantified by a cell counting kit-8 assay. Cell morphology and mitochondrial membrane potential (MMP) were detected by a high content screening (HCS) assay. Cell migration and invasion were detected by a Transwell chamber. Moreover, UHPLC-Q-TOF/MS was performed to investigate the potential metabolites and signaling pathway affecting the protective effects of DHE on MNNG-induced cell migration and invasion of GES-1. Furthermore, in view of the key role of angiogenesis in the transformation of inflammation and cancer, this study explored relative mRNA and protein expression levels of HIF-1α-mediated VEGF pathway in vivo and in vitro by RT-PCR and Western Blotting, respectively. RESULTS The results showed that the therapeutic effects of DHE on CAG rats were presented in down-regulation serum biochemical indices and alleviating histological damage of gastric tissue. Besides, DHE has an effect on increasing cell proliferation of GES-1 cells, ameliorating MNNG-induced gastric epithelial cell damage and mitochondrial dysfunction. In addition, DHE could inhibit MNNG induced migration and invasion of GES-1 cells. Cell metabolomics analyses showed that the protective effect of DHE on GES-1 cells is mainly associated with the regulation of inflammation metabolites and energy metabolism related pathways. It was found that DHE has a regulating effect on tumor angiogenesis and can inhibit the relative gene and protein expression of HIF-1α-mediated VEGF signaling pathway. CONCLUSIONS The present work highlighted the role of DHE ameliorated gastric injury in MNNG-induced CAG rats in vivo and GES-1 cell migration in vitro by inhibiting HIF-1α/VEGF angiogenesis pathway. These results suggest that DHE may be the effective components of Euodiae Fructus, which provides a new agent for the treatment of CAG.
Collapse
Affiliation(s)
- Jian-Xia Wen
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Yu-Ling Tong
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Xiao Ma
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rui-Lin Wang
- Department of Integrative Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Rui-Sheng Li
- Research Center for Clinical and Translational Medicine, Chinese PLA General Hospital, Beijing, China
| | - Hong-Tao Song
- Department of Pharmacy, 900 Hospital of the Joint Logistics Team, Fuzhou, China.
| | - Yan-Ling Zhao
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
17
|
Huang MP, Gu SZ, Huang B, Li GW, Xiong ZP, Tang T, Zeng SN. Apatinib Inhibits Angiogenesis in Intrahepatic Cholangiocarcinoma by Regulating the Vascular Endothelial Growth Factor Receptor-2/Signal Transducer and Activator of Transcription Factor 3/Hypoxia Inducible Factor 1 Subunit Alpha Signaling Axis. Pharmacology 2021; 106:509-519. [PMID: 34412054 DOI: 10.1159/000514410] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 11/24/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Intrahepatic cholangiocarcinoma (ICC), which is difficult to diagnose and is usually fatal due to its late clinical presentation and a lack of effective treatment, has risen over the past decades but without much improvement in prognosis. OBJECTIVE The study aimed to investigate the role of apatinib that targets vascular endothelial growth factor receptor-2 (VEGFR2) in ICC. METHODS MTT assays, cell scratch assays, and tube formation assays were used to assess the effect of apatinib on human ICC cell line (HuCCT-1) and RBE cells proliferation, migration, and angiogenic capacity, respectively. Expression of vascular endothelial growth factor (VEGF), VEGFR2, signal transducer and activator of transcription factor 3 (STAT3), pSTAT3, and hypoxia inducible factor 1 subunit alpha (HIF-1α) pathway proteins was assessed using Western blotting and mRNA expression analysis in HuCCT-1 was performed using RT-qPCR assays. The pcDNA 3.1(-)-VEGFR2 and pcDNA 3.1(-)-HIF-1α were transfected into HuCCT-1 and RBE cells using Lipofectamine 2,000 to obtain overexpressed HuCCT-1 and RBE cells. RESULTS We found that apatinib-inhibited proliferation, migration, and angiogenesis of HuCCT-1 and RBE cells in vitro in a dose-dependent manner. We also proved that apatinib effectively inhibits angiogenesis in tumor cells by blocking the expression of VEGF and VEGFR2 in these cells. In addition, we demonstrated that apatinib regulates the expression of STAT3 phosphorylation by inhibiting VEGFR2. Finally, we showed that apatinib regulates ICC angiogenesis and HIF-1α/VEGF expression via STAT3. CONCLUSIONS Based on the above findings, we conclude that apatinib inhibits HuCCT-1 and RBE cell proliferation, migration, and tumor angiogenesis by inhibiting the VEGFR2/STAT3/HIF-1α axis signaling pathway. Apatinib can be a promising drug for ICC-targeted molecular therapy.
