1
|
Zhang J, Li Z, Han J, Tian Z, Meng Q, Niu W. KLF7 enhances the invasion and migration of colorectal cancer cells via the miR-139-5p/TPD52 axis. Cancer Biol Ther 2024; 25:2385172. [PMID: 39097779 PMCID: PMC11299624 DOI: 10.1080/15384047.2024.2385172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 07/07/2024] [Accepted: 07/23/2024] [Indexed: 08/05/2024] Open
Abstract
In this study, we aimed to investigate the molecular mechanism of Krüppel-like factor 7 (KLF7) in colorectal cancer (CRC) cell invasion and migration. The expression pattern of KLF7 in CRC tissues and the correlation between KLF7 expression and clinical symptoms of CRC were analyzed. CRC cell lines were transfected with si-KLF7, followed by qRT-PCR or western blot detection of KLF7, miR-139-5p, and tumor protein D52 (TPD52) expression, cell counting kit-8 (CCK-8) assay to detect cell viability, and transwell detection of invasion and migration. Chromatin immunoprecipitation (ChIP) analyzed the enrichment KLF7 in the miR-139-5p promoter. The dual-luciferase reporter assay verified the binding relationship between KLF7 and miR-139-5p, and between miR-139-5p and TPD52. In the subcutaneous tumorigenesis experiment, tumor growth was observed and ki67-positive expression was detected. KLF7 is abundantly expressed in CRC cells KLF7 silencing inhibits CRC cell viability, invasion, and migration. KLF7 represses miR-139-5p expression by binding to the miR-139-5p promoter. miR-139-5p targets TPD52 expression. miR-13-5p inhibition or TPD52 overexpression partially counteracted the effect of KLF7 silencing in CRC cells. KLF7 silencing suppresses tumor growth in vivo. In conclusion, KLF7 suppresses miR-139-5p expression by binding to the miR-139-5p promoter, thereby upregulating TPD52 expression and enhancing CRC cell invasion and migration.
Collapse
Affiliation(s)
- Juan Zhang
- Department of External Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhihan Li
- Department of External Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiaxu Han
- Department of External Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhongtao Tian
- Department of External Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Qingyu Meng
- Department of External Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wenbo Niu
- Department of External Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
2
|
Liu Y, Su Y, Chen L, Li A, Ma Z. Exploring the roles and therapeutic implications of melatonin-mediated KLF6 in the development of intracranial aneurysm. Ann Med 2024; 56:2397568. [PMID: 39215680 PMCID: PMC11370671 DOI: 10.1080/07853890.2024.2397568] [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: 02/06/2024] [Revised: 04/29/2024] [Accepted: 05/04/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Intracranial aneurysm (IA) is a cerebrovascular disease with a high mortality rate due to ruptured subarachnoid hemorrhage. While Krüppel-like factor 6 (KLF6) dysregulation has been implicated in cancer and cardiovascular diseases, its role in IA remains unclear. MATERIALS AND METHODS The GSE122897 and GSE15629 datasets were downloaded from the Gene Expression Omnibus database. Immune cell infiltration and hypoxia analysis were performed to explore the effects of KLF6 on IA. Weighted gene co-expression network analysis was used to identify hub genes related to KLF6 expression for subsequent analyses. Hypoxia-related genes were identified. Drug prediction was performed for IA. Samples from healthy individuals and patients with IA were collected to detect the expression of endothelin-1 (ET-1), vascular hematoma factor (vWF), and KLF6. A model of H2O2-induced human brain vascular smooth muscle cells (HBVSMC) injury was constructed to explore the effects of KLF6 and melatonin to treat IA. RESULTS T cells CD4 memory resting and monocytes were significantly different in the KLF6 high and low expression groups. Four hypoxia-related gene sets were significantly enriched in the KLF6 high-expression group. Six hypoxia-related hub genes were obtained, which were significantly associated with KLF6. Drug prediction showed that melatonin may be a potential drug for IA. The levels of ET-1, vWF, and KLF6 were significantly upregulated in patients with IA. KLF6 exacerbates H2O2-induced injury in HBVSMC, ameliorated by melatonin. CONCLUSION KLF6 may be a potential target for IA treatment, with melatonin-mediated KLF6 effects playing a crucial role in the development of IA.
