1
|
Tian M, Zhou Y, Guo Y, Xia Q, Wang Z, Zheng X, Shen J, Guo J, Duan S, Wang L. MicroRNAs in adipose tissue fibrosis: Mechanisms and therapeutic potential. Genes Dis 2025; 12:101287. [PMID: 40242037 PMCID: PMC12002615 DOI: 10.1016/j.gendis.2024.101287] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 03/07/2024] [Indexed: 04/18/2025] Open
Abstract
Adipose tissue fibrosis, characterized by abnormal extracellular matrix deposition within adipose tissue, signifies a crucial indicator of adipose tissue malfunction, potentially leading to organ tissue dysfunction. Various factors, including a high-fat diet, non-alcoholic fatty liver disease, and insulin resistance, coincide with adipose tissue fibrosis. MicroRNAs (miRNAs) represent a class of small non-coding RNAs with significant influence on tissue fibrosis through diverse signaling pathways. For instance, in response to a high-fat diet, miRNAs can modulate signaling pathways such as TGF-β/Smad, PI3K/AKT, and PPAR-γ to impact adipose tissue fibrosis. Furthermore, miRNAs play roles in inhibiting fibrosis in different contexts: suppressing corneal fibrosis via the TGF-β/Smad pathway, mitigating cardiac fibrosis through the VEGF signaling pathway, reducing wound fibrosis via regulation of the MAPK signaling pathway, and diminishing fibrosis post-fat transplantation via involvement in the PDGFR-β signaling pathway. Notably, the secretome released by miRNA-transfected adipose-derived stem cells facilitates targeted delivery of miRNAs to evade host immune rejection, enhancing their anti-fibrotic efficacy. Hence, this study endeavors to elucidate the role and mechanism of miRNAs in adipose tissue fibrosis and explore the mechanisms and advantages of the secretome released by miRNA-transfected adipose-derived stem cells in combating fibrotic diseases.
Collapse
Affiliation(s)
- Mei Tian
- College of Pharmacy, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
- Geriatric Medicine Center, Department of Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Yang Zhou
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Yitong Guo
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Qing Xia
- College of Pharmacy, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Zehua Wang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Xinying Zheng
- College of Pharmacy, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Jinze Shen
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Junping Guo
- Rainbowfish Rehabilitation and Nursing School, Hangzhou Vocational & Technical College, Hangzhou, Zhejiang 310018, China
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Shiwei Duan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang 310015, China
| | - Lijun Wang
- Geriatric Medicine Center, Department of Endocrinology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| |
Collapse
|
2
|
Luo Y, Jin X, Huang L, Zeng D, Zhang N, Tang S, Luo S, Syed SE, Dai R, Li Q, Liang S. RUNX1/SLAMF3 Axis Drives Immunosuppression to Contribute to Colorectal Cancer Liver Metastasis by Blocking Phagocytosis and Depleting C1QC + Tumor-Associated Macrophages. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e06641. [PMID: 40448626 DOI: 10.1002/advs.202506641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2025] [Revised: 05/15/2025] [Indexed: 06/02/2025]
Abstract
Colorectal cancer liver metastasis (CRLM) is a leading cause of death in colorectal cancer (CRC) patients and is characterized by an immunosuppressive tumor microenvironment (TME). This study employs mouse in vivo selection to isolate highly metastatic CRLM derivatives for profiling their transcriptomic, proteomic, and metabolomic alterations associated with CRLM. Notably, the expression of SLAMF3 is significantly upregulated in CRLM derivatives and its knockdown effectively suppresses CRLM in mice. RUNX1 transcriptionally upregulates SLAMF3 expression and combined targeting of the RUNX1/SLAMF3 axis synergistically suppresses liver metastasis in mice. In parallel, SLAMF3 suppresses macrophage-mediated phagocytosis of CRC cells through the SHP-1/2/mTORC1 pathway. Conversely, SLAMF3 knockdown promotes M1 polarization in liver metastases and activates the CCL signaling pathway between macrophages and CD8+ T cells. It also reduces the exhausted CD8+ T cells in liver metastases and the expression of inhibitory receptors PD-1 and TIM-3, thus alleviating the immunosuppressive TME. Clinically, activation of the RUNX1/SLAMF3 axis is closely associated with CRLM progression and correlates with a reduced proportion of clinically beneficial C1QC⁺ tumor-associated macrophages (TAMs). Collectively, these findings identify the RUNX1/SLAMF3 axis as a key driver of immunosuppressive TME remodeling and CRLM progression, highlighting its potential as a promising therapeutic target for CRLM.
Collapse
Affiliation(s)
- Yinheng Luo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Xiaoli Jin
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Lan Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Dejia Zeng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| | - Nan Zhang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Shiyu Tang
- The Second Department of Gastrointestinal Surgery, The Affiliated Hospital of North Sichuan Medical College, Sichuan, P. R. China
| | - Shu Luo
- Department of Medical Oncology, Suining First People's Hospital, Suining, Sichuan, P. R. China
| | - Samina Ejaz Syed
- Department of Biochemistry and Biotechnology, Baghdad Campus, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Ruiwu Dai
- Department of General Surgery, The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, P. R. China
| | - Shufang Liang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, P. R. China
| |
Collapse
|
3
|
Yang Y, Deng S, Yin Q, Zhang T, Liu T, Wu D, Xu Y. RUNX1 promotes colorectal cancer progression by activating SERPINE1 transcription. Biochem Biophys Res Commun 2025; 760:151732. [PMID: 40164015 DOI: 10.1016/j.bbrc.2025.151732] [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: 01/17/2025] [Revised: 03/04/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) is among the most prevalent malignancies affecting the digestive system globally, considerably endangering public health. Runt-related transcription factor 1 (RUNX1) is a key regulator that influences the progression of cancer. In this study, we investigated the involvement of RUNX1 in CRC progression and its underlying mechanisms. METHODS The expression levels and prognostic significance of RUNX1 in CRC were analyzed based on data from the TNMplot, Gene Expression Profiling Interactive Analysis, and Gene Expression Omnibus databases as well as tissue microarrays. RUNX1 was knocked down or overexpressed in HT29 and HCT116 cells. The proliferation, migration, and invasion of CRC cells were evaluated. The target gene of RUNX1 was identified using PCR array analysis and validated via chromatin immunoprecipitation and luciferase reporter assays. The effects of RUNX1 on CRC cells were verified in vivo. RESULTS RUNX1 expression in CRC tissues was significantly higher than that in adjacent non-tumorous tissues and was associated with an unfavorable prognosis. Silencing RUNX1 significantly suppressed the proliferation, migratory capability, and invasive potential of CRC cells; overexpressing RUNX1 had the opposite effect. SERPINE1 was identified as a direct transcriptional target of RUNX1. In vivo experiments further validated that RUNX1-promoted CRC tumor growth. Mechanistically, RUNX1 promotes CRC progression by promoting the transcription of SERPINE1. CONCLUSIONS Our findings revealed that RUNX1 promotes CRC progression by activating SERPINE1 transcription, suggesting that RUNX1 is a potential prognostic biomarker and therapeutic target in CRC.
Collapse
Affiliation(s)
- Yueyan Yang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China; The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Shihua Deng
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China; The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Qing Yin
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China; The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Ting Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China; The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Teng Liu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China; The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Dongming Wu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China; The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China.
| | - Ying Xu
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China; The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China; Sichuan Clinical Research Center for Radiation and Therapy, The First Affiliated Hospital of Chengdu Medical College, 610500, PR China.
| |
Collapse
|
4
|
Guo B, Wang K, Wu J, Yu H, Geng C, Jin Y, Song Y. Runx1 activates the transformation of adipocytes into cancer-associated adipocytes by downregulating Plin1. Exp Cell Res 2025; 448:114573. [PMID: 40280321 DOI: 10.1016/j.yexcr.2025.114573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 03/23/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Adipocytes play a pivotal role in the breast tumor microenvironment, with the capacity to differentiate into cancer-associated adipocytes (CAAs) under the influence of breast cancer cells. This transformation significantly contributes to the formation and progression of breast cancer; however, the mechanisms underlying this interaction remain poorly understood. This study aims to illuminate these interactions by establishing an in vitro co-culture system of mature adipocytes and breast cancer cells. RNA sequencing analysis identified elevated runt-related transcription factor 1 (Runx1) expression in CAAs. Furthermore, Runx1 expression was also increased in the peritumoral adipose tissue of both breast cancer mouse models and clinical patient samples. Overexpression of Runx1 in 3T3-L1 preadipocytes resulted in reduced adipocyte volume, decreased lipid droplet size, diminished expression of mature adipocyte markers, and an increase in pro-inflammatory factor levels. These findings suggest that Runx1 overexpression facilitates the transformation of adipocytes into CAAs, thereby enhancing breast cancer cell migration and invasion. Conversely, Runx1 knockdown in CAAs diminished their supportive role in cancer progression. Mechanically, Runx1 enhances breast cancer development by regulating perilipin 1 (Plin1) levels, the overexpression of Plin1 in adipocytes inhibited the effect of Runx1 to promote the transition of adipocytes into CAAs. Our findings propose that targeting Runx1 in CAAs may represent a novel therapeutic strategy for breast cancer intervention.
Collapse
Affiliation(s)
- Boning Guo
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Provincial Hospital, Jinan, 250021, China
| | - Kai Wang
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Provincial Hospital, Jinan, 250021, China; Department of Endocrinology, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China
| | - Jing Wu
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Provincial Hospital, Jinan, 250021, China
| | - Huimin Yu
- Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Provincial Hospital, Jinan, 250021, China; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Chong Geng
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Yi Jin
- Department of Endocrinology, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China.
| | - Yongfeng Song
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education, Jinan, Shandong, 250021, China; Shandong Key Laboratory of Endocrinology and Lipid Metabolism, Shandong Provincial Hospital, Jinan, 250021, China; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China; Department of Endocrinology, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China.
| |
Collapse
|
5
|
Wang R, Yao X, Yu J, Wan X, Li S, Tian Y, Liu G, Yang Z, Yang X, Cheng S, Pan W, Cao Y, Luo H. A novel L-shaped ortho-quinone analog targeting adenosine A2b receptor to inhibit epithelial-mesenchymal transition in colorectal cancer cells. Med Oncol 2025; 42:197. [PMID: 40325273 DOI: 10.1007/s12032-025-02767-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Colorectal cancer (CRC) is a common malignancy of the gastrointestinal tract, with its incidence and mortality rates rising significantly in recent decades. In this study, we identified a compound (TC4) from a series of L-shaped ortho-quinone analog with notable inhibitory effects on epithelial-mesenchymal transition (EMT) in CRC cells. In vitro studies demonstrated that TC4 induces apoptosis, thereby suppressing CRC cell growth, invasion, and metastasis. Target analysis suggested that adenosine A2b receptor (ADORA2B) is a key molecular target of TC4, which was further confirmed by thermodynamic experiments showing direct binding to ADORA2B in living cells. Using ADORA2B overexpression and knockdown models, we found that abnormal expression of ADORA2B significantly affects CRC cell growth, invasion, metastasis, and sensitivity to TC4, confirming ADORA2B as a critical target for the compound's anti-tumor activity. TC4 was shown to markedly influence EMT, downregulating E-cadherin while upregulating N-cadherin, Vimentin, and Snail, with these effects dependent on ADORA2B overexpression. This indicates that the regulation of EMT by TC4 is closely associated with its interaction with ADORA2B. The present study confirms that TC4, a newly discovered compound with the ability to inhibit the growth and metastasis of CRC cells, can target ADORA2B to significantly regulate EMT in cancer cells.
Collapse
Affiliation(s)
- Rui Wang
- Medical College of GuiZhou University, Guiyang, 550025, Guizhou Province, China
| | - Xingsheng Yao
- Medical College of GuiZhou University, Guiyang, 550025, Guizhou Province, China
| | - Jia Yu
- Medical College of GuiZhou University, Guiyang, 550025, Guizhou Province, China
| | - Xinwei Wan
- Medical College of GuiZhou University, Guiyang, 550025, Guizhou Province, China
| | - Shengyou Li
- School of Pharmaceutical Sciences, GuiZhou University, Guiyang, 550025, Guizhou Province, China
| | - Yuxuan Tian
- Medical College of GuiZhou University, Guiyang, 550025, Guizhou Province, China
| | - Guangyang Liu
- Medical College of GuiZhou University, Guiyang, 550025, Guizhou Province, China
| | - Ziqi Yang
- Medical College of GuiZhou University, Guiyang, 550025, Guizhou Province, China
| | - Xianhui Yang
- Medical College of GuiZhou University, Guiyang, 550025, Guizhou Province, China
| | - Sha Cheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, Guizhou Province, China
| | - Weidong Pan
- School of Pharmaceutical Sciences, GuiZhou University, Guiyang, 550025, Guizhou Province, China
| | - Ying Cao
- Medical College of GuiZhou University, Guiyang, 550025, Guizhou Province, China.
| | - Heng Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, Guizhou Province, China.
| |
Collapse
|
6
|
Zhou S, Zhang Y, Belmar J, Hou C, Zhang Y, Peng C, Meng Y, Li Z, Mughal MJ, Gao Y, Seto E, Shen M, Hall MD, Ma J, Ma CX, Li S, Zhu W. Stabilization of RUNX1 Induced by O-GlcNAcylation Promotes PDGF-BB-Mediated Resistance to CDK4/6 Inhibitors in Breast Cancer. Cancer Res 2025; 85:1708-1724. [PMID: 39937190 DOI: 10.1158/0008-5472.can-24-2492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/01/2024] [Accepted: 02/06/2025] [Indexed: 02/13/2025]
Abstract
Cyclin-dependent kinases 4 and 6 (CDK4/6) are crucial in regulating cell-cycle progression and cancer development. Targeting CDK4/6 has shown considerable promise in treating various cancers, including breast cancer. Despite significant therapeutic efficacy, resistance to CDK4/6 inhibitors (CDK4/6i), such as palbociclib, remains a substantial hurdle in clinical practice. Using a coculture system, cytokine array, and quantitative high-throughput combinatorial screening, we discovered a mechanism by which the Runt-related transcription factor (RUNX) 1-platelet-derived growth factor (PDGF)-BB axis regulates palbociclib resistance in breast cancer cells. Specifically, RUNX1 functioned as a transcription factor to drive expression of PDGFB, leading to resistance to palbociclib by enhancing the Akt pathway and suppressing senescence. Furthermore, in resistant cells, RUNX1 was O-GlcNAcylated at serine 252 by O-GlcNAc transferase, resulting in the stabilization of RUNX1 by preventing ubiquitin-mediated degradation. Inhibition of the RUNX1-PDGF-BB axis by specific inhibitors overcame palbociclib resistance both in vitro and in vivo. Notably, the RUNX1-PDGF-BB axis was upregulated in resistant patient-derived xenograft lines and in patients with breast cancer following treatment with CDK4/6i. These findings not only unveil O-GlcNAcylation-mediated activation of a RUNX1-PDGF-BB pathway as a driver of palbociclib resistance but also provide clinical evidence supporting the repurposing of FDA-approved PDGFR inhibitors as a therapeutic strategy to treat patients with CDK4/6i-resistant breast cancer. Significance: RUNX1-PDGF-BB signaling drives resistance to CDK4/6 inhibition in breast cancer, providing the foundation to develop approaches to target the RUNX1-PDGF-BB axis to overcome CDK4/6 inhibitor resistance in breast cancer patients.
