1
|
Gao H, Wang J, Liu J, Wang H, Wang T, Li S, Niu L, Wei Y. FOXD1 activates KIFC1 to modulate aerobic glycolysis and reinforce cisplatin resistance of breast cancer. Reprod Biol 2024; 25:100969. [PMID: 39541848 DOI: 10.1016/j.repbio.2024.100969] [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: 09/02/2024] [Revised: 10/24/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Breast cancer (BC) is the most prevalent invasive malignant tumor. Cisplatin (DDP) is a prototype of platinum-based chemotherapy drugs, its resistance severely hinders its clinical application. This project intended to figure out the exact mechanism of KIFC1 in the DDP resistance of BC. METHODS The levels of KIFC1 and FOXD1 in BC as well as their binding sites were investigated by bioinformatics analysis. The signaling pathways regulated by FOXD1 were analyzed. Chromatin immunoprecipitation (ChIP) and dual-luciferase reporter assays verified the binding relationship between the two. Through quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blot (WB), we assessed the expression of FOXD1, KIFC1, and glycolysis-related genes. CCK-8 assay was applied in the determination of cell viability to assess the efficacy of DDP resistance. Extracellular acidification rate (ECAR), glucose consumption, lactate synthesis, Adenosine triphosphate (ATP) content, and oxygen consumption rate (OCR) were measured to evaluate glycolysis. RESULTS FOXD1 and KIFC1 were significantly upregulated in BC, with KIFC1 being significantly enriched in the glycolysis pathway. Overexpression of KIFC1 significantly enhanced the DDP resistance of BC cells, while promoting aerobic glycolysis. Mechanistically, FOXD1 was bound to the promoter of KIFC1 to activate its transcription. Its overexpression counteracted the inhibitory effect of KIFC1 knockdown on the DDP resistance of BC cells. CONCLUSION FOXD1 activates the glycolysis pathway by upregulating KIFC1, thereby facilitating BC cells' DDP resistance. Therefore, the FOXD1/KIFC1 axis linked the glycolysis pathway to DDP resistance and may be a promising new target for reinforcing DDP resistance in BC.
Collapse
Affiliation(s)
- Haitao Gao
- General Surgery Department, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Jing Wang
- General Surgery Department, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Jiacai Liu
- General Surgery Department, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Huihua Wang
- General Surgery Department, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Tiantian Wang
- General Surgery Department, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Sha Li
- General Surgery Department, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Lili Niu
- General Surgery Department, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Ya Wei
- General Surgery Department, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China.
| |
Collapse
|
2
|
Tang T, Pei M, Xiao Y, Deng Y, Lu Y, Yu XQ, Wen L, Hu Q. Functional Analysis of Forkhead Transcription Factor Fd59a in the Spermatogenesis of Drosophila melanogaster. INSECTS 2024; 15:480. [PMID: 39057213 PMCID: PMC11277555 DOI: 10.3390/insects15070480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/16/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024]
Abstract
Spermatogenesis is critical for insect reproduction and is regulated by many different genes. In this study, we found that Forkhead transcription factor Fd59a functions as a key factor in the spermatogenesis of Drosophila melanogaster. Fd59a contains a conversed Forkhead domain, and it is clustered to the FoxD subfamily with other FoxD members from some insect and vertebrate species. Mutations in Fd59a caused swelling in the apical region of the testis. More importantly, fewer mature sperm were present in the seminal vesicle of Fd59a mutant flies compared to the control flies, and the fertility of Fd59a2/2 mutant males was significantly lower than that of the control flies. Immunofluorescence staining showed that the homeostasis of the testis stem cell niche in Fd59a2/2 mutant and Fd59a RNAi flies was disrupted and the apoptosis of sperm bundles was increased. Furthermore, results from RNA sequencing and qRT-PCR suggested that Fd59a can regulate the expression of genes related to reproductive process and cell death. Taken together, our results indicated that Fd59a plays a key role in the spermatogenesis of Drosophila.
Collapse
Affiliation(s)
- Ting Tang
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China; (T.T.); (M.P.); (Y.X.); (Y.D.); (Y.L.); (X.-Q.Y.)
| | - Mengyuan Pei
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China; (T.T.); (M.P.); (Y.X.); (Y.D.); (Y.L.); (X.-Q.Y.)
| | - Yanhong Xiao
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China; (T.T.); (M.P.); (Y.X.); (Y.D.); (Y.L.); (X.-Q.Y.)
| | - Yingshan Deng
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China; (T.T.); (M.P.); (Y.X.); (Y.D.); (Y.L.); (X.-Q.Y.)
| | - Yuzhen Lu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China; (T.T.); (M.P.); (Y.X.); (Y.D.); (Y.L.); (X.-Q.Y.)
| | - Xiao-Qiang Yu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China; (T.T.); (M.P.); (Y.X.); (Y.D.); (Y.L.); (X.-Q.Y.)
| | - Liang Wen
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China; (T.T.); (M.P.); (Y.X.); (Y.D.); (Y.L.); (X.-Q.Y.)
- National Key Laboratory of Green Pesticide, College of Plant Protection, South China Agricultural University, Guangzhou 510642, China
| | - Qihao Hu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China; (T.T.); (M.P.); (Y.X.); (Y.D.); (Y.L.); (X.-Q.Y.)
| |
Collapse
|
3
|
Shi ZY, Li CY, Chen RY, Shi JJ, Liu YJ, Lu JF, Yang GJ, Chen J. The emerging role of deubiquitylating enzyme USP21 as a potential therapeutic target in cancer. Bioorg Chem 2024; 147:107400. [PMID: 38688196 DOI: 10.1016/j.bioorg.2024.107400] [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: 03/17/2024] [Revised: 04/15/2024] [Accepted: 04/24/2024] [Indexed: 05/02/2024]
Abstract
Although certain members of the Ubiquitin-specific peptidases (USPs) have been recognized as promising therapeutic targets for various diseases, research progress regarding USP21 has been relatively sluggish in its early stages. USP21 is a crucial member of the USPs subfamily, involved in diverse cellular processes such as apoptosis, DNA repair, and signal transduction. Research findings from the past decade demonstrate that USP21 mediates the deubiquitination of multiple well-known target proteins associated with critical cellular processes relevant to both disease and homeostasis, particularly in various cancers.This reviewcomprehensively summarizes the structure and biological functions of USP21 with an emphasis on its role in tumorigenesis, and elucidates the advances on the discovery of tens of small-molecule inhibitors targeting USP21, which suggests that targeting USP21 may represent a potential strategy for cancer therapy.
Collapse
Affiliation(s)
- Zhen-Yuan Shi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Chang-Yun Li
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Ru-Yi Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Jin-Jin Shi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Yan-Jun Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Jian-Fei Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Guan-Jun Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China.
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, Zhejiang, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
4
|
Liu X, Min S, Zhang Q, Liu Y, Zou Z, Wang N, Zhou B. Prognostic and clinicopathological significance of FOXD1 in various cancers: a meta and bioinformation analysis. Future Sci OA 2024; 10:FSO901. [PMID: 38827805 PMCID: PMC11140636 DOI: 10.2144/fsoa-2023-0085] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/21/2023] [Indexed: 06/05/2024] Open
Abstract
Aim: To examine both predictive and clinicopathological importance underlying FOXD1 in malignant tumors, our study adopts meta-analysis. Methods: We searched from PubMed, Embase, WOS, Wanfang and CNKI. Stata SE15.1 was used to calculate the risk ratio (HR) as well as relative risk (RR) with 95% of overall CIs to assess FOXD1 and overall survival rate (OS), disease-free survival rate as well as clinicopathological parameters. Results: 3808 individuals throughout 17 trials showed high FOXD1 expression was linked to disadvantaged OS (p < 0.001) and disease-free survival (p < 0.001) and higher TNM stage (p < 0.001). Conclusion: Elevated FOXD1 had worse predictions and clinicopathological parameters in most cancers. The GEPIA database findings also support our results.
Collapse
Affiliation(s)
- Xiaohan Liu
- Department of general surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
- Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Shengyun Min
- Department of general surgery, Changzheng Hospital, Nanchang, Jiangxi, 330100, P.R. China
| | - Qin Zhang
- Department of general surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
- Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Yan Liu
- Department of general surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
- Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Zhenhong Zou
- Department of general surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
| | - Nanye Wang
- Department of ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, P.R. China
| | - Bin Zhou
- Department of orthopedics, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
5
|
Tsujimoto H, Hoshina A, Mae SI, Araoka T, Changting W, Ijiri Y, Nakajima-Koyama M, Sakurai S, Okita K, Mizuta K, Niwa A, Saito MK, Saitou M, Yamamoto T, Graneli C, Woollard KJ, Osafune K. Selective induction of human renal interstitial progenitor-like cell lineages from iPSCs reveals development of mesangial and EPO-producing cells. Cell Rep 2024; 43:113602. [PMID: 38237600 DOI: 10.1016/j.celrep.2023.113602] [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: 05/11/2022] [Revised: 06/13/2023] [Accepted: 12/05/2023] [Indexed: 03/02/2024] Open
Abstract
Recent regenerative studies using human pluripotent stem cells (hPSCs) have developed multiple kidney-lineage cells and organoids. However, to further form functional segments of the kidney, interactions of epithelial and interstitial cells are required. Here we describe a selective differentiation of renal interstitial progenitor-like cells (IPLCs) from human induced pluripotent stem cells (hiPSCs) by modifying our previous induction method for nephron progenitor cells (NPCs) and analyzing mouse embryonic interstitial progenitor cell (IPC) development. Our IPLCs combined with hiPSC-derived NPCs and nephric duct cells form nephrogenic niche- and mesangium-like structures in vitro. Furthermore, we successfully induce hiPSC-derived IPLCs to differentiate into mesangial and erythropoietin-producing cell lineages in vitro by screening differentiation-inducing factors and confirm that p38 MAPK, hypoxia, and VEGF signaling pathways are involved in the differentiation of mesangial-lineage cells. These findings indicate that our IPC-lineage induction method contributes to kidney regeneration and developmental research.