Collapse
Affiliation(s)
- Yan Liu
- Department of Neurology, Suzhou Hospital of Anhui Medical University, Suzhou, Anhui, P.R. China
| | - Yongxing Su
- Department of Neurology, Suzhou Hospital of Anhui Medical University, Suzhou, Anhui, P.R. China
| | - Le Chen
- Department of Neurology, Suzhou Hospital of Anhui Medical University, Suzhou, Anhui, P.R. China
| | - Anzhi Li
- Department of Neurology, Suzhou Hospital of Anhui Medical University, Suzhou, Anhui, P.R. China
| | - Zhengfei Ma
- Department of Neurology, Suzhou Hospital of Anhui Medical University, Suzhou, Anhui, P.R. China
| |
Collapse
|
3
|
Li M, Hou Y, Chen Y, Sun C, Liang M, Chu X, Wen X, Yuan F, Peng C, Wang C, Xie J, Zhang J. Palmitic acid promotes miRNA release from adipocyte exosomes by activating NF-κB/ER stress. Nutr Diabetes 2024; 14:75. [PMID: 39271650 PMCID: PMC11399118 DOI: 10.1038/s41387-024-00334-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
OBJECTIVE The release of adipose tissue-derived miRNAs is increased under conditions of obesity, but the exact molecular mechanisms involved have not been elucidated. This study investigated whether obesity-induced increases in palmitic acid (PA) content could activate the NF-κB/endoplasmic reticulum stress (ER stress) pathway and promote the expression and release of exosomal miRNAs in adipocytes. METHODS Abdominal adipose tissue and serum samples were collected from normal weight individuals and people with obesity to clarify the correlation of serum PA content with NF-κB/ER stress and the release of exosomal miRNAs. NF-κB and ER stress were blocked in obese mice and in vitro cultured adipocytes to demonstrate the molecular mechanisms by which PA promotes the release of exosomal miRNAs.The morphology, particle size and distribution of the exosomes were observed via transmission electron microscopy and NTA. RESULTS Accompanied by increased serum PA levels, the NF-κB/ER stress pathway was activated in the adipose tissue of people with obesity and in high-fat diet (HFD)-induced obese mice; moreover, the levels of miRNAs in both adipose tissue and serum were increased. P-p65 (Bay11-7082) and ER stress (TUDCA) blockers significantly reduced the levels of miRNAs in abdominal adipose tissue and serum, decreased blood glucose levels, and improved glucose tolerance and insulin sensitivity in obese mice. In 3T3-L1 adipocytes, high concentrations of PA activated the NF-κB/ER stress pathway and increased the expression and release of miRNAs in exosomes. P-p65 (Bay11-7082) and ER stress (TUDCA) blockers significantly reversed the increased release exosomal miRNAs cause by PA. CONCLUSIONS Obesity-induced increases in PA content increase the expression and release of miRNAs in adipocyte exosomes by activating the NF-κB/ER stress pathway.
Collapse
Affiliation(s)
- Menghuan Li
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China
| | - Yanting Hou
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China
| | - Yao Chen
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China
| | - Chaoyue Sun
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China
| | - Maodi Liang
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China
| | - Xiaolong Chu
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China
- Medical College of Tarim University, Tarim Road, Alaer, Xinjiang, China
| | - Xin Wen
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China
| | - Fangyuan Yuan
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China
| | - Chaoling Peng
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China
| | - Cuizhe Wang
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China.
| | - Jianxin Xie
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China.
| | - Jun Zhang
- Medical College of Shihezi University, Bei-Er-Road, Shihezi, Xinjiang, China.
| |
Collapse
|
4
|
Song L, Xu Q, Chen R, Sun W, Zhan J. KLF1 Activates RAC3 to Mediate Fatty Acid Synthesis and Enhance Cisplatin Resistance in Bladder Cancer Cells. Am J Mens Health 2024; 18:15579883241273305. [PMID: 39376007 PMCID: PMC11459471 DOI: 10.1177/15579883241273305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/10/2024] [Accepted: 07/05/2024] [Indexed: 10/09/2024] Open
Abstract
While cisplatin remains a frontline treatment for bladder cancer (BCa), the onset of resistance greatly hampers its effectiveness. RAC3 is closely linked to chemoresistance in cancer cells, but its specific role in cisplatin resistance within BCa is still elusive. RAC3 expression in BCa was analyzed using bioinformatics and quantitative polymerase chain reaction (qPCR). The gene set enrichment analysis (GSEA) identified RAC3-enriched pathways and the correlation between RAC3 and fatty acid synthase (FASN), a gene involved in fatty acid synthesis. Potential upstream transcription factors of RAC3 were predicted and their interaction with RAC3 was confirmed via dual-luciferase and chromatin immunoprecipitation (ChIP) assays. T24/DDP, a cisplatin-resistant BCa cell line, was established to probe into the regulatory role of RAC3 in cisplatin resistance. Cell proliferation was evaluated by colony formation and the IC50 values after cisplatin treatment were determined using cell counting kit-8 (CCK-8). The levels of free fatty acids and triglycerides (TGs), as well as the expression of DGAT2 and FASN proteins, were measured to gauge the extent of fatty acid synthesis in cells. Elevated expression of RAC3 was observed in BCa and the cisplatin-resistant BCa cells (T24/DDP). The knockdown of RAC3 within T24/DDP cells was demonstrated to counteract cisplatin resistance. Subsequent analyses identified RAC3 as being notably enriched in the fatty acid synthesis pathway, with Kruppel-like factor 1 (KLF1) emerging as a key upstream regulator. The overexpression of RAC3 was correlated with increased cisplatin resistance in T24/DDP cells, an effect that was mitigated by the addition of the FASN inhibitor, Orlistat. Furthermore, the downregulation of KLF1 suppressed RAC3 expression, disrupted fatty acid synthesis, and attenuated cisplatin resistance in T24/DDP cells. Conversely, the co-overexpression of RAC3 counteracted the effects conferred by KLF1 knockdown. Our study has validated that KLF1 activates RAC3 to mediate fatty acid synthesis and promote cisplatin resistance in BCa, suggesting the KLF1/RAC3 axis as a potential target for combating cisplatin-resistant BCa.