Collapse
Affiliation(s)
- Shuyan Zhou
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Yi Zhang
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Julie Belmar
- Department of Medicine, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Chunyan Hou
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Yaqin Zhang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland
| | - Changmin Peng
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Yunxiao Meng
- Laboratory & Molecular and Genomic Pathology, Department of Laboratory and Transfusion Services, The George Washington University Hospital, Washington, District of Columbia
| | - Zhuqing Li
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Muhammad Jameel Mughal
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Yanjun Gao
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Edward Seto
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| | - Min Shen
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland
| | - Matthew D Hall
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), National Institutes of Health (NIH), Rockville, Maryland
| | - Junfeng Ma
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, District of Columbia
| | - Cynthia X Ma
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine in St Louis, St. Louis, Missouri
| | - Shunqiang Li
- Department of Medicine, Washington University School of Medicine in St Louis, St Louis, Missouri
| | - Wenge Zhu
- Department of Biochemistry and Molecular Medicine, GWU Cancer Center, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia
| |
Collapse
|
7
|
Feng Y, Mao T, Yi J, Zhang N, Gu Y, Shen H, Chen J. Runt-related transcription factors: from pathogenesis to therapeutic targets in multiple-organ fibrosis. Front Cell Dev Biol 2025; 13:1528645. [PMID: 40356603 PMCID: PMC12066561 DOI: 10.3389/fcell.2025.1528645] [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: 11/15/2024] [Accepted: 03/26/2025] [Indexed: 05/15/2025] Open
Abstract
Fibrosis is a partially manageable process that leads to scarring and tissue hardening by prompting myofibroblasts to deposit significant amounts of extracellular matrix (ECM) following injury. It results in detrimental consequences and pathological characteristics, which hinder the functioning of associated organs and increase mortality rates. Runt-related transcription factors (RUNX) are part of a highly conserved family of heterodimer transcription factors, comprising RUNX1, RUNX2, and RUNX3. They are involved in several biological processes and undergo various forms of post-translational modification. RUNX regulates multiple targets and pathways to impact fibrosis, indicating promise for clinical application. Therefore, its significance in the fibrosis process should not be disregarded. The review begins with an objective description of the structure, transcriptional mechanism, and biological function of RUNX1, RUNX2, and RUNX3. A subsequent analysis is made of their physiological relationship with heart, lung, kidney, and liver, followed by a focus on the signaling mechanism of RUNX in regulating fibrosis of these organs. Furthermore, potential agents or drugs targeting RUNX for treating organ fibrosis are summarized, along with an evaluation of the therapeutic prospects and potential value of RUNX in fibrosis. Further research into RUNX could contribute to the development of novel therapeutic approaches for fibrosis.
Collapse
Affiliation(s)
- Yuan Feng
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Tianshi Mao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jifei Yi
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Na Zhang
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Yinying Gu
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Huifen Shen
- Suzhou Wujiang District Hospital of Traditional Chinese Medicine, Suzhou, China
| | - Jie Chen
- Department of Chinese Integrative Medicine Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Department of Integrated Traditional Chinese and Western Medicine, Anhui Medical University, Hefei, China
| |
Collapse
|
8
|
Tang Y, Cheng C, Ding R, Qian J, Liu M, Guo Y, Li Q. MSC exosomes and MSC exosomes loaded with LncRNA H19 as nanotherapeutics regulate the neurogenetic potential of Müller Glial Cells in dry age-related macular degeneration. Free Radic Biol Med 2025; 231:178-192. [PMID: 40015462 DOI: 10.1016/j.freeradbiomed.2025.02.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
In retinal degeneration diseases such as dry age-related macular degeneration (AMD), Müller Glial Cells (MGCs) in mammals undergo a process of reactive gliosis leading to the progression of dry AMD. Here, It is demonstrated that exosomes derived from mesenchymal stem cells (MSC exosomes) and MSC exosomes loaded with LncRNA H19, acting as nanotherapeutics, can be regulated by MGCs in dry AMD. In the in vivo study, MSC exosomes were administered via intravitreal injection. MSC exosomes effectively redirected MGCs from gliosis to dedifferentiation and alleviated MGCs-to-epithelial transition by inhibiting oxidative stress in mice with dry AMD induced by NaIO3. In the in vitro study, MSC exosomes promoted MGCs dedifferentiation by activating Wnt/β-catenin signaling pathway and prevented oxidative stress-induced MGCs gliosis and MGCs-to-epithelial transition by inhibiting TGFβ1 signaling pathway. MSC exosomes loaded with LncRNA H19 enhanced the activation of Wnt/β-catenin signaling pathway and the inhibition of the TGFβ1 signaling pathway compared with MSC exosomes. These results suggest that MSC exosomes regulate the neurogenetic potential of MGCs by redirecting MGCs from gliosis to dedifferentiation and alleviating the transformation of MGCs to epithelial cells through regulating oxidative stress. Regulating LncRNA H19 in MGCs to promote mammalian retinal regeneration in dry AMD was suggested for the first time.
Collapse
Affiliation(s)
- Yue Tang
- China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Caiyi Cheng
- China Pharmaceutical University, Nanjing, 211198, PR China
| | - Rui Ding
- China Pharmaceutical University, Nanjing, 211198, PR China
| | - Jingyuan Qian
- China Pharmaceutical University, Nanjing, 211198, PR China
| | - Min Liu
- China Pharmaceutical University, Nanjing, 211198, PR China
| | - Yuzun Guo
- China Pharmaceutical University, Nanjing, 211198, PR China
| | - Qian Li
- China Pharmaceutical University, Nanjing, 211198, PR China.
| |
Collapse
|
9
|
Wang B, Zhang S, Guo Y, Gao W, Wu H, Wang J, Wang Y, Tang C, Liu L. CBX2 as a therapeutic target in colorectal cancer: insights into the altered chromatin accessibility via RUNX1-CBX2-MAP4K1 axis. Oncogene 2025; 44:909-926. [PMID: 40082555 DOI: 10.1038/s41388-025-03331-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/06/2025] [Accepted: 02/24/2025] [Indexed: 03/16/2025]
Abstract
Chromobox homolog 2 (CBX2), a component of the polycomb repressive complex 1, is overexpressed in various cancers, but its specific role in colorectal cancer (CRC) is not fully understood. This study aimed to characterize the functional and regulatory roles of CBX2 in CRC. Tissue microarray analysis revealed the elevated CBX2 levels in tumor compared to adjacent normal tissues, which is significantly correlated with poor prognosis. Gain and loss of function studies demonstrated that CBX2 significantly promoted CRC progression and chemoresistance in cell lines, patient-derived CRC organoids and xenografts. In the AOM/DSS mouse model, treatment with the innovatively-developed cy5-PBAE/siCBX2 nanoparticle significantly reduced tumor aggressiveness. Mechanistic studies unveiled that the transcription factor RUNX1 is the positive regulator of CBX2. RNA-seq, ATAC-seq and CUT & RUN results indicated CBX2 knockdown induced epigenetic changes, especially alterations in chromatin accessibility. Moreover, we further identified MAP4K1 as a target gene of RUNX1-CBX2, with significant clinical and prognostic relevance in CRC. Collectively, these findings suggest the pivotal role of RUNX1-CBX2-MAP4K1 axis in CRC progression and underscore CBX2 as a promising biomarker and therapeutic target. The regulatory function of CBX2 on chromatin accessibility and the role of the RUNX1-CBX2-MAP4K1-pERK axis in the progression of colorectal cancer.
Collapse
Affiliation(s)
- Bangting Wang
- Digestive Endoscopy Department, The First Affiliated Hospital with Nanjing Medical University and Jiangsu Province Hospital, Nanjing, Jiangsu, China
- The Friendship Hospital of Ili Kazkh Autonomous Prefecture, Ili & Jiangsu Joint Institute of Health, Yining, China
| | - Shijie Zhang
- Digestive Endoscopy Department, The First Affiliated Hospital with Nanjing Medical University and Jiangsu Province Hospital, Nanjing, Jiangsu, China
| | - Yumeng Guo
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Wenqing Gao
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Hao Wu
- Digestive Endoscopy Department, The First Affiliated Hospital with Nanjing Medical University and Jiangsu Province Hospital, Nanjing, Jiangsu, China
| | - Jiankun Wang
- Digestive Endoscopy Department, The First Affiliated Hospital with Nanjing Medical University and Jiangsu Province Hospital, Nanjing, Jiangsu, China
| | - Yan Wang
- Digestive Endoscopy Department, The First Affiliated Hospital with Nanjing Medical University and Jiangsu Province Hospital, Nanjing, Jiangsu, China.
- The Friendship Hospital of Ili Kazkh Autonomous Prefecture, Ili & Jiangsu Joint Institute of Health, Yining, China.
| | - Chunming Tang
- College of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Li Liu
- Digestive Endoscopy Department, The First Affiliated Hospital with Nanjing Medical University and Jiangsu Province Hospital, Nanjing, Jiangsu, China.
| |
Collapse
|
10
|
Stojchevski R, Sutanto EA, Sutanto R, Hadzi-Petrushev N, Mladenov M, Singh SR, Sinha JK, Ghosh S, Yarlagadda B, Singh KK, Verma P, Sengupta S, Bhaskar R, Avtanski D. Translational Advances in Oncogene and Tumor-Suppressor Gene Research. Cancers (Basel) 2025; 17:1008. [PMID: 40149342 PMCID: PMC11940485 DOI: 10.3390/cancers17061008] [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: 02/10/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
Cancer, characterized by the uncontrolled proliferation of cells, is one of the leading causes of death globally, with approximately one in five people developing the disease in their lifetime. While many driver genes were identified decades ago, and most cancers can be classified based on morphology and progression, there is still a significant gap in knowledge about genetic aberrations and nuclear DNA damage. The study of two critical groups of genes-tumor suppressors, which inhibit proliferation and promote apoptosis, and oncogenes, which regulate proliferation and survival-can help to understand the genomic causes behind tumorigenesis, leading to more personalized approaches to diagnosis and treatment. Aberration of tumor suppressors, which undergo two-hit and loss-of-function mutations, and oncogenes, activated forms of proto-oncogenes that experience one-hit and gain-of-function mutations, are responsible for the dysregulation of key signaling pathways that regulate cell division, such as p53, Rb, Ras/Raf/ERK/MAPK, PI3K/AKT, and Wnt/β-catenin. Modern breakthroughs in genomics research, like next-generation sequencing, have provided efficient strategies for mapping unique genomic changes that contribute to tumor heterogeneity. Novel therapeutic approaches have enabled personalized medicine, helping address genetic variability in tumor suppressors and oncogenes. This comprehensive review examines the molecular mechanisms behind tumor-suppressor genes and oncogenes, the key signaling pathways they regulate, epigenetic modifications, tumor heterogeneity, and the drug resistance mechanisms that drive carcinogenesis. Moreover, the review explores the clinical application of sequencing techniques, multiomics, diagnostic procedures, pharmacogenomics, and personalized treatment and prevention options, discussing future directions for emerging technologies.
Collapse
Affiliation(s)
- Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | - Edward Agus Sutanto
- CUNY School of Medicine, The City College of New York, 160 Convent Avenue, New York, NY 10031, USA;
| | - Rinni Sutanto
- New York Institute of Technology College of Osteopathic Medicine, Glen Head, NY 11545, USA;
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia; (N.H.-P.)
| | - Sajal Raj Singh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Jitendra Kumar Sinha
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, Uttar Pradesh, India (J.K.S.)
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune 411057, Maharashtra, India;
| | - Prashant Verma
- School of Management, BML Munjal University, NH8, Sidhrawali, Gurugram 122413, Haryana, India
| | - Sonali Sengupta
- Department of Gastroenterology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA;
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
11
|
Hu H, Li W, Ma P, Song J, Zhang X, Ruan L, Zhang J, Zheng Y. TRIM22 inhibits the metastasis of colorectal cancer through facilitating β-Catenin degradation. Exp Cell Res 2025; 446:114473. [PMID: 39978715 DOI: 10.1016/j.yexcr.2025.114473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/15/2025] [Accepted: 02/16/2025] [Indexed: 02/22/2025]
Abstract
Tripartite motif-containing 22 (TRIM22), a member of the tripartite motif protein family, has emerged as a putative tumor suppressor in various cancers. Nevertheless, its specific role and clinical significance in colorectal cancer (CRC) remain poorly characterized. Herein, we observed that TRIM22 expression was frequently downregulated in primary CRC tissues and was significantly correlated with better prognosis. Functional assays demonstrated that TRIM22 overexpression substantially attenuated the metastatic potential of CRC cells both in vitro and in vivo. Mechanistically, our results revealed that TRIM22 directly interacts with and ubiquitinates β-Catenin, a crucial transcription factor that drives CRC metastasis by modulating the epithelial-mesenchymal transition (EMT) process. Additionally, our data indicated that the anti-metastatic effect of TRIM22 relies on the degradation of β-catenin. In summary, this study is the first to deliberate the vital anti-tumor role of TRIM22 in CRC metastasis. We also provide new evidence suggesting that TRIM22 could be a prognostic marker and therapeutic target for inhibiting CRC progression.
Collapse
Affiliation(s)
- Haiyang Hu
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Wensheng Li
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Pengfei Ma
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Junxin Song
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Xiaobo Zhang
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Longhui Ruan
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Jing Zhang
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Youwei Zheng
- The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China.
| |
Collapse
|
12
|
Yan Y, Gong Y, Liang X, Xiong Q, Lin J, Wu Y, Zhang L, Chen H, Jin J, Luan X. Decoding β-catenin associated protein-protein interactions: Emerging cancer therapeutic opportunities. Biochim Biophys Acta Rev Cancer 2025; 1880:189232. [PMID: 39643250 DOI: 10.1016/j.bbcan.2024.189232] [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: 05/26/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024]
Abstract
The hyperactive Wnt/β-catenin signaling circuit has been proven to be closely related to the progression of various cancers, with β-catenin serving as a central regulator of pro-tumorigenic processes. Preclinical evidences strongly support β-catenin as a promising therapeutic target. However, it has long been considered "undruggable" due to challenges such as the lack of crystal structures for its N- and C-terminal domains, high mutation rates, and limited availability of inhibitors. Notably, the network of β-catenin-associated protein-protein interactions (PPIs) is vital in the progression of multiple diseases. These interactions form a cancer-specific network that participates in all phases of oncogenesis, from cell metastasis to immunosuppressive microenvironment formation. Thus, researches on these PPIs are essential for unraveling the molecular mechanisms behind tumors with aberrant β-catenin activation, as well as for developing new targeted therapies. In this review, we delve into how β-catenin's PPIs orchestrate cancer progression and highlight biological and clinical dilemmas, proposing frontier technologies and potential challenges in targeting β-catenin for cancer therapy.