Collapse
Affiliation(s)
- Hiraku Tsujimoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Rege Nephro Co., Ltd., Med-Pharm Collaboration Building, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Azusa Hoshina
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Shin-Ichi Mae
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Toshikazu Araoka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Wang Changting
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Yoshihiro Ijiri
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - May Nakajima-Koyama
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Satoko Sakurai
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Kazusa Okita
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Ken Mizuta
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Akira Niwa
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Megumu K Saito
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan
| | - Mitinori Saitou
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan; Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto 606-8501, Japan
| | - Takuya Yamamoto
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan; Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto 606-8501, Japan; Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Kyoto 606-8507, Japan
| | - Cecilia Graneli
- BioPharmaceuticals R&D Cell Therapy, Research and Early Development, Cardiovascular, Renal and Metabolic (CVRM), BioPharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden
| | - Kevin J Woollard
- Bioscience Renal, Research and Early Development, Cardiovascular, Renal and Metabolic, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | - Kenji Osafune
- Center for iPS Cell Research and Application (CiRA), Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto 606-8507, Japan.
| |
Collapse
|
6
|
Cheng W, Cai C, Xu Y, Xiao X, Shi T, Liao Y, Wang X, Chen S, Zhou M, Liao Z. The TRIM21-FOXD1-BCL-2 axis underlies hyperglycaemic cell death and diabetic tissue damage. Cell Death Dis 2023; 14:825. [PMID: 38092733 PMCID: PMC10719266 DOI: 10.1038/s41419-023-06355-1] [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: 12/14/2022] [Revised: 11/26/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023]
Abstract
Chronic hyperglycaemia is a devastating factor that causes diabetes-induced damage to the retina and kidney. However, the precise mechanism by which hyperglycaemia drives apoptotic cell death is incompletely known. Herein, we found that FOXD1, a FOX family transcription factor specifically expressed in the retina and kidney, regulated the transcription of BCL-2, a master regulator of cell survival. Intriguingly, the protein level of FOXD1, which responded negatively to hyperglycaemic conditions, was controlled by the TRIM21-mediated K48-linked polyubiquitination and subsequent proteasomal degradation. The TRIM21-FOXD1-BCL-2 signalling axis was notably active during diabetes-induced damage to murine retinal and renal tissues. Furthermore, we found that tartary buckwheat flavonoids effectively reversed the downregulation of FOXD1 protein expression and thus restored BCL-2 expression and facilitated the survival of retinal and renal tissues. In summary, we identified a transcription factor responsible for BCL-2 expression, a signalling axis (TRM21-FOXD1-BCL-2) underlying hyperglycaemia-triggered apoptosis, and a potential treatment for deleterious diabetic complications.
Collapse
Affiliation(s)
- Wenwen Cheng
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Cifeng Cai
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Yifan Xu
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Xueqi Xiao
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Tiantian Shi
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Yueling Liao
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Xiaoyi Wang
- First Affiliated Hospital of Huzhou University, Huzhou, 313000, China
| | - Shasha Chen
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China.
| | - Meiliang Zhou
- Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Beijing, 100081, China.
| | - Zhiyong Liao
- College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China.
| |
Collapse
|
7
|
Cao S, Wang D, Wang P, Liu Y, Dong W, Ruan X, Liu L, Xue Y, E T, Lin H, Liu X. SUMOylation of RALY promotes vasculogenic mimicry in glioma cells via the FOXD1/DKK1 pathway. Cell Biol Toxicol 2023; 39:3323-3340. [PMID: 37906341 PMCID: PMC10693529 DOI: 10.1007/s10565-023-09836-3] [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: 05/28/2023] [Accepted: 10/11/2023] [Indexed: 11/02/2023]
Abstract
Human malignant gliomas are the most common and aggressive primary malignant tumors of the human central nervous system. Vasculogenic mimicry (VM), which refers to the formation of a tumor blood supply system independently of endothelial cells, contributes to the malignant progression of glioma. Therefore, VM is considered a potential target for glioma therapy. Accumulated evidence indicates that alterations in SUMOylation, a reversible post-translational modification, are involved in tumorigenesis and progression. In the present study, we found that UBA2 and RALY were upregulated in glioma tissues and cell lines. Downregulation of UBA2 and RALY inhibited the migration, invasion, and VM of glioma cells. RALY can be SUMOylated by conjugation with SUMO1, which is facilitated by the overexpression of UBA2. The SUMOylation of RALY increases its stability, which in turn increases its expression as well as its promoting effect on FOXD1 mRNA. The overexpression of FOXD1 promotes DKK1 transcription by activating its promoter, thereby promoting glioma cell migration, invasion, and VM. Remarkably, the combined knockdown of UBA2, RALY, and FOXD1 resulted in the smallest tumor volumes and the longest survivals of nude mice in vivo. UBA2/RALY/FOXD1/DKK1 axis may play crucial roles in regulating VM in glioma, which may contribute to the development of potential strategies for the treatment of gliomas.
Collapse
Affiliation(s)
- Shuo Cao
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning Province, China
| | - Di Wang
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China
| | - Ping Wang
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
| | - Yunhui Liu
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China
| | - Weiwei Dong
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China
| | - Xuelei Ruan
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
| | - Libo Liu
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
| | - Yixue Xue
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurobiology, School of Life Sciences, China Medical University, Shenyang, 110122, China
| | - Tiange E
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China
| | - Hongda Lin
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China
| | - Xiaobai Liu
- Key Laboratory of Neuro-Oncology in Liaoning Province, Shenyang, 110004, China.
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
- Liaoning Medical Surgery and Rehabilitation Robot Technology Engineering Research Center, Shenyang, 110004, China.
| |
Collapse
|
8
|
Huang Y, Zhang L, Liu T, Liang E. LMNB1 targets FOXD1 to promote progression of prostate cancer. Exp Ther Med 2023; 26:513. [PMID: 37840569 PMCID: PMC10570766 DOI: 10.3892/etm.2023.12212] [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: 09/02/2022] [Accepted: 06/09/2023] [Indexed: 10/17/2023] Open
Abstract
Forkhead box D1 (FOXD1) expression is upregulated in various types of human cancer. To the best of our knowledge, the roles of FOXD1 in prostate cancer (PC) remain largely unknown. The Cancer Genome Atlas dataset was used for the bioinformatics analysis of FOXD1 in PC. FOXD1 expression levels in normal immortalized human prostate epithelial cells (RWPE-1) and prostate cancer cells were detected by reverse transcription-quantitative PCR. PC cell viability was detected using Cell Counting Kit-8 assay. Transwell assays were performed to assess the migration and invasion of PC cells. Luciferase reporter gene assay was used to validate the association between FOXD1 and lamin (LMN)B1. LMNB1 is an important part of the cytoskeleton, which serves an important role in the process of tumor occurrence and development, regulating apoptosis and DNA repair. FOXD1 expression was upregulated in PC tissues, with its high expression being associated with clinical stage and survival in PC. Knockdown of FOXD1 inhibited viability, migration and invasion of PC cells. FOXD1 positively regulated LMNB1 expression. The effect of FOXD1 knockdown on PC cells was reversed by LMNB1 overexpression. In conclusion, FOXD1, positively regulated by LMNB1, served as an oncogene in PC and may be a potential biomarker and treatment target for PC.
Collapse
Affiliation(s)
- Yuanshe Huang
- Agriculture College, Innovation Center for Efficient Agricultural of Guizhou Mountain Characteristics, Anshun University, Anshun, Guizhou 561000, P.R. China
| | - Lai Zhang
- Agriculture College, Innovation Center for Efficient Agricultural of Guizhou Mountain Characteristics, Anshun University, Anshun, Guizhou 561000, P.R. China
| | - Tianlei Liu
- Agriculture College, Innovation Center for Efficient Agricultural of Guizhou Mountain Characteristics, Anshun University, Anshun, Guizhou 561000, P.R. China
| | - E Liang
- Agriculture College, Innovation Center for Efficient Agricultural of Guizhou Mountain Characteristics, Anshun University, Anshun, Guizhou 561000, P.R. China
| |
Collapse
|
9
|
Cheng L, Yan H, Liu Y, Guan G, Cheng P. Dissecting multifunctional roles of forkhead box transcription factor D1 in cancers. Biochim Biophys Acta Rev Cancer 2023; 1878:188986. [PMID: 37716516 DOI: 10.1016/j.bbcan.2023.188986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/18/2023]
Abstract
As a member of the forkhead box (FOX) family of transcription factors (TF), FOXD1 has recently been implicated as a crucial regulator in a variety of human cancers. Accumulating evidence has established dysregulated and aberrant FOXD1 signaling as a prominent feature in cancer development and progression. However, there is a lack of systematic review on this topic. Here, we summarized the present understanding of FOXD1 functions in cancer biology and reviewed the downstream targets and upstream regulatory mechanisms of FOXD1 as well as the related signaling pathways within the context of current reports. We highlighted the functional features of FOXD1 in cancers to identify the future research consideration of this multifunctional transcription factor and potential therapeutic strategies targeting its oncogenic activity.
Collapse
Affiliation(s)
- Lin Cheng
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Haixu Yan
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yang Liu
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China
| | - Gefei Guan
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China.
| | - Peng Cheng
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, China; Institute of Health Sciences, China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
10
|
Luo Y, Ni R, Jin X, Feng P, Dai C, Jiang L, Chen P, Yang L, Zhu Y. FOXD1 expression-based prognostic model for uveal melanoma. Heliyon 2023; 9:e21333. [PMID: 38027647 PMCID: PMC10651470 DOI: 10.1016/j.heliyon.2023.e21333] [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: 03/30/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 12/01/2023] Open
Abstract
FOXD1, a new member of the FOX transcription factor family, serves as a mediator and biomarker for cell reprogramming. But its contribution to prognosis of uveal melanoma (UVM) is unclear. This study demonstrated that FOXD1 might promote tumor growth and invasion, because FOXD1 expression was negatively correlated with overall survival, progression-free survival, and disease-specific survival in UVM patients. This conjecture was verified in cell culture with human uveal melanoma cell line (MUM2B) as model cells. Additionally, the biological mechanisms of FOXD1 based on FOXD1-related genomic spectrum, molecular pathways, tumor microenvironment, and drug treatment sensitivity were examined using The Cancer Genome Atlas (TCGA) database, aiming to reasonably explain why FOXD1 leads to poor prognosis of UVM. On these bases, a novel tumor prognostic model was established using the FOXD1-related immunomodulators TMEM173, TNFRSF4, TNFSF13, and ULBP1, which will enable the stratification of disease seriousness and clinical treatment for patients.
Collapse
Affiliation(s)
- Yang Luo
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Renhao Ni
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Xiaojun Jin
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Peipei Feng
- Ningbo Institute of Innovation for Combined Medicine and Engineering, Ningbo Medical Centre Lihuili Hospital, Ningbo, 315000, China
| | - Chenyi Dai
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Lingjing Jiang
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | | | - Lu Yang
- The First Affiliated Hospital of Ningbo University, Ningbo, 315010, China
| | - Yabin Zhu
- Health Science Center, Ningbo University, Ningbo, 315211, China
| |
Collapse
|
11
|
Wang Q, Fan X, Sheng Q, Yang M, Zhou P, Lu S, Gao Y, Kong Z, Shen N, Lv Z, Wang R. N6-methyladenosine methylation in kidney injury. Clin Epigenetics 2023; 15:170. [PMID: 37865763 PMCID: PMC10590532 DOI: 10.1186/s13148-023-01586-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/11/2023] [Indexed: 10/23/2023] Open
Abstract
Multiple mechanisms are involved in kidney damage, among which the role of epigenetic modifications in the occurrence and development of kidney diseases is constantly being revealed. However, N6-methyladenosine (M6A), a well-known post-transcriptional modification, has been regarded as the most prevalent epigenetic modifications in higher eukaryotic, which is involved in various biological processes of cells such as maintaining the stability of mRNA. The role of M6A modification in the mechanism of kidney damage has attracted widespread attention. In this review, we mainly summarize the role of M6A modification in the progression of kidney diseases from the following aspects: the regulatory pattern of N6-methyladenosine, the critical roles of N6-methyladenosine in chronic kidney disease, acute kidney injury and renal cell carcinoma, and then reveal its potential significance in the diagnosis and treatment of various kidney diseases. A better understanding of this field will be helpful for future research and clinical treatment of kidney diseases.