Collapse
Affiliation(s)
- Lide Song
- Department of Urology, Zhuji People’s Hospital, Zhuji, China
| | - Qi Xu
- Department of Urology, Zhuji People’s Hospital, Zhuji, China
| | - Rong Chen
- Department of Urology, Zhuji People’s Hospital, Zhuji, China
| | - Wanghong Sun
- Department of Urology, Zhuji People’s Hospital, Zhuji, China
| | - Jianfei Zhan
- Department of Urology, Zhuji People’s Hospital, Zhuji, China
| |
Collapse
|
5
|
Carpanese V, Festa M, Prosdocimi E, Bachmann M, Sadeghi S, Bertelli S, Stein F, Velle A, Abdel-Salam MAL, Romualdi C, Pusch M, Checchetto V. Interactomic exploration of LRRC8A in volume-regulated anion channels. Cell Death Discov 2024; 10:299. [PMID: 38909013 PMCID: PMC11193767 DOI: 10.1038/s41420-024-02032-0] [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: 11/27/2023] [Revised: 05/08/2024] [Accepted: 05/16/2024] [Indexed: 06/24/2024] Open
Abstract
Ion channels are critical in enabling ion movement into and within cells and are important targets for pharmacological interventions in different human diseases. In addition to their ion transport abilities, ion channels interact with signalling and scaffolding proteins, which affects their function, cellular positioning, and links to intracellular signalling pathways. The study of "channelosomes" within cells has the potential to uncover their involvement in human diseases, although this field of research is still emerging. LRRC8A is the gene that encodes a crucial protein involved in the formation of volume-regulated anion channels (VRACs). Some studies suggest that LRRC8A could be a valuable prognostic tool in different types of cancer, serving as a biomarker for predicting patients' outcomes. LRRC8A expression levels might be linked to tumour progression, metastasis, and treatment response, although its implications in different cancer types can be varied. Here, publicly accessible databases of cancer patients were systematically analysed to determine if a correlation between VRAC channel expression and survival rate exists across distinct cancer types. Moreover, we re-evaluated the impact of LRRC8A on cellular proliferation and migration in colon cancer via HCT116 LRRC8A-KO cells, which is a current topic of debate in the literature. In addition, to investigate the role of LRRC8A in cellular signalling, we conducted biotin proximity-dependent identification (BioID) analysis, revealing a correlation between VRAC channels and cell-cell junctions, mechanisms that govern cellular calcium homeostasis, kinases, and GTPase signalling. Overall, this dataset improves our understanding of LRRC8A/VRAC and explores new research avenues while identifying promising therapeutic targets and promoting inventive methods for disease treatment.
