101
|
Yang X, Chen C, Li L, Xiao T, Zou YD, Zheng D. Current research advances in microRNA-mediated regulation of Krüppel-like factor 4 in cancer: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:948. [PMID: 34350263 PMCID: PMC8263881 DOI: 10.21037/atm-21-2347] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 05/28/2021] [Indexed: 11/06/2022]
Abstract
Objective The purpose of this study was to investigate the miRNAs and related mechanisms that regulates KLF4 in different cancers. Furthermore, we summarized the potential targets of miRNAs regulating the KLF4 pathway in cancer research. Background MiRNAs are single-stranded, endogenous non-coding small RNAs, some of which are related to human cancers. miRNAs carry out post-transcriptional gene regulation through translation inhibition and degradation of target messenger RNAs (mRNAs) via complementarily pairing with their 3' untranslated regions. KLF4 is an important transcription factor with complex involvement in cancer. Increasing evidence shows that miRNAs are dysregulated in cancer and can regulate cancer-related signaling pathways, thereby affecting tumor progression. Methods Systematic scientific literature searches were undertaken on PubMed using the following terms: "miRNAs and KLF4", "KLF4 and cancer", "miRNAs and cancer", and "miRNAs, KLF4 and cancer". Relevant papers were retrieved and further results were found by reviewing related papers and the references of the retrieved papers. We then conducted a narrative overview of the literature to summarize the results of the papers. Conclusions The role of KLF4 in cancer varies in a context-dependent manner. KLF4-regulating miRNAs in different tumors include miR-124, miR-9-5p, miR-10b, miR-18a, miR-25-3p, miR-10b, miR-92a, miR-103, miR-155, miR-135b-5p, miR-32-5p, miR-148-3p, miR-152-3p, miR-10b, miR-25, miR-3120-5p, miR-7, miR-1233-3p, miR-10b, miR-145, miR-139-5p, miR-16, miR-152, miR-375, and miR-145.
Collapse
Affiliation(s)
- Xi Yang
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China.,Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Cheng Chen
- Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Li Li
- Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Tian Xiao
- Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Yong-Dong Zou
- Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Duo Zheng
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China.,Shenzhen University International Cancer Center, Department of Cell Biology and Genetics, School of Medicine, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| |
Collapse
|
102
|
An ZJ, Li Y, Tan BB, Zhao Q, Fan LQ, Zhang ZD, Zhao XF, Li SY. Up-regulation of KLF17 expression increases the sensitivity of gastric cancer to 5-fluorouracil. Int J Immunopathol Pharmacol 2021; 35:20587384211010925. [PMID: 33960231 PMCID: PMC8113926 DOI: 10.1177/20587384211010925] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
It has been reported that the expression of Krüppel-like factor 17 (KLF17) was
associated with the occurrence, development, invasion, metastasis and
chemotherapy resistance of various tumors. However, the detailed mechanisms by
which KLF17 promotes chemotherapy resistance in gastric cancer (GC) have not
been fully investigated. In the present study, we collected the GC tissues and
non-tumor tissues (matched adjacent normal tissues with corresponding GC
tissues) of 60 GC patients, used qRT-PCR, Western blot and immunohistochemistry
assay to analyze the relationship between the expression of KLF17 and the
clinical pathological data of the patients. The effect of KLF17 on the
sensitivity of GC cell lines to 5-fluorouracil (5-FU), and the potential
mechanism were detected by MTS assay, Flow cytometry assay, and Western blot.
Compared with non-tumor tissues, the expression level of KLF17 in GC tissue was
significantly down-regulated, and the expression level of KLF17 in GES-1 cell
line and GC cell lines also had a similar trend. Down-regulated expression of
KLF17 is related to tumor size, invasion, regional lymph node metastasis, and
TNM staging. Furthermore, through upregulating the expression of KLF17, the
sensitivity of BGC-823 and SGC-7901 cell lines to 5-FU was obviously increased.
