1
|
El-Meguid MA, Lotaif LM, Salum GM, Fotouh BE, Salama RM, Salem MISE, El Awady MK, Abdel Aziz AO, Dawood RM. Evaluation of the expression of fibrosis-related genes as non-invasive diagnostic biomarkers for cirrhotic HCV-infected patients. Cytokine 2024; 182:156714. [PMID: 39068734 DOI: 10.1016/j.cyto.2024.156714] [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: 02/13/2024] [Revised: 07/02/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Liver cirrhosis is a condition with high mortality that poses a significant health and economic burden worldwide. The clinical characteristics of liver cirrhosis are complex and varied. Therefore, the evaluation of immune infiltration-involved genes incirrhosis has become mandatory in liver disease research, not only to identify the potential biomarkers but also to provide important insights into the underlying mechanisms of the disease. In this study, we aimed to investigate the expression profile of cytokine genes in peripheral blood mononuclear cells (PBMCs) of HCV patients and identify the gene expression signature associated with advanced cirrhosis. A cross-sectional study of 90 HCV genotype 4 patients, including no fibrosis patients (F0, n = 24), fibrotic patients (F1-F3, n = 36), and cirrhotic patients (F4, n = 30) has been conducted. The expression of cytokine genes was analyzed by quantitative real-time PCR in the subjects' PBMCs, and the serum level of TGFβ2 was measured by ELISA. Our findings showed that the expression level of the TGIF1 transcript was lower in cirrhotic and fibrotic patients compared to no fibrosis patients (p = 0.046 and 0.022, respectively). Also, there was an upregulation of the TGFβ1 gene in cirrhotic patients relative to fibrotic patients (p = 0.015). Additionally, the cirrhotic patients had higher expression levels of the TGF-β2 transcript and elevated levels of the TGF-β2 protein than patients with no cirrhosis or fibrosis. According to the ROC analysis, TGFβ1, TGIF1 transcripts, and TGFβ2 protein have a good discriminatory performance in distinguishing between cirrhotic, fibrotic, and non-fibrotic patients. Our results suggested that the expression of TGIF1, TGF-β1, and TGF-β2 genes in PBMCs may provide a valuable tool for identifying patients with advanced cirrhosis and that TGF-β and TGIF1 may be potential biomarkers for cirrhosis. These findings may have implications for the diagnosis and treatment of cirrhosis in HCV patients.
Collapse
Affiliation(s)
- Mai Abd El-Meguid
- Microbial Biotechnology Department, Biotechnology Research Institute, National Research Centre, P.O. 12622, Dokki, Giza, Egypt
| | - Lotaif Mostafa Lotaif
- Gastroenterology & Infectious Diseases Department, Ahmed Maher Teaching Hospital, Cairo, Egypt
| | - Ghada M Salum
- Microbial Biotechnology Department, Biotechnology Research Institute, National Research Centre, P.O. 12622, Dokki, Giza, Egypt
| | - Basma E Fotouh
- Microbial Biotechnology Department, Biotechnology Research Institute, National Research Centre, P.O. 12622, Dokki, Giza, Egypt
| | - Rabab Maamoun Salama
- Endemic Medicine and Hepatogastroenterology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | - Mostafa K El Awady
- Microbial Biotechnology Department, Biotechnology Research Institute, National Research Centre, P.O. 12622, Dokki, Giza, Egypt
| | - Ashraf Omar Abdel Aziz
- Endemic Medicine and Hepatogastroenterology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Reham M Dawood
- Microbial Biotechnology Department, Biotechnology Research Institute, National Research Centre, P.O. 12622, Dokki, Giza, Egypt.