Collapse
Affiliation(s)
- Qimeng Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Xiaoting Fan
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Qinghao Sheng
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Meilin Yang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Ping Zhou
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Shangwei Lu
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Ying Gao
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Zhijuan Kong
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Ning Shen
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Zhimei Lv
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
| | - Rong Wang
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
| |
Collapse
|
12
|
You L, Han Z, Chen H, Chen L, Lin Y, Wang B, Fan Y, Zhang M, Luo J, Peng F, Ma Y, Wang Y, Yuan L, Han Z. The role of N6-methyladenosine (m 6A) in kidney diseases. Front Med (Lausanne) 2023; 10:1247690. [PMID: 37841018 PMCID: PMC10569431 DOI: 10.3389/fmed.2023.1247690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/01/2023] [Indexed: 10/17/2023] Open
Abstract
Chemical modifications are a specific and efficient way to regulate the function of biological macromolecules. Among them, RNA molecules exhibit a variety of modifications that play important regulatory roles in various biological processes. More than 170 modifications have been identified in RNA molecules, among which the most common internal modifications include N6-methyladenine (m6A), n1-methyladenosine (m1A), 5-methylcytosine (m5C), and 7-methylguanine nucleotide (m7G). The most widely affected RNA modification is m6A, whose writers, readers, and erasers all have regulatory effects on RNA localization, splicing, translation, and degradation. These functions, in turn, affect RNA functionality and disease development. RNA modifications, especially m6A, play a unique role in renal cell carcinoma disease. In this manuscript, we will focus on the biological roles of m6A in renal diseases such as acute kidney injury, chronic kidney disease, lupus nephritis, diabetic kidney disease, and renal cancer.
Collapse
Affiliation(s)
- Luling You
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhongyu Han
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Haoran Chen
- Science and Education Department, Chengdu Xinhua Hospital, Chengdu, China
| | - Liuyan Chen
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yumeng Lin
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Binjian Wang
- Eye School of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yiyue Fan
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Meiqi Zhang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ji Luo
- School of Medical Information Engineering, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fang Peng
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yue Ma
- School of Clinical Medicine, Southeast University, Nanjing, China
| | - Yanmei Wang
- Institute of Traditional Chinese Medicine, Sichuan College of Traditional Chinese Medicine (Sichuan Second Hospital of TCM), Chengdu, China
| | - Lan Yuan
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhongyu Han
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
13
|
Taghehchian N, Lotfi M, Zangouei AS, Akhlaghipour I, Moghbeli M. MicroRNAs as the critical regulators of Forkhead box protein family during gynecological and breast tumor progression and metastasis. Eur J Med Res 2023; 28:330. [PMID: 37689738 PMCID: PMC10492305 DOI: 10.1186/s40001-023-01329-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 08/29/2023] [Indexed: 09/11/2023] Open
Abstract
Gynecological and breast tumors are one of the main causes of cancer-related mortalities among women. Despite recent advances in diagnostic and therapeutic methods, tumor relapse is observed in a high percentage of these patients due to the treatment failure. Late diagnosis in advanced tumor stages is one of the main reasons for the treatment failure and recurrence in these tumors. Therefore, it is necessary to assess the molecular mechanisms involved in progression of these tumors to introduce the efficient early diagnostic markers. Fokhead Box (FOX) is a family of transcription factors with a key role in regulation of a wide variety of cellular mechanisms. Deregulation of FOX proteins has been observed in different cancers. MicroRNAs (miRNAs) as a group of non-coding RNAs have important roles in post-transcriptional regulation of the genes involved in cellular mechanisms. They are also the non-invasive diagnostic markers due to their high stability in body fluids. Considering the importance of FOX proteins in the progression of breast and gynecological tumors, we investigated the role of miRNAs in regulation of the FOX proteins in these tumors. MicroRNAs were mainly involved in progression of these tumors through FOXM, FOXP, and FOXO. The present review paves the way to suggest a non-invasive diagnostic panel marker based on the miRNAs/FOX axis in breast and gynecological cancers.
Collapse
Affiliation(s)
- Negin Taghehchian
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Malihe Lotfi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Sadra Zangouei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
14
|
Jiang W, Li Y, Li R, Chen W, Song M, Zhang Q, Chen S. The Prognostic Significance of FOXD1 Expression in Head and Neck Squamous Cell Carcinoma. J Pers Med 2023; 13:530. [PMID: 36983712 PMCID: PMC10053205 DOI: 10.3390/jpm13030530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 03/09/2023] [Accepted: 03/10/2023] [Indexed: 03/18/2023] Open
Abstract
It has been reported that forkhead box D1 (FOXD1) plays an established role in human early embryonic development and is broadly involved in various malignancies. However, there is limited information regarding FOXD1 expression in head and neck squamous cell carcinoma (HNSCC). This present study aimed to explore the clinical significance of FOXD1 in patients with HNSCC. Tissue microarrays of 334 primary HNSCC patients who underwent surgery between 2008 and 2010 at Sun Yat-sen University Cancer Center were investigated by immunohistochemistry regarding FOXD1 expression. χ2 test was used to estimate the relationship of FOXD1 expression with clinicopathologic characteristics. Univariate and multivariate analyses were performed to identify FOXD1 expression as an independent prognostic indicator of overall survival (OS) and disease-free survival (DFS). FOXD1 expression is closely associated with postoperative recurrence. HNSCC patients with high FOXD1 expression have poorer prognoses than the low-expression group (p < 0.05). According to multivariate analysis, FOXD1 was an independent prognostic factor for OS and DFS. The results revealed that FOXD1 could be a prognostic factor for HNSCC and might serve as a potential target for novel therapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Quan Zhang
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China (M.S.)
| | - Shuwei Chen
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China (M.S.)
| |
Collapse
|
15
|
Mun JY, Baek SW, Jeong MS, Jang IH, Lee SR, You JY, Kim JA, Yang GE, Choi YH, Kim TN, Chu IS, Leem SH. Stepwise molecular mechanisms responsible for chemoresistance in bladder cancer cells. Cell Death Dis 2022; 8:450. [DOI: 10.1038/s41420-022-01242-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 11/09/2022]
Abstract
AbstractChemotherapy resistance is an obstacle to cancer therapy and is considered a major cause of recurrence. Thus, understanding the mechanisms of chemoresistance is critical to improving the prognosis of patients. Here, we have established a stepwise gemcitabine-resistant T24 bladder cancer cell line to understand the molecular mechanisms of chemoresistance within cancer cells. The characteristics of the stepwise chemoresistance cell line were divided into 4 phases (parental, early, intermediate, and late phases). These four phase cells showed increasingly aggressive phenotypes in vitro and in vivo experiments with increasing phases and revealed the molecular properties of the biological process from parent cells to phased gemcitabine-resistant cell line (GRC). Taken together, through the analysis of gene expression profile data, we have characterized gene set of each phase indicating the response to anticancer drug treatment. Specifically, we identified a multigene signature (23 genes including GATA3, APOBEC3G, NT5E, MYC, STC1, FOXD1, SMAD9) and developed a chemoresistance score consisting of that could predict eventual responsiveness to gemcitabine treatment. Our data will contribute to predicting chemoresistance and improving the prognosis of bladder cancer patients.
Collapse
|
16
|
FOXD1 facilitates pancreatic cancer cell proliferation, invasion, and metastasis by regulating GLUT1-mediated aerobic glycolysis. Cell Death Dis 2022; 13:765. [PMID: 36057597 PMCID: PMC9440910 DOI: 10.1038/s41419-022-05213-w] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/19/2022] [Accepted: 08/23/2022] [Indexed: 01/21/2023]
Abstract
Although FOXD1 has been found to be involved in the malignant processes of several types of cancers, its role in pancreatic cancer (PC) is not well understood. This study aimed to investigate the expression and function of FOXD1 in PC. We found that FOXD1 mRNA and protein expression were upregulated in PC tissues compared with non-tumor tissues, and high expression level of FOXD1 was associated with an adverse prognostic index of PC. The results of in vitro and in vivo assays indicate that overexpression of FOXD1 promotes aerobic glycolysis and the capacity of PC cells to proliferate, invade, and metastasize, whereas FOXD1 knockdown inhibits these functions. The results of mechanistic experiments suggest that FOXD1 can not only directly promote SLC2A1 transcription but also inhibit the degradation of SLC2A1 through the RNA-induced silencing complex. As a result, FOXD1 enhances GLUT1 expression and ultimately facilitates PC cell proliferation, invasion, and metastasis by regulating aerobic glycolysis. Taken together, FOXD1 is suggested to be a potential therapeutic target for PC.
Collapse
|
17
|
Zhang Q, Chen Z, Tang Q, Wang Z, Lu J, You Y, Wang H. USP21 promotes self-renewal and tumorigenicity of mesenchymal glioblastoma stem cells by deubiquitinating and stabilizing FOXD1. Cell Death Dis 2022; 13:712. [PMID: 35974001 PMCID: PMC9381540 DOI: 10.1038/s41419-022-05163-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 07/09/2022] [Accepted: 08/04/2022] [Indexed: 01/21/2023]
Abstract
Recent studies suggest that Forkhead box D1 (FOXD1) plays an indispensable role in maintaining the mesenchymal (MES) properties of glioblastoma (GBM) stem cells (GSCs). Thus, understanding the mechanisms that control FOXD1 protein expression is critical for guiding GBM treatment, particularly in patients with therapy-resistant MES subtypes. In this study, we identify the ubiquitin-specific peptidase 21 (USP21) as a critical FOXD1 deubiquitinase in MES GSCs. We find that USP21 directly interacts with and stabilizes FOXD1 by reverting its proteolytic ubiquitination. Silencing of USP21 enhances polyubiquitination of FOXD1, promotes its proteasomal degradation, and ultimately attenuates MES identity in GSCs, while these effects could be largely restored by reintroduction of FOXD1. Remarkably, we show that disulfiram, a repurposed drug that could block the enzymatic activities of USP21, suppresses GSC tumorigenicity in MES GSC-derived GBM xenograft model. Additionally, we demonstrate that USP21 is overexpressed and positively correlated with FOXD1 protein levels in GBM tissues, and its expression is inversely correlated with patient survival. Collectively, our work reveals that USP21 maintains MES identity by antagonizing FOXD1 ubiquitination and degradation, suggesting that USP21 is a potential therapeutic target for the MES subtype of GBM.