Mechanistically, upregulation the expression of KLF17 can inhibit the
expressions of P-glycoprotein (P-gp), multidrug resistance protein 1 (MRP1), and
B-Cell lymphoma-2 (BCL-2), which have been reported to be associated with drug
resistance and cell proliferation. Collectively, these data implied that KLF17
has the biological effect of inhibiting chemotherapy resistance of GC, and it
could be a potential strategy for the GC chemotherapy resistance.
Collapse
Affiliation(s)
- Zhao-Jie An
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yong Li
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Bi-Bo Tan
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Qun Zhao
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Li-Qiao Fan
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhi-Dong Zhang
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xue-Feng Zhao
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shao-Yi Li
- Department of General Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
103
|
ITIH5, a p53-responsive gene, inhibits the growth and metastasis of melanoma cells by downregulating the transcriptional activity of KLF4. Cell Death Dis 2021; 12:438. [PMID: 33935281 PMCID: PMC8089095 DOI: 10.1038/s41419-021-03707-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 04/08/2021] [Accepted: 04/12/2021] [Indexed: 02/08/2023]
Abstract
ITIH5, a member of the inter-α-trypsin inhibitory (ITI) gene family, acts as a putative tumour-suppressor gene in many cancers. However, its role and the regulatory mechanism in melanoma are still unclear. Here, we found that the expression of ITIH5 was decreased in melanoma tissues compared with normal skin tissues. Decreased expression of ITIH5 was correlated with clinicopathological features and predicted poor prognosis in patients with melanoma. Forced expression of ITIH5 significantly inhibited melanoma cell proliferation and metastasis in vitro and ex vivo while knockdown of ITIH5 expression enhanced the malignant behaviour of melanoma cells. In further mechanistic studies, we showed that p53 can directly bind to the promoter of ITIH5 and thus promotes transcription of ITIH5 in melanoma cells. Additionally, we found that ITIH5 interacted with Krüppel-like factor 4 (KLF4) and inhibited its transcriptional activity. Collectively, our data not only identified a tumour-suppressive role of ITIH5 in melanoma but also revealed that upregulation of ITIH5 by p53 suppressed melanoma cell growth and migration likely by downmodulating the transcriptional activity of KLF4.
Collapse
|
104
|
METTL3-mediated m 6A methylation of SPHK2 promotes gastric cancer progression by targeting KLF2. Oncogene 2021; 40:2968-2981. [PMID: 33758320 DOI: 10.1038/s41388-021-01753-1] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 02/27/2021] [Accepted: 03/08/2021] [Indexed: 02/01/2023]
Abstract
N6-methyladenosine (m6A) RNA methylation is profoundly involved in epigenetic regulation, especially for carcinogenesis and tumor progression. Mounting evidence suggests that methyltransferase METTL3 regulates malignant behaviors of gastric cancer (GC). However, the clinical significance and biological implication of SPHK2 and its related m6A modification in GC remain unclear. In this study, quantitative real-time PCR (qRT-PCR), western blot and immunohistochemistry were utilized to detect the expression profiles and prognostic significance of SPHK2 in GC. Here, we showed that increased SPHK2 was signified a poor prognosis of GC patients. Phosphorylation and ubiquitination assays were used to investigate the possible mechanisms of SPHK2-mediated KLF2 expression. SPHK2 can promote the phosphorylation of KLF2, which triggers the ubiquitination and degradation of KLF2 protein in GC. Methylated RNA immunoprecipitation (MeRIP) was performed to uncover the m6A modification of SPHK2 mRNA. METTL3 promotes translation of SPHK2 mRNA via an m6A-YTHDF1-dependent manner. Functionally, SPHK2 facilitates GC cell proliferation, migration and invasion by inhibiting KLF2 expression. SPHK2/KLF2 regulates the cell proliferation, migration, and invasion induced by METTL3 in GC. Overall, our findings reveal that METTL3-mediated m6A modification of SPHK2 contributes to GC progression, which extends the understanding of the importance m6A methylation in GC and represents a potential target for GC therapy.