| |
Collapse
|
2
|
Zhang B, Liu M, Mai F, Li X, Wang W, Huang Q, Du X, Ding W, Li Y, Barwick BG, Ni JJ, Osunkoya AO, Chen Y, Zhou W, Xia S, Dong JT. Interruption of KLF5 acetylation promotes PTEN-deficient prostate cancer progression by reprogramming cancer-associated fibroblasts. J Clin Invest 2024; 134:e175949. [PMID: 38781024 PMCID: PMC11245161 DOI: 10.1172/jci175949] [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: 09/19/2023] [Accepted: 05/21/2024] [Indexed: 05/25/2024] Open
Abstract
Inactivation of phosphatase and tensin homolog (PTEN) is prevalent in human prostate cancer and causes high-grade adenocarcinoma with a long latency. Cancer-associated fibroblasts (CAFs) play a pivotal role in tumor progression, but it remains elusive whether and how PTEN-deficient prostate cancers reprogram CAFs to overcome the barriers for tumor progression. Here, we report that PTEN deficiency induced Krüppel-like factor 5 (KLF5) acetylation and that interruption of KLF5 acetylation orchestrated intricate interactions between cancer cells and CAFs that enhance FGF receptor 1 (FGFR1) signaling and promote tumor growth. Deacetylated KLF5 promoted tumor cells to secrete TNF-α, which stimulated inflammatory CAFs to release FGF9. CX3CR1 inhibition blocked FGFR1 activation triggered by FGF9 and sensitized PTEN-deficient prostate cancer to the AKT inhibitor capivasertib. This study reveals the role of KLF5 acetylation in reprogramming CAFs and provides a rationale for combined therapies using inhibitors of AKT and CX3CR1.
Collapse
Affiliation(s)
- Baotong Zhang
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, School of Medicine, Shenzhen, Guangdong, China
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Mingcheng Liu
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, School of Medicine, Shenzhen, Guangdong, China
| | - Fengyi Mai
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, School of Medicine, Shenzhen, Guangdong, China
| | - Xiawei Li
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, School of Medicine, Shenzhen, Guangdong, China
- Inner Mongolia Institute of Quality and Standardization, Inner Mongolia Administration for Market Regulation, Hohhot, China
| | - Wenzhou Wang
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, School of Medicine, Shenzhen, Guangdong, China
| | - Qingqing Huang
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, School of Medicine, Shenzhen, Guangdong, China
| | - Xiancai Du
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, School of Medicine, Shenzhen, Guangdong, China
| | - Weijian Ding
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, School of Medicine, Shenzhen, Guangdong, China
| | - Yixiang Li
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Benjamin G. Barwick
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Jianping Jenny Ni
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Adeboye O. Osunkoya
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
- Departments of Pathology and Urology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Yuanli Chen
- Key Laboratory of Major Metabolic Diseases and Nutritional Regulation of Anhui Department of Education, School of Food and Biological Engineering, Hefei University of Technology, Hefei, Anhui, China
| | - Wei Zhou
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Siyuan Xia
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, School of Medicine, Shenzhen, Guangdong, China
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Jin-Tang Dong
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, School of Medicine, Shenzhen, Guangdong, China
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, Georgia, USA
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
3
|
Qu X, Xu C, Yang W, Li Q, Tu S, Gao C. KLF5 inhibits the migration and invasion in cervical cancer cell lines by regulating SNAI1. Cancer Biomark 2024; 39:231-243. [PMID: 38217587 PMCID: PMC11191462 DOI: 10.3233/cbm-230175] [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: 05/19/2023] [Accepted: 11/21/2023] [Indexed: 01/15/2024]
Abstract
BACKGROUND Epithelial-mesenchymal transition (EMT) is an important biological process by which malignant tumor cells to acquire migration and invasion abilities. This study explored the role of KLF5 in the EMT process of in cervical cancer cell lines. OBJECTIVE Krüpple-like factor 5 (KLF5) is a basic transcriptional factor that plays a key role in cell-cycle arrest and inhibition of apoptosis. However, the molecular mechanism by which KLF5 mediates the biological functions of cervical cancer cell lines has not been elucidated. Here, we focus on the potential function of ELF5 in regulating the EMT process in in vitro model of cervical cancer cell lines. METHOD Western-blot and real-time quantitative PCR were used to detect the expression of EMT-related genes in HeLa cells. MTT assays, cell scratch and Transwell assays were used to assess HeLa cells proliferation and invasion capability. Using the bioinformatics tool JASPAR, we identified a high-scoring KLF5-like binding sequence in the SNAI1 gene promoter. Luciferase reporter assays was used to detect transcriptional activity for different SNAI1 promoter truncates. RESULT After overexpressing the KLF5 gene in HeLa cells, KLF5 not only significantly inhibited the invasion and migration of HeLa cells, but also increased the expression of E-cadherin and decreased the expression of N-cadherin and MMP9. In addition, the mRNA expression of upstream regulators of E-cadherin, such as SNAI1, SLUG, ZEB1/2 and TWIST1 was also decreased. Furthermore, KLF5 inhibiting the expression of the SNAI1 gene via binding its promoter region, and the EMT of Hela cells was promoted after overexpression of the SNAI1 gene. CONCLUSION These results indicate that KLF5 can downregulate the EMT process of HeLa cells by decreasing the expression of the SNAI1 gene, thereby inhibiting the migration and invasion of HeLa cervical cancer cells.