Collapse
Affiliation(s)
- Man-Ping Huang
- Department of Intervention Hunan, Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University, Changsha, China
| | - Shan-Zhi Gu
- Department of Intervention Hunan, Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University, Changsha, China
| | - Bin Huang
- Department of Intervention Hunan, Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University, Changsha, China
| | - Guo-Wen Li
- Department of Intervention Hunan, Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University, Changsha, China
| | - Zheng-Ping Xiong
- Department of Intervention Hunan, Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University, Changsha, China
| | - Tian Tang
- Department of Intervention Hunan, Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine Central South University, Changsha, China
| | - Sai-Nan Zeng
- Infection Controlling Center, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
18
|
Gastrin mediates cardioprotection through angiogenesis after myocardial infarction by activating the HIF-1α/VEGF signalling pathway. Sci Rep 2021; 11:15836. [PMID: 34349170 PMCID: PMC8339006 DOI: 10.1038/s41598-021-95110-7] [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: 02/15/2021] [Accepted: 07/16/2021] [Indexed: 11/08/2022] Open
Abstract
Acute myocardial infarction (MI) is one of the leading causes of death in humans. Our previous studies showed that gastrin alleviated acute myocardial ischaemia-reperfusion injury. We hypothesize that gastrin might protect against heart injury after MI by promoting angiogenesis. An MI model was simulated by ligating the anterior descending coronary artery in adult male C57BL/6J mice. Gastrin was administered twice daily by intraperitoneal injection for 2 weeks after MI. We found that gastrin reduced mortality, improved myocardial function with reduced infarct size and promoted angiogenesis. Gastrin increased HIF-1α and VEGF expression. Downregulation of HIF-1α expression by siRNA reduced the proliferation, migration and tube formation of human umbilical vein endothelial cells. These results indicate that gastrin restores cardiac function after MI by promoting angiogenesis via the HIF-1α/VEGF pathway.
Collapse
|
19
|
Zhou Y, Wan Y, He M, Li Y, Wu Q, Yao H. Determination of Vascular Endothelial Growth Factor (VEGF) in Cell Culture Medium by Gold-Coated Magnetic Nanoparticle Based Label-Free Electrochemical Impedance Spectroscopy (EIS). ANAL LETT 2021. [DOI: 10.1080/00032719.2021.1951750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Yaping Zhou
- College of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Yao Wan
- College of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Mingyu He
- College of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Ying Li
- College of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Qimei Wu
- College of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Hui Yao
- College of Pharmacy, Zunyi Medical University, Zunyi, China
| |
Collapse
|
20
|
Uz Z, Ergin B, Shen L, van Lienden KP, Rassam F, Olthof PB, Bennink RJ, Ince C, van Gulik TM. Increased Hepatic Microvascular Density, Oxygenation, and VEGF in the Hypertrophic Lobe following Portal Vein Embolization in Rabbits. Eur Surg Res 2021; 63:9-18. [PMID: 34265760 DOI: 10.1159/000517025] [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: 02/15/2021] [Accepted: 04/12/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION The microvascular events following portal vein embolization (PVE) are poorly understood despite the pivotal role of the microcirculation in liver regeneration and tumor progression. We aimed to assess the changes in hepatic microvascular perfusion and neo-angiogenesis after experimental PVE. METHODS PVE of the cranial liver lobes was performed in 12 New Zealand White rabbits divided into 2 groups of permanent (P-PVE) and reversible PVE (R-PVE), respectively. Hepatobiliary scintigraphy and CT were used to evaluate hepatic function and volume. Hepatic microcirculation was assessed using a handheld vital microscope (Cytocam) to measure microvascular density (total vessel density; TVD) before PVE, right after PVE, and 20 min after PVE, as well as at 14 days (D14 post-PVE) and 35 days (D35 post-PVE). Additionally, on D35, microvascular PO2 and liver parenchymal VEGF were assessed. RESULTS Eleven