Collapse
Affiliation(s)
- Qixiang Zhang
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Zhengxin Chen
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Qikai Tang
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Zhangjie Wang
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Jiacheng Lu
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Yongping You
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| | - Huibo Wang
- grid.412676.00000 0004 1799 0784Department of Neurosurgery, First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029 China
| |
Collapse
|
18
|
Minemura C, Asai S, Koma A, Kikkawa N, Kato M, Kasamatsu A, Uzawa K, Hanazawa T, Seki N. Identification of Antitumor miR-30e-5p Controlled Genes; Diagnostic and Prognostic Biomarkers for Head and Neck Squamous Cell Carcinoma. Genes (Basel) 2022; 13:genes13071225. [PMID: 35886008 PMCID: PMC9322981 DOI: 10.3390/genes13071225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 01/27/2023] Open
Abstract
Analysis of microRNA (miRNA) expression signatures in head and neck squamous cell carcinoma (HNSCC) has revealed that the miR-30 family is frequently downregulated in cancer tissues. The Cancer Genome Atlas (TCGA) database confirms that all members of the miR-30 family (except miR-30c-5p) are downregulated in HNSCC tissues. Moreover, low expression of miR-30e-5p and miR-30c-1-3p significantly predicts shorter survival of HNSCC patients (p = 0.0081 and p = 0.0224, respectively). In this study, we focused on miR-30e-5p to investigate its tumor-suppressive roles and its control of oncogenic genes in HNSCC cells. Transient expression of miR-30e-5p significantly attenuated cancer cell migration and invasive abilities in HNSCC cells. Nine genes (DDIT4, FOXD1, FXR1, FZD2, HMGB3, MINPP1, PAWR, PFN2, and RTN4R) were identified as putative targets of miR-30e-5p control. Their expression levels significantly predicted shorter survival of HNSCC patients (p < 0.05). Among those targets, FOXD1 expression appeared to be an independent factor predicting patient survival according to multivariate Cox regression analysis (p = 0.049). Knockdown assays using siRNAs corresponding to FOXD1 showed that malignant phenotypes (e.g., cell proliferation, migration, and invasive abilities) of HNSCC cells were significantly suppressed. Overexpression of FOXD1 was confirmed by immunostaining of HNSCC clinical specimens. Our miRNA-based approach is an effective strategy for the identification of prognostic markers and therapeutic target molecules in HNSCC. Moreover, these findings led to insights into the molecular pathogenesis of HNSCC.
Collapse
Affiliation(s)
- Chikashi Minemura
- Department of Oral Science, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (C.M.); (A.K.); (A.K.); (K.U.)
| | - Shunichi Asai
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; (S.A.); (N.K.); (M.K.)
- Department of Otorhinolaryngology/Head and Neck Surgery, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan;
| | - Ayaka Koma
- Department of Oral Science, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (C.M.); (A.K.); (A.K.); (K.U.)
| | - Naoko Kikkawa
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; (S.A.); (N.K.); (M.K.)
- Department of Otorhinolaryngology/Head and Neck Surgery, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan;
| | - Mayuko Kato
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; (S.A.); (N.K.); (M.K.)
| | - Atsushi Kasamatsu
- Department of Oral Science, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (C.M.); (A.K.); (A.K.); (K.U.)
| | - Katsuhiro Uzawa
- Department of Oral Science, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; (C.M.); (A.K.); (A.K.); (K.U.)
| | - Toyoyuki Hanazawa
- Department of Otorhinolaryngology/Head and Neck Surgery, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan;
| | - Naohiko Seki
- Department of Functional Genomics, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan; (S.A.); (N.K.); (M.K.)
- Correspondence: ; Tel.: +81-43-226-2971; Fax: +81-43-227-3442
| |
Collapse
|
19
|
Hashem Boroojerdi M, Hosseinpour Sarmadi V, Maqbool M, Ling KH, Safarzadeh Kozani P, Safarzadeh Kozani P, Ramasamy R. Directional capacity of human mesenchymal stem cells to support hematopoietic stem cell proliferation in vitro. Gene 2022; 820:146218. [PMID: 35134469 DOI: 10.1016/j.gene.2022.146218] [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: 08/04/2021] [Revised: 12/16/2021] [Accepted: 01/13/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Hematopoietic stem cells (HSCs) reside in a specialised microenvironment in the bone marrow, which is majorly composed of mesenchymal stem cells (MSCs) and its' derivatives. This study aimed to investigate the regulatory role of MSCs to decipher the cellular and humoral communications on HSCs' proliferation, self-renewal, and differentiation at the transcriptomic level. MATERIALS AND METHODS Microarray assay was employed to analyse the gene expression profile of HSCs that imparted by MSCs during co-culture. RESULTS The proliferation of human umbilical cord blood-derived HSCs (hUC-HSCs) markedly propagated when MSCs were used as the feeder layer, without disturbing the undifferentiated state of HSCs, and reduced the cell death of HSCs. Upon co-culture with MSCs, the global microarray analysis of HSCs disclosed 712 differentially expressed genes (DEGs) (561 up-regulated and 151 down-regulated). The dysregulations of various transcripts were enriched for cellular functions such as cell cycle (including CCND1), apoptosis (including TNF), and genes related to signalling pathways governing self-renewal, as well as WNT5A from the Wnt signalling pathway, MAPK, Hedgehog, FGF2 from FGF, Jak-STAT, and PITX2 from the TGF-β signalling pathway. To concur this, real-time quantitative PCR (RT-qPCR) was utilised for corroborating the microarray results from five of the most dysregulated genes. CONCLUSION This study elucidates the underlying mechanisms of the mitogenic influences of MSCs on the propagation of HSCs. The exploitation of such mechanisms provides a potential means for achieving larger quantities of HSCs in vitro, thus obviating the need for manipulating their differentiation potential for clinical application.
Collapse
Affiliation(s)
- Mohadese Hashem Boroojerdi
- Stem Cell & Immunity Research Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine and Health Science, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia; Genetics and Regenerative Medicine Research Centre, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Vahid Hosseinpour Sarmadi
- Institute of Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Science, Tehran, Iran; Cellular and Molecular Research Center, Iran University of Medical Science, Tehran, Iran
| | - Maryam Maqbool
- Stem Cell & Immunity Research Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine and Health Science, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia; Genetics and Regenerative Medicine Research Centre, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - King-Hwa Ling
- Genetics and Regenerative Medicine Research Centre, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia; Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | - Pooria Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Pouya Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Paramedicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Rajesh Ramasamy
- Stem Cell & Immunity Research Group, Immunology Laboratory, Department of Pathology, Faculty of Medicine and Health Science, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia; Genetics and Regenerative Medicine Research Centre, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia; Department of Dental Radiology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya 60132, Indonesia.
| |
Collapse
|
20
|
Ma Q, Yang T. E2F transcription factor 1/small nucleolar RNA host gene 18/microRNA-338-5p/forkhead box D1: an important regulatory axis in glioma progression. Bioengineered 2021; 13:418-430. [PMID: 34937497 PMCID: PMC8805867 DOI: 10.1080/21655979.2021.2005990] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
This study aims to probe the biological functions of long non-coding RNA small nucleolar RNA host gene 18 (SNHG18) on glioma cells and its underlying mechanism. In this study, SNHG18 expression in glioma tissues was quantified employing GEPIA database; quantitative real-time PCR was adopted to examine the expressions of SNHG18, microRNA-338-5p (miR-338-5p) and forkhead box D1 (FOXD1) mRNA in glioma tissues and cell lines; cell proliferation, migration and invasion were detected utilizing cell counting kit-8, EdU and Transwell assays; Western blot was utilized to quantify the protein expressions of E-cadherin, N-cadherin, Vimentin and FOXD1; dual-luciferase reporter gene and RNA immunoprecipitation experiments were utilized to validate the targeting relationships between SNHG18 and miR-338-5p, as well as miR-338-5p and FOXD1 mRNA 3ʹUTR; dual-luciferase reporter gene and chromatin immunoprecipitation assays were utilized to verify the binding of E2F transcription factor 1 (E2F1) to the SNHG18 promoter region. It was revealed that, SNHG18 expression in glioma was up-regulated and associated with unfavorable prognosis of the patients; knockdown of SNHG18 repressed the malignant biological behaviors of glioma cells, enhanced E-cadherin expression and repressed N-cadherin and Vimentin expressions. MiR-338-5p was a target of SNHG18, and SNHG18 promoted the expression of FOXD1 by decoying miR-338-5p. Additionally, E2F1 could bind to the promoter of SNHG18 to elevate its expression. In conclusion, SNHG18 accelerates glioma progression via regulating the miR-338-5p/FOXD1 axis.
Collapse
Affiliation(s)
- Quanfeng Ma
- Department of Neurosurg, Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurg Institution, Tianjin China
| | - Tianhao Yang
- Department of Radiology, Tianjin Huanhu Hospital, Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin China
| |
Collapse
|
21
|
Fang G, Fan J, Ding Z, Li R, Lin K, Fu J, Huang Q, Zeng Y, Liu J. Prognostic and Predictive Value of Transcription Factors Panel for Digestive System Carcinoma. Front Oncol 2021; 11:670129. [PMID: 34745933 PMCID: PMC8566925 DOI: 10.3389/fonc.2021.670129] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Accepted: 09/24/2021] [Indexed: 12/23/2022] Open
Abstract
Purpose Digestive system carcinoma is one of the most devastating diseases worldwide. Lack of valid clinicopathological parameters as prognostic factors needs more accurate and effective biomarkers for high-confidence prognosis that guide decision-making for optimal treatment of digestive system carcinoma. The aim of the present study was to establish a novel model to improve prognosis prediction of digestive system carcinoma, with a particular interest in transcription factors (TFs). Materials and Methods A TF-related prognosis model of digestive system carcinoma with data from TCGA database successively were processed by univariate and multivariate Cox regression analyses. Then, for evaluating the prognostic prediction value of the model, ROC curve and survival analysis were performed by external data from GEO database. Furthermore, we verified the expression of TFs expression by qPCR in digestive system carcinoma tissue. Finally, we constructed a TF clinical characteristics nomogram to furtherly predict digestive system carcinoma patient survival probability with TCGA database. Results By Cox regression analysis, a panel of 17 TFs (NFIC, YBX2, ZBTB47, ZNF367, CREB3L3, HEYL, FOXD1, TIGD1, SNAI1, HSF4, CENPA, ETS2, FOXM1, ETV4, MYBL2, FOXQ1, ZNF589) was identified to present with powerful predictive performance for overall survival of digestive system carcinoma patients based on TCGA database. A nomogram that integrates TFs was established, allowing efficient prediction of survival probabilities and displaying higher clinical utility. Conclusion The 17-TF panel is an independent prognostic factor for digestive system carcinoma, and 17 TFs based nomogram might provide implication an effective approach for digestive system carcinoma patient management and treatment.