Collapse
Affiliation(s)
- Xinjian Qu
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of Marine Drugs, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Chang Xu
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of Marine Drugs, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Wenbo Yang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Dalian, Liaoning, China
| | - Qianqian Li
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Dalian, Liaoning, China
| | - Simei Tu
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Dalian, Liaoning, China
| | - Chenghai Gao
- Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
- Guangxi Key Laboratory of Marine Drugs, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| |
Collapse
|
4
|
Wang F, Luo M, Cheng Y. KLF5 promotes esophageal squamous cell cancer through the transcriptional activation of FGFBP1. Med Oncol 2023; 41:17. [PMID: 38087142 PMCID: PMC10716083 DOI: 10.1007/s12032-023-02244-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/19/2023] [Indexed: 12/18/2023]
Abstract
Krüpple-like factor 5 (KLF5) is a zinc-finger-containing transcription factor implicated in several human malignancies, but its potential regulatory mechanisms implicated in esophageal squamous cell carcinoma (ESCC) remain elusive. Here, we show that KLF5 is upregulated in ESCC, where its level was significantly associated with tumor differentiation and lymph node metastasis status. Upregulated KLF5 expression promoted the proliferation, migration, and invasion of ESCC cells. Reduced KLF5 showed the opposite effects. Mechanistically, KLF5 exerts its tumor promotion effect by up-regulating fibroblast growth factor binding protein 1 (FGF-BP1) and snail family transcriptional repressor 2 (SNAIL2). KLF5 binds to the promoter regions of FGF-BP1 and transcriptionally activates its expression. Our study indicated that KLF5 could promote esophageal squamous cell cancer proliferation, migration, and invasion by upregulating FGF-BP1/SNAIL2 signaling. Our work suggests that KLF5 might be a proto-oncogene in ESCC and implicated in ESCC metastasis.
Collapse
Affiliation(s)
- Fengyun Wang
- Department of Oncology, First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science & Technology, Baotou, Inner Mongolia, China
| | - Ming Luo
- Imaging Department, Third Affiliated Hospital of Baotou Medical College, Baotou, Inner Mongolia, China
| | - Yufeng Cheng
- Department of Radiotherapy, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No.107, West Wenhua Road, Lixia District, Jinan, Shandong, China.
| |
Collapse
|
5
|
Tuano NK, Beesley J, Manning M, Shi W, Perlaza-Jimenez L, Malaver-Ortega LF, Paynter JM, Black D, Civitarese A, McCue K, Hatzipantelis A, Hillman K, Kaufmann S, Sivakumaran H, Polo JM, Reddel RR, Band V, French JD, Edwards SL, Powell DR, Chenevix-Trench G, Rosenbluh J. CRISPR screens identify gene targets at breast cancer risk loci. Genome Biol 2023; 24:59. [PMID: 36991492 PMCID: PMC10053147 DOI: 10.1186/s13059-023-02898-w] [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: 09/18/2022] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) have identified > 200 loci associated with breast cancer risk. The majority of candidate causal variants are in non-coding regions and likely modulate cancer risk by regulating gene expression. However, pinpointing the exact target of the association, and identifying the phenotype it mediates, is a major challenge in the interpretation and translation of GWAS. RESULTS Here, we show that pooled CRISPR screens are highly effective at identifying GWAS target genes and defining the cancer phenotypes they mediate. Following CRISPR mediated gene activation or suppression, we measure proliferation in 2D, 3D, and in immune-deficient mice, as well as the effect on DNA repair. We perform 60 CRISPR screens and identify 20 genes predicted with high confidence to be GWAS targets that promote cancer by driving proliferation or modulating the DNA damage response in breast cells. We validate the regulation of a subset of these genes by breast cancer risk variants. CONCLUSIONS We demonstrate that phenotypic CRISPR screens can accurately pinpoint the gene target of a risk locus. In addition to defining gene targets of risk loci associated with increased breast cancer risk, we provide a platform for identifying gene targets and phenotypes mediated by risk variants.