Collapse
Affiliation(s)
| | - Margherita Festa
- DiBio, Unipd, via Ugo Bassi 58/B, 35131, Padova, Italy
- Institute of Biophysics, CNR, Via De Marini, 6 16149, Genova, Italy
| | | | - Magdalena Bachmann
- DiBio, Unipd, via Ugo Bassi 58/B, 35131, Padova, Italy
- Daba Farber Cancer Research Institute, Boston, MA, USA
| | - Soha Sadeghi
- DiBio, Unipd, via Ugo Bassi 58/B, 35131, Padova, Italy
| | - Sara Bertelli
- Institute of Biophysics, CNR, Via De Marini, 6, 16149, Genova, Italy
- Humboldt Universität Berlin, AG Zelluläre Biophysik, Dorotheenstr, 19-21 10099, Berlin, Germany
| | - Frank Stein
- Proteomics Core Facility, EMBL Heidelberg, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Angelo Velle
- DiBio, Unipd, via Ugo Bassi 58/B, 35131, Padova, Italy
| | - Mostafa A L Abdel-Salam
- DiBio, Unipd, via Ugo Bassi 58/B, 35131, Padova, Italy
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Chiara Romualdi
- DiBio, Unipd, via Ugo Bassi 58/B, 35131, Padova, Italy
- Padua Center for Network Medicine, University of Padua, Via F. Marzolo 8, 315126, Padova, Italy
| | - Michael Pusch
- Institute of Biophysics, CNR, Via De Marini, 6, 16149, Genova, Italy
- RAISE Ecosystem, Genova, Italy
| | | |
Collapse
|
6
|
Cao Y, Li J, Zhang G, Fang H, Du Y, Liang Y. KLF15 transcriptionally activates LINC00689 to inhibit colorectal cancer development. Commun Biol 2024; 7:130. [PMID: 38273088 PMCID: PMC10810960 DOI: 10.1038/s42003-023-05757-3] [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: 04/21/2023] [Accepted: 12/29/2023] [Indexed: 01/27/2024] Open
Abstract
Colorectal cancer is a grievous health concern, we have proved long non-coding RNA LINC00689 is considered as a potential diagnosis biomarker for colorectal cancer, and it is necessary to further investigate its upstream and downstream mechanisms. Here, we show that KLF15, a transcription factor, exhibits the reduced expression in colorectal cancer. KLF15 suppresses the proliferative and metastatic capacities of colorectal cancer cells both in vitro and in vivo by transcriptionally activating LINC00689. Subsequently, LINC00689 recruits PTBP1 protein to enhance the stability of LATS2 mRNA in the cytoplasm. This stabilization causes the suppression of the YAP1/β-catenin pathway and its target downstream genes. Our findings highlight a regulatory network involving KLF15, LINC00689, PTBP1, LATS2, and the YAP1/β-catenin pathway in colorectal cancer, shedding light on potential therapeutic targets for colorectal cancer therapy.
Collapse
Affiliation(s)
- Yan Cao
- Department of Nuclear Medicine, Xiangya Third Hospital, Central South University, Changsha, 410013, Hunan Province, PR China
| | - Jian Li
- Department of Nuclear Medicine, Xiangya Third Hospital, Central South University, Changsha, 410013, Hunan Province, PR China
| | - Gang Zhang
- Department 2 of Gastrointestinal Surgery, Haikou Hospital Affiliated to Xiangya Medical College of Central South University, Haikou People's Hospital, Haikou, 570208, Hainan Province, PR China
| | - Hao Fang
- Department of Nuclear Medicine, Xiangya Third Hospital, Central South University, Changsha, 410013, Hunan Province, PR China
| | - Yongliang Du
- Department of Nuclear Medicine, Xiangya Third Hospital, Central South University, Changsha, 410013, Hunan Province, PR China
| | - Yan Liang
- Department of Nuclear Medicine, Xiangya Third Hospital, Central South University, Changsha, 410013, Hunan Province, PR China.
| |
Collapse
|
7
|
Wan B, Zhang W, Deng X, Lu Y, Zhang Z, Yang Y. Molecular Expression and Prognostic Implications of Krüppel-Like Factor 3 (KLF3) in Clear Cell Renal Cell Carcinoma. Crit Rev Eukaryot Gene Expr 2024; 34:45-59. [PMID: 38073441 DOI: 10.1615/critreveukaryotgeneexpr.2023049010] [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: 12/18/2023]
Abstract
A major subtype of renal cancer is clear cell renal cell carcinoma (ccRCC). Krüppel-like factor 3 (KLF3) dysfunction is also revealed leading to poor prognosis in multiple cancer types. However, dysregulation and molecular dynamics of KLF3 underlying ccRCC progression still remains elusive. Here KLF3 gene and protein expressions in ccRCC were explored using data cohorts from The Cancer Genome Atlas (TCGA), Human Protein Atlas (HPA), Clinical Proteomic Tumor Analysis Consortium (CPTAC) and verified them in our patient cohort. Correlations of KLF3 expression with clinicopathological features, epigenetic modification, and immune microenvironment characteristics were further investigated. KLF3 was significantly down-regulated expressed in ccRCC tissues compared to adjacent normal controls. Adverse pathological parameters and poor prognosis were associated with lower expression of KLF3. Mechanically, KLF3 regulation was mainly attributed to CpG island methylation. KLF3-high expression subgroup was significantly enriched in cell signaling pathways most associated with EMT markers, angiogenesis, inflammatory response, apoptosis, TGF-β, degradation of ECM, G2M checkpoint, and PI3K-AKT-mTOR. Based on GDSC database, KLF3 upregulation was identified to be associated with higher sensitivities towards PI3K-Akt-mTOR pathway inhibitors such as PI-103, PIK-93, and OSI-027. In addition, patients with down-regulated KLF3 expressions were found more sensitive towards Trametinib, Cetuximab, and Erlotinib. Collectively, our findings suggest that KLF3 may act as a suitable biomarker for prognosis prediction, tumor microenvironment (TME) phenotype identification, thereby helping ccRCC patients to make better therapeutic decisions.