Collapse
Affiliation(s)
- Guoxu Fang
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Big Data Institute of Southeast Hepatobiliary Health Information, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Jianhui Fan
- Department of Hepatology for Pregnancy, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Zongren Ding
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Big Data Institute of Southeast Hepatobiliary Health Information, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Rong Li
- Department of Pathology, Eastern Hepatobiliary Surgery Hospital of Naval Medical University, Shanghai, China
| | - Kongying Lin
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Big Data Institute of Southeast Hepatobiliary Health Information, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Jun Fu
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Big Data Institute of Southeast Hepatobiliary Health Information, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Qizhen Huang
- The Big Data Institute of Southeast Hepatobiliary Health Information, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,Department of Radiation Oncology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Yongyi Zeng
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Jingfeng Liu
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,The Big Data Institute of Southeast Hepatobiliary Health Information, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China.,Department of Hepatopancreatobiliary Surgery, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, China
| |
Collapse
|
22
|
Chen Y, Liang W, Liu K, Shang Z. FOXD1 promotes EMT and cell stemness of oral squamous cell carcinoma by transcriptional activation of SNAI2. Cell Biosci 2021; 11:154. [PMID: 34348789 PMCID: PMC8335989 DOI: 10.1186/s13578-021-00671-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/26/2021] [Indexed: 12/15/2022] Open
Abstract
Background Epithelial-mesenchymal transition (EMT) and cell stemness are implicated in the initiation and progression of oral squamous cell carcinoma (OSCC). Revealing the intrinsic regulatory mechanism may provide effective therapeutic targets for OSCC. Results In this study, we found that Forkhead box D1 (FOXD1) was upregulated in OSCC compared with normal samples. Patients with a higher FOXD1 expression had a poorer overall survival and disease-free survival. Immunohistochemical staining results showed that FOXD1 expression was related to the clinical stage and relapse status of OSCC patients. When FOXD1 expression was knocked down in CAL27 and SCC25 cells, the migration, invasion, colony formation, sphere formation, and proliferation abilities decreased. Moreover, EMT and stemness-related markers changed remarkably, which indicated that the EMT process and cell stemness were inhibited. Conversely, overexpression of FOXD1 promoted EMT and cell stemness. Further study demonstrated that FOXD1 could bind to the promoter region and activate the transcription of SNAI2. In turn, the elevated SNAI2 affected EMT and cell stemness. An in vivo study showed that FOXD1-overexpressing CAL27 cells possessed a stronger tumorigenic ability. Conclusions Our findings revealed a novel mechanism in regulating EMT and cell stemness and proposed FOXD1 as a potential marker for the diagnosis and treatment of OSCC. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00671-9.
Collapse
Affiliation(s)
- Yang Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology, Hubei Province and Key Laboratory of Oral Biomedicine (Wuhan University), Ministry of Education (Hubei-MOST KLOS & KLOBM), Wuhan, China
| | - Weilian Liang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology, Hubei Province and Key Laboratory of Oral Biomedicine (Wuhan University), Ministry of Education (Hubei-MOST KLOS & KLOBM), Wuhan, China
| | - Ke Liu
- Department of Oral and Maxillofacial-Head and Neck Oncology, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Hongshan District, Wuhan, 430079, China.
| | - Zhengjun Shang
- Department of Oral and Maxillofacial-Head and Neck Oncology, School and Hospital of Stomatology, Wuhan University, 237 Luoyu Road, Hongshan District, Wuhan, 430079, China.
| |
Collapse
|
23
|
Determination of the dynamic cellular transcriptional profiles during kidney development from birth to maturity in rats by single-cell RNA sequencing. Cell Death Discov 2021; 7:162. [PMID: 34226524 PMCID: PMC8257621 DOI: 10.1038/s41420-021-00542-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 04/27/2021] [Accepted: 06/03/2021] [Indexed: 01/02/2023] Open
Abstract
Recent single-cell RNA sequencing (scRNA-seq) analyses have offered much insight into the gene expression profiles in early-stage kidney development. However, comprehensive gene expression profiles from mid- and late-stage kidney development are lacking. In the present study, by using the scRNA-seq technique, we analyzed 54,704 rat kidney cells from just after birth to adulthood (six time points: postnatal days 0, 2, 5, 10, 20, and 56) including the mid and late stages of kidney development. Twenty-five original clusters and 13 different cell types were identified during these stages. Gene expression in these 13 cell types was mapped, and single cell atlas of the rat kidney from birth to maturity ( http://youngbearlab.com ) was built to enable users to search for a gene of interest and to evaluate its expression in different cells. The variation trend of six major types of kidney cells-intercalated cells of the collecting duct (CD-ICs), principal cells of the collecting duct (CD-PCs), cells of the distal convoluted tubules (DCTs), cells of the loop of Henle (LOH), podocytes (PDs), and cells of the proximal tubules (PTs)-during six postnatal time points was demonstrated. The trajectory of rat kidney development and the order of induction of the six major types of kidney cells from just after birth to maturity were determined. In addition, features of the dynamically changing genes as well as transcription factors during postnatal rat kidney development were identified. The present study provides a resource for achieving a deep understanding of the molecular basis of and regulatory events in the mid and late stages of kidney development.
Collapse
|
24
|
Zhang D, Zhang Y, Sun X. LINC01133 promotes the progression of cervical cancer via regulating miR-30a-5p/FOXD1. Asia Pac J Clin Oncol 2021; 17:253-263. [PMID: 33078907 DOI: 10.1111/ajco.13451] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 07/30/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The prognosis of patients with recurrent or metastatic cervical cancer (CC) remains poor, and its incidence is especially high in developing countries. Multiple long noncoding RNAs are recently identified as crucial oncogenic factors or tumor suppressors. In this study, we explored the function and mechanism of LINC01133 during the progression of CC. METHODS Expression levels of LINC01133 and miR-30a-5p in 50 CC tissue samples were measured using quantitative real-time polymerase chain reaction. Immunohistochemistry and Western blot analysis were used to detect the expression of oncogene forkhead box D1 (FOXD1). The association between pathological indices and the expression level of LINC01133 was also analyzed. Human CC cell lines HeLa and SiHa were used as cell models. CCK-8 and bromodeoxyuridine assays were used to assess the effect of LINC01133 on CC cell line proliferation. Flow cytometry was used to study the effect of LINC01133 on CC apoptosis. Transwell assay was conducted to detect the effect of LINC01133 on migration and invasion. Furthermore, luciferase reporter assay was used to confirm the targeting relationship between miR-30a-5p to LINC01133. RESULTS We observed that LINC01133 expression in CC clinical samples was significantly increased, with high expression associated with higher T stage and negative HPV infection of the patients. Its overexpression remarkably accelerated proliferation and metastasis of CC cells, with reduced apoptosis. LINC01133 knockdown suppressed the malignant phenotypes of CC cells. Overexpression of LINC01133 significantly reduced the expression of miR-30a-5p by sponging it and enhanced the expression of FOXD1. CONCLUSIONS We report the overexpression of LINC01133 in CC sample and cell lines, which correlated with unfavorable pathological indices. LINC01133 was a sponge of tumor suppressor miR-30a-5p, and it enhanced the expression of FOXD1 indirectly and functioned as an oncogenic lncRNA in CC.
Collapse
Affiliation(s)
- Dan Zhang
- Department of Gynecology, General Hospital of Fushun Mining Bureau of Liaoning Health Industry Group, Fushun, Liaoning Province, China
| | - Yuyang Zhang
- The Second Department of Oncology, General Hospital of Fushun Mining Bureau of Liaoning Health Industry Group, Fushun, Liaoning Province, China
| | - Xiuyun Sun
- Department of Gynecology, General Hospital of Fushun Mining Bureau of Liaoning Health Industry Group, Fushun, Liaoning Province, China
| |
Collapse
|
25
|
Bond KH, Fetting JL, Lary CW, Emery IF, Oxburgh L. FOXD1 regulates cell division in clear cell renal cell carcinoma. BMC Cancer 2021; 21:312. [PMID: 33761914 PMCID: PMC7988646 DOI: 10.1186/s12885-021-07957-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 02/23/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Forkhead transcription factors control cell growth in multiple cancer types. Foxd1 is essential for kidney development and mitochondrial metabolism, but its significance in renal cell carcinoma (ccRCC) has not been reported. METHODS Transcriptome data from the TCGA database was used to correlate FOXD1 expression with patient survival. FOXD1 was knocked out in the 786-O cell line and known targets were analyzed. Reduced cell growth was observed and investigated in vitro using growth rate and Seahorse XF metabolic assays and in vivo using a xenograft model. Cell cycle characteristics were determined by flow cytometry and immunoblotting. Immunostaining for TUNEL and γH2AX was used to measure DNA damage. Association of the FOXD1 pathway with cell cycle progression was investigated through correlation analysis using the TCGA database. RESULTS FOXD1 expression level in ccRCC correlated inversely with patient survival. Knockout of FOXD1 in 786-O cells altered expression of FOXD1 targets, particularly genes involved in metabolism (MICU1) and cell cycle progression. Investigation of metabolic state revealed significant alterations in mitochondrial metabolism and glycolysis, but no net change in energy production. In vitro growth rate assays showed a significant reduction in growth of 786-OFOXD1null. In vivo, xenografted 786-OFOXD1null showed reduced capacity for tumor formation and reduced tumor size. Cell cycle analysis showed that 786-OFOXD1null had an extended G2/M phase. Investigation of mitosis revealed a deficiency in phosphorylation of histone H3 in 786-OFOXD1null, and increased DNA damage. Genes correlate with FOXD1 in the TCGA dataset associate with several aspects of mitosis, including histone H3 phosphorylation. CONCLUSIONS We show that FOXD1 regulates the cell cycle in ccRCC cells by control of histone H3 phosphorylation, and that FOXD1 expression governs tumor formation and tumor growth. Transcriptome analysis supports this role for FOXD1 in ccRCC patient tumors and provides an explanation for the inverse correlation between tumor expression of FOXD1 and patient survival. Our findings reveal an important role for FOXD1 in maintaining chromatin stability and promoting cell cycle progression and provide a new tool with which to study the biology of FOXD1 in ccRCC.