Collapse
Affiliation(s)
- Natasha K Tuano
- Cancer Research Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Jonathan Beesley
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Murray Manning
- Cancer Research Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Functional Genomics Platform, Monash University, Clayton, VIC, Australia
| | - Wei Shi
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Laura Perlaza-Jimenez
- Cancer Research Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Bioinformatics Platform, Monash University, Clayton, VIC, Australia
| | | | - Jacob M Paynter
- Cancer Research Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
| | - Debra Black
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Andrew Civitarese
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Karen McCue
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Aaron Hatzipantelis
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Kristine Hillman
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Susanne Kaufmann
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Haran Sivakumaran
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Jose M Polo
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
| | - Roger R Reddel
- Cancer Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW, Australia
| | - Vimla Band
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, USA
| | - Juliet D French
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Stacey L Edwards
- Cancer Program, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - David R Powell
- Bioinformatics Platform, Monash University, Clayton, VIC, Australia
| | | | - Joseph Rosenbluh
- Cancer Research Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia.
- Functional Genomics Platform, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
6
|
Ali S, Rehman MU, Yatoo AM, Arafah A, Khan A, Rashid S, Majid S, Ali A, Ali MN. TGF-β signaling pathway: Therapeutic targeting and potential for anti-cancer immunity. Eur J Pharmacol 2023; 947:175678. [PMID: 36990262 DOI: 10.1016/j.ejphar.2023.175678] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 03/07/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023]
Abstract
Transforming growth factor-β (TGFβ) is a pleiotropic secretory cytokine exhibiting both cancer-inhibitory and promoting properties. It transmits its signals via Suppressor of Mother against Decapentaplegic (SMAD) and non-SMAD pathways and regulates cell proliferation, differentiation, invasion, migration, and apoptosis. In non-cancer and early-stage cancer cells, TGFβ signaling suppresses cancer progression via inducing apoptosis, cell cycle arrest, or anti-proliferation, and promoting cell differentiation. On the other hand, TGFβ may also act as an oncogene in advanced stages of tumors, wherein it develops immune-suppressive tumor microenvironments and induces the proliferation of cancer cells, invasion, angiogenesis, tumorigenesis, and metastasis. Higher TGFβ expression leads to the instigation and development of cancer. Therefore, suppressing TGFβ signals may present a potential treatment option for inhibiting tumorigenesis and metastasis. Different inhibitory molecules, including ligand traps, anti-sense oligo-nucleotides, small molecule receptor-kinase inhibitors, small molecule inhibitors, and vaccines, have been developed and clinically trialed for blocking the TGFβ signaling pathway. These molecules are not pro-oncogenic response-specific but block all signaling effects induced by TGFβ. Nonetheless, targeting the activation of TGFβ signaling with maximized specificity and minimized toxicity can enhance the efficacy of therapeutic approaches against this signaling pathway. The molecules that are used to target TGFβ are non-cytotoxic to cancer cells but designed to curtail the over-activation of invasion and metastasis driving TGFβ signaling in stromal and cancer cells. Here, we discussed the critical role of TGFβ in tumorigenesis, and metastasis, as well as the outcome and the promising achievement of TGFβ inhibitory molecules in the treatment of cancer.
Collapse
|
7
|
Zhang Y, Yao C, Ju Z, Jiao D, Hu D, Qi L, Liu S, Wu X, Zhao C. Krüppel-like factors in tumors: Key regulators and therapeutic avenues. Front Oncol 2023; 13:1080720. [PMID: 36761967 PMCID: PMC9905823 DOI: 10.3389/fonc.2023.1080720] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
Krüppel-like factors (KLFs) are a group of DNA-binding transcriptional regulators with multiple essential functions in various cellular processes, including proliferation, migration, inflammation, and angiogenesis. The aberrant expression of KLFs is often found in tumor tissues and is essential for tumor development. At the molecular level, KLFs regulate multiple signaling pathways and mediate crosstalk among them. Some KLFs may also be molecular switches for specific biological signals, driving their transition from tumor suppressors to promoters. At the histological level, the abnormal expression of KLFs is closely associated with tumor cell stemness, proliferation, apoptosis, and alterations in the tumor microenvironment. Notably, the role of each KLF in tumors varies according to tumor type and different stages of tumor development rather than being invariant. In this review, we focus on the advances in the molecular biology of KLFs, particularly the regulations of several classical signaling pathways by these factors, and the critical role of KLFs in tumor development. We also highlight their strong potential as molecular targets in tumor therapy and suggest potential directions for clinical translational research.