Collapse
Affiliation(s)
- Bin Wan
- Department of Urology, The First People's Hospital of Jiujiang in Jiangxi Province, Jiujiang City, 332000, Jiangxi Province, China
| | - Wensheng Zhang
- Department of Urology, The First People's Hospital of Jiujiang in Jiangxi Province, Jiujiang City, 332000, Jiangxi Province, China
| | - Xinxi Deng
- Department of Urology, The First People's Hospital of Jiujiang in Jiangxi Province, Jiujiang City, 332000, Jiangxi Province, China
| | - Yigang Lu
- Department of Urology, The First People's Hospital of Jiujiang in Jiangxi Province, Jiujiang City, 332000, Jiangxi Province, China
| | - Zhuo Zhang
- Department of Urology, The First People's Hospital of Jiujiang in Jiangxi Province, Jiujiang City, 332000, Jiangxi Province, China
| | - Yang Yang
- Department of Urology, The First People's Hospital of Jiujiang in Jiangxi Province, Jiujiang City, 332000, Jiangxi Province, China
| |
Collapse
|
8
|
Zhu J, Teng H, Zhu X, Yuan J, Zhang Q, Zou Y. Pan-cancer analysis of Krüppel-like factor 3 and its carcinogenesis in pancreatic cancer. Front Immunol 2023; 14:1167018. [PMID: 37600783 PMCID: PMC10435259 DOI: 10.3389/fimmu.2023.1167018] [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: 02/15/2023] [Accepted: 07/10/2023] [Indexed: 08/22/2023] Open
Abstract
Background Krüppel-like factor 3 (KLF3) is a key transcriptional repressor, which is involved in various biological functions such as lipogenesis, erythropoiesis, and B cell development, and has become one of the current research hotspots. However, the role of KLF3 in the pan-cancer and tumor microenvironment remains unclear. Methods TCGA and GTEx databases were used to evaluate the expression difference of KLF3 in pan-cancer and normal tissues. The cBioPortal database and the GSCALite platform analyzed the genetic variation and methylation modification of KLF3. The prognostic role of KLF3 in pan-cancer was identified using Cox regression and Kaplan-Meier analysis. Correlation analysis was used to explore the relationship between KLF3 expression and tumor mutation burden, microsatellite instability, and immune-related genes. The relationship between KLF3 expression and tumor immune microenvironment was calculated by ESTIMATE, EPIC, and MCPCOUNTER algorithms. TISCH and CancerSEA databases analyzed the expression distribution and function of KLF3 in the tumor microenvironment. TIDE, GDSC, and CTRP databases evaluated KLF3-predicted immunotherapy response and sensitivity to small molecule drugs. Finally, we analyzed the role of KLF3 in pancreatic cancer by in vivo and in vitro experiments. Results KLF3 was abnormally expressed in a variety of tumors, which could effectively predict the prognosis of patients, and it was most obvious in pancreatic cancer. Further experiments verified that silencing KLF3 expression inhibited pancreatic cancer progression. Functional analysis and gene set enrichment analysis found that KLF3 was involved in various immune-related pathways and tumor progression-related pathways. In addition, based on single-cell sequencing analysis, it was found that KLF3 was mainly expressed in CD4Tconv, CD8T, monocytes/macrophages, endothelial cells, and malignant cells in most of the tumor microenvironment. Finally, we assessed the value of KLF3 in predicting response to immunotherapy and predicted a series of sensitive drugs targeting KLF3. Conclusion The role of KLF3 in the tumor microenvironment of various types of tumors cannot be underestimated, and it has significant potential as a biomarker for predicting the response to immunotherapy. In particular, it plays an important role in the progression of pancreatic cancer.