Collapse
|
105
|
Dai DL, Li X, Wang L, Xie C, Jin Y, Zeng MS, Zuo Z, Xia TL. Identification of an N6-methyladenosine-mediated positive feedback loop that promotes Epstein-Barr virus infection. J Biol Chem 2021; 296:100547. [PMID: 33741341 PMCID: PMC8063736 DOI: 10.1016/j.jbc.2021.100547] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/10/2021] [Accepted: 03/15/2021] [Indexed: 12/11/2022] Open
Abstract
N6-methyladenosine (m6A) is among the most abundant mRNA modifications, particularly in eukaryotes, and is found in mammals, plants, and even some viruses. Although essential for the regulation of many biological processes, the exact role of m6A modification in virus–host interaction remains largely unknown. Here, using m6A -immunoprecipitation and sequencing, we find that Epstein–Barr virus (EBV) infection decreases the m6A modification of transcriptional factor KLF4 mRNA and subsequently increases its protein level. Mechanistically, EBV immediate-early protein BZLF1 interacts with the promoter of m6A methyltransferase METTL3, inhibiting its expression. Subsequently, the decrease of METTL3 reduces the level of KLF4 mRNA m6A modification, preventing its decay by the m6A reader protein YTHDF2. As a result, KLF4 protein level is upregulated and, in turn, promotes EBV infection of nasopharyngeal epithelial cells. Thus, our results suggest the existence of a positive feedback loop formed between EBV and host molecules via cellular mRNA m6A levels, and this feedback loop acts to facilitate viral infection. This mechanism contains multiple potential targets for controlling viral infectious diseases.
Collapse
Affiliation(s)
- Dan-Ling Dai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Xingyang Li
- Department of Temporomandibular Joint Surgery, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, P. R. China
| | - Lin Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China; Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Chu Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Yanan Jin
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China; Department of VIP Region, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Mu-Sheng Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Zhixiang Zuo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.
| | - Tian-Liang Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.
| |
Collapse
|
106
|
Gao L, Yang X, Liang B, Jia Y, Tan S, Chen A, Cao P, Zhang Z, Zheng S, Sun L, Zhang F, Shao J. Autophagy-induced p62 accumulation is required for curcumol to regulate KLF5-mediated angiogenesis in liver sinusoidal endothelial cells. Toxicology 2021; 452:152707. [PMID: 33549628 DOI: 10.1016/j.tox.2021.152707] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/20/2021] [Accepted: 02/01/2021] [Indexed: 02/07/2023]
Abstract
Liver pathological angiogenesis is considered to be one of the key events in the development of liver fibrosis. Autophagy is a defense and stress regulation mechanism. However, whether autophagy regulates pathological angiogenesis in liver fibrosis is still questionable. Here, we aimed to study how curcumol regulated liver sinusoidal endothelial cells (LSECs) angiogenesis through autophagy. We found that curcumol (10, 20 and 40 μM) could inhibit the expression of angiogenesis markers in the LSECs. Importantly, we showed that curcumol might influence LSEC pathological angiogenesis by regulating autophagy level. Furthermore, we indicated that the transcription factor Krüppel-like factor 5 (KLF5) was considered as a key target for curcumol to regulate LSEC angiogenesis. Interestingly, we also suggested that autophagy was as a potential mechanism for curcumol to restrain KLF5 expression. Increased autophagy level could impair the suppression effect of curcumol on KLF5. Fascinatingly, our results indicated that curcumol inhibited autophagy and led to p62 accumulation, which might be a regulation mechanism of KLF5 degradation. Finally, in mice liver fibrosis model, we unanimously showed that curcumol (30 mg/kg) inhibited pathological angiogenesis by reducing LSEC autophagy level and suppressing KLF5 expression. Collectively, these results provided a deeper insight into the molecular mechanism of curcumol to inhibit LSEC pathological angiogenesis during liver fibrosis.