Collapse
Affiliation(s)
- Yue Yan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yiting Gong
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaohui Liang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qingyi Xiong
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiayi Lin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ye Wu
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lijun Zhang
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hongzhuan Chen
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Jinmei Jin
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Xin Luan
- Shanghai Frontiers Science Center for Chinese Medicine Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research and Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
13
|
Han M, Zhou X, Cheng H, Qiu M, Qiao M, Geng X. Chitosan and hyaluronic acid in colorectal cancer therapy: A review on EMT regulation, metastasis, and overcoming drug resistance. Int J Biol Macromol 2025; 289:138800. [PMID: 39694373 DOI: 10.1016/j.ijbiomac.2024.138800] [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: 11/03/2024] [Revised: 12/04/2024] [Accepted: 12/14/2024] [Indexed: 12/20/2024]
Abstract
Up to 90% of cancer-related fatalities could be attributed to metastasis. Therefore, understanding the mechanisms that facilitate tumor cell metastasis is beneficial for improving patient survival and results. EMT is considered the main process involved in the invasion and spread of CRC. Essential molecular components like Wnt, TGF-β, and PI3K/Akt play a role in controlling EMT in CRC, frequently triggered by various factors such as Snail, Twist, and ZEB1. These factors affect not only the spread of CRC but also determine the reaction to chemotherapy. The influence of non-coding RNAs, especially miRNAs and lncRNAs, on the regulation of EMT is clear in CRC. Exosomes, involved in cell-to-cell communication, can affect the TME and metastasis of CRC. Pharmacological substances and nanoparticles demonstrate promise as efficient modulators of EMT in CRC. Chitosan and HA are two major carbohydrate polymers with considerable potential in inhibiting CRC. Chitosan and HA can be employed to modify nanoparticles to enhance cargo transport for reducing CRC. Additionally, chitosan and HA-modified nanocarriers, which can be utilized as potential approaches in suppressing EMT and reversing drug resistance in CRC, can inhibit EMT and chemoresistance, crucial components in tumorigenesis.
Collapse
Affiliation(s)
- Mingming Han
- Department of Pharmacy and Medical Devices, Shandong Academy of Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China
| | - Xi Zhou
- Department of Occupational Pulmonology, Shandong Academy of Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China
| | - Hang Cheng
- Department of Bioanalytical Laboratory (ClinicalLaboratory), Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China
| | - Mengru Qiu
- Department of Occupational Pulmonology, Shandong Academy of Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China.
| | - Meng Qiao
- Department of Bioanalytical Laboratory (ClinicalLaboratory), Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China.
| | - Xiao Geng
- Department of Party Committee Office, Shandong Academy of Occupational Health and Occupational Medicine, Occupational Disease Hospital of Shandong First Medical University, Shandong Province Hospital Occupational Disease Hospital, Jinan, Shandong, China.
| |
Collapse
|
14
|
Wang S, Xie D, Yue H, Li G, Jiang B, Gao Y, Zheng Z, Zheng X, Wu G. Phospholipase C Beta 2 as a Key Regulator of Tumor Progression and Epithelial-Mesenchymal Transition via PI3K/AKT Signaling in Renal Cell Carcinoma. Biomedicines 2025; 13:304. [PMID: 40002717 PMCID: PMC11853330 DOI: 10.3390/biomedicines13020304] [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: 12/05/2024] [Revised: 12/24/2024] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Renal cell carcinoma (RCC) represents the most common form of invasive kidney cancer in adults. Among the components critical to cellular regulation is Phospholipase C Beta 2 (PLCB2), a member of the phospholipase C enzyme family. This enzyme plays a vital role in managing key cellular functions such as growth, differentiation, migration, and survival. Despite its significant importance, the specific expression patterns and molecular mechanisms of PLCB2 in the progression of RCC are not well understood. Methods: This investigation employed a combination of bioinformatics analyses, scRNA-seq, functional assays, transcriptome sequencing, real-time quantitative PCR (RT-PCR), immunofluorescence, rescue experiments, and Western blotting to explore the regulatory function of PLCB2 in driving the epithelial-mesenchymal transition (EMT) in RCC through the PI3K/AKT signaling pathway. Results:PLCB2 expression is significantly elevated in RCC samples, and this increase is inversely correlated with patient prognosis. The knockdown of PLCB2 in RCC cell lines leads to a marked reduction in cell proliferation, invasion, migration, and EMT. Transcriptome sequencing further revealed that PLCB2 is significantly associated with the PI3K/AKT pathway. Notably, the PI3K activator 740Y-P was able to reverse the reductions in migration, invasion, and EMT caused by the PLCB2 knockdown. Conclusions: Our findings underscore the pivotal role of PLCB2 in regulating RCC invasion and metastasis by modulating the EMT via the PI3K/AKT signaling pathway. This highlights PLCB2 not only as a key prognostic biomarker, but also as a promising therapeutic target in the treatment of advanced-stage RCC, offering new avenues for more effective interventions.
Collapse
Affiliation(s)
- Shijin Wang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Deqian Xie
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Hongzhe Yue
- Department of Emergency, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Guandu Li
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Bowen Jiang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yaru Gao
- Department of Nursing, The Second Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Zunwen Zheng
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xu Zheng
- Department of Cell Biology, College of Basic Medical Science, Dalian Medical University, Dalian 116011, China
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| |
Collapse
|
15
|
Jia Y, Yang N, Tang S, Deng L, Wang Y, Cai X. RUNX1 promotes proliferation of cervical cancer through TGFB2-MAPK pathway. Sci Rep 2025; 15:497. [PMID: 39747496 PMCID: PMC11696507 DOI: 10.1038/s41598-024-84254-x] [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: 08/21/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025] Open
Abstract
As cervical cancer (CC) caused more than 300,000 deaths in the world, it is urgent to identify therapeutic targets to improve survival. Though RUNX1 is overexpressed in CC, its specific role and underlying molecular mechanisms remain incompletely understood. Here we presented that RUNX1 was upregulated in CC and associated with poor prognosis. Functional studies demonstrated that RUNX1 acts as an oncogene in CC, with overexpression accelerating cell cycle progression and promoting cell proliferation. Mechanistically, RUNX1 regulates the MAPK pathway by modulating TGFB2 expression, while TGFB2 inhibition impaired MAPK pathway activation and the proliferation driven by RUNX1 overexpression. Comprehensive analyses also suggested that RUNX1 may modulate the immune microenvironment in CC through TGFB2. These findings indicate that RUNX1 promotes CC progression by activating the MAPK pathway through upregulation of TGFB2. Our study provides new insights into the role of RUNX1 in CC proliferation and suggests RUNX1 as a potential therapeutic target in CC.
Collapse
Affiliation(s)
- Yongqin Jia
- Department of Obstetrics and Gynecology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Neng Yang
- Department of Obstetrics and Gynecology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shuai Tang
- Department of Obstetrics and Gynecology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Li Deng
- Department of Obstetrics and Gynecology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yanzhou Wang
- Department of Obstetrics and Gynecology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Xiongwei Cai
- Department of Gynecology, Chongqing Health Center for Women and Children (Women and Children's Hospital of Chongqing Medical University), Chongqing, China.
| |
Collapse
|
16
|
Farombi EO, Ajayi BO, Ajeigbe OF, Maruf OR, Anyebe DA, Opafunso IT, Adedara IA. Mechanistic exploration of 6-shogaol's preventive effects on azoxymethane and dextran sulfate sodium -induced colorectal cancer: involvement of cell proliferation, apoptosis, carcinoembryonic antigen, wingless-related integration site signaling, and oxido-inflammation. Toxicol Mech Methods 2025; 35:1-10. [PMID: 39034841 DOI: 10.1080/15376516.2024.2381798] [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: 02/01/2024] [Revised: 06/04/2024] [Accepted: 07/14/2024] [Indexed: 07/23/2024]
Abstract
Colorectal cancer (CRC) poses a significant global health burden, being the third most prevalent cancer and the second most significant contributor to cancer-related deaths worldwide. Preventive strategies are crucial to combat this rising incidence. 6-shogaol, derived from ginger, has shown promise in preventing and treating various cancers. This study investigated the preventive effects of 6-shogaol on azoxymethane (AOM) and dextran sulfate sodium (DSS)-induced CRC in mice. Forty male BALB/c mice were randomly divided into control, 6-shogaol, AOM + DSS, and 6-shogaol + AOM + DSS. Mice in the control group received corn oil for 16 weeks, while those in the 6-Shogaol group were administered 20 mg/kg of 6-shogaol for 16 weeks. The AOM + DSS group received a single intraperitoneal dose (ip) of 10 mg/kg of AOM, followed by three cycles of 2.5% DSS in drinking water. The 6-shogaol + AOM + DSS group received both 6-shogaol for 16 weeks and a single ip of 10 mg/kg of AOM, followed by three cycles of 2.5% DSS in drinking water. The AOM + DSS-treated mice exhibited reduced food consumption, colon weight, and colon length, along with increased tumor formation. Co-administration of 6-shogaol effectively reversed these changes, inhibiting CRC development. Histopathological analysis revealed protective effects of 6-shogaol against colonic insults and modulation of inflammatory responses. 6-shogaol significantly reduced Carcinoembryonic antigen and Kiel 67 levels, indicating inhibition of tumor cell proliferation. Mechanistically, 6-shogaol promoted apoptosis by upregulating protein 53 and caspase-3 expression, and it effectively restored the balance of the Wingless-related integration site signaling pathway by regulating β-catenin and adenomatous polyposis coli levels. Moreover, 6-shogaol demonstrated anti-inflammatory effects, reducing myeloperoxidase, Tumor necrosis factor alpha, and cyclooxygenase-2 levels in AOM/DSS-treated mice. Additionally, 6-shogaol restored redox homeostasis by reducing lipid peroxidation and nitrosative stress and enhancing antioxidant enzyme activities. The findings suggest that 6-shogaol inhibits cell proliferation, induces apoptosis, regulates Wnt signaling, suppresses inflammation, and restores redox homeostasis, providing comprehensive insights into its potential therapeutic benefits for CRC.
Collapse
Affiliation(s)
- Ebenezer Olatunde Farombi
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Department of Biochemistry, College of Natural and Applied Sciences, Chrisland University, Abeokuta, Nigeria
| | - Babajide Oluwaseun Ajayi
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Oncopreventives and Systems Oncology Research Laboratory, Biochemistry Unit, Department of Chemical Sciences, Ajayi Crowther University, Oyo, Nigeria
| | - Olufunke Florence Ajeigbe
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Opeyemi Rabiat Maruf
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Daniel Abu Anyebe
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Ifeoluwa Tobi Opafunso
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Isaac Adegboyega Adedara
- Drug Metabolism & Toxicology Research Laboratories, Department of Biochemistry, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
17
|
Wu H, Luo H, Wang M, Du Y, Li J. NAP1L5 promotes epithelial-mesenchymal transition by regulating PEG10 expression in acute myeloid leukaemia. Leuk Res 2025; 148:107623. [PMID: 39579659 DOI: 10.1016/j.leukres.2024.107623] [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: 07/31/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 11/25/2024]
Abstract
Acute myeloid leukaemia (AML) is a haematological malignancy that poses a serious threat to human health. Studies have shown that the expression of NAP1L5 is elevated in patients with AML; however, the specific molecular mechanism remains unknown. Therefore, in this study, we aimed to investigate the pathogenic mechanisms of NAP1L5 in AML. The expression level of NAP1L5 was increased in AML, and the upregulation of NAP1L5 was related to tumour growth and epithelial-mesenchymal transition. Furthermore, PEG10 is a downstream regulatory factor of NAP1L5, and its overexpression promotes tumour growth and epithelial-mesenchymal transition. More importantly, the loss of PEG10 inhibited the negative effects induced by NAP1L5 overexpression. Our results suggest that NAP1L5 is a novel therapeutic target for AML treatment.
Collapse
Affiliation(s)
- Huan Wu
- School of Mental Health, Bengbu Medical University, Bengbu, Anhui 233000, China; Department of Hematology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233000, China
| | - Hang Luo
- Department of Hematology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233000, China; Department of Clinical Medicine, Bengbu Medical University, Bengbu, Anhui 233000, China
| | - Meng Wang
- Department of Hematology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233000, China
| | - YuQing Du
- Department of Hematology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233000, China
| | - Jiajia Li
- Department of Hematology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui 233000, China.
| |
Collapse
|
18
|
Zhang Y, Yang J, Shao T, Chen J, Shu Q, Shou L. Exploration of genetic characterization in hyperprogressive disease after immunotherapy retreatment in a patient with LCNEC: A case report. Hum Vaccin Immunother 2024; 20:2313281. [PMID: 38348622 PMCID: PMC10865920 DOI: 10.1080/21645515.2024.2313281] [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: 09/12/2023] [Accepted: 01/30/2024] [Indexed: 02/15/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have emerged as a promising therapeutic option for large cell neuroendocrine carcinoma (LCNEC). However, various studies have suggested a potential risk of hyperprogressive disease (HPD) in patients receiving ICI, which might be associated with gene alterations. Here, this is the first report on an unknown primary LCNEC patient who had achieved a long-term response from ICI treatment (atezolizumab), but developed HPD after tumor progression due to receiving another ICI agent (serplulimab). The mutation region of FAT4, SMARCA4, CYLD, CTNNB1, and KIT was altered prior to serplulimab treatment compared to before atezolizumab treatment. This case suggested a potential association between these mutated genes and HPD. Patients with the aforementioned genes should caution when selecting ICI treatment. These findings required further confirmation in a larger study cohort.