rabbits were included after PVE (R-PVE, n = 5; P-PVE, n = 6). TVD in the nonembo-lized (hypertrophic) lobes was higher than in the embolized (atrophic) lobes of the P-PVE group at D35 post-PVE (36.7 ± 7.2 vs. 23.4 ± 4.9 mm/mm2; p < 0.05). In the R-PVE group, TVD in the nonembolized lobes was not increased at D35. Function and volume were increased in the nonembolized lobes of the P-PVE group compared to the embolized lobes, but not in the R-PVE group. Likewise, the mmicrovascular PO2 and VEGF staining rate were higher in the nonembolized lobes of the P-PVE group at D35 post-PVE. DISCUSSION/CONCLUSION Successful volumetric and functional hypertrophy of the nonembolized lobe was accompanied by microvascular alterations featuring increased neo-angiogenesis, microvascular density, and microvascular oxygen pressure following P-PVE.
Collapse
Affiliation(s)
- Zühre Uz
- Department of Surgery, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Translational Physiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Bülent Ergin
- Department of Intensive Care Adults, Laboratory of Translational Intensive Care, Erasmus MC, Rotterdam, The Netherlands
| | - Lucinda Shen
- Department of Intensive Care Adults, Laboratory of Translational Intensive Care, Erasmus MC, Rotterdam, The Netherlands
| | - Krijn P van Lienden
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Fadi Rassam
- Department of Surgery, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Pim B Olthof
- Department of Surgery, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Surgery, Erasmus MC, Rotterdam, The Netherlands
| | - Roel J Bennink
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Can Ince
- Department of Translational Physiology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.,Department of Intensive Care Adults, Laboratory of Translational Intensive Care, Erasmus MC, Rotterdam, The Netherlands
| | - Thomas M van Gulik
- Department of Surgery, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
21
|
Knockdown of Gastrin Promotes Apoptosis of Gastric Cancer Cells by Decreasing ROS Generation. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5590037. [PMID: 33937399 PMCID: PMC8062189 DOI: 10.1155/2021/5590037] [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: 01/08/2021] [Accepted: 04/01/2021] [Indexed: 12/27/2022]
Abstract
Overexpressed gastrin is reported to promote oncogenesis and development of gastric cancer by inhibiting apoptosis of cancer cells; however, the underlying mechanism remains unclear. Our study is aimed at revealing the mechanism underlying the effect of gastrin on apoptosis of gastric cancer cells. Gastrin-interfering cell line was constructed by stably transfecting gastrin-specific pshRNA plasmid to gastric cancer cell line BGC-823. Then, differentially expressed proteins between untreated BGC-823 and gastrin-interfering BGC-823 cell lines were detected by the iTRAQ technique. GO and KEGG analysis was used to analyze the differentially expressed genes that code these differentially expressed proteins. The Annexin V-FITC staining assay was used to detect gastric cancer cell apoptosis. The DCFH-DA fluorescent probe staining assay was used to measure intracellular ROS. Mitochondrial membrane potential was detected by flow cytometry. Western blot was used to analyze the mitochondria respiratory chain proteins and apoptosis-related proteins. A total of 107 differentially expressed proteins were identified by iTRAQ. GO and KEGG analysis showed that proteins coded by the corresponding differentially expressed genes were mainly enriched in the mitochondrial oxidative respiratory chain, and the expression of three proteins (COX17, COX5B, ATP5J) was upregulated. The three proteins with higher scores were verified by Western blot. The apoptosis rate of the gastrin knockdown cancer cell was significantly increased; meanwhile, gastrin knockdown leads to increase of membrane potential and decrease of intracellular ROS production. Additionally, Bax was significantly increased, whereas NF-κB-p65 and Bcl-2 were downregulated after knockdown of gastrin. Concomitantly, pretreatment with NAC reversed the effect of gastrin on the Bax and Bcl-2 expression. Gastrin promotes the production of ROS from mitochondria, activates NF-κB, and inhibits apoptosis via modulating the expression level of Bcl-2 and Bax.