Collapse
Affiliation(s)
- Kyle H Bond
- The Rogosin Institute, 310 East 67th Street, New York, NY, 10065, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, 168 College Ave, Orono, 04469, ME, USA
| | - Jennifer L Fetting
- Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA
- Current affiliation: ICON Plc, 2100 Pembrook Parkway, North Wales, 19446, PA, USA
| | - Christine W Lary
- Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Ivette F Emery
- Maine Medical Center Research Institute, 81 Research Drive, Scarborough, ME, 04074, USA
| | - Leif Oxburgh
- The Rogosin Institute, 310 East 67th Street, New York, NY, 10065, USA.
| |
Collapse
|
26
|
Dou J, Puttabyatappa M, Padmanabhan V, Bakulski KM. Developmental programming: Adipose depot-specific transcriptional regulation by prenatal testosterone excess in a sheep model of PCOS. Mol Cell Endocrinol 2021; 523:111137. [PMID: 33359827 PMCID: PMC7854529 DOI: 10.1016/j.mce.2020.111137] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/16/2020] [Accepted: 12/14/2020] [Indexed: 02/08/2023]
Abstract
Prenatal testosterone (T)-treated female sheep manifest adipose depot-specific disruptions in inflammatory/oxidative state, adipocyte differentiation and thermogenic adipocyte distribution. The objective of this study was to identify common and divergent gene pathways underlying prenatal T excess-induced adipose depot-specific disruptions. RNA sequencing and network analyses were undertaken with visceral (VAT), subcutaneous (SAT), epicardiac (ECAT) and perirenal (PRAT) adipose tissues from control and prenatal T-treated (100 mg T propionate twice a week from days 30-90 of gestation) female sheep at 21 months of age. Increased expression of adiposity and inflammation-related genes in VAT and genes that promote differentiation of white adipocytes in SAT were congruous with their metabolic roles with SAT favoring uptake/storage of free fatty acids and triglycerides and VAT favoring higher rate of fatty acid turnover and lipolysis. Selective upregulation of cardiac muscle and renoprotection genes in ECAT and PRAT respectively are suggestive of protective paracrine actions. Expression profile in prenatal T-treated sheep paralleled depot-specific dysfunctions with increased proinflammatory genes in VAT, reduced adipocyte differentiation genes in VAT and SAT and increased vascular related gene expression in PRAT. The high expression of genes involved in cardiomyocyte function in ECAT is suggestive of cardioprotective function being maintained to overcome the prenatal T-induced cardiac dysfunction and hypertension. These findings coupled with changes in gene pathways and networks involved in chromatin modification, extracellular matrix, immune and mitochondrial function, and endoplasmic reticulum to Golgi transport suggest that dysregulation in gene expression underlie prenatal T-treatment induced functional differences among adipose depots and manifestation of metabolic dysfunction.
Collapse
Affiliation(s)
- John Dou
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Kelly M Bakulski
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
27
|
Liu Q, Song X, Liu Z, Yu Z. Investigation of Candidate Genes and Pathways in Basal/TNBC Patients by Integrated Analysis. Technol Cancer Res Treat 2021; 20:15330338211019506. [PMID: 34184566 PMCID: PMC8246569 DOI: 10.1177/15330338211019506] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 03/09/2021] [Accepted: 04/19/2021] [Indexed: 12/11/2022] Open
Abstract
PURPOSE This study aims to identify the key pathway and related genes and to further explore the potential molecular mechanisms of triple negative breast cancer (TNBC). METHODS The transcriptome data and clinical information of breast cancer patients were downloaded from the TCGA database, including 94 cases of paracancerous tissue, 225 cases of Basal like type, 151 cases of Her2 type, 318 cases of Luminal type A, 281 cases of Luminal type B, and 89 cases of Normal Like type. The differentially expressed genes (DEGs) were identified based on the criteria of |logFC|≥1.5 and adjust P < 0.001.Their functions were annotated by gene ontology (GO) analysis and Kyoto Encyclopedia of differentially expressed genes & Genomes (KEGG) pathway analysis. Cox regression univariate analysis and Kaplan-Meier survival curves (Log-rank method) were used for survival analysis. FOXD1, DLL3 and LY6D were silenced in breast cancer cell lines, and cell viability was assessed by CCK-8 assay. Further, the expression of FOXD1, DLL3 and LY6D were explored by immunohistochemistry on triple negative breast tumor tissue and normal breast tissue. RESULTS A total of 533 DEGs were identified. Functional annotation showed that DEGs were significantly enriched in intermediate filament cytoskeleton, DNA-binding transcription activator activity, epidermis development, and Neuroactive ligand-receptor interaction. Survival analysis found that FOXD1, DLL3, and LY6D showed significant correlation with the prognosis of patients with the Basal-like type (P < 0.05). CCK-8 assay showed that compared with Doxorubicin alone group, the cytotoxicity of Doxorubicin combined with siRNA-knockdown of FOXD1, DLL3, or LY6D was much significant. CONCLUSION The DEGs and their enriched functions and pathways identified in this study contribute to the understanding of the molecular mechanisms of TNBC. In addition, FOXD1, DLL3, and LY6D may be defined as the prognostic markers and potential therapeutic targets for TNBC patients.
Collapse
Affiliation(s)
- Qi Liu
- School of Medicine, Shandong University, Jinan, People’s Republic of China
- Department of Breast Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People’s Republic of China
- Department of Breast and Thyroid Surgery, Weifang Traditional Chinese Hospital, Weifang, Shandong, People’s Republic of China
| | - Xiang Song
- Department of Breast Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People’s Republic of China
| | - Zhaoyun Liu
- School of Medicine, Shandong University, Jinan, People’s Republic of China
- Department of Breast Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People’s Republic of China
| | - Zhiyong Yu
- Department of Breast Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
28
|
Li Z, Yan T, Wu X, Zhang W, Li J, Wang L, Yang J. Increased expression of FOXD1 is associated with cervical node metastasis and unfavorable prognosis in oral squamous cell carcinoma. J Oral Pathol Med 2020; 49:1030-1036. [PMID: 32808339 DOI: 10.1111/jop.13098] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/20/2020] [Accepted: 07/20/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Previous studies suggest that FOXD1 is involved in tumorigenesis and closely related to the patients' poor outcome in human cancer. However, its expression pattern in primary oral squamous cell carcinoma (OSCC) remains uncovered. In this study, we tried to explore the expression pattern of FOXD1 and its clinicopathological significance in primary OSCC. METHODS Data mining and analysis on FOXD1 mRNA expression in OSCC samples were performed using publicly available databases. Its protein expression was supervised by immunohistochemistry in a retrospective cohort containing 58 primary OSCC samples. Furthermore, the potential associations between FOXD1 expression and various clinicopathological characteristics and patients' survival were further investigated. RESULTS Bioinformatic analysis indicated that FOXD1 mRNA abundance was obviously up-regulated in OSCC cohorts. Immunohistochemical staining results showed that FOXD1 protein was significantly up-regulated in OSCC specimens as compared to normal counterparts and its aberrant up-regulation was remarkably related to cervical lymph node metastasis (P = .0198) and decreased overall survival (P = .0281) and disease-free survival (P = .0312). Both univariate and multivariate Cox regression analysis further revealed the expression pattern of FOXD1 as an independent prognostic factor for overall survival of patients. CONCLUSION Taken together, these findings indicate that the aberrant up-regulation of FOXD1 is related to cervical node metastasis and unfavorable prognosis in OSCC and it also may play a key role during OSCC tumorigenesis and regard as a novel diagnostic and prognostic biomarker for OSCC.
Collapse
Affiliation(s)
- Zhongwu Li
- Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital, Nanjing Medical University, Nanjing, China
| | - Tingyuan Yan
- Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital, Nanjing Medical University, Nanjing, China
| | - Xiang Wu
- Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China
| | - Wei Zhang
- Department of Oral Pathology, Affiliated Stomatological Hospital, Nanjing Medical University, Nanjing, China
| | - Jin Li
- Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China
| | - Laijie Wang
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital, Nanjing Medical University, Nanjing, China
| | - Jianrong Yang
- Jiangsu Key Laboratory of Oral Disease, Nanjing Medical University, Nanjing, China.,Department of Oral and Maxillofacial Surgery, Affiliated Stomatological Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
29
|
Xu X, Cheng J, Luo S, Huang D, Xu J, Qian Y, Zhou H, Wan X. Deoxycholic acid-stimulated macrophage-derived exosomes promote intestinal metaplasia and suppress proliferation in human gastric epithelial cells. Eur J Pharmacol 2020; 887:173566. [PMID: 32950501 DOI: 10.1016/j.ejphar.2020.173566] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023]
Abstract
The crosstalk between macrophages and gastric epithelial cells has emerged as a player in chronic inflammation during intestinal metaplasia. However, the role of bile acid on this modulation remains to be studied. We hypothesized that deoxycholic acid-induced macrophages secreted exosomes to mediate intercellular communication and promoted intestinal metaplasia in human gastric epithelial cells (GES-1 cells). Macrophage-derived exosomes (M-Exos) and deoxycholic acid-induced macrophage-derived exosomes (D-Exos) were isolated by ultracentrifugation. EdU staining and CCK-8 assay were utilized to evaluate the effects of exosomes on the proliferation of GES-1 cells. Intestinal metaplasia was assessed by the expression of caudal-related homeobox transcription factor 2 (CDX2) at both mRNA and protein level. MicroRNA sequencing revealed the microRNA (miRNA) expression profiles of M-Exos and D-Exos. The role of a specific miRNA and mRNA was analyzed by using miRNA mimics, miRNA inhibitors and siRNAs. D-Exos promoted the expression of CDX2 and suppressed the proliferation of GES-1 cells, compared to M-Exos. The miRNA profiles and quantitative real-time PCR examination showed D-Exos enriched a higher level of hsa-miR-30a-5p than M-Exos. Overexpressed has-miR-30a-5p increased CDX2 expression and inhibited the proliferation in GES-1 cells via targeted Forkhead Box D1 (FOXD1), a potential regulatory factor in the process of intestinal metaplasia. D-Exos may promote intestinal metaplasia and suppress proliferation of GES-1 cells via hsa-miR-30a-5p targeting FOXD1, which may be involved in the action mechanism of bile acid on gastric mucosa.
Collapse
Affiliation(s)
- Xianjun Xu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinnian Cheng
- Department of Gastroenterology, Shanghai General Hospital, Nanjing Medical University, Nanjing, China
| | - Shengzheng Luo
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan Huang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jingxian Xu
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yueqin Qian
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Hui Zhou
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Key Laboratory of Pancreatic Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Gastroenterology, Shanghai General Hospital, Nanjing Medical University, Nanjing, China.
| | - Xinjian Wan
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Gastroenterology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
30
|
He W, Kang Y, Zhu W, Zhou B, Jiang X, Ren C, Guo W. FOXF2 acts as a crucial molecule in tumours and embryonic development. Cell Death Dis 2020; 11:424. [PMID: 32503970 PMCID: PMC7275069 DOI: 10.1038/s41419-020-2604-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 02/14/2020] [Accepted: 02/14/2020] [Indexed: 12/24/2022]
Abstract
As a key member of the forkhead box transcription factors, forkhead box F2 (FOXF2) serves as a transcriptional regulator and regulates downstream gene expression in embryonic development, metabolism and in some common diseases, such as stroke and gastroparesis. Recent studies have shown that aberrant expression of FOXF2 is associated with a variety of tumorigenic processes, such as proliferation, invasion and metastasis. The role of FOXF2 in the development of many different organs has been confirmed by studies and has been speculated about in case reports. We focus on the mechanisms and signal pathways of tumour development initiated by aberrant expression of FOXF2, and we summarize the diseases and signal pathways caused by aberrant expression of FOXF2 in embryogenesis. This article highlights the differences in the role of FOXF2 in different tumours and demonstrates that multiple factors can regulate FOXF2 levels. In addition, FOXF2 is considered a biomarker for the diagnosis or prognosis of various tumours. Therefore, regulating the level of FOXF2 is an ideal treatment for tumours. FOXF2 could also affect the expression of some organ-specific genes to modulate organogenesis and could serve as a biomarker for specific differentiated cells. Finally, we present prospects for the continued research focus of FOXF2.