Collapse
Affiliation(s)
- Yuchen Zhang
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chongjie Yao
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ziyong Ju
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Danli Jiao
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dan Hu
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Qi
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shimin Liu
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Shanghai Research Institute of Acupuncture and Meridian, Shanghai, China
| | - Xueqing Wu
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,*Correspondence: Chen Zhao, ; Xueqing Wu,
| | - Chen Zhao
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China,*Correspondence: Chen Zhao, ; Xueqing Wu,
| |
Collapse
|
8
|
Zakeri S, Aminian H, Sadeghi S, Esmaeilzadeh-Gharehdaghi E, Razmara E. Krüppel-like factors in bone biology. Cell Signal 2022; 93:110308. [PMID: 35301064 DOI: 10.1016/j.cellsig.2022.110308] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 03/07/2022] [Accepted: 03/09/2022] [Indexed: 12/27/2022]
Abstract
The krüppel-like factor (KLF) family is a group of zinc finger transcription factors and contributes to different cellular processes such as differentiation, proliferation, migration, and apoptosis. While different studies show the roles of this family in skeletal development-specifically in chondrocyte and osteocyte development and bone homeostasis-there are few reviews summarizing their importance. To fill this gap, this review discusses current knowledge on different functions of the KLF family during skeletal development, including their roles in stem cell maintenance and differentiation, cell apoptosis, and cell cycle. To understand the importance of the KLF family, we also review genotype-phenotype correlations in different animal models. We also discuss how KLF proteins function through different signaling pathways and display their paramount importance in skeletal development. To highlight their roles in cartilage- or bone-related cells, we also use single-cell RNA sequencing publicly available data on mouse hindlimb. We also challenge our knowledge of how the KLF family is epigenetically regulated-e.g., using DNA methylation, histone modifications, and noncoding RNAs-during chondrocyte and osteocyte development.
Collapse
Affiliation(s)
- Sina Zakeri
- Department of Veterinary Science, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| | - Hesam Aminian
- Department of Biology, Faculty of Sciences, Nour Danesh Institute of Higher Education, Meymeh, Isfahan, Iran
| | - Soheila Sadeghi
- Department of Biology, Faculty of Basic Sciences, Sanandaj Branch, Islamic Azad University, Kurdistan, Iran
| | | | - Ehsan Razmara
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
9
|
The Partial Role of KLF4 and KLF5 in Gastrointestinal Tumors. Gastroenterol Res Pract 2021; 2021:2425356. [PMID: 34367275 PMCID: PMC8337138 DOI: 10.1155/2021/2425356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/08/2021] [Accepted: 07/12/2021] [Indexed: 12/11/2022] Open
Abstract
Background KLF4 and KLF5 are members of the KLF transcription factor family, which play an important role in many gastrointestinal tumors. To gain a deeper insight into its function and role, bioinformatics was used to analyze the function and role of KLF4 and KLF5 in gastrointestinal tumors. Methods Data were collected from several online databases. Gene Expression Profiling Interactive Analysis (GEPIA), UALCAN database analysis, Kaplan-Meier Plotter analysis, LOGpc system, the Pathology Atlas, and the STRING website were used to analyze the data. We download relevant data from TCGA and then perform GO enrichment and KEGG enrichment analysis. The effects of KLF5 on gastric cancer cell proliferation were measured by CCK-8 assay. The effect of KLF5 on the expression of CyclinD1 and MMP9 was detected by Western blot. Results KLF4 and KLF5 were differentially expressed in normal and tumor tissues of the gastrointestinal tract, and their differential expression is related to several genes or pathways. KEGG analysis showed that KLF5 was coexpressed with endocytosis-related genes. KLF5 promotes the proliferation of gastric cancer cells and the expression of metastasis-related molecules. Conclusion KLF4 and KLF5 are of great significance for developing gastrointestinal tumors and can be used as therapeutic targets.