Collapse
Affiliation(s)
- Yao Zhang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiayao Yang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Tianyu Shao
- Department of Oncology, Guang’ Anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jialu Chen
- Department of Oncology, Hangzhou Third People’s Hospital, Hangzhou, China
| | - Qijin Shu
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Liumei Shou
- Department of Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| |
Collapse
|
19
|
Zhou J, Zhou D, Zhang Q, Zhang X, Liu X, Ding L, Wen J, Xu X, Cheng Z. DCLK1 mediated cooperative acceleration of EMT by avian leukosis virus subgroup J and Marek's disease virus via the Wnt/β-catenin pathway promotes tumor metastasis. J Virol 2024; 98:e0111224. [PMID: 39445786 PMCID: PMC11575233 DOI: 10.1128/jvi.01112-24] [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: 07/26/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024] Open
Abstract
Co-infection with oncogenic retrovirus and herpesvirus significantly facilitates tumor metastasis in human and animals. Co-infection with avian leukosis virus subgroup J (ALV-J) and Marek's disease virus (MDV), which are typical oncogenic retrovirus and herpesvirus, respectively, leads to enhanced oncogenicity and accelerated tumor formation, resulting in increased mortality of affected chickens. Previously, we found that ALV-J and MDV cooperatively promoted tumor metastasis. However, the molecular mechanism remains elusive. Here, we found that doublecortin-like kinase 1 (DCLK1) mediated cooperative acceleration of epithelial-mesenchymal transition (EMT) by ALV-J and MDV promoted tumor metastasis. Mechanistically, DCLK1 induced EMT via activating Wnt/β-catenin pathway by interacting with β-catenin, thereby cooperatively promoting tumor metastasis. Initially, we screened and found that DCLK1 was a potential mediator for the cooperative activation of EMT by ALV-J and MDV, and enhanced cell proliferation, migration, and invasion. Subsequently, we revealed that DCLK1 physically interacted with β-catenin to promote the formation of the β-catenin-TCF4 complex, inducing transcription of the Wnt target gene, c-Myc, promoting EMT by increasing the expression of N-cadherin, Vimentin, and Snail, and decreasing the expression of E-cadherin. Taken together, we discovered that jointly activated DCLK1 by ALV-J and MDV accelerated cell proliferation, migration and invasion, and ultimately activated EMT, paving the way for tumor metastasis. This study elucidated the molecular mechanism underlying cooperative metastasis induced by co-infection with retrovirus and herpesvirus. IMPORTANCE Tumor metastasis, a complex phenomenon in which tumor cells spread to new organs, is one of the greatest challenges in cancer research and is the leading cause of cancer-induced death. Numerous studies have shown that oncoviruses and their encoded proteins significantly affect metastasis, especially the EMT process. ALV-J and MDV are classic tumorigenic retrovirus and herpesvirus, respectively. We found that ALV-J and MDV synergistically promoted EMT. Further, we identified the tumor stem cell marker DCLK1 in ALV-J and MDV co-infected cells. DCLK1 directly interacted with β-catenin, promoting the formation of the β-catenin-TCF4 complex. This interaction activated the Wnt/β-catenin pathway, thereby inducing EMT and paving the way for synergistic tumor metastasis. Exploring the molecular mechanisms by which ALV-J and MDV cooperate during EMT will contribute to our understanding of tumor progression and metastasis. This study provides new insights into the cooperative induced tumor metastasis by retroviruses and herpesviruses.
Collapse
Affiliation(s)
- Jing Zhou
- College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| | - Defang Zhou
- College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| | - Qian Zhang
- Department of Neurology, The Affiliated Taian City Central Hospital of Qingdao University, Taian, Shandong, China
| | - Xinyue Zhang
- College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| | - Xiaoyang Liu
- College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| | - Longying Ding
- College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| | - Jing Wen
- College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| | - Xiaoyu Xu
- College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| | - Ziqiang Cheng
- College of Veterinary Medicine, Shandong Agricultural University, Taian, Shandong, China
| |
Collapse
|
20
|
Kalaei Z, Shekarchi AA, Hojjat-Farsangi M, Jalali P, Jadidi-Niaragh F. The prognostic and therapeutic potential of vimentin in colorectal cancer. Mol Biol Rep 2024; 51:1027. [PMID: 39347868 DOI: 10.1007/s11033-024-09965-w] [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: 06/15/2024] [Accepted: 09/20/2024] [Indexed: 10/01/2024]
Abstract
Several cells and molecules in the tumor microenvironment have been introduced as effective factors in the prognosis and progression of colorectal cancer. As a key element of the intermediate filament family, vimentin is expressed by mesenchymal cells in a ubiquitous manner and contributes significantly to cellular integrity and stress resistance in colorectal cancer. Recent studies have shown that alterations in the expression patterns of intermediate filaments are significantly related to cancer progression, especially in phenotypes associated with cellular migration and invasion. In addition to its multiple biological roles, vimentin also has a substantial function in mediating the epithelial-mesenchymal transition. Therefore, evaluating vimentin as an effective factor involved in the prognosis of colorectal cancer and targeting it as a novel approach to cancer therapy have become one of the main goals of many researchers worldwide. In this article, we will review the various biological functions of vimentin, as well as its relationship with colorectal cancer with the aim of providing novel insights into its clinical importance in the prognosis and treatment of colorectal cancer.
Collapse
Affiliation(s)
- Zahra Kalaei
- Department of Biology, Faculty of Natural Sciences, Tabriz University, Tabriz, Iran
| | - Ali Akbar Shekarchi
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
21
|
Forbes AN, Xu D, Cohen S, Pancholi P, Khurana E. Discovery of therapeutic targets in cancer using chromatin accessibility and transcriptomic data. Cell Syst 2024; 15:824-837.e6. [PMID: 39236711 PMCID: PMC11415227 DOI: 10.1016/j.cels.2024.08.004] [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: 01/07/2022] [Revised: 09/22/2023] [Accepted: 08/08/2024] [Indexed: 09/07/2024]
Abstract
Most cancer types lack targeted therapeutic options, and when first-line targeted therapies are available, treatment resistance is a huge challenge. Recent technological advances enable the use of assay for transposase-accessible chromatin with sequencing (ATAC-seq) and RNA sequencing (RNA-seq) on patient tissue in a high-throughput manner. Here, we present a computational approach that leverages these datasets to identify drug targets based on tumor lineage. We constructed gene regulatory networks for 371 patients of 22 cancer types using machine learning approaches trained with three-dimensional genomic data for enhancer-to-promoter contacts. Next, we identified the key transcription factors (TFs) in these networks, which are used to find therapeutic vulnerabilities, by direct targeting of either TFs or the proteins that they interact with. We validated four candidates identified for neuroendocrine, liver, and renal cancers, which have a dismal prognosis with current therapeutic options.
Collapse
Affiliation(s)
- Andre Neil Forbes
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Duo Xu
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Sandra Cohen
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Priya Pancholi
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Ekta Khurana
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY 10065, USA; Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; Caryl and Israel Englander Institute for Precision Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
22
|
Chen X, Tu J, Yang M, Wang Y, Liu B, Qiu H, Yuan X. RUNX1-MUC13 Interaction Activates Wnt/β-Catenin Signaling Implications for Colorectal Cancer Metastasis. Int J Biol Sci 2024; 20:4999-5026. [PMID: 39309442 PMCID: PMC11414392 DOI: 10.7150/ijbs.98396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/07/2024] [Indexed: 09/25/2024] Open
Abstract
Background: Colorectal cancer (CRC) remains a significant global health challenge, often characterized by late-stage metastasis and poor prognosis. The Runt-related transcription factor 1 (RUNX1) plays a dual role as both an oncogene and a tumor suppressor in various cancers, including CRC. However, the specific regulatory mechanisms of RUNX1 in CRC, particularly its direct roles, are not fully understood. Objective: This study aimed to investigate the role of RUNX1 in CRC progression and its interaction with Mucin 13 (MUC13) as a potential regulatory target. Methods: RUNX1 expression was analyzed in CRC tissues and cell lines compared to controls. In vitro and in vivo assays were conducted to assess the effects of RUNX1 overexpression and knockdown on cell behavior. ChIP-seq and RNA-seq analyses were performed to identify RUNX1 targets, with a focus on MUC13. Results: RUNX1 expression was significantly upregulated in CRC tissues and cells, correlating with advanced pathological characteristics and poor patient outcomes. RUNX1 overexpression enhanced CRC cell proliferation, migration, invasion, and G2/M phase arrest, while its knockdown had the opposite effects. MUC13 was identified as a direct transcriptional target of RUNX1, with its expression contributing to the activation of the Wnt/β-catenin signaling pathway. Disruption of MUC13 partially reversed the malignant phenotypes induced by RUNX1. Conclusion: RUNX1 promotes CRC progression by upregulating MUC13 and activating the Wnt/β-catenin pathway. This RUNX1-MUC13 axis represents a potential therapeutic target for managing CRC.
Collapse
Affiliation(s)
| | | | | | | | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hong Qiu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
23
|
Ren Y, Sun X, Chen X, Shao S, Tang J, Xu Z, Xu Y, Kang H, Wang L. The transcription factor ZNF248 promotes colorectal cancer metastasis by binding to ZEB1. J Cancer 2024; 15:5440-5450. [PMID: 39247604 PMCID: PMC11375549 DOI: 10.7150/jca.92886] [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: 12/04/2023] [Accepted: 07/05/2024] [Indexed: 09/10/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors globally, with metastasis emerging as the leading cause of mortality in CRC patients. Transcription factors play pivotal roles in the metastatic process. Using bioinformatics tools, we analyzed the TCGA-COAD and GES146587 datasets and identified ZNF248 participating in tumor progression. By analyzing 100 CRC patient tissues, it is found that ZNF248 is highly expressed in cancer tissue as well as in CRC cell lines identified by qRT-PCR. Our study discovered that ZNF248 enhances CRC cell migratory and invasive capabilities. A positive correlation was found between ZNF248 and epithelial-mesenchymal transition (EMT)-related markers (ZEB1, snail1), while E-cadherin exhibited a negative correlation with ZNF248. In addition, the analysis of the TCGA dataset demonstrated a strong correlation between the mRNA level of ZNF248 and ZEB1 expressions. Furthermore, it is found that the overexpression of ZEB1 could reverse CRC cell invasion and migration, along with the inhibition on EMT marker expressions induced by the RNA interference with ZNF248. Immunohistochemical analysis indicated a substantial association of ZNF248 expression with the lymph node metastasis, and with the liver metastasis (P =0.01, P =0.01), and a positive correlation between ZNF248 and ZEB1 expression (P =0.021) was also identified. Using Chip-PCR assay, it is found that ZNF248 bound to the ZEB1 promoter region. These findings showed that ZNF248 promotes CRC metastasis in vivo, revealed its role as an oncogene in CRC by targeting ZEB1 and activating the EMT pathway, which provided novel and promising biomarkers for CRC therapy through targeting ZEB1.
Collapse
Affiliation(s)
- Yanying Ren
- The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- Dalian Medical University, Dalian, Liaoning Province, China
| | - Xiaoxu Sun
- Dalian Medical University, Dalian, Liaoning Province, China
| | - Xin Chen
- The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Shuai Shao
- The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - JingTong Tang
- Department of Gastrointestinal Surgery, the first Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Zhaohui Xu
- The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Yang Xu
- The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- Dalian Medical University, Dalian, Liaoning Province, China
| | - Haonan Kang
- The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- Dalian Medical University, Dalian, Liaoning Province, China
| | - Liming Wang
- The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
- Dalian Medical University, Dalian, Liaoning Province, China
| |
Collapse
|
24
|
Chang Z, Liu B, He H, Li X, Shi H. High expression of RUNX1 in colorectal cancer subtype accelerates malignancy by inhibiting HMGCR. Pharmacol Res 2024; 206:107293. [PMID: 38971271 DOI: 10.1016/j.phrs.2024.107293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/09/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Colorectal cancer (CRC) presents a complex landscape, characterized by both inter-tumor and intra-tumor heterogeneity. RUNX1, a gene implicated in modulating tumor cell growth, survival, and differentiation, remains incompletely understood regarding its impact on CRC prognosis. In our investigation, we discerned a positive correlation between elevated RUNX1 expression and aggressive phenotypes across various CRC subtypes. Notably, knockdown of RUNX1 demonstrated efficacy in restraining CRC proliferation both in vitro and in vivo, primarily through inducing apoptosis and impeding cell proliferation. Mechanistically, we unveiled a direct regulatory link between RUNX1 and cholesterol synthesis, mediated by its control over HMGCR expression. Knockdown of RUNX1 in CRC cells triggered HMGCR transcriptional activation, culminating in elevated cholesterol levels that subsequently hindered cancer progression. Clinically, heightened RUNX1 expression emerged as a prognostic marker for adverse outcomes in CRC patients. Our findings underscore the pivotal involvement of RUNX1 in CRC advancement and its potential as a therapeutic target. The unique influence of RUNX1 on cholesterol synthesis and HMGCR transcriptional regulation uncovers a novel pathway contributing to CRC progression.
Collapse
Affiliation(s)
- Zhilin Chang
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Bing Liu
- Laboratory of Experimental Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China; State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Han He
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China.
| | - Xiaoyan Li
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Hui Shi
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
25
|
Wang Q, Lu W, Lu L, Wu R, Wu D. miR-575/RIPK4 axis modulates cell cycle progression and proliferation by inactivating the Wnt/β-catenin signaling pathway through inhibiting RUNX1 in colon cancer. Mol Cell Biochem 2024; 479:1747-1766. [PMID: 38480605 DOI: 10.1007/s11010-024-04938-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 01/10/2024] [Indexed: 07/18/2024]
Abstract
Receptor interacting protein serine/threonine kinase 4 (RIPK4) is widely involved in human cancer development. Nevertheless, its role in colon cancer (COAD) has not been elucidated till now. Our research aimed at exploring the function and underlying molecular mechanism of RIPK4 in COAD progression. Through bioinformatic analyses and RT-qPCR, RIPK4 was discovered to be increased in COAD cells and tissues, and its high level predicted poor prognosis. Loss-of-function assays revealed that RIPK4 silencing suppressed COAD cell growth, induced cell cycle arrest, and enhanced cell apoptosis. In vivo experiments also proved that tumor growth was inhibited by silencing of RIPK4. Luciferase reporter assay validated that RIPK4 was targeted and negatively regulated by miR-575. Western blotting demonstrated that Wnt3a, phosphorylated (p)-GSK-3β, and cytoplasmic and nuclear β-catenin protein levels, β-catenin nuclear translocation, and Cyclin D1, CDK4, Cyclin E, and c-Myc protein levels were reduced by RIPK4 knockdown, which however was reversed by treatment with LiCl, the Wnt/β-catenin pathway activator. LiCl also offset the influence of RIPK4 knockdown on COAD cell growth, cell cycle process, and apoptosis. Finally, RIPK4 downregulation reduced RUNX1 level, which was upregulated in COAD and its high level predicted poor prognosis. RIPK4 is positively associated with RUNX1 in COAD. Overexpressing RUNX1 antagonized the suppression of RIPK4 knockdown on RUNX1, Wnt3a, p-GSK-3β, cytoplasmic β-catenin, nuclear β-catenin, Cyclin D1, CDK4, Cyclin E, and c-Myc levels. Collectively, miR-575/RIPK4 axis repressed COAD progression via inactivating the Wnt/β-catenin pathway through downregulating RUNX1.
Collapse
Affiliation(s)
- Qun Wang
- Department of Hepatopancreatobiliary Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 16 Zhuodaoquan South Road, Hongshan District, Wuhan, 430079, China.
- Colorectal Cancer Clinical Research Center of Wuhan, Wuhan, 430079, China.