Collapse
Affiliation(s)
- Weihan He
- Cancer Research Institute, Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Kaifu District, Changsha, 410008, China.,Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, Changsha, Hunan, China.,The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yuanbo Kang
- Cancer Research Institute, Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Kaifu District, Changsha, 410008, China.,Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, Changsha, Hunan, China.,The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Wei Zhu
- Cancer Research Institute, Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Kaifu District, Changsha, 410008, China.,Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Bolun Zhou
- Cancer Research Institute, Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Kaifu District, Changsha, 410008, China.,Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Xingjun Jiang
- Cancer Research Institute, Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Kaifu District, Changsha, 410008, China.,Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Caiping Ren
- Cancer Research Institute, Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Kaifu District, Changsha, 410008, China. .,Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, Changsha, Hunan, China. .,The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Weihua Guo
- Cancer Research Institute, Department of Neurosurgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Kaifu District, Changsha, 410008, China. .,Cancer Research Institute, Collaborative Innovation Center for Cancer Medicine, School of Basic Medical Science, Central South University, Changsha, Hunan, China. .,The NHC Key Laboratory of Carcinogenesis and The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
31
|
Li X, Jiao M, Hu J, Qi M, Zhang J, Zhao M, Liu H, Xiong X, Dong X, Han B. miR-30a inhibits androgen-independent growth of prostate cancer via targeting MYBL2, FOXD1, and SOX4. Prostate 2020; 80:674-686. [PMID: 32294305 DOI: 10.1002/pros.23979] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/22/2020] [Accepted: 03/26/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Castrate-resistant prostate cancer (CRPC) is an aggressive and lethal disease. The pathogenesis of CRPC is not fully understood and novel therapeutic targets need to be identified to improve the patients' prognosis. MicroRNA-30a (miR-30a) has been demonstrated to be a tumor suppressor in many types of solid malignancies. However, its role in androgen-independent (AI) growth of prostate cancer (PCa) received limited attention as yet. METHODS The clinical association of miR-30a and its potential targets with AI growth was characterized by bioinformatics analyses. Regulation of cell proliferation and colony formation rates by miR-30a were tested using PCa cell models. Xenograft models were used to measure the regulation of prostate tumor growth by miR-30a. The real-time quantitative polymerase chain reaction was used to validate whether miR-30a and its targets regulate cell cycle control genes and androgen receptor (AR)-dependent transcription. Bioinformatics tools, Western blot, and luciferase reporter assays were utilized to identify miR-30a targets. RESULTS Bioinformatic analysis showed that low expression of miR-30a is associated with castration resistance of PCa patients and poor outcomes. Transfection of miR-30a mimics inhibited the AI growth of PCa cells in vitro and in vivo. Upregulation of miR-30a in 22RV1 cells altered the expression of cell cycle control genes and AR-mediated transcription, while downregulation of miR-30a in LNCaP cells had the opposite effects to AR-mediated transcription. MYBL2, FOXD1, and SOX4 were identified as miR-30a targets. Downregulation of MYBL2, FOXD1, and SOX4 affected the expression of cell cycle control genes and AR-mediated transcription and suppressed the AI growth of 22RV1 cells. CONCLUSIONS Our results suggest that miR-30a inhibits AI growth of PCa by targeting MYBL2, FOXD1, and SOX4. They provide novel insights into developing new treatment strategies for CRPC.
Collapse
Affiliation(s)
- Xinjun Li
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, China
- Department of Pathology, Binzhou People's Hospital, Binzhou, China
- School of Medicine, Shandong University, Jinan, China
| | - Meng Jiao
- Department of Pathology, The Second Hospital of Shandong University, Jinan, China
| | - Jing Hu
- Department of Pathology, Shandong University QiLu Hospital, Jinan, China
| | - Mei Qi
- Department of Pathology, Shandong University QiLu Hospital, Jinan, China
| | - Jing Zhang
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Mingfeng Zhao
- Department of Pathology, Binzhou Medical University, Binzhou, China
| | - Hui Liu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xueting Xiong
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Xuesen Dong
- Department of Urologic Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Bo Han
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, China
- Department of Pathology, Shandong University QiLu Hospital, Jinan, China
| |
Collapse
|
32
|
Laissue P, Vaiman D. Exploring the Molecular Aetiology of Preeclampsia by Massive Parallel Sequencing of DNA. Curr Hypertens Rep 2020; 22:31. [PMID: 32172383 DOI: 10.1007/s11906-020-01039-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW This manuscript aims to review (for the first time) studies describing NGS sequencing of preeclampsia (PE) women's DNA. RECENT FINDINGS Describing markers for the early detection of PE is an essential task because, although associated molecular dysfunction begins early on during pregnancy, the disease's clinical signs usually appear late in pregnancy. Although several biochemical biomarkers have been proposed, their use in clinical environments is still limited, thereby encouraging research into PE's genetic origin. Hundreds of genes involved in numerous implantation- and placentation-related biological processes may be coherent candidates for PE aetiology. Next-generation sequencing (NGS) offers new technical possibilities for PE studying, as it enables large genomic regions to be analysed at affordable cost. This technique has facilitated the description of genes contributing to the molecular origin of a significant amount of monogenic and complex diseases. Regarding PE, NGS of DNA has been used in familial and isolated cases, thereby enabling new genes potentially related to the phenotype to be proposed. For a better understanding of NGS, technical aspects, applications and limitations are presented initially. Thereafter, NGS studies of DNA in familial and non-familial cases are described, including pitfalls and positive findings. The information given here should enable scientists and clinicians to analyse and design new studies permitting the identification of novel clinically useful molecular PE markers.
Collapse
Affiliation(s)
- Paul Laissue
- Biopas Laboratoires, Biopas Group, Bogotá, Colombia. .,Inserm U1016, CNRS UMR8104, Institut Cochin, équipe FGTB, 24, rue du faubourg Saint-Jacques, 75014, Paris, France. .,CIGGUR Genetics Group, School of Medicine and Health Sciences, El Rosario University, Bogotá, Colombia.
| | - Daniel Vaiman
- Inserm U1016, CNRS UMR8104, Institut Cochin, équipe FGTB, 24, rue du faubourg Saint-Jacques, 75014, Paris, France
| |
Collapse
|
33
|
Estrada SM, Thagard AS, Dehart MJ, Damicis JR, Dornisch EM, Ippolito DL, Burd I, Napolitano PG, Ieronimakis N. The orphan nuclear receptor Nr4a1 mediates perinatal neuroinflammation in a murine model of preterm labor. Cell Death Dis 2020; 11:11. [PMID: 31907354 PMCID: PMC6944691 DOI: 10.1038/s41419-019-2196-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/06/2019] [Accepted: 12/09/2019] [Indexed: 12/25/2022]
Abstract
Prematurity is associated with perinatal neuroinflammation and injury. Screening for genetic modulators in an LPS murine model of preterm birth revealed the upregulation of Nr4a1, an orphan nuclear transcription factor that is normally absent or limited in embryonic brains. Concurrently, Nr4a1 was downregulated with magnesium sulfate (MgSO4) and betamethasone (BMTZ) treatments administered to LPS exposed dams. To understand the role of Nr4a1 in perinatal brain injury, we compared the preterm neuroinflammatory response in Nr4a1 knockout (KO) versus wild type (wt) mice. Key inflammatory factors Il1b, Il6 and Tnf, and Iba1+ microglia were significantly lower in Nr4a1 KO versus wt brains exposed to LPS in utero. Treatment with MgSO4/BMTZ mitigated the neuroinflammatory process in wt but not Nr4a1 KO brains. These results correspond with a reduction in cerebral hemorrhage in wt but not mutant embryos from dams given MgSO4/BMTZ. Further analysis with Nr4a1-GFP-Cre × tdTomato loxP reporter mice revealed that the upregulation of Nr4a1 with perinatal neuroinflammation occurs in the cerebral vasculature. Altogether, this study implicates Nr4a1 in the developing vasculature as a potent mediator of neuroinflammatory brain injury that occurs with preterm birth. It is also possible that MgSO4/BMTZ mitigates this process by direct or indirect inhibition of Nr4a1.
Collapse
Affiliation(s)
- Sarah M Estrada
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Madigan Army Medical Center, Tacoma, WA, USA
| | - Andrew S Thagard
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Madigan Army Medical Center, Tacoma, WA, USA
| | - Mary J Dehart
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA, USA
| | - Jennifer R Damicis
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA, USA
| | - Elisabeth M Dornisch
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA, USA
| | | | - Irina Burd
- Integrated Research Center for Fetal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter G Napolitano
- Department of Obstetrics and Gynecology, University of Washington Medical Center, Seattle, WA, USA
| | - Nicholas Ieronimakis
- Department of Clinical Investigation, Madigan Army Medical Center, Tacoma, WA, USA.
| |
Collapse
|
34
|
Li CH, Chang YC, Hsiao M, Liang SM. FOXD1 and Gal-3 Form a Positive Regulatory Loop to Regulate Lung Cancer Aggressiveness. Cancers (Basel) 2019; 11:cancers11121897. [PMID: 31795213 PMCID: PMC6966623 DOI: 10.3390/cancers11121897] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 11/22/2019] [Accepted: 11/25/2019] [Indexed: 01/15/2023] Open
Abstract
Dysregulation of forkhead box D1 (FOXD1) is known to promote tumor progression; however, its molecular mechanism of action is unclear. Based on microarray analysis, we identified galectin-3/LGALS3 (Gal-3) as a potential downstream target of FOXD1, as FOXD1 transactivated Gal-3 by interacting with the Gal-3 promoter to upregulate Gal-3 in FOXD1-overexpressing CL1-0 lung cancer cells. Ectopic expression of FOXD1 increased the expression of Gal-3 and the growth and motility of lung cancer cells, whereas depletion of Gal-3 attenuated FOXD1-mediated tumorigenesis. ERK1/2 interacted with FOXD1 in the cytosol and translocated FOXD1 into the nucleus to activate Gal-3. Gal-3 in turn upregulated FOXD1 via the transcription factor proto-oncogene 1 (ETS-1) to transactivate FOXD1. The increase in ETS-1/FOXD1 expression by Gal-3 was through Gal-3-mediated integrin-β1 (ITGβ1) signaling. The overexpression of both FOXD1 and Gal-3 form a positive regulatory loop to promote lung cancer aggressiveness. Moreover, both FOXD1 and Gal-3 were positively correlated in human lung cancer tissues. Our findings demonstrated that FOXD1 and Gal-3 form a positive feedback loop in lung cancer, and interference of this loop may serve as an effective therapeutic target for the treatment of lung cancers, particularly those related to dysregulation of Gal-3.