Collapse
|
10
|
Luo Y, Chen C. The roles and regulation of the KLF5 transcription factor in cancers. Cancer Sci 2021; 112:2097-2117. [PMID: 33811715 PMCID: PMC8177779 DOI: 10.1111/cas.14910] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/27/2021] [Accepted: 03/30/2021] [Indexed: 12/11/2022] Open
Abstract
Krüppel‐like factor 5 (KLF5) is a member of the KLF family. Recent studies have suggested that KLF5 regulates the expression of a large number of new target genes and participates in diverse cellular functions, such as stemness, proliferation, apoptosis, autophagy, and migration. In response to multiple signaling pathways, various transcriptional modulation and posttranslational modifications affect the expression level and activity of KLF5. Several transgenic mouse models have revealed the physiological and pathological functions of KLF5 in different cancers. Studies of KLF5 will provide prognostic biomarkers, therapeutic targets, and potential drugs for cancers.
Collapse
Affiliation(s)
- Yao Luo
- Medical Faculty of Kunming University of Science and Technology, Kunming, China.,Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| |
Collapse
|
11
|
Effects of TG interaction factor 1 on synthesis of estradiol and progesterone in granulosa cells of goats through SMAD2/3-SP1 signaling pathway. Anim Reprod Sci 2021; 229:106750. [PMID: 33940561 DOI: 10.1016/j.anireprosci.2021.106750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 12/31/2022]
Abstract
The TG interaction factor 1 (TGIF1) is of the TALE homologue domain protein family and is considered as a transcriptional repressor of SMAD protein that interacts with DNA through a specific consensus-binding site for TG and recruits mSin3A and histone deacetylases to the SMAD complex. In this study, there is the first detailed description of TGIF1 on steroidogenesis in goat granulosa cells. When there is a relatively greater expression of the TGIF1 gene, there is a lesser abundance of CYP11A1, CYP19A1, and StAR mRNA transcript and protein and 3β-HSD mRNA transcript in granulosa cells of goats. Furthermore, there were lesser concentrations of 17β-estradiol (E2) and progesterone (P4) in culture medium when there was greater TGIF1 gene expression and there were greater concentrations of these hormones in the culture medium when there was lesser TGIF1 gene expression. There may be functions of TGIF1, therefore, in suppression of SMAD-induced E2 and P4 production and in decreasing the phosphorylation of SMAD2/3 in granulosa cells of goats and relative abundance of the SMAD2/3 protein transcription factor, SP1. With suppression of TGIF1 gene expression, there was a reversal of SP1-induced suppression of steroidogenesis-related genes. Results of the present study provide insights about the potential mechanism underlying the regulation of granulosa cell steroidogenesis of goats by TGIF1.
Collapse
|
12
|
Xie Z, Chen J, Wang C, Zhang J, Wu Y, Yan X. Current knowledge of Krüppel-like factor 5 and vascular remodeling: providing insights for therapeutic strategies. J Mol Cell Biol 2021; 13:79-90. [PMID: 33493334 PMCID: PMC8104942 DOI: 10.1093/jmcb/mjaa080] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/23/2020] [Accepted: 12/08/2020] [Indexed: 12/18/2022] Open
Abstract
Vascular remodeling is a pathological basis of various disorders. Therefore, it is necessary to understand the occurrence, prevention, and treatment of vascular remodeling. Krüppel-like factor 5 (KLF5) has been identified as a significant factor in cardiovascular diseases during the last two decades. This review provides a mechanism network of function and regulation of KLF5 in vascular remodeling based on newly published data and gives a summary of its potential therapeutic applications. KLF5 modulates numerous biological processes, which play essential parts in the development of vascular remodeling, such as cell proliferation, phenotype switch, extracellular matrix deposition, inflammation, and angiogenesis by altering downstream genes and signaling pathways. Considering its essential functions, KLF5 could be developed as a potent therapeutic target in vascular disorders.
Collapse
Affiliation(s)
- Ziyan Xie
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Junye Chen
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Chenyu Wang
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jiahao Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yanxiang Wu
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xiaowei Yan
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
13
|
Xue VW, Chung JYF, Córdoba CAG, Cheung AHK, Kang W, Lam EWF, Leung KT, To KF, Lan HY, Tang PMK. Transforming Growth Factor-β: A Multifunctional Regulator of Cancer Immunity. Cancers (Basel) 2020. [PMID: 33114183 DOI: 10.3390/cancers12113099.pmid:33114183;pmcid:pmc7690808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
Transforming growth factor-β (TGF-β) was originally identified as an anti-tumour cytokine. However, there is increasing evidence that it has important roles in the tumour microenvironment (TME) in facilitating cancer progression. TGF-β actively shapes the TME via modulating the host immunity. These actions are highly cell-type specific and complicated, involving both canonical and non-canonical pathways. In this review, we systemically update how TGF-β signalling acts as a checkpoint regulator for cancer immunomodulation. A better appreciation of the underlying pathogenic mechanisms at the molecular level can lead to the discovery of novel and more effective therapeutic strategies for cancer.