Collapse
Affiliation(s)
- Jinfeng Zhu
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Hong Teng
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Department of Medical Genetics, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- School of Public Health, Nanchang University, Nanchang, Jiangxi, China
| | - Xiaojian Zhu
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Jingxuan Yuan
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Department of Medical Genetics, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- School of Public Health, Nanchang University, Nanchang, Jiangxi, China
| | - Qiong Zhang
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Department of Medical Genetics, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- School of Public Health, Nanchang University, Nanchang, Jiangxi, China
| | - Yeqing Zou
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- Department of Medical Genetics, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- School of Public Health, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
9
|
Chen XQ, Ma J, Xu D, Xiang ZL. Comprehensive analysis of KLF2 as a prognostic biomarker associated with fibrosis and immune infiltration in advanced hepatocellular carcinoma. BMC Bioinformatics 2023; 24:270. [PMID: 37386390 PMCID: PMC10308631 DOI: 10.1186/s12859-023-05391-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/17/2023] [Indexed: 07/01/2023] Open
Abstract
PURPOSE Most Hepatocellular carcinoma (HCC) patients are in advanced or metastatic stage at the time of diagnosis. Prognosis for advanced HCC patients is dismal. This study was based on our previous microarray results, and aimed to explore the promising diagnostic and prognostic markers for advanced HCC by focusing on the important function of KLF2. METHODS The Cancer Genome Atlas (TCGA), Cancer Genome Consortium database (ICGC), and the Gene Expression Comprehensive Database (GEO) provided the raw data of this study research. The cBioPortal platform, CeDR Atlas platform, and the Human Protein Atlas (HPA) website were applied to analyze the mutational landscape and single-cell sequencing data of KLF2. Basing on the results of single-cell sequencing analyses, we further explored the molecular mechanism of KLF2 regulation in the fibrosis and immune infiltration of HCC. RESULTS Decreased KLF2 expression was discovered to be mainly regulated by hypermethylation, and indicated a poor prognosis of HCC. Single-cell level expression analyses revealed KLF2 was highly expressed in immune cells and fibroblasts. The function enrichment analysis of KLF2 targets indicated the crucial association between KLF2 and tumor matrix. 33-genes related with cancer associated fibroblasts (CAFs) were collected to identify the significant association of KLF2 with fibrosis. And SPP1 was validated as a promising prognostic and diagnostic marker for advanced HCC patients. CXCR6 CD8+ T cells were noted as a predominant proportion in the immune microenvironment, and T cell receptor CD3D was discovered to be a potential therapeutic biomarker for HCC immunotherapy. CONCLUSION This study identified that KLF2 is an important factor promoting HCC progression by affecting the fibrosis and immune infiltration, highlighting its great potential as a novel prognostic biomarker for advanced HCC.
Collapse
Affiliation(s)
- Xue-Qin Chen
- Department of Radiation Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Jie Ma
- Department of Radiation Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Di Xu
- Department of Radiation Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Zuo-Lin Xiang
- Department of Radiation Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
- Department of Radiation Oncology, Shanghai East Hospital Ji'an hospital, Jiangxi, 343000, China.
| |
Collapse
|
10
|
Li Y, Wang Y, Zou Q, Li S, Zhang F. KLF3 Transcription Activates WNT1 and Promotes the Growth and Metastasis of Gastric Cancer via Activation of the WNT/β-Catenin Signaling Pathway. J Transl Med 2023; 103:100078. [PMID: 36827869 DOI: 10.1016/j.labinv.2023.100078] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/09/2023] [Accepted: 01/24/2023] [Indexed: 02/04/2023] Open
Abstract
The transcription factor Krüppel-like factor (KLF) 3 is one of the members of the KLF family, which plays an important role in tumor progression. Nevertheless, the role of KLF3 in the growth and metastasis of gastric cancer (GC) still needs to be elucidated. Bioinformatics analysis showed that KLF3 was overexpressed in patients with GC, and the high expression of KLF3 was correlated with poor survival. KLF3 was also overexpressed in GC clinical samples and cell lines. In vitro functional role of KLF3 in GC cells was explored by a gain-of-function and loss-of-function assay. Overexpressed KLF3 promoted the cell proliferation, migration, invasion, and epithelial-mesenchymal transition of GC cells, whereas suppressed KLF3 inhibited these biological behaviors. The clinical samples and bioinformatics analysis showed that WNT1 was also highly expressed in GC tumor tissues and positively correlated with KLF3 expression. The luciferase reporter assay and chromatin immunoprecipitation result confirmed that KLF3 could directly bind to the WNT1 promoter to increase the transcriptional activity of WNT1, thus regulating its expression. Overexpressed KLF3 enhanced the protein expression level of p-GSK3β(Ser9) and β-catenin, the key elements in the WNT/β-catenin signaling pathway. Repression of KLF3 decreased the level of p-GSK3β(Ser9) and β-catenin. Immunofluorescence images showed that KLF3 promoted nuclear β-catenin accumulation. Inhibition of WNT1 attenuated the proliferation, migration, and invasiveness of KLF3-overexpressing GC cells. Moreover, the xenograft mouse model confirmed that KLF3 promotes GC tumor growth and metastasis in vivo. Our results demonstrated that KLF3 activates the WNT/β-catenin signaling pathway via WNT1 to promote GC tumor growth and metastasis, indicating that repression of KLF3 may act as a potential therapeutic target for patients with GC.