- Colorectal Cancer Clinical Research Center of Hubei Province, Wuhan, 430079, China.
| | - Weijun Lu
- Department of Hepatopancreatobiliary Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 16 Zhuodaoquan South Road, Hongshan District, Wuhan, 430079, China
- Colorectal Cancer Clinical Research Center of Wuhan, Wuhan, 430079, China
| | - Li Lu
- Colorectal Cancer Clinical Research Center of Wuhan, Wuhan, 430079, China
- Department of Gastrointestinal Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, China
| | - Ruopu Wu
- Tianjin Medical University, Tianjin, 300070, China
| | - Dongde Wu
- Department of Hepatopancreatobiliary Surgery, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 16 Zhuodaoquan South Road, Hongshan District, Wuhan, 430079, China.
| |
Collapse
|
26
|
Wang H, Zhang L, Hu C, Li H, Jiang M. Wnt signaling and tumors (Review). Mol Clin Oncol 2024; 21:45. [PMID: 38798312 PMCID: PMC11117032 DOI: 10.3892/mco.2024.2743] [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: 02/02/2023] [Accepted: 04/23/2024] [Indexed: 05/29/2024] Open
Abstract
Wnt signaling is a highly conserved evolutionary pathway that plays a key role in regulation of embryonic development, as well as tissue homeostasis and regeneration. Abnormalities in Wnt signaling are associated with tumorigenesis and development, leading to poor prognosis in patients with cancer. However, the pharmacological effects and mechanisms underlying Wnt signaling and its inhibition in cancer treatment remain unclear. In addition, potential side effects of inhibiting this process are not well understood. Therefore, the present review outlines the role of Wnt signaling in tumorigenesis, development, metastasis, cancer stem cells, radiotherapy resistance and tumor immunity. The present review further identifies inhibitors that target Wnt signaling to provide a potential novel direction for cancer treatment. This may facilitate early application of safe and effective drugs targeting Wnt signaling in clinical settings. An in-depth understanding of the mechanisms underlying inhibition of Wnt signaling may improve the prognosis of patients with cancer.
Collapse
Affiliation(s)
- Huaishi Wang
- Department of Pulmonary and Critical Care Medicine, Xiangtan Central Hospital, Xiangtan, Hunan 411100, P.R. China
| | - Lihai Zhang
- Department of Pulmonary and Critical Care Medicine, Xiangtan Central Hospital, Xiangtan, Hunan 411100, P.R. China
| | - Chao Hu
- Department of Pulmonary and Critical Care Medicine, Xiangtan Central Hospital, Xiangtan, Hunan 411100, P.R. China
| | - Hui Li
- Department of Pulmonary and Critical Care Medicine, Xiangtan Central Hospital, Xiangtan, Hunan 411100, P.R. China
| | - Mingyan Jiang
- Department of Pulmonary and Critical Care Medicine, Xiangtan Central Hospital, Xiangtan, Hunan 411100, P.R. China
| |
Collapse
|
27
|
Lu X, Yang Y, Chen J, Zhao T, Zhao X. RUNX1/miR-429 feedback loop promotes growth, metastasis, and epithelial-mesenchymal transition in oral squamous cell carcinoma by targeting ITGB1. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:5289-5302. [PMID: 38277041 DOI: 10.1007/s00210-024-02960-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/15/2024] [Indexed: 01/27/2024]
Abstract
This study aimed to explore the role of miR-429 on the progression of oral squamous cell carcinoma (OSCC). OSCC cell lines were transfected with miR-429 mimic, pcDNA3.1-RUNX1, or pcDNA3.1-ITGB1, and their cell viability, apoptosis, migration, and invasion abilities were analyzed by cell counting, terminal deoxynucleotidyl transferase dUTP nick-end labeling staining, wound healing, and transwell assays, respectively. Furthermore, luciferase reporter assay, RNA pull-down, and ChIP were used to assess the regulation of miR-429, RUNX1, and ITGB1 expression in OSCC. Lastly, the biological role of the RUNX1/miR-429 feedback loop was explored in nude mice. The results revealed that miR-429 level was down-regulated, while RUNX1 and ITGB1 levels were up-regulated in OSCC tissues and that miR-429 was negatively correlated with RUNX1 and ITGB1 in OSCC tissues. Transfection of miR-429 mimic suppressed OSCC cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT). Moreover, we found that miR-429 participated in OSCC progression by directly targeting ITGB1. Additionally, we found that RUNX1 negatively regulated miR-429 expression by binding to its promoter. Our results also revealed that the RUNX1/miR-429 feedback loop regulated ITGB1 expression and that RUNX1 overexpression rescued the inhibitory effects of miR-429 mimic on OSCC cells. In addition, miR-429 mimic significantly suppressed tumor growth, inflammatory cell infiltration, EMT, and ITGB1 expression in vivo, which were inhibited by RUNX1 overexpression. Altogether, these results indicate that the RUNX1/miR-429 feedback loop promoted growth, metastasis, and EMT in OSCC by targeting ITGB1.
Collapse
Affiliation(s)
- Xun Lu
- Hospital of Stomatology, General Hospital of Ningxia Medical University, 804 Shengli South Street, Xingqing District, Yinchuan City, Ningxia, 750004, China
| | - Yiqiang Yang
- Hospital of Stomatology, General Hospital of Ningxia Medical University, 804 Shengli South Street, Xingqing District, Yinchuan City, Ningxia, 750004, China
| | - Jia Chen
- Hospital of Stomatology, General Hospital of Ningxia Medical University, 804 Shengli South Street, Xingqing District, Yinchuan City, Ningxia, 750004, China
| | - Tian Zhao
- Hospital of Stomatology, General Hospital of Ningxia Medical University, 804 Shengli South Street, Xingqing District, Yinchuan City, Ningxia, 750004, China
| | - Xiaofan Zhao
- Hospital of Stomatology, General Hospital of Ningxia Medical University, 804 Shengli South Street, Xingqing District, Yinchuan City, Ningxia, 750004, China.
| |
Collapse
|
28
|
Xu F, Wang W, Li Q, Zou L, Miao H. The roles and mechanisms of APOL1 in the development of colorectal cancer. J Gastrointest Oncol 2024; 15:974-986. [PMID: 38989412 PMCID: PMC11231876 DOI: 10.21037/jgo-24-275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 05/28/2024] [Indexed: 07/12/2024] Open
Abstract
Background Research has demonstrated that apolipoprotein L1 (APOL1) has a role in the emergence and progression of a number of malignant cancers. It is unclear, however, how APOL1 functions in colorectal cancer (CRC). In this study, we examined the possible molecular processes underlying APOL1's biological role in CRC. Methods Quantitative real-time polymerase chain reaction (qRT-PCR) was used to identify APOL1 expression in patients with CRC and the cell line of cancer tissue. Following transfection of human colon carcinoma cells (HCT116) and human colon adenocarcinoma cells (SW1116) with sh-APOL1, the effects of APOL1 on the biological behavior of CRC cell lines were examined. In nude mice, the effect of APOL1 on tumor growth was noted. The protein interaction between APOL1 and RUNX1 was detected via coimmunoprecipitation. The expression of relevant proteins and cell biological behaviors were examined to confirm the APOL1-RUNX1 pathway in CRC cell lines. Results The CRC tissues and cells exhibited elevated expression of APOL1. HCT116 and SW1116 cells' proliferation, migration, and invasion were suppressed by sh-APOL1, and sh-APOL1 also increased the expression of E-cadherin and decreased the expression of RUNX1, cyclin D1, β-catenin, N-cadherin, and vimentin. APOL1 bound to the RUNX1 protein and regulated its protein levels. The counteractive effect of sh-APOL1 epithelial-mesenchymal transition (EMT), proliferation, migration, and invasion of CRC cells was counteracted by the overexpression of RUNX1. By silencing APOL1, the Wnt-β-catenin pathway was able to restrain EMT and regulate the biological behavior processes in CRC cells. Conclusions APOL1 has potential as a diagnostic biomarker for CRC. By preventing the Wnt-β-catenin pathway from being activated, the sh-APOL1-binding protein RUNX1 inhibited the EMT and biological behavior of CRC cells.
Collapse
Affiliation(s)
- Feipeng Xu
- Department of General Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Weiwei Wang
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Qidong Li
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lirui Zou
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Huilai Miao
- Department of General Surgery, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Department of Gastrointestinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
29
|
Tian W, Zhao J, Zhang X, Li P, Li X, Hong Y, Li S. RUNX1 regulates MCM2/CDC20 to promote COAD progression modified by deubiquitination of USP31. Sci Rep 2024; 14:13906. [PMID: 38886545 PMCID: PMC11183096 DOI: 10.1038/s41598-024-64726-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
Colon adenocarcinoma (COAD) is the second leading cause of cancer death, and there is still a lack of diagnostic biomarkers and therapeutic targets. In this study, bioinformatics analysis of the TCGA database was used to obtain RUNX1, a gene with prognostic value in COAD. RUNX1 plays an important role in many malignancies, and its molecular regulatory mechanisms in COAD remain to be fully understood. To explore the physiological role of RUNX1, we performed functional analyses, such as CCK-8, colony formation and migration assays. In addition, we investigated the underlying mechanisms using transcriptome sequencing and chromatin immunoprecipitation assays. RUNX1 is highly expressed in COAD patients and significantly correlates with survival. Silencing of RUNX1 significantly slowed down the proliferation and migratory capacity of COAD cells. Furthermore, we demonstrate that CDC20 and MCM2 may be target genes of RUNX1, and that RUNX1 may be physically linked to the deubiquitinating enzyme USP31, which mediates the upregulation of RUNX1 protein to promote transcriptional function. Our results may provide new insights into the mechanism of action of RUNX1 in COAD and reveal potential therapeutic targets for this disease.
Collapse
Affiliation(s)
- Wei Tian
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Dalian Medical University, Dalian, China
| | - Jingyuan Zhao
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xinyu Zhang
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Dalian Medical University, Dalian, China
| | - Pengfei Li
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Dalian Medical University, Dalian, China
| | - Xuening Li
- Dalian Medical University, Dalian, China
| | - Yuan Hong
- Clinical Laboratory Center, Dalian Municipal Central Hospital, Dalian, China.
| | - Shuai Li
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
30
|
Hirose Y, Sato S, Hashiya K, Ooga M, Bando T, Sugiyama H. Chb-M', an Inhibitor of the RUNX Family Binding to DNA, Induces Apoptosis in p53-Mutated Non-Small Cell Lung Cancer and Inhibits Tumor Growth and Repopulation In Vivo. J Med Chem 2024; 67:9165-9172. [PMID: 38803164 DOI: 10.1021/acs.jmedchem.4c00315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Runt-related transcription factor (RUNX) proteins are considered to play various roles in cancer. Here, we evaluated the anticancer activity of Chb-M', a compound that specifically and covalently binds to the consensus sequence for RUNX family proteins, in p53-mutated non-small cell lung cancer cells. Chb-M' killed the cancer cells by inducing apoptosis. The compound showed an anticancer effect comparable to that of the clinically used drugs alectinib and ceritinib in vivo. Notably, Chb-M' extended the cancer-free survival of mice after ending treatment more effectively than did the other two drugs. The results presented here suggest that Chb-M' is an attractive candidate as an anticancer drug applicable to the treatment of non-small cell lung cancer and various other types of cancers.
Collapse
Affiliation(s)
- Yuki Hirose
- Department of Chemistry, Graduate School of Science, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Shinsuke Sato
- Department of Chemistry, Graduate School of Science, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Kaori Hashiya
- Department of Chemistry, Graduate School of Science, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Mitsuharu Ooga
- Department of Chemistry, Graduate School of Science, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Toshikazu Bando
- Department of Chemistry, Graduate School of Science, Kyoto University, Sakyo, Kyoto 606-8502, Japan
| | - Hiroshi Sugiyama
- Department of Chemistry, Graduate School of Science, Kyoto University, Sakyo, Kyoto 606-8502, Japan
- Institute for Integrated Cell-Material Science (WPI-iCeMS), Kyoto University, Sakyo, Kyoto 606-8501, Japan
| |
Collapse
|
31
|
TANG CHAO, ZHONG CHUNYU, ZHU JUNHAO, YUAN FENG, YANG JIN, XU YONG, MA CHIYUAN. GNAS mutations suppress cell invasion by activating MEG3 in growth hormone-secreting pituitary adenoma. Oncol Res 2024; 32:1079-1091. [PMID: 38827318 PMCID: PMC11136687 DOI: 10.32604/or.2024.046007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 12/08/2023] [Indexed: 06/04/2024] Open
Abstract
Approximately 30%-40% of growth hormone-secreting pituitary adenomas (GHPAs) harbor somatic activating mutations in GNAS (α subunit of stimulatory G protein). Mutations in GNAS are associated with clinical features of smaller and less invasive tumors. However, the role of GNAS mutations in the invasiveness of GHPAs is unclear. GNAS mutations were detected in GHPAs using a standard polymerase chain reaction (PCR) sequencing procedure. The expression of mutation-associated maternally expressed gene 3 (MEG3) was evaluated with RT-qPCR. MEG3 was manipulated in GH3 cells using a lentiviral expression system. Cell invasion ability was measured using a Transwell assay, and epithelial-mesenchymal transition (EMT)-associated proteins were quantified by immunofluorescence and western blotting. Finally, a tumor cell xenograft mouse model was used to verify the effect of MEG3 on tumor growth and invasiveness. The invasiveness of GHPAs was significantly decreased in mice with mutated GNAS compared with that in mice with wild-type GNAS. Consistently, the invasiveness of mutant GNAS-expressing GH3 cells decreased. MEG3 is uniquely expressed at high levels in GHPAs harboring mutated GNAS. Accordingly, MEG3 upregulation inhibited tumor cell invasion, and conversely, MEG3 downregulation increased tumor cell invasion. Mechanistically, GNAS mutations inhibit EMT in GHPAs. MEG3 in mutated GNAS cells prevented cell invasion through the inactivation of the Wnt/β-catenin signaling pathway, which was further validated in vivo. Our data suggest that GNAS mutations may suppress cell invasion in GHPAs by regulating EMT through the activation of the MEG3/Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- CHAO TANG
- Department of Neurosurgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - CHUNYU ZHONG
- Department of Neurosurgery, Children’s Hospital of Nanjing Medical University, Nanjing, 210019, China
| | - JUNHAO ZHU
- Department of Neurosurgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - FENG YUAN
- Department of Neurosurgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - JIN YANG
- Department of Neurosurgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - YONG XU
- Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210029, China
| | - CHIYUAN MA
- Department of Neurosurgery, Affiliated Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
- Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210029, China
- Department of Neurosurgery, Jinling Hospital, Southern Medical University, Nanjing, 210002, China
- Department of Neurosurgery, Jinling Hospital, Nanjing Medical University, Nanjing, 210002, China
| |
Collapse
|
32
|
Zheng W, Guo Y, Kahar A, Bai J, Zhu Q, Huang X, Li Y, Xu B, Jia X, Wu G, Zhang C, Zhu Y. RUNX1-induced upregulation of PTGS2 enhances cell growth, migration and invasion in colorectal cancer cells. Sci Rep 2024; 14:11670. [PMID: 38778047 PMCID: PMC11111780 DOI: 10.1038/s41598-024-60296-z] [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: 11/09/2022] [Accepted: 04/21/2024] [Indexed: 05/25/2024] Open
Abstract
Colorectal cancer (CRC) arises via the progressive accumulation of dysregulation in key genes including oncogenes and tumor-suppressor genes. Prostaglandin-endoperoxide synthase 2 (PTGS2, also called COX2) acts as an oncogenic driver in CRC. Here, we explored the upstream transcription factors (TFs) responsible for elevating PTGS2 expression in CRC cells. The results showed that PTGS2 silencing repressed cell growth, migration and invasion in HCT116 and SW480 CRC cells. The two fragments (499-981 bp) and (1053-1434 bp) were confirmed as the core TF binding profiles of the PTGS2 promoter. PTGS2 expression positively correlated with RUNX1 level in colon adenocarcinoma (COAD) samples using the TCGA-COAD dataset. Furthermore, RUNX1 acted as a positive regulator of PTGS2 expression by promoting transcriptional activation of the PTGS2 promoter via the 1086-1096 bp binding motif. In conclusion, our study demonstrates that PTGS2 upregulation induced by the TF RUNX1 promotes CRC cell growth, migration and invasion, providing an increased rationale for the use of PTGS2 inhibitors in CRC prevention and treatment.