Collapse
Affiliation(s)
- Vivian Weiwen Xue
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Jeff Yat-Fai Chung
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Cristina Alexandra García Córdoba
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Alvin Ho-Kwan Cheung
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | - Kam-Tong Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong 999077, China
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
14
|
Transforming Growth Factor-β: A Multifunctional Regulator of Cancer Immunity. Cancers (Basel) 2020; 12:cancers12113099. [PMID: 33114183 PMCID: PMC7690808 DOI: 10.3390/cancers12113099] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/12/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Transforming growth factor beta (TGF-β) is a multifunctional cytokine that can restrict cancer onset but also promote cancer progression at late stages of cancer. The ability of TGF-β in producing diverse and sometimes opposing effects relies on its potential to control different cellular signalling and gene expression in distinct cell types, and environmental settings. The tumour promoting role of TGF-β is primarily mediated through its effects on the local tumour microenvironment (TME) of the cancer cells. In this review, we discuss the most recent research on the role and regulation of TGF-β, with a specific focus on its functions on promoting cancer progression through targeting different immune cells in the TME as well as its therapeutic perspectives. Abstract Transforming growth factor-β (TGF-β) was originally identified as an anti-tumour cytokine. However, there is increasing evidence that it has important roles in the tumour microenvironment (TME) in facilitating cancer progression. TGF-β actively shapes the TME via modulating the host immunity. These actions are highly cell-type specific and complicated, involving both canonical and non-canonical pathways. In this review, we systemically update how TGF-β signalling acts as a checkpoint regulator for cancer immunomodulation. A better appreciation of the underlying pathogenic mechanisms at the molecular level can lead to the discovery of novel and more effective therapeutic strategies for cancer.
Collapse
|
15
|
Shan TD, Tian ZB, Li Q, Jiang YP, Liu FG, Sun XG, Han Y, Sun LJ, Chen L. Long intergenic noncoding RNA 00908 promotes proliferation and inhibits apoptosis of colorectal cancer cells by regulating KLF5 expression. J Cell Physiol 2020; 236:889-899. [PMID: 33020901 DOI: 10.1002/jcp.29899] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 06/06/2020] [Accepted: 06/13/2020] [Indexed: 12/14/2022]
Abstract
Long intergenic noncoding RNAs (lincRNAs) play a vital role in the occurrence and progression of cancer. The mechanism of lincRNAs in colorectal cancer (CRC) has not been fully elucidated. In this context, an integrated comparative long noncoding RNA (lncRNA) microarray technology was used to determine the expression profile of lncRNAs in CRC. The roles of LINC00908 are unclear. We found that LINC00908 was significantly upregulated in CRC. Inhibition of LINC00908 resulted in reduced cell proliferation and G1 cell cycle arrest, which was mediated by cyclin D1, cyclin-dependent kinase 4, and phosphorylated retinoblastoma. Moreover, inhibition of LINC00908-induced apoptosis through the intrinsic apoptosis signaling pathway, as shown by the activation of caspase-9 and caspase-3. Mechanistically, miR-143-3p directly bound to LINC00908. miR-143-3p expression was negatively correlated with LINC00908 expression in CRC tissue. Functional experiments revealed opposing roles for miR-143-3p and LINC00908, suggesting that LINC00908 negatively regulates miR-143-3p. Mechanistically, miR-143-3p directly targets LINC00908. The KLF5 inhibitor ML264 affected proliferation and apoptosis, indicating that LINC00908 may act as a competing endogenous RNA to facilitate the expression of the miR-143-3p target gene KLF5. Thus, LINC00908 has an important proliferative and antiapoptotic role in CRC by regulating the cell cycle and intrinsic apoptosis. LINC00908 could be a potential biomarker and a new therapeutic target for CRC.
Collapse
Affiliation(s)
- Ti-Dong Shan
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Zi-Bin Tian
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Qian Li
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Yue-Ping Jiang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Fu-Guo Liu
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Xue-Guo Sun
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Yue Han
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Li-Juan Sun
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| | - Li Chen
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, China
| |
Collapse
|