Collapse
|
22
|
Kietzmann T, Mäkelä VH. The hypoxia response and nutritional peptides. Peptides 2021; 138:170507. [PMID: 33577839 DOI: 10.1016/j.peptides.2021.170507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/29/2021] [Accepted: 02/03/2021] [Indexed: 10/22/2022]
Abstract
Hypoxia controls metabolism at several levels, e.g., via mitochondrial ATP production, glucose uptake and glycolysis. Hence it is likely that hypoxia also affects the action and/or production of many peptide hormones linked to food intake and appetite control. Many of those are produced in the gastrointestinal tract, endocrine pancreas, adipose tissue, and selective areas in the brain which modulate and concert their actions. However, the complexity of the hypoxia response and the links to peptides/hormones involved in food intake and appetite control in the different organs are not well known. This review summarizes the role of the hypoxia response and its effects on major peptides linked to appetite regulation, nutrition and metabolism.
Collapse
Affiliation(s)
- Thomas Kietzmann
- University of Oulu, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Oulu, Finland.
| | - Ville H Mäkelä
- University of Oulu, Faculty of Biochemistry and Molecular Medicine, Biocenter Oulu, Oulu, Finland
| |
Collapse
|
23
|
He RP, Jin Z, Ma RY, Hu FD, Dai JY. Network pharmacology unveils spleen-fortifying effect of Codonopsis Radix on different gastric diseases based on theory of “same treatment for different diseases” in traditional Chinese medicine. CHINESE HERBAL MEDICINES 2021; 13:189-201. [PMID: 36117498 PMCID: PMC9476800 DOI: 10.1016/j.chmed.2020.12.005] [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: 07/13/2020] [Revised: 10/13/2020] [Accepted: 10/23/2020] [Indexed: 12/08/2022] Open
Abstract
Objective Methods Results Conclusion
Collapse
|
24
|
Camacho-Cardenosa M, Quesada-Gómez JM, Camacho-Cardenosa A, Leal A, Dorado G, Torrecillas-Baena B, Casado-Díaz A. Effects of normobaric cyclic hypoxia exposure on mesenchymal stem-cell differentiation-pilot study on bone parameters in elderly. World J Stem Cells 2020; 12:1667-1690. [PMID: 33505607 PMCID: PMC7789125 DOI: 10.4252/wjsc.v12.i12.1667] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/30/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSC) of bone marrow are the progenitor of osteoblasts and adipocytes. MSC tend to differentiate into adipocytes, instead of osteoblasts, with aging. This favors the loss of bone mass and development of osteoporosis. Hypoxia induces hypoxia inducible factor 1α gene encoding transcription factor, which regulates the expression of genes related to energy metabolism and angiogenesis. That allows a better adaptation to low O2 conditions. Sustained hypoxia has negative effects on bone metabolism, favoring bone resorption. Yet, surprisingly, cyclic hypoxia (CH), short times of hypoxia followed by long times in normoxia, can modulate MSC differentiation and improve bone health in aging. AIM To evaluate the CH effect on MSC differentiation, and whether it improves bone mineral density in elderly. METHODS MSC cultures were induced to differentiate into osteoblasts or adipocytes, in CH (3% O2 for 1, 2 or 4 h, 4 d a week). Extracellular-matrix mineralization and lipid-droplet formation were studied in MSC induced to differentiate into osteoblast or adipocytes, respectively. In addition, gene expression of marker genes, for osteogenesis or adipogenesis, have been quantified by quantitative real time polymerase chain reaction. The in vivo studies with elderly (> 75 years old; n = 10) were carried out in a hypoxia chamber, simulating an altitude of 2500 m above sea level, or in normoxia, for 18 wk (36 CH sessions of 16 min each). Percentages of fat mass and bone mineral density from whole body, trunk and right proximal femur (femoral, femoral neck and trochanter) were assessed, using dual-energy X-ray absorptiometry. RESULTS CH (4 h of hypoxic exposure) inhibited extracellular matrix mineralization and lipid-droplet formation in MSC induced to differentiate into osteoblasts or adipocytes, respectively. However, both parameters were not significantly affected by the other shorter hypoxia times assessed. The longest