Collapse
Affiliation(s)
- Weiwei Zheng
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, 450003, Henan, China
- Hepatobiliary Gastrointestinal Surgery Department, Red Star Hospital of the 13th Division of Xinjiang Production and Construction Corps, Hami, 839000, The Xinjiang Uygur Autonomous Region, China, China
- The Affiliated People's Hospital of Xinxiang Medical College, Xinxiang, 453000, Henan, China
| | - Yingchang Guo
- Department of Interventional Therapy, The First Affiliated Hospital of Xinxiang Medical College, Xinxiang, 453000, Henan, China
| | - Aihemaiti Kahar
- Hepatobiliary Gastrointestinal Surgery Department, Red Star Hospital of the 13th Division of Xinjiang Production and Construction Corps, Hami, 839000, The Xinjiang Uygur Autonomous Region, China, China
| | - Junwei Bai
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, 450003, Henan, China
| | - Qinhui Zhu
- Department of General Surgery, Shangcai People's Hospital, Zhumadian, 463800, Henan, China
| | - Xinli Huang
- Department of General Surgery, Suiping People's Hospital, Zhumadian, 463100, Henan, China
| | - Yuan Li
- Department of Anesthesiology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Bingyi Xu
- Weihui People's Hospital, Weihui, 453100, Henan, China
| | - Xueshan Jia
- Development Department, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| | - Gang Wu
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, 450003, Henan, China.
| | - Chao Zhang
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, 450003, Henan, China.
| | - Yuanzeng Zhu
- Department of Gastrointestinal Surgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, No. 7 Weiwu Road, Zhengzhou, 450003, Henan, China.
| |
Collapse
|
33
|
Jokelainen O, Rintala TJ, Fortino V, Pasonen-Seppänen S, Sironen R, Nykopp TK. Differential expression analysis identifies a prognostically significant extracellular matrix-enriched gene signature in hyaluronan-positive clear cell renal cell carcinoma. Sci Rep 2024; 14:10626. [PMID: 38724670 PMCID: PMC11082176 DOI: 10.1038/s41598-024-61426-3] [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: 10/28/2023] [Accepted: 05/06/2024] [Indexed: 05/12/2024] Open
Abstract
Hyaluronan (HA) accumulation in clear cell renal cell carcinoma (ccRCC) is associated with poor prognosis; however, its biology and role in tumorigenesis are unknown. RNA sequencing of 48 HA-positive and 48 HA-negative formalin-fixed paraffin-embedded (FFPE) samples was performed to identify differentially expressed genes (DEG). The DEGs were subjected to pathway and gene enrichment analyses. The Cancer Genome Atlas Kidney Renal Clear Cell Carcinoma (TCGA-KIRC) data and DEGs were used for the cluster analysis. In total, 129 DEGs were identified. HA-positive tumors exhibited enhanced expression of genes related to extracellular matrix (ECM) organization and ECM receptor interaction pathways. Gene set enrichment analysis showed that epithelial-mesenchymal transition-associated genes were highly enriched in the HA-positive phenotype. A protein-protein interaction network was constructed, and 17 hub genes were discovered. Heatmap analysis of TCGA-KIRC data identified two prognostic clusters corresponding to HA-positive and HA-negative phenotypes. These clusters were used to verify the expression levels and conduct survival analysis of the hub genes, 11 of which were linked to poor prognosis. These findings enhance our understanding of hyaluronan in ccRCC.
Collapse
Affiliation(s)
- Otto Jokelainen
- Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, Kuopio Campus, P.O. Box 1627, 70211, Kuopio, Finland.
- Department of Clinical Pathology, Kuopio University Hospital, Kuopio, Finland.
| | - Teemu J Rintala
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Vittorio Fortino
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | | | - Reijo Sironen
- Institute of Clinical Medicine, Pathology and Forensic Medicine, University of Eastern Finland, Kuopio Campus, P.O. Box 1627, 70211, Kuopio, Finland
- Department of Clinical Pathology, Kuopio University Hospital, Kuopio, Finland
| | - Timo K Nykopp
- Department of Surgery, Kuopio University Hospital, Kuopio, Finland
- Institute of Clinical Medicine, Surgery, University of Eastern Finland, Kuopio, Finland
| |
Collapse
|
34
|
Gadewal N, Natu A, Sen S, Rauniyar S, Bastikar V, Gupta S. Integrative epigenome-transcriptome analysis unravels cancer-specific over-expressed genes potentially regulating immune microenvironment in clear cell renal cell carcinoma. Biochim Biophys Acta Gen Subj 2024; 1868:130596. [PMID: 38471632 DOI: 10.1016/j.bbagen.2024.130596] [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: 06/16/2023] [Revised: 10/19/2023] [Accepted: 03/08/2024] [Indexed: 03/14/2024]
Abstract
BACKGROUND Clear cell Renal Cell Carcinoma (ccRCC) is the frequently diagnosed histological life-threatening tumor subtype in the urinary system. Integrating multi-omics data is emerging as a tool to provide a comprehensive view of biology and disease for better therapeutic interventions. METHOD We have integrated freely available ccRCC data sets of genome-wide DNA methylome, transcriptome, and active histone modification marks, H3K27ac, H3K4me1, and H3K4me3 specific ChIP-seq data to screen genes with higher expression. Further, these genes were filtered based on their effect on survival upon alteration in expression. RESULTS The six multi-omics-based identified genes, RUNX1, MSC, ADA, TREML1, TGFA, and VWF, showed higher expression with enrichment of active histone marks and hypomethylated CpG in ccRCC. In continuation, the identified genes were validated by an independent dataset and showed a correlation with nodal and metastatic status. Furthermore, gene ontology and pathway analysis revealed that immune-related pathways are activated in ccRCC patients. CONCLUSIONS The network analysis of six overexpressed genes suggests their potential role in an immunosuppressive environment, leading to tumor progression and poor prognosis. Our study shows that the multi-omics approach helps unravel complex biology for patient subtyping and proposes combination strategies with epi-drugs for more precise immunotherapy in ccRCC.
Collapse
Affiliation(s)
- Nikhil Gadewal
- Bioinformatics & Computational Biology Facility, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, MH, India; Center for Computational Biology & Translational Research, Amity Institute of Biotechnology, Amity University, Mumbai - Pune Expressway, Bhatan, Post - Somathne, Panvel, Mumbai, 410206, MH, India
| | - Abhiram Natu
- Epigenetics and Chromatin Biology Group, Gupta Lab, Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, MH, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400094, MH, India
| | - Siddhartha Sen
- Bioinformatics & Computational Biology Facility, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, MH, India
| | - Sukanya Rauniyar
- Epigenetics and Chromatin Biology Group, Gupta Lab, Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, MH, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400094, MH, India
| | - Virupaksha Bastikar
- Center for Computational Biology & Translational Research, Amity Institute of Biotechnology, Amity University, Mumbai - Pune Expressway, Bhatan, Post - Somathne, Panvel, Mumbai, 410206, MH, India
| | - Sanjay Gupta
- Epigenetics and Chromatin Biology Group, Gupta Lab, Cancer Research Institute, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, MH, India; Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400094, MH, India.
| |
Collapse
|
35
|
Huang H, Li Q, Tu X, Yu D, Zhou Y, Ma L, Wei K, Gao Y, Zhao G, Han R, Ye F, Ke C. DNA hypomethylation patterns and their impact on the tumor microenvironment in colorectal cancer. Cell Oncol (Dordr) 2024:10.1007/s13402-024-00933-x. [PMID: 38520647 DOI: 10.1007/s13402-024-00933-x] [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] [Accepted: 03/02/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND Recent research underscores the pivotal role of immune checkpoints as biomarkers in colorectal cancer (CRC) therapy, highlighting the dynamics of resistance and response to immune checkpoint inhibitors. The impact of epigenetic alterations in CRC, particularly in relation to immune therapy resistance, is not fully understood. METHODS We integrated a comprehensive dataset encompassing TCGA-COAD, TCGA-READ, and multiple GEO series (GSE14333, GSE37892, GSE41258), along with key epigenetic datasets (TCGA-COAD, TCGA-READ, GSE77718). Hierarchical clustering, based on Euclidean distance and Ward's method, was applied to 330 primary tumor samples to identify distinct clusters. The immune microenvironment was assessed using MCPcounter. Machine learning algorithms were employed to predict DNA methylation patterns and their functional enrichment, in addition to transcriptome expression analysis. Genomic mutation profiles and treatment response assessments were also conducted. RESULTS Our analysis delineated a specific tumor cluster with CpG Island (CGI) methylation, termed the Demethylated Phenotype (DMP). DMP was associated with metabolic pathways such as oxidative phosphorylation, implicating increased ATP production efficiency in mitochondria, which contributes to tumor aggressiveness. Furthermore, DMP showed activation of the Myc target pathway, known for tumor immune suppression, and exhibited downregulation in key immune-related pathways, suggesting a tumor microenvironment characterized by diminished immunity and increased fibroblast infiltration. Six potential therapeutic agents-lapatinib, RDEA119, WH.4.023, MG.132, PD.0325901, and AZ628-were identified as effective for the DMP subtype. CONCLUSION This study unveils a novel epigenetic phenotype in CRC linked to resistance against immune checkpoint inhibitors, presenting a significant step toward personalized medicine by suggesting epigenetic classifications as a means to identify ideal candidates for immunotherapy in CRC. Our findings also highlight potential therapeutic agents for the DMP subtype, offering new avenues for tailored CRC treatment strategies.
Collapse
Grants
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- 2021YDZ03 Medical Products Administration of Guangdong Province
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- QN2021012 Science and Technology Research Project of Hebei Higher Education Institutions
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 81902498,H2022405002 National Natural Science Foundation of China
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- 2019CFB177 Hubei Provincial Natural Science Foundation
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- Q20182105 Natural Science Foundation of Hubei Provincial Department of Education
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- CXPJJH11800001-2018333 Chen Xiao-ping Foundation for the development of science and technology of Hubei Provincial
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- WJ2021Q007 The Foundation of Health and Family planning Commission of Hubei Province
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 201810929005, 201810929009, 201810929068, 201813249010, S201910929009, S201910929045, S202013249005, S202013249008 and 202010929009 Innovation and entrepreneurship training program
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
- 2021JJXM009 The Scientific and Technological Project of Taihe hospital
Collapse
Affiliation(s)
- He Huang
- Department of General Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Qian Li
- Department of Oncology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Xusheng Tu
- Department of Emergency Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China
| | - Dongyue Yu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Yundong Zhou
- Shanghai Medical Innovation Fusion Biomedical Research Center, Shanghai, China
| | - Lifei Ma
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China
- College of Lab Medicine, Hebei North University, Zhangjiakou, Hebei, 075000, China
| | - Kongyuan Wei
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 420, 69120, Heidelberg, Germany
| | - Yuzhen Gao
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310016, China
| | - Guodong Zhao
- Faculty of Hepatopancreatobiliary Surgery, The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, 100853, China
| | - Ruiqin Han
- State Key Laboratory of Common Mechanism Research for Major Diseas, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Fangdie Ye
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, 200000, China.
| | - Chunlian Ke
- Department of General Surgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, 510630, China.
| |
Collapse
|
36
|
Wang P, Pan Y, Zhang Y, Chen C, Hu J, Wang X. Role of interferon-induced transmembrane protein family in cancer progression: a special focus on pancreatic cancer. Med Oncol 2024; 41:85. [PMID: 38472606 DOI: 10.1007/s12032-024-02308-6] [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: 12/26/2023] [Accepted: 01/19/2024] [Indexed: 03/14/2024]
Abstract
Human interferon-induced transmembrane protein family (IFITMs) consists of five main proteins. IFITM1, IFITM2, and IFITM3 can be induced by interferon, while IFITM5 and IFITM10 are insensitive to interferon. IFITMs has various functions, including well-researched antiviral effects. As a molecule whose expression is significantly increased by interferon in the immune microenvironment, IFITMs has drawn growing interest in recent years for their role in the cancer progression. Unlike antiviral effects, the role and mechanism of IFITMs in cancer progression have not been clearly studied, especially the role and molecular mechanism of IFITMs in pancreatic cancer are rarely reported in the literature. This article focuses on the role and potential mechanism of IFITMs in pancreatic cancer progression by analyzing the function and mechanism of IFITM1-3 in other cancers and conducting bioinformatics analysis using the databases, so as to provide a new target for pancreatic cancer therapy.
Collapse
Affiliation(s)
- Peipei Wang
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, 3-17 Renmin South Rd, Chengdu, 610041, Sichuan, China
| | - Yan Pan
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, 3-17 Renmin South Rd, Chengdu, 610041, Sichuan, China
| | - Yu Zhang
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, 3-17 Renmin South Rd, Chengdu, 610041, Sichuan, China
| | - Congliang Chen
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, 3-17 Renmin South Rd, Chengdu, 610041, Sichuan, China
| | - Junmei Hu
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, 3-17 Renmin South Rd, Chengdu, 610041, Sichuan, China
| | - Xia Wang
- Department of Immunology, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, 3-17 Renmin South Rd, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
37
|
Cao Q, Zhao M, Su Y, Liu S, Lin Y, Da H, Yue C, Liu Y, Jing D, Zhao Q, Liu N, Du J, Zuo Z, Fu Y, Chen A, Birnbaumer L, Yang Y, Dai B, Gao X. Chronic Stress Dampens Lactobacillus Johnsonii-Mediated Tumor Suppression to Enhance Colorectal Cancer Progression. Cancer Res 2024; 84:771-784. [PMID: 38190716 DOI: 10.1158/0008-5472.can-22-3705] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 09/30/2023] [Accepted: 01/04/2024] [Indexed: 01/10/2024]
Abstract
Colorectal cancer development and outcome are impacted by modifiable risk factors, including psychologic stress. The gut microbiota has also been shown to be linked to psychologic factors. Here, we found a marked deteriorative effect of chronic stress in multiple colorectal cancer models, including chemically induced (AOM/DSS), genetically engineered (APCmin/+), and xenograft tumor mouse models. RNA sequencing data from colon tissues revealed that expression of stemness-related genes was upregulated in the stressed colorectal cancer group by activated β-catenin signaling, which was further confirmed by results from ex vivo organoid analyses as well as in vitro and in vivo cell tumorigenicity assays. 16S rRNA sequencing of the gut microbiota showed that chronic stress disrupted gut microbes, and antibiotic treatment and fecal microbiota transplantation abolished the stimulatory effects of chronic stress on colorectal cancer progression. Stressed colorectal cancer mice displayed a significant decrease in Lactobacillus johnsonii (L. johnsonii) abundance, which was inversely correlated with tumor load. Moreover, protocatechuic acid (PCA) was identified as a beneficial metabolite produced by L. johnsonii based on metabolome sequencing and LC/MS-MS analysis. Replenishment of L. johnsonii or PCA blocked chronic stress-induced colorectal cancer progression by decreasing β-catenin expression. Furthermore, PCA activated the cGMP pathway, and the cGMP agonist sildenafil abolished the effects of chronic stress on colorectal cancer. Altogether, these data identify that stress impacts the gut microbiome to support colorectal cancer progression. SIGNIFICANCE Chronic stress stimulates cancer stemness by reducing the intestinal abundance of L. johnsonii and its metabolite PCA to enhance β-catenin signaling, forming a basis for potential strategies to circumvent stress-induced cancer aggressiveness. See related commentary by McCollum and Shah, p. 645.