Collapse
Affiliation(s)
- Chien-Hsiu Li
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan;
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Yu-Chan Chang
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan;
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan;
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence: (M.H.); (S.-M.L.); Tel.: +886-227-871-243 (M.H.); +886-227-872-082 (S.-M.L.)
| | - Shu-Mei Liang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan;
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei 115, Taiwan
- Correspondence: (M.H.); (S.-M.L.); Tel.: +886-227-871-243 (M.H.); +886-227-872-082 (S.-M.L.)
| |
Collapse
|
35
|
Quintero-Ronderos P, Jiménez KM, Esteban-Pérez C, Ojeda DA, Bello S, Fonseca DJ, Coronel MA, Moreno-Ortiz H, Sierra-Díaz DC, Lucena E, Barbaux S, Vaiman D, Laissue P. FOXD1 mutations are related to repeated implantation failure, intra-uterine growth restriction and preeclampsia. Mol Med 2019; 25:37. [PMID: 31395028 PMCID: PMC6688323 DOI: 10.1186/s10020-019-0104-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 07/16/2019] [Indexed: 12/14/2022] Open
Abstract
Background Human reproductive disorders consist of frequently occurring dysfunctions including a broad range of phenotypes affecting fertility and women’s health during pregnancy. Several female-related diseases have been associated with hypofertility/infertility phenotypes, such as recurrent pregnancy loss (RPL). Other occurring diseases may be life-threatening for the mother and foetus, such as preeclampsia (PE) and intra-uterine growth restriction (IUGR). FOXD1 was defined as a major molecule involved in embryo implantation in mice and humans by regulating endometrial/placental genes. FOXD1 mutations in human species have been functionally linked to RPL’s origin. Methods FOXD1 gene mutation screening, in 158 patients affected by PE, IUGR, RPL and repeated implantation failure (RIF), by direct sequencing and bioinformatics analysis. Plasmid constructs including FOXD1 mutations were used to perform in vitro gene reporter assays. Results Nine non-synonymous sequence variants were identified. Functional experiments revealed that p.His267Tyr and p.Arg57del led to disturbances of promoter transcriptional activity (C3 and PlGF genes). The FOXD1 p.Ala356Gly and p.Ile364Met deleterious mutations (previously found in RPL patients) have been identified in the present work in women suffering PE and IUGR. Conclusions Our results argue in favour of FOXD1 mutations’ central role in RPL, RIF, IUGR and PE pathogenesis via C3 and PlGF regulation and they describe, for the first time, a functional link between FOXD1 and implantation/placental diseases. FOXD1 could therefore be used in clinical environments as a molecular biomarker for these diseases in the near future. Keywords Recurrent pregnancy loss, Preeclampsia, Intra-uterine growth restriction, FOXD1 Electronic supplementary material The online version of this article (10.1186/s10020-019-0104-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Paula Quintero-Ronderos
- Center For Research in Genetics and Genomics-CIGGUR. GENIUROS Research Group. School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Karen Marcela Jiménez
- Center For Research in Genetics and Genomics-CIGGUR. GENIUROS Research Group. School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Clara Esteban-Pérez
- Fertility and Sterility Colombian Center, Department of Reproductive Genetics, Bogotá, Colombia
| | - Diego A Ojeda
- Center For Research in Genetics and Genomics-CIGGUR. GENIUROS Research Group. School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia.,Clinical Neurosciences and Psychiatry, Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK., Southampton, United Kingdom
| | - Sandra Bello
- Center For Research in Genetics and Genomics-CIGGUR. GENIUROS Research Group. School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Dora Janeth Fonseca
- Center For Research in Genetics and Genomics-CIGGUR. GENIUROS Research Group. School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - María Alejandra Coronel
- Center For Research in Genetics and Genomics-CIGGUR. GENIUROS Research Group. School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Harold Moreno-Ortiz
- Fertility and Sterility Colombian Center, Department of Reproductive Genetics, Bogotá, Colombia
| | - Diana Carolina Sierra-Díaz
- Center For Research in Genetics and Genomics-CIGGUR. GENIUROS Research Group. School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Elkin Lucena
- Fertility and Sterility Colombian Center, Department of Reproductive Genetics, Bogotá, Colombia
| | - Sandrine Barbaux
- Inserm U1016, CNRS UMR8104, Institut Cochin, équipe FGTB, 24, rue du faubourg Saint-Jacques, 75014, Paris, France
| | - Daniel Vaiman
- Inserm U1016, CNRS UMR8104, Institut Cochin, équipe FGTB, 24, rue du faubourg Saint-Jacques, 75014, Paris, France
| | - Paul Laissue
- Center For Research in Genetics and Genomics-CIGGUR. GENIUROS Research Group. School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia.
| |
Collapse
|
36
|
Belair C, Sim S, Kim KY, Tanaka Y, Park IH, Wolin SL. The RNA exosome nuclease complex regulates human embryonic stem cell differentiation. J Cell Biol 2019; 218:2564-2582. [PMID: 31308215 PMCID: PMC6683745 DOI: 10.1083/jcb.201811148] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 05/21/2019] [Accepted: 06/12/2019] [Indexed: 01/09/2023] Open
Abstract
This work shows that the exosome modulates the levels of LINE-1 retrotransposons and specific miRNAs, lncRNAs, and mRNAs that encode developmental regulators or affect their expression. The exosome restrains stem cell differentiation in part by degrading transcripts encoding FOXH1, a transcription factor crucial for mesendoderm formation. A defining feature of embryonic stem cells (ESCs) is the ability to differentiate into all three germ layers. Pluripotency is maintained in part by a unique transcription network that maintains expression of pluripotency-specific transcription factors and represses developmental genes. While the mechanisms that establish this transcription network are well studied, little is known of the posttranscriptional surveillance pathways that degrade differentiation-related RNAs. We report that the surveillance pathway mediated by the RNA exosome nuclease complex represses ESC differentiation. Depletion of the exosome expedites differentiation of human ESCs into all three germ layers. LINE-1 retrotransposons and specific miRNAs, lncRNAs, and mRNAs that encode developmental regulators or affect their expression are all bound by the exosome and increase in level upon exosome depletion. The exosome restrains differentiation in part by degrading transcripts encoding FOXH1, a transcription factor crucial for mesendoderm formation. Our studies establish the exosome as a regulator of human ESC differentiation and reveal the importance of RNA decay in maintaining pluripotency.
Collapse
Affiliation(s)
- Cedric Belair
- Department of Cell Biology, Yale School of Medicine, New Haven, CT.,RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD
| | - Soyeong Sim
- Department of Cell Biology, Yale School of Medicine, New Haven, CT.,RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD
| | - Kun-Yong Kim
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT
| | - Yoshiaki Tanaka
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT
| | - In-Hyun Park
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT
| | | | - Sandra L Wolin
- Department of Cell Biology, Yale School of Medicine, New Haven, CT .,RNA Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD
| |
Collapse
|
37
|
Laissue P. The forkhead-box family of transcription factors: key molecular players in colorectal cancer pathogenesis. Mol Cancer 2019; 18:5. [PMID: 30621735 PMCID: PMC6325735 DOI: 10.1186/s12943-019-0938-x] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 01/01/2019] [Indexed: 12/18/2022] Open
Abstract
Colorectal cancer (CRC) is the third most commonly occurring cancer worldwide and the fourth most frequent cause of death having an oncological origin. It has been found that transcription factors (TF) dysregulation, leading to the significant expression modifications of genes, is a widely distributed phenomenon regarding human malignant neoplasias. These changes are key determinants regarding tumour’s behaviour as they contribute to cell differentiation/proliferation, migration and metastasis, as well as resistance to chemotherapeutic agents. The forkhead box (FOX) transcription factor family consists of an evolutionarily conserved group of transcriptional regulators engaged in numerous functions during development and adult life. Their dysfunction has been associated with human diseases. Several FOX gene subgroup transcriptional disturbances, affecting numerous complex molecular cascades, have been linked to a wide range of cancer types highlighting their potential usefulness as molecular biomarkers. At least 14 FOX subgroups have been related to CRC pathogenesis, thereby underlining their role for diagnosis, prognosis and treatment purposes. This manuscript aims to provide, for the first time, a comprehensive review of FOX genes’ roles during CRC pathogenesis. The molecular and functional characteristics of most relevant FOX molecules (FOXO, FOXM1, FOXP3) have been described within the context of CRC biology, including their usefulness regarding diagnosis and prognosis. Potential CRC therapeutics (including genome-editing approaches) involving FOX regulation have also been included. Taken together, the information provided here should enable a better understanding of FOX genes’ function in CRC pathogenesis for basic science researchers and clinicians.
Collapse
Affiliation(s)
- Paul Laissue
- Center For Research in Genetics and Genomics-CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 N° 63C-69, Bogotá, Colombia.
| |
Collapse
|
38
|
Li P, Zhou Y, Goodwin AJ, Cook JA, Halushka PV, Zhang XK, Wilson CL, Schnapp LM, Zingarelli B, Fan H. Fli-1 Governs Pericyte Dysfunction in a Murine Model of Sepsis. J Infect Dis 2018; 218:1995-2005. [PMID: 30053030 PMCID: PMC6217724 DOI: 10.1093/infdis/jiy451] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 07/17/2018] [Indexed: 12/17/2022] Open
Abstract
Background Pericytes are vascular mural cells and are embedded in the basement membrane of the microvasculature. Recent studies suggest a role for pericytes in lipopolysaccharide (LPS)-induced microvascular dysfunction and mortality, but the mechanisms of pericyte loss in sepsis are largely unknown. Methods By using a cecal ligation and puncture (CLP)-induced murine model of sepsis, we observed that CLP led to lung and renal pericyte loss and reduced lung pericyte density and pericyte/endothelial cell (EC) coverage. Results Up-regulated Friend leukemia virus integration 1 (Fli-1) messenger ribonucleic acid (RNA) and protein levels were found in lung pericytes from CLP mice in vivo and in LPS-stimulated lung pericytes in vitro. Knockout of Fli-1 in Foxd1-derived pericytes prevented CLP-induced pericyte loss, vascular leak, and improved survival. Disrupted Fli-1 expression by small interfering RNA inhibited LPS-induced inflammatory cytokines and chemokines in cultured lung pericytes. Furthermore, CLP-induced pericyte pyroptosis was mitigated in pericyte Fli-1 knockout mice. Conclusions Our findings suggest that Fli-1 is a potential therapeutic target in sepsis.
Collapse
Affiliation(s)
- Pengfei Li
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston
| | - Yue Zhou
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston
- Department of Biopharmaceutics, College of Pharmacy, Nanjing University of Chinese Medicine, China
| | - Andrew J Goodwin
- Divisions of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Medical University of South Carolina, Charleston
| | - James A Cook
- Department of Neurosciences, Medical University of South Carolina, Charleston
| | - Perry V Halushka
- Department of Medicine, Medical University of South Carolina, Charleston
- Department of Pharmacology, Medical University of South Carolina, Charleston
| | - Xian K Zhang
- Rheumatology and Immunology, Department of Medicine, Medical University of South Carolina, Charleston
| | - Carole L Wilson
- Divisions of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Medical University of South Carolina, Charleston
| | - Lynn M Schnapp
- Divisions of Pulmonary, Critical Care, Allergy, and Sleep Medicine, Medical University of South Carolina, Charleston
| | - Basilia Zingarelli
- Division of Critical Care Medicine, Cincinnati Children’s Hospital Medical Center, Ohio
| | - Hongkuan Fan
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston
- Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston
| |
Collapse
|