periods of hypoxia downregulated the expression of genes related to extracellular matrix formation, in MSC induced to differentiate into osteoblasts. Interestingly, osteocalcin (associated to energy metabolism) was upregulated. Vascular endothelial growth factor an expression and low-density lipoprotein receptor related protein 5/6/dickkopf Wnt signaling pathway inhibitor 1 (associated to Wnt/β-catenin pathway activation) increased in osteoblasts. Yet, they decreased in adipocytes after CH treatments, mainly with the longest hypoxia times. However, the same CH treatments increased the osteoprotegerin/receptor activator for nuclear factor kappa B ligand ratio in both cell types. An increase in total bone mineral density was observed in elderly people exposed to CH, but not in specific regions. The percentage of fat did not vary between groups. CONCLUSION CH may have positive effects on bone health in the elderly, due to its possible inhibitory effect on bone resorption, by increasing the osteoprotegerin / receptor activator for nuclear factor kappa B ligand ratio.
Collapse
Affiliation(s)
| | - José Manuel Quesada-Gómez
- CIBER De Fragilidad Y Envejecimiento Saludable (CIBERFES), Unidad De Gestión Clínica De Endocrinología Y Nutrición, Instituto Maimónides De Investigación Biomédica De Córdoba, Hospital Universitario Reina Sofía, Córdoba 14004, Spain
| | | | - Alejo Leal
- Servicio de Traumatología, Hospital de Cáceres, Cáceres 10004, Spain
| | - Gabriel Dorado
- Departamento Bioquímica y Biología Molecular, Campus Rabanales C6-1-E17, Campus de Excelencia Internacional Agroalimentario (ceiA3), Universidad de Córdoba-CIBERFES, 14071 Córdoba, Spain
| | - Bárbara Torrecillas-Baena
- CIBER De Fragilidad Y Envejecimiento Saludable (CIBERFES), Unidad De Gestión Clínica De Endocrinología Y Nutrición, Instituto Maimónides De Investigación Biomédica De Córdoba, Hospital Universitario Reina Sofía, Córdoba 14004, Spain
| | - Antonio Casado-Díaz
- CIBER De Fragilidad Y Envejecimiento Saludable (CIBERFES), Unidad De Gestión Clínica De Endocrinología Y Nutrición, Instituto Maimónides De Investigación Biomédica De Córdoba, Hospital Universitario Reina Sofía, Córdoba 14004, Spain
| |
Collapse
|
25
|
Forma A, Tyczyńska M, Kędzierawski P, Gietka K, Sitarz M. Gastric carcinogenesis: a comprehensive review of the angiogenic pathways. Clin J Gastroenterol 2020; 14:14-25. [PMID: 33206367 PMCID: PMC7886717 DOI: 10.1007/s12328-020-01295-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 10/31/2020] [Indexed: 12/12/2022]
Abstract
Gastric cancer (GC) is undoubtedly one of the most prevalent malignancies worldwide. Since GC is the second leading cause of cancer-related deaths with nearly one million new diagnoses reported every year, there is a need for the development of new, effective treatment strategies of GC. Gastric carcinogenesis is a complex process that is induced by numerous factors and further stimulated by many pro-oncogenic pathways. Angiogenesis is the process of the new blood vessels formation from the already existing ones and it significantly contributes to the progression of gastric tumorigenesis and the growth of the cancerous tissues. The newly formed vessels provide cancer cells with proper nutrition, growth factors, and oxygen supply that are crucial for tumor growth and progression. Tumor-associated vessels differ from the physiological ones both morphologically and functionally. They are usually inefficient and unevenly distributed due to structural transformations. Thus, the development of the angiogenesis inhibitors that possess therapeutic effects has been the main focus of recent studies. Angiogenesis inhibitors mostly affect the vascular endothelial growth factor (VEGF) pathway since it is a major factor that stimulates the pro-angiogenic pathways. The aim of this review was to describe and summarize other promising molecular pathways that might be crucial in further improvements in GC therapies. This article provides an overview of how a meaningful role in tumor progression the angiogenetic process has. Furthermore, this review includes a description of the most important angiogenic factors as well as pathways and their involvement in gastric carcinogenesis.