Collapse
Affiliation(s)
- Liyuan Gao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiang Yang
- Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, China
| | - Baoyu Liang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Jia
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shanzhong Tan
- Department of Integrated TCM and Western Medicine, Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Anping Chen
- Department of Pathology, School of Medicine, Saint Louis University, St Louis, USA
| | - Peng Cao
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zili Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lixia Sun
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Jiangjuan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
107
|
Abstract
IMPORTANCE Clinical decision and immunosuppression dosing in kidney transplantation rely on transplant biopsy tissue histology even though histology has low specificity, sensitivity, and reproducibility for rejection diagnosis. The inclusion of stable allografts in mechanistic and clinical studies is vital to provide a normal, noninjured comparative group for all interrogative studies on understanding allograft injury. OBJECTIVE To refine the definition of a stable allograft as one that is clinically, histologically, and molecularly quiescent using publicly available transcriptomics data. DESIGN, SETTING, AND PARTICIPANTS In this prognostic study, the National Center for Biotechnology Information Gene Expression Omnibus was used to search for microarray gene expression data from kidney transplant tissues, resulting in 38 studies from January 1, 2017, to December 31, 2018. The diagnostic annotations included 510 acute rejection (AR) samples, 1154 histologically stable (hSTA) samples, and 609 normal samples. Raw fluorescence intensity data were downloaded and preprocessed followed by data set merging and batch correction. MAIN OUTCOMES AND MEASURES The primary measure was area under the receiver operating characteristics curve from a set of feature selected genes and cell types for distinguishing AR from normal kidney tissue. RESULTS Within the 28 data sets, the feature selection procedure identified a set of 6 genes (KLF4, CENPJ, KLF2, PPP1R15A, FOSB, TNFAIP3) (area under the curve [AUC], 0.98) and 5 immune cell types (CD4+ T-cell central memory [Tcm], CD4+ T-cell effector memory [Tem], CD8+ Tem, natural killer [NK] cells, and Type 1 T helper [TH1] cells) (AUC, 0.92) that were combined into 1 composite Instability Score (InstaScore) (AUC, 0.99). The InstaScore was applied to the hSTA samples: 626 of 1154 (54%) were found to be immune quiescent and redefined as histologically and molecularly stable (hSTA/mSTA); 528 of 1154 (46%) were found to have molecular evidence of rejection (hSTA/mAR) and should not have been classified as stable allografts. The validation on an independent cohort of 6 months of protocol biopsy samples in December 2019 showed that hSTA/mAR samples had a significant change in graft function (r = 0.52, P < .001) and graft loss at 5-year follow-up (r = 0.17). A drop by 10 mL/min/1.73m2 in estimated glomerular filtration rate was estimated as a threshold in allograft transitioning from hSTA/mSTA to hSTA/mAR. CONCLUSIONS AND RELEVANCE The results of this prognostic study suggest that the InstaScore could provide an important adjunct for comprehensive and highly quantitative phenotyping of protocol kidney transplant biopsy samples and could be integrated into clinical care for accurate estimation of subsequent patient clinical outcomes.
Collapse
Affiliation(s)
- Dmitry Rychkov
- Division of Multi-Organ Transplantation, Department of Surgery, University of California, San Francisco
- Bakar Computational Health Sciences Institute, University of California, San Francisco
| | - Swastika Sur
- Division of Multi-Organ Transplantation, Department of Surgery, University of California, San Francisco
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco
- Department of Pediatrics, University of California, San Francisco
| | - Minnie M. Sarwal
- Division of Multi-Organ Transplantation, Department of Surgery, University of California, San Francisco
| |
Collapse
|
108
|
Shi X, Zheng Y, Jiang L, Zhou B, Yang W, Li L, Ding L, Huang M, Gery S, Lin DC, Koeffler HP. EWS-FLI1 regulates and cooperates with core regulatory circuitry