Collapse
Affiliation(s)
- Ying Li
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yu Wang
- Endoscopy Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Qinguang Zou
- Department of Thoracic Surgery, Jilin Cancer Hospital, Changchun, Jilin, China
| | - Shouqing Li
- Tumor Integrative Medicine Center, Jilin Province People's Hospital, Changchun, Jilin, China
| | - Fan Zhang
- Department of Gastroenterology, The First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
11
|
Lee E, Cheung J, Bialkowska AB. Krüppel-like Factors 4 and 5 in Colorectal Tumorigenesis. Cancers (Basel) 2023; 15:cancers15092430. [PMID: 37173904 PMCID: PMC10177156 DOI: 10.3390/cancers15092430] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023] Open
Abstract
Krüppel-like factors (KLFs) are transcription factors regulating various biological processes such as proliferation, differentiation, migration, invasion, and homeostasis. Importantly, they participate in disease development and progression. KLFs are expressed in multiple tissues, and their role is tissue- and context-dependent. KLF4 and KLF5 are two fascinating members of this family that regulate crucial stages of cellular identity from embryogenesis through differentiation and, finally, during tumorigenesis. They maintain homeostasis of various tissues and regulate inflammation, response to injury, regeneration, and development and progression of multiple cancers such as colorectal, breast, ovarian, pancreatic, lung, and prostate, to name a few. Recent studies broaden our understanding of their function and demonstrate their opposing roles in regulating gene expression, cellular function, and tumorigenesis. This review will focus on the roles KLF4 and KLF5 play in colorectal cancer. Understanding the context-dependent functions of KLF4 and KLF5 and the mechanisms through which they exert their effects will be extremely helpful in developing targeted cancer therapy.
Collapse
Affiliation(s)
- Esther Lee
- Department of Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Jacky Cheung
- Department of Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Agnieszka B Bialkowska
- Department of Medicine, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
12
|
Weidle UH, Nopora A. Up-regulated Circular RNAs in Colorectal Cancer: New Entities for Therapy and Tools for Identification of Therapeutic Targets. Cancer Genomics Proteomics 2023; 20:132-153. [PMID: 36870691 PMCID: PMC9989668 DOI: 10.21873/cgp.20369] [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: 11/22/2022] [Revised: 12/20/2022] [Accepted: 12/28/2022] [Indexed: 03/06/2023] Open
Abstract
Patients with disseminated colorectal cancer have a dismal prognosis with a 5-year survival rate of only 13%. In order to identify new treatment modalities and new targets, we searched the literature for up-regulated circular RNAs in colorectal cancer which induce tumor growth in corresponding preclinical in vivo models. We identified nine circular RNAs that mediate resistance against chemotherapeutic agents, seven that up-regulate transmembrane receptors, five that induce secreted factors, nine that activate signaling components, five which up-regulate enzymes, six which activate actin-related proteins, six which induce transcription factors and two which up-regulate the MUSASHI family of RNA binding proteins. All of the circular RNAs discussed in this paper induce the corresponding targets by sponging microRNAs (miRs) and can be inhibited by RNAi or shRNA in vitro and in xenograft models. We have focused on circular RNAs with demonstrated activity in preclinical in vivo models because the latter is an important milestone in drug development. All circular RNAs with in vitro activity only data are not referenced in this review. The translational impact of inhibition of these circular RNAs and of the identified targets for treatment of colorectal cancer (CRC) are discussed.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Adam Nopora
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
13
|
Zhang Y, Yao C, Ju Z, Jiao D, Hu D, Qi L, Liu S, Wu X, Zhao C. Krüppel-like factors in tumors: Key regulators and therapeutic avenues. Front Oncol 2023; 13:1080720. [PMID: 36761967 PMCID: PMC9905823 DOI: 10.3389/fonc.2023.1080720] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/09/2023] [Indexed: 01/26/2023] Open
Abstract
Krüppel-like factors (KLFs) are a group of DNA-binding transcriptional regulators with multiple essential functions in various cellular processes, including proliferation, migration, inflammation, and angiogenesis. The aberrant expression of KLFs is often found in tumor tissues and is essential for tumor development. At the molecular level, KLFs regulate multiple signaling pathways and mediate crosstalk among them. Some KLFs may also be molecular switches for specific biological signals, driving their transition from tumor suppressors to promoters. At the histological level, the abnormal expression of KLFs is closely associated with tumor cell stemness, proliferation, apoptosis, and alterations in the tumor microenvironment. Notably, the role of each KLF in tumors varies according to tumor type and different stages of tumor development rather than being invariant. In this review, we focus on the advances in the molecular biology of KLFs, particularly the regulations of several classical signaling pathways by these factors, and the critical role of KLFs in tumor development. We also highlight their strong potential as molecular targets in tumor therapy and suggest potential directions for clinical translational research.