Collapse
Affiliation(s)
- Alicja Forma
- Department of Forensic Medicine, Medical University of Lublin, 20-090, Lublin, Poland.
| | - Magdalena Tyczyńska
- Department of Human Anatomy, Medical University of Lublin, 20-090, Lublin, Poland
| | - Paweł Kędzierawski
- Department of Forensic Medicine, Medical University of Lublin, 20-090, Lublin, Poland
| | - Klaudyna Gietka
- Department of Forensic Medicine, Medical University of Lublin, 20-090, Lublin, Poland
| | - Monika Sitarz
- Department of Conservative Dentistry with Endodontics, Medical University of Lublin, 20-090, Lublin, Poland
| |
Collapse
|
26
|
He S, Tang S. WNT/β-catenin signaling in the development of liver cancers. Biomed Pharmacother 2020; 132:110851. [PMID: 33080466 DOI: 10.1016/j.biopha.2020.110851] [Citation(s) in RCA: 212] [Impact Index Per Article: 42.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/27/2020] [Accepted: 10/04/2020] [Indexed: 02/06/2023] Open
Abstract
The WNT/β-catenin signaling pathway is a highly conserved and tightly controlled molecular mechanism that regulates embryonic development, cellular proliferation and differentiation. Of note, accumulating evidence has shown that the aberrant of WNT/β-catenin signaling promotes the development and/or progression of liver cancer, including hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), the two most prevalent primary liver tumours in adults. There are two different WNT signaling pathways have been identified, which were termed non-canonical and canonical pathways, the latter involving the activation of β-catenin. β-catenin, acting as an intracellular signal transducer in the WNT signaling pathway, is encoded by CTNNB1 and plays a critical role in tumorigenesis. In the past research, most liver tumors have mutations in genes encoding key components of the WNT/β-catenin signaling pathway. In addition, several of other signaling pathways also can crosswalk with β-catenin. In this review, we discuss the most relevant molecular mechanisms of action and regulation of WNT/β-catenin signaling in the development and pathophysiology of liver cancers, as well as in the development of therapeutics.
Collapse
Affiliation(s)
- Shuai He
- Department of General Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China
| | - Shilei Tang
- Department of General Surgery, the Fourth Affiliated Hospital, China Medical University, Shenyang 110032, China.
| |
Collapse
|
27
|
Cheng C, Shou Q, Lang J, Jin L, Liu X, Tang D, Yang Z, Fu H. Gehua Jiecheng Decoction Inhibits Diethylnitrosamine-Induced Hepatocellular Carcinoma in Mice by Improving Tumor Immunosuppression Microenvironment. Front Pharmacol 2020; 11:809. [PMID: 32547401 PMCID: PMC7272686 DOI: 10.3389/fphar.2020.00809] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 05/18/2020] [Indexed: 12/11/2022] Open
Abstract
Gehua Jiecheng Decoction (GHJCD), a famous traditional Chinese medicine, has been used in the prevention and treatment of precancerous lesion of liver cancer, but its active mechanism has not been reported. This study aimed to evaluate the therapeutic effect of GHJCD on diethylnitrosamine (DEN)-induced hepatocellular carcinoma (HCC) in mice and the mechanism of this effect. We found that GHJCD effectively inhibited the occurrence of liver cancer and reduced the tumor area. The ratio of regulatory cells (Tregs), tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs) in HCC microenvironment was down-regulated, whereas that of CD8 T and effective CD8 T cells was up-regulated. In addition, the expression levels of inflammatory factors IL-6, IL-10, TNF-α, and CCL-2 in the liver were inhibited, whereas those of the angiogenesis related molecules CD31 and VEGF were decreased. Moreover, WNT1, β-catenin, NF-kB, p-MAPK, p-AKT, and p-SRC content in the liver decreased, whereas APC content increased. These results suggested that GHJCD exerted a good inhibitory effect on liver cancer induced by DEN and thus may have a multi-target effect; GHJCD not only antagonized the immunosuppressive effect of the microenvironment of liver cancer but also exerted strong anti-inflammatory and antiangiogenesis effects.