Collapse
Affiliation(s)
- Qiuhua Cao
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
- Vaccine Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, P.R. China
| | - Mingrui Zhao
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Yali Su
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Siliang Liu
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Yanting Lin
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Huijuan Da
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Chongxiu Yue
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Yiming Liu
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Dongquan Jing
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Qixiang Zhao
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Ning Liu
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, P.R. China
| | - Juan Du
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, P.R. China
| | - Zhanjie Zuo
- Thoracic Cancer Treatment Center, Armed Police Beijing Corps Hospital, Beijing, P.R. China
| | - Yao Fu
- Department of Pathology, Drum Tower Hospital, Medical School of Nanjing University, Nanjing, P.R. China
| | - Anqi Chen
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| | - Lutz Birnbaumer
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
- Institute of Biomedical Research (BIOMED), Catholic University of Argentina, Buenos Aires, Argentina
| | - Yong Yang
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
- School of Pharmacy, Xuzhou Medical University, Xuzhou, P.R. China
| | - Beiying Dai
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| | - Xinghua Gao
- Center for New Drug Safety Evaluation and Research, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, P.R. China
| |
Collapse
|
38
|
Chen X, Wang L, Yang M, Zhao W, Tu J, Liu B, Yuan X. RUNX transcription factors: biological functions and implications in cancer. Clin Exp Med 2024; 24:50. [PMID: 38430423 PMCID: PMC10908630 DOI: 10.1007/s10238-023-01281-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 11/10/2023] [Indexed: 03/03/2024]
Abstract
Runt-related transcription factors (RUNX) are a family of transcription factors that are essential for normal and malignant hematopoietic processes. Their most widely recognized role in malignancy is to promote the occurrence and development of acute myeloid leukemia. However, it is worth noting that during the last decade, studies of RUNX proteins in solid tumors have made considerable progress, suggesting that these proteins are directly involved in different stages of tumor development, including tumor initiation, progression, and invasion. RUNX proteins also play a role in tumor angiogenesis, the maintenance of tumor cell stemness, and resistance to antitumor drugs. These findings have led to the consideration of RUNX as a tumor biomarker. All RUNX proteins are involved in the occurrence and development of solid tumors, but the role of each RUNX protein in different tumors and the major signaling pathways involved are complicated by tumor heterogeneity and the interacting tumor microenvironment. Understanding how the dysregulation of RUNX in tumors affects normal biological processes is important to elucidate the molecular mechanisms by which RUNX affects malignant tumors.
Collapse
Affiliation(s)
- Xinyi Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei Province, China
| | - Lu Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei Province, China
| | - Mu Yang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei Province, China
| | - Weiheng Zhao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei Province, China
| | - Jingyao Tu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei Province, China.
| | - Bo Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei Province, China.
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang Road 1095, Wuhan, Hubei Province, China.
| |
Collapse
|
39
|
Xian Y, Wang X, Yu Y, Chen X. The mechanism of EGFL7 regulating neovascularization in diabetic retinopathy through the PI3K/AKT/VEGFA pathway. Life Sci 2024; 340:122483. [PMID: 38307238 DOI: 10.1016/j.lfs.2024.122483] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/04/2024]
Abstract
Diabetic retinopathy (DR) is a blinding disease caused by diabetes, characterized by neovascularization of the retina. The aim of this study was to investigate the roles of epidermal growth factor-like structural domain 7 (EGFL7) on human retinal vascular endothelial cells (HRECS) and retinas from rats with DR. An in vitro model of DR was established through culturing HRECS in high glucose. The in vivo model of DR was established by injecting SD rats with streptozotocin (STZ) to induce diabetes. The differences in the expressed levels of EGFL7, PI3K, AKT, P-AKT and VEGFA in high-glucose cultured cells and retinal tissues of diabetic rats were detected in compared to those in the control group. Stable EGFL7 knockdown cell lines were generated by transfecting HRECS with lentiviral vectors and the effects of EGFL7 knockdown on angiogenesis, cell migration and proliferation were investigated. The results showed that EGFL7, PI3K, P-AKT and VEGFA was increased in cells and tissues under high glucose conditions. Knockdown of EGFL7 downregulated the proliferation, migration and angiogenesis capacity of HRECS, and blocked the PI3K/AKT/VEGFA signaling pathway. Furthermore, overexpression of PI3K reversed the effects of EGFL7 inhibition. These findings provide new ideas for the treatment of neovascularisation in DR.
Collapse
Affiliation(s)
- Yang Xian
- Department of Ophthalmology, Shengjing Hospital of China Medical University, China
| | - XingLi Wang
- Department of Ophthalmology, Shengjing Hospital of China Medical University, China
| | - Yong Yu
- Department of Ophthalmology, Shengjing Hospital of China Medical University, China
| | - XiaoLong Chen
- Department of Ophthalmology, Shengjing Hospital of China Medical University, China.
| |
Collapse
|
40
|
Guo X, Zhang H, He C, Qin K, Lai Q, Fang Y, Chen Q, Li W, Wang Y, Wang X, Li A, Liu S, Li Q. RUNX1 promotes angiogenesis in colorectal cancer by regulating the crosstalk between tumor cells and tumor associated macrophages. Biomark Res 2024; 12:29. [PMID: 38419056 PMCID: PMC10903076 DOI: 10.1186/s40364-024-00573-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024] Open
Abstract
Colorectal cancer (CRC) is a common malignancy worldwide. Angiogenesis and metastasis are the critical hallmarks of malignant tumor. Runt-related transcription factor 1 (RUNX1), an efficient transcription factor, facilitates CRC proliferation, metastasis and chemotherapy resistance. We aimed to investigate the RUNX1 mediated crosstalk between tumor cells and M2 polarized tumor associated macrophages (TAMs) in CRC, as well as its relationship with neoplastic angiogenesis. We found that RUNX1 recruited macrophages and induced M2 polarized TAMs in CRC by promoting the production of chemokine 2 (CCL2) and the activation of Hedgehog pathway. In addition, we found that the M2 macrophage-specific generated cytokine, platelet-derived growth factor (PDGF)-BB, promoted vessel formation both in vitro and vivo. PDGF-BB was also found to enhance the expression of RUNX1 in CRC cell lines, and promote its migration and invasion in vitro. A positive feedback loop of RUNX1 and PDGF-BB was thus formed. In conclusion, our data suggest that RUNX1 promotes CRC angiogenesis by regulating M2 macrophages during the complex crosstalk between tumor cells and TAMs. This observation provides a potential combined therapy strategy targeting RUNX1 and TAMs-related PDGF-BB in CRC.
Collapse
Affiliation(s)
- Xuxue Guo
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, People's Republic of China
- Department of Gastroenterology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Haonan Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, People's Republic of China
- Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Chengcheng He
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, People's Republic of China
- Department of Gastroenterology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Kaiwen Qin
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, People's Republic of China
- The First School of Clinical Medicine), Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qiuhua Lai
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, People's Republic of China
| | - Yuxin Fang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, People's Republic of China
| | - Qianhui Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Hepatology Unit and Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Weize Li
- The First School of Clinical Medicine), Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yiqing Wang
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Xinke Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, People's Republic of China
| | - Aimin Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, People's Republic of China
| | - Side Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, People's Republic of China.
- Pazhou Lab, Guangzhou, Guangdong, China.
| | - Qingyuan Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, People's Republic of China.
| |
Collapse
|
41
|
Cai F, Li J, Zhang Y, Huang S, Liu W, Zhuo W, Qiu C. Interaction between Wnt/β-catenin signaling pathway and EMT pathway mediates the mechanism of sunitinib resistance in renal cell carcinoma. BMC Cancer 2024; 24:175. [PMID: 38317072 PMCID: PMC10840195 DOI: 10.1186/s12885-024-11907-5] [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: 04/04/2023] [Accepted: 01/22/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Targeted drugs are the main methods of RCC treatment. However, drug resistance is common in RCC patients, in-depth study of the drug-resistant mechanism is essential. METHODS We constructed sunitinib resistant and Twist overexpressed A498 cells, and studied its mechanisms in vitro and in vivo. RESULTS In cell research, we found that either sunitinib resistance or Twist overexpression can activate Wnt/β-catenin and EMT signaling pathway, and the sunitinib resistance may work through β-catenin/TWIST/TCF4 trimer. In zebrafish research, we confirmed the similarity of Twist overexpression and sunitinib resistance, and the promoting effect of Twist overexpression on drug resistance. CONCLUSIONS Sunitinib resistance and Twist overexpression can activate Wnt/β-catenin signaling pathway and EMT to promote the growth and metastasis of RCC cells.
Collapse
Affiliation(s)
- Fangzhen Cai
- Department of Urology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Jianwei Li
- Department of Urology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Yanmei Zhang
- Department of Urology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Sihuai Huang
- Department of Urology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Wenbin Liu
- Department of Urology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Weifeng Zhuo
- Department of Urology, JinJiang Municipal Hospital, Quanzhou, Fujian, China
| | - Chengzhi Qiu
- Department of General Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China.
| |
Collapse
|
42
|
Yu X, Zhao P, Luo Q, Wu X, Wang Y, Nan Y, Liu S, Gao W, Li B, Liu Z, Cui Z. RUNX1-IT1 acts as a scaffold of STAT1 and NuRD complex to promote ROS-mediated NF-κB activation and ovarian cancer progression. Oncogene 2024; 43:420-433. [PMID: 38092960 DOI: 10.1038/s41388-023-02910-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 11/23/2023] [Accepted: 11/28/2023] [Indexed: 02/04/2024]
Abstract
Dysregulated expression of long-stranded non-coding RNAs is strongly associated with carcinogenesis. However, the precise mechanisms underlying their involvement in ovarian cancer pathogenesis remain poorly defined. Here, we found that lncRNA RUNX1-IT1 plays a crucial role in the progression of ovarian cancer. Patients with high RUNX1-IT1 expression had shorter survival and poorer outcomes. Notably, knockdown of RUNX1-IT1 suppressed the proliferation, migration and invasion of ovarian cancer cells in vitro, and reduced the formation of peritoneum metastasis in vivo. Mechanistically, RUNX1-IT1 bound to HDAC1, the core component of the NuRD complex, and STAT1, acting as a molecular scaffold of the STAT1 and NuRD complex to regulate intracellular reactive oxygen homeostasis by altering the histone modification status of downstream targets including GPX1. Consequently, RUNX1-IT1 activated NF-κB signaling and altered the biology of ovarian cancer cells. In conclusion, our findings demonstrate that RUNX1-IT1 promotes ovarian malignancy and suggest that targeting RUNX1-IT1 represents a promising therapeutic strategy for ovarian cancer treatment.
Collapse
Affiliation(s)
- Xiao Yu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Pengfei Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qingyu Luo
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Xiaowei Wu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02215, USA
| | - Yating Wang
- Department of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yabing Nan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shi Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wenyan Gao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Bin Li
- Department of Gynecological Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhihua Liu
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Zhumei Cui
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China.
| |
Collapse
|
43
|
Gao J, Pan H, Li J, Jiang J, Wang W. A peptide encoded by the circular form of the SHPRH gene induces apoptosis in neuroblastoma cells. PeerJ 2024; 12:e16806. [PMID: 38282862 PMCID: PMC10812589 DOI: 10.7717/peerj.16806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 12/28/2023] [Indexed: 01/30/2024] Open
Abstract
Background Circular RNAs (circRNAs) and their derived peptides represent largely unchartered areas in cellular biology, with many potential roles yet to be discovered. This study aimed to elucidate the role and molecular interactions of circSHPRH and its peptide derivative SHPRH-146aa in the pathogenesis of neuroblastoma (NB). Methods NB samples in the GSE102285 dataset were analyzed to measure circSHPRH expression, followed by in vitro experiments for validation. The role of SHPRH-146aa in NB cell proliferation, migration, and invasion was then examined, and luciferase activity assay was performed after SHPRH-146aa and RUNX1 transfection. Finally, the regulation of NB cell apoptosis by SHPRH-146aa combined with NFKBIA was tested. Results The GSE102285 dataset indicated overexpression of circSHPRH in NB samples, further supported by in vitro findings. Overexpression of circ-SHPRH and SHPRH-146aa inhibited proliferation, migration, and invasion of NB cells. A significant increase in apoptosis was observed, with upregulation of Caspase-3 and downregulation of Bcl-2. Furthermore, the peptide derivative SHPRH-146aa, derived from circSHPRH, suppressed NB cell malignancy traits, suggesting its role as a therapeutic target. A direct interaction between SHPRH-146aa and the transcription factor RUNX1 was identified, subsequently leading to increased NFKBIA expression. Notably, NFKBIA knockdown inhibited the pro-apoptotic effect of SHPRH-146aa on NB cells. Conclusion The study demonstrates that circ-SHPRH and SHPRH-146aa play significant roles in inhibiting the malignant progression of NB. They induce apoptosis primarily by modulating key apoptotic proteins Caspase-3 and Bcl-2, a process that appears to be regulated by NFKBIA. The SHPRH-146aa-RUNX1 interaction further elucidates a novel pathway in the regulation of apoptosis in NB. These findings indicate that circ-SHPRH and its derived peptide SHPRH-146aa could be potential therapeutic targets for NB treatment.