Collapse
Affiliation(s)
- Yuchen Zhang
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chongjie Yao
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ziyong Ju
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Danli Jiao
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dan Hu
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Qi
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shimin Liu
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China,Shanghai Research Institute of Acupuncture and Meridian, Shanghai, China
| | - Xueqing Wu
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China,*Correspondence: Chen Zhao, ; Xueqing Wu,
| | - Chen Zhao
- School of Acupuncture-moxibustion and Tuina, Shanghai University of Traditional Chinese Medicine, Shanghai, China,*Correspondence: Chen Zhao, ; Xueqing Wu,
| |
Collapse
|
14
|
Yu Z, Chen Z, Zhou G, Zhou X, Ma H, Yu Y, Wang X, Cao X. miR-92a-3p promotes breast cancer proliferation by regulating the KLF2/BIRC5 axis. Thorac Cancer 2022; 13:2992-3000. [PMID: 36100919 PMCID: PMC9626348 DOI: 10.1111/1759-7714.14648] [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: 08/07/2022] [Revised: 08/25/2022] [Accepted: 08/28/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Breast cancer remains the most common malignancy in females around the world. Recently, a growing number of studies have focused on gene dysregulation. In our previous study, Krüppel-like factors (KLFs) were found to play essential roles in breast cancer development, among which KLF2 could function as a tumor suppressor. Nevertheless, the underlying molecular mechanism remains unclear. METHODS miR-92a-3p was identified as the upstream regulator of KLF2 by starBase v.3.0. The regulation of KLF2 by miR-92a-3p was verified by a series of in vitro and in vivo assays. Further exploration revealed that Baculoviral IAP Repeat Containing 5 (BIRC5) was the target of KLF2. ChIP assay, dual-luciferase reporter analysis, quantitative real-time PCR, and western blot were performed for verification. RESULTS miR-92a-3p functioned as a tumor promoter by inhibiting KLF2 by binding to its 3'-untranslated region (3'-UTR). In addition, KLF2 could transcriptionally suppress the expression of BIRC5. CONCLUSION Collectively, our results uncovered the miR-92a-3p/KLF2/BIRC5 axis in breast cancer and provided a potential mechanism for breast cancer development, which may serve as promising strategies for breast cancer therapy.
Collapse
Affiliation(s)
- Zhi‐Hao Yu
- The First Department of Breast CancerTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina,Key Laboratory of Cancer Prevention and TherapyTianjinChina,Tianjin's Clinical Research Center for CancerTianjinChina,Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical University, Ministry of EducationTianjinChina
| | - Zhao‐Hui Chen
- The First Department of Breast CancerTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina,Key Laboratory of Cancer Prevention and TherapyTianjinChina,Tianjin's Clinical Research Center for CancerTianjinChina,Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical University, Ministry of EducationTianjinChina
| | - Guang‐Lei Zhou
- The First Department of Breast CancerTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina,Key Laboratory of Cancer Prevention and TherapyTianjinChina,Tianjin's Clinical Research Center for CancerTianjinChina,Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical University, Ministry of EducationTianjinChina
| | - Xue‐Jie Zhou
- The First Department of Breast CancerTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina,Key Laboratory of Cancer Prevention and TherapyTianjinChina,Tianjin's Clinical Research Center for CancerTianjinChina,Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical University, Ministry of EducationTianjinChina
| | - Hai‐Yan Ma
- The First Department of Breast CancerTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina,Key Laboratory of Cancer Prevention and TherapyTianjinChina,Tianjin's Clinical Research Center for CancerTianjinChina,Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical University, Ministry of EducationTianjinChina
| | - Yue Yu
- The First Department of Breast CancerTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina,Key Laboratory of Cancer Prevention and TherapyTianjinChina,Tianjin's Clinical Research Center for CancerTianjinChina,Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical University, Ministry of EducationTianjinChina
| | - Xin Wang
- The First Department of Breast CancerTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina,Key Laboratory of Cancer Prevention and TherapyTianjinChina,Tianjin's Clinical Research Center for CancerTianjinChina,Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical University, Ministry of EducationTianjinChina
| | - Xu‐Chen Cao
- The First Department of Breast CancerTianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for CancerTianjinChina,Key Laboratory of Cancer Prevention and TherapyTianjinChina,Tianjin's Clinical Research Center for CancerTianjinChina,Key Laboratory of Breast Cancer Prevention and TherapyTianjin Medical University, Ministry of EducationTianjinChina
| |
Collapse
|