Collapse
Affiliation(s)
- Changpei Cheng
- Affiliated First Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China.,Affiliated Secondary Hospital, Zhejiang Chinese Medical University, Hangzhou, China.,Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qiyang Shou
- Affiliated Secondary Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiali Lang
- Affiliated Secondary Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Jin
- Affiliated Secondary Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xia Liu
- Affiliated Secondary Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| | - Dongxin Tang
- Affiliated First Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Zhu Yang
- Affiliated First Hospital, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Huiying Fu
- Affiliated Secondary Hospital, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
28
|
Mei F, Liu J, Wu J, Duan Z, Chen M, Meng K, Chen S, Shen X, Xia G, Zhao M. Collagen Peptides Isolated from Salmo salar and Tilapia nilotica Skin Accelerate Wound Healing by Altering Cutaneous Microbiome Colonization via Upregulated NOD2 and BD14. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:1621-1633. [PMID: 31967468 DOI: 10.1021/acs.jafc.9b08002] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Collagen peptides can promote wound healing and are closely related to microbiome colonization. We investigated the relationship among collagen peptides, wound healing, and wound microflora colonization by administering the murine wound model with Salmo salar skin collagen peptides (Ss-SCPs) and Tilapia nilotica skin collagen peptides (Tn-SCPs). We analyzed the vascular endothelial growth factor (VEGF), fibroblast growth factors (β-FGF), pattern recognition receptor (NOD2), antimicrobial peptides (β-defence14, BD14), proinflammatory (TNF-α, IL-6, and IL-8) and anti-inflammatory (IL-10) cytokines, macrophages, neutrophil infiltration levels, and microbial communities in the rat wound. The healing rates of the Ss-SCP- and Tn-SCP-treated groups were significantly accelerated, associated with decreased TNF-α, IL-6, and IL-8 and upregulated BD14, NOD2, IL-10, VEGF, and β-FGF. Accelerated healing in the collagen peptide group shows that the wound microflora such as Leuconostoc, Enterococcus, and Bacillus have a positive effect on wound healing (P < 0.01). Other microbiome species such as Stenotrophomonas, Bradyrhizobium, Sphingomonas, and Phyllobacterium had a negative influence and decreased colonization (P < 0.01). Altogether, these studies show that collagen peptide could upregulate wound NOD2 and BD14, which were implicated in microflora colonization regulation in the wound tissue and promoted wound healing by controlling the inflammatory reaction and increasing wound angiogenesis and collagen deposition.