Collapse
Affiliation(s)
- Jingjing Gao
- Department of Nutrition, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hong Pan
- Department of Nutrition, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Li
- Department of Nutrition, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Jiang
- Endoscopy Center, Minhang District Central Hospital of Fudan University, Shanghai, China
| | - Wenxian Wang
- Department of Nutrition, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
44
|
Liang Y, Wei X, Yue PJ, Zhang HC, Li ZN, Wang XX, Sun YY, Fu WN. MYCT1 inhibits hematopoiesis in diffuse large B-cell lymphoma by suppressing RUNX1 transcription. Cell Mol Biol Lett 2024; 29:5. [PMID: 38172714 PMCID: PMC10763471 DOI: 10.1186/s11658-023-00522-0] [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: 08/15/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND The abnormality of chromosomal karyotype is one factor causing poor prognosis of lymphoma. In the analysis of abnormal karyotype of lymphoma patients, three smallest overlap regions were found, in which MYCT1 was located. MYCT1 is the first tumor suppressor gene cloned by our research team, but its studies relating to the occurrence and development of lymphoma have not been reported. METHODS R banding analyses were employed to screen the abnormality of chromosomal karyotype in clinical specimen and MYCT1 over-expression cell lines. FISH was to monitor MYCT1 copy number aberration. RT-PCR and Western blot were to detect the mRNA and protein levels of the MYCT1 and RUNX1 genes, respectively. The MYCT1 and RUNX1 protein levels in clinical specimen were evaluated by immunohistochemical DAB staining. The interaction between MYCT1 and MAX proteins was identified via Co-IP and IF. The binding of MAX on the promoter of the RUNX1 gene was detected by ChIP and Dual-luciferase reporter assay, respectively. Flow cytometry and CCK-8 assay were to explore the effects of MYCT1 and RUNX1 on the cell cycle and proliferation, respectively. RESULTS MYCT1 was located in one of three smallest overlap regions of diffuse large B-cell lymphoma, it altered chromosomal instability of diffuse large B-cell lymphoma cells. MYCT1 negatively correlated with RUNX1 in lymphoma tissues of the patients. MAX directly promoted the RUNX1 gene transcription by binding to its promoter region. MYCT1 may represses RUNX1 transcription by binding MAX in diffuse large B-cell lymphoma cells. MYCT1 binding to MAX probably suppressed RUNX1 transcription, leading to the inhibition of proliferation and cell cycle of the diffuse large B-cell lymphoma cells. CONCLUSION This study finds that there is a MYCT1-MAX-RUNX1 signaling pathway in diffuse large B-cell lymphoma. And the study provides clues and basis for the in-depth studies of MYCT1 in the diagnosis, treatment and prognosis of lymphoma.
Collapse
Affiliation(s)
- Ying Liang
- Department of Medical Genetics, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, People's Republic of China
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Xin Wei
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Peng-Jie Yue
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - He-Cheng Zhang
- Department of Medical Genetics, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, People's Republic of China
| | - Zhen-Ning Li
- Department of Oromaxillofacial-Head and Neck Surgery, Liaoning Province Key Laboratory of Oral Disease, School and Hospital of Stomatology, China Medical University, Shenyang, People's Republic of China
| | - Xiao-Xue Wang
- Department of Hematology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, People's Republic of China
| | - Yuan-Yuan Sun
- Department of Medical Genetics, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, People's Republic of China.
| | - Wei-Neng Fu
- Department of Medical Genetics, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province, 110122, People's Republic of China.
| |
Collapse
|
45
|
Feng YL. A New Frontier in Phytotherapy: Harnessing the Therapeutic Power of Medicinal Herb-derived miRNAs. Curr Pharm Des 2024; 30:3009-3017. [PMID: 39162273 DOI: 10.2174/0113816128310724240730072626] [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: 02/16/2024] [Revised: 06/18/2024] [Accepted: 06/26/2024] [Indexed: 08/21/2024]
Abstract
Medicinal herbs have been utilized in the treatment of various pathologic conditions, including neoplasms, organ fibrosis, and diabetes mellitus. However, the precise pharmacological actions of plant miRNAs in animals remain to be fully elucidated, particularly in terms of their therapeutic efficacy and mechanism of action. In this review, some important miRNAs from foods and medicinal herbs are presented. Plant miRNAs exhibit a range of pharmacological properties, such as anti-cancer, anti-fibrosis, anti-viral, anti-inflammatory effects, and neuromodulation, among others. These results have not only demonstrated a cross-species regulatory effect, but also suggested that the miRNAs from medicinal herbs are their bioactive components. This shows a promising prospect for plant miRNAs to be used as drugs. Here, the pharmacological properties of plant miRNAs and their underlying mechanisms have been highlighted, which can provide new insights for clarifying the therapeutic mechanisms of medicinal herbs and suggest a new way for developing therapeutic drugs.
Collapse
Affiliation(s)
- Ya-Long Feng
- Department of Life Science, Xianyang Normal University, No.43 Wenlin Road, Xianyang 712000, Shaanxi, China
| |
Collapse
|
46
|
Jiang Z, Yi P, You J, Cai E. Differentially expressed miR-4310 functions as a tumor suppressor in colorectal cancer. Technol Health Care 2024; 32:1043-1053. [PMID: 37545284 DOI: 10.3233/thc-230472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is a digestive tract malignancy microRNAs (miRNAs) have attracted much attention as biomarkers in tumor studies. OBJECTIVE This work focused on the predictive potential and mechanism of miR-4310 in CRC. METHODS The miRNA expression profile sets were obtained from the Gene Expression Omnibus (GEO) database, and the appropriate miRNA was screened by GEO2R. The CRC tissues and control tissues of 88 patients with CRC were collected, and the expression of miR-4310 was detected by quantitative real-time PCR, and the efficacy of miR-4310 in diagnosing CRC was evaluated by the receiver operating characteristic curve (ROC). The effects of miR-4310 on the proliferation, migration, and invasion of CRC cells were explored by Cell Counting Kit-8 (CCK-8) and Transwell experiments. Predicting the potential binding sites of miR-4310 and Runt-related transcription factor 1 (RUNX1) by four predictive websites. The relationship between miR-4310 and RUNX1 was confirmed by a double luciferase reporter gene experiment. RESULTS The bioinformatics analysis found that miR-4310 was differentially expressed in CRC tissues and this finding was certified by the expression of miR-4310 in CRC tissues of collected patients and cultured CRC cell lines. The expression of miR-4310 had a predictive possibility for CRC patients. MiR-4310/RUNX1 pathway had effects on CRC viability, migration, and invasion. CONCLUSION MiR-4310 had the potential to be a biomarker for early screening of CRC. MiR-4310 and RUNX1 participated in the regulation of CRC cells.
Collapse
|
47
|
Sun M, Peng Z, Shen W, Guo X, Liao Y, Huang Y, Ye P, Hu M, Lin Q, Liu R. Synergism of Fusobacterium periodonticum and N-nitrosamines promote the formation of EMT subtypes in ESCC by modulating Wnt3a palmitoylation. Gut Microbes 2024; 16:2391521. [PMID: 39193618 PMCID: PMC11364064 DOI: 10.1080/19490976.2024.2391521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 07/17/2024] [Accepted: 08/08/2024] [Indexed: 08/29/2024] Open
Abstract
N-Nitrosamine disinfection by-products (NAs-DBPs) have been well proven for its role in esophageal carcinogenesis. However, the role of intratumoral microorganisms in esophageal squamous cell carcinoma (ESCC) has not yet been well explored in the context of exposure to NAs-DBPs. Here, the multi-omics integration reveals F. periodonticum (Fp) as "facilitators" is highly enriched in cancer tissues and promotes the epithelial mesenchymal transition (EMT)-like subtype formation of ESCC. We demonstrate that Fp potently drives de novo synthesis of fatty acids, migration, invasion and EMT phenotype through its unique FadAL adhesin. However, N-nitrosomethylbenzylamine upregulates the transcription level of FadAL. Mechanistically, co-immunoprecipitation coupled to mass spectrometry shows that FadAL interacts with FLOT1. Furthermore, FLOT1 activates PI3K-AKT/FASN signaling pathway, leading to triglyceride and palmitic acid (PA) accumulation. Innovatively, the results from the acyl-biotin exchange demonstrate that FadAL-mediated PA accumulation enhances Wnt3A palmitoylation on a conserved cysteine residue, Cys-77, and promotes Wnt3A membrane localization and the translocation of β-catenin into the nucleus, further activating Wnt3A/β-catenin axis and inducing EMT phenotype. We therefore propose a "microbiota-cancer cell subpopulation" interaction model in the highly heterogeneous tumor microenvironment. This study unveils a mechanism by which Fp can drive ESCC and identifies FadAL as a potential diagnostic and therapeutic target for ESCC.
Collapse
Affiliation(s)
- Mingjun Sun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Zhenyan Peng
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Weitao Shen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Xinxin Guo
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Yinghao Liao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Yang Huang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Ping Ye
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Mohan Hu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
| | - Qiang Lin
- Department of Oncology, North China Petroleum Bureau General Hospital, Hebei Medical University, Renqiu, China
| | - Ran Liu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, China
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, China
| |
Collapse
|
48
|
Zhao H, Feng L, Cheng R, Wu M, Bai X, Fan L, Liu Y. miR-29c-3p acts as a tumor promoter by regulating β-catenin signaling through suppressing DNMT3A, TET1 and HBP1 in ovarian carcinoma. Cell Signal 2024; 113:110936. [PMID: 37925048 DOI: 10.1016/j.cellsig.2023.110936] [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: 07/12/2023] [Revised: 10/05/2023] [Accepted: 10/19/2023] [Indexed: 11/06/2023]
Abstract
Ovarian Carcinoma (OvCa) is characterized by rapid and sustained growth, activated invasion and metastasis. Studies have shown that microRNAs recruit and alter the expression of key regulators to modulate carcinogenesis. Here, we find that miR-29c-3p is increased in benign OvCa and malignant OvCa compared to normal ovary. Univariate and multivariate analyses report that miR-29c-3p overexpression is associated with poor prognosis in OvCa. Furthermore, we investigate that expression of miR-29c-3p is inversely correlated to DNA methyltransferase (DNMT) 3 A and Ten-Eleven-Translocation enzyme TET1. The high-throughput mRNA sequencing, bioinformatics analysis and pharmacological studies confirm that aberrant miR-29c-3p modulates tumorigenesis in OvCa cells, including epithelial-mesenchymal transition (EMT), proliferation, migration, and invasion. This modulation occurs through the regulation of β-catenin signaling by directly targeting 3'UTR of DNMT3A, TET1 and the HMG box transcription factor HBP1 and suppressing their expression. The further 3D spheres assay clearly shows the regulatory effects of miR-29c-3p on OvCa tumorigenesis. Additionally, the receiver operating characteristic (ROC) curve analysis of miR-29c-3p and the clinical detection/diagnostic biomarker CA125 suggests that miR-29c-3p may be conducive for clinical diagnosis or co-diagnosis of OvCa. These findings support miR-29c-3p functions as a tumor promoter by targeting its functional targets, providing new potential biomarker (s) for precision medicine strategies in OvCa.
Collapse
Affiliation(s)
- Haile Zhao
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, State Key Laboratory of Reproductive Regulation & Breeding of Grassland livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia 010020, PR China
| | - Lijuan Feng
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, State Key Laboratory of Reproductive Regulation & Breeding of Grassland livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia 010020, PR China
| | - Rui Cheng
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, State Key Laboratory of Reproductive Regulation & Breeding of Grassland livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia 010020, PR China
| | - Man Wu
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, State Key Laboratory of Reproductive Regulation & Breeding of Grassland livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia 010020, PR China
| | - Xiaozhou Bai
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, State Key Laboratory of Reproductive Regulation & Breeding of Grassland livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia 010020, PR China
| | - Lifei Fan
- Inner Mongolia Key Laboratory for Molecular Regulation of the Cell, State Key Laboratory of Reproductive Regulation & Breeding of Grassland livestock, School of Life Sciences, Inner Mongolia University, Hohhot, Inner Mongolia 010020, PR China.
| | - Yaping Liu
- Department of Gynecology and Obstetrics, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, PR China.
| |
Collapse
|
49
|
Chen H, Zhai C, Xu X, Wang H, Han W, Shen J. Multilevel Heterogeneity of Colorectal Cancer Liver Metastasis. Cancers (Basel) 2023; 16:59. [PMID: 38201487 PMCID: PMC10778489 DOI: 10.3390/cancers16010059] [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: 11/03/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Colorectal cancer liver metastasis (CRLM) is a highly heterogeneous disease. Therapies that target both primary foci and liver metastasis are severely lacking. Therefore, understanding the features of metastatic tumor cells in the liver is valuable for the overall control of CRLM patients. In this review, we summarize the heterogeneity exhibited in CRLM from five aspects (gene, transcriptome, protein, metabolism, and immunity). In addition to genetic heterogeneity, the other four aspects exhibit significant heterogeneity. Compared to primary CRC, the dysregulation of epithelial-mesenchymal transition (EMT)-related proteins, the enhanced metabolic activity, and the increased infiltration of immunosuppressive cells are detected in CRLM. Preclinical evidence shows that targeting the EMT process or enhancing cellular metabolism may represent a novel approach to increasing the therapeutic efficacy of CRLM.
Collapse
Affiliation(s)
| | | | | | | | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; (H.C.); (C.Z.); (X.X.); (H.W.)
| | - Jiaying Shen
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; (H.C.); (C.Z.); (X.X.); (H.W.)
| |
Collapse
|
50
|
Zhou S, Zhao T, Chen X, Zhang W, Zou X, Yang Y, Wang Q, Zhang P, Zhou T, Feng T. Runx1 Deficiency Promotes M2 Macrophage Polarization Through Enhancing STAT6 Phosphorylation. Inflammation 2023; 46:2241-2253. [PMID: 37530929 DOI: 10.1007/s10753-023-01874-7] [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: 05/09/2023] [Revised: 07/03/2023] [Accepted: 07/06/2023] [Indexed: 08/03/2023]
Abstract
Our previous study had demonstrated that Runx1 promoted LPS-induced macrophage inflammatory response, however, the role of Runx1 in M2 macrophage polarization still remains largely unknown. This study was conducted to investigate the role of Runx1 in IL-4/IL-13-induced M2 macrophage polarization and its potential regulatory mechanism. We found that exposure of macrophages to IL-4/IL-13 induced a remarkable increasement in Runx1 expression level. Specifically, we established genetically modified mice lacking Runx1 in myeloid cells, including macrophages. RNA-Seq was performed to identify differentially expressed genes (DEGs) between Runx1 knockout and WT control bone marrow-derived macrophages (BMDMs). We identified 686 DEGs, including many genes which were highly expressed in M2 macrophage. In addition, bioinformatics analysis indicated that these DEGs were significantly enriched in extracellular matrix-related processes. Moreover, RT-qPCR analysis showed that there was an obvious upregulation in the relative expression levels of M2 marker genes, including Arg1, Ym1, Fizz1, CD71, Mmp9, and Tgm2, in Runx1 knockout macrophages, as compared to WT controls. Consistently, similar results were obtained in the protein and enzymatic activity levels of Arg1. Finally, we found that the STAT6 phosphorylation level was significantly enhanced in Runx1 knockout macrophages, and the STAT6 inhibitor AS1517499 partly reduced the upregulated effect of Runx1 deficiency on the M2 macrophage polarization. Taken together, Runx1 deficiency facilitates IL-4/IL-13-induced M2 macrophage polarization through enhancing STAT6 phosphorylation.
Collapse
Affiliation(s)
- Siyuan Zhou
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Ting Zhao
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Xuqiong Chen
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Wuwen Zhang
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Xiaoyi Zou
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Yi Yang
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Qinshi Wang
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Ping Zhang
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China
| | - Tong Zhou
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Tongbao Feng
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China.
| |
Collapse
|