Collapse
Affiliation(s)
- Fengfeng Mei
- Hainan Engineering Research Center of Aquatic Resources Efficient Utilization in South China Sea , Hainan University , Hainan 570228 , China
- College of Food Science and Technology , Hainan University , Hainan 570228 , China
| | - Jingjie Liu
- College of Food Science and Technology , Hainan University , Hainan 570228 , China
| | - Jintao Wu
- College of Food Science and Technology , Hainan University , Hainan 570228 , China
| | - Zhouwei Duan
- Institute of Processing & Design of Agroproducts , Hainan Academy of Agricultural Science , Hainan 570228 , China
| | - Muxue Chen
- Hainan Engineering Research Center of Aquatic Resources Efficient Utilization in South China Sea , Hainan University , Hainan 570228 , China
- College of Food Science and Technology , Hainan University , Hainan 570228 , China
| | - Keke Meng
- Hainan Engineering Research Center of Aquatic Resources Efficient Utilization in South China Sea , Hainan University , Hainan 570228 , China
- College of Food Science and Technology , Hainan University , Hainan 570228 , China
| | - Shenjun Chen
- South China Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences , Guangdong 510000 , China
| | - Xuanri Shen
- Hainan Engineering Research Center of Aquatic Resources Efficient Utilization in South China Sea , Hainan University , Hainan 570228 , China
- College of Food Science and Technology , Hainan University , Hainan 570228 , China
- Collaborative Innovation Center of Marine Food Deep Processing , Dalian Polytechnic University , Liaoning 116000 , China
| | - Guanghua Xia
- Hainan Engineering Research Center of Aquatic Resources Efficient Utilization in South China Sea , Hainan University , Hainan 570228 , China
- College of Food Science and Technology , Hainan University , Hainan 570228 , China
- Collaborative Innovation Center of Marine Food Deep Processing , Dalian Polytechnic University , Liaoning 116000 , China
| | - Meihui Zhao
- College of Food Science and Technology , Hainan University , Hainan 570228 , China
| |
Collapse
|
29
|
Zhang H, Liao Y, Zhang H, Wu J, Zheng D, Chen Z. Cytotoxin-associated gene A increases carcinogenicity of helicobacter pylori in colorectal adenoma. Int J Biol Markers 2020; 35:19-25. [PMID: 31971064 DOI: 10.1177/1724600819877193] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE This study aimed to investigate the correlation of Helicobacter pylori (Hp) infection with disease risk and severity of colorectal adenoma, also to explore the association of cytotoxin-associated gene A (CagA) positive (CagA+)-Hp infection with gastrin and ki-67 expressions in colorectal adenoma patients. METHODS There were 1000 colorectal adenoma patients and 1500 controls consecutively enrolled, then Hp infection status was determined by 14C urea breath test and rapid urease test. Also, serum CagA expression and gastrin expression of colorectal adenoma patients were determined by enzyme-linked immunosorbent assay. Ki-67 expression in adenoma tissue of colorectal adenoma patients was assessed using immunohistochemistry. RESULTS Hp+ rate in colorectal adenoma patients (623 (62.3%)) was more elevated than that in controls (814 (54.3%)). Multivariate logistic regression model analysis disclosed that Hp+ was an independent risk factor for colorectal adenoma. Additionally, Hp+ was positively associated with tumor size and high-grade intraepithelial neoplasia in colorectal adenoma patients. Also, serum gastrin expression and intratumoral ki-67 expression were higher in Hp+ CagA+ patients and Hp+ CagA- patients compared to Hp- patients, and they were also higher in Hp+ CagA+ patients compared to Hp+ CagA- patients. CONCLUSION Hp infection positively associates with higher disease risk and worse disease conditions of colorectal adenoma, and CagA enhances the carcinogenicity of Hp in colorectal adenoma.
Collapse
Affiliation(s)
- Heng Zhang
- Department of Gastroenterology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology; Clinical Research Center for Intestinal & Colorectal Diseases of Hubei Province; Key Laboratory for Molecular Diagnosis of Hubei Province, Wuhan, China
| | - Yusheng Liao
- Department of Gastroenterology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology; Clinical Research Center for Intestinal & Colorectal Diseases of Hubei Province; Key Laboratory for Molecular Diagnosis of Hubei Province, Wuhan, China
| | - Hongfeng Zhang
- Department of Pathology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Wu
- Department of Gastroenterology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology; Clinical Research Center for Intestinal & Colorectal Diseases of Hubei Province; Key Laboratory for Molecular Diagnosis of Hubei Province, Wuhan, China
| | - Dan Zheng
- Department of Gastroenterology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology; Clinical Research Center for Intestinal & Colorectal Diseases of Hubei Province; Key Laboratory for Molecular Diagnosis of Hubei Province, Wuhan, China
| | - Zhitao Chen
- Department of Gastroenterology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology; Clinical Research Center for Intestinal & Colorectal Diseases of Hubei Province; Key Laboratory for Molecular Diagnosis of Hubei Province, Wuhan, China
| |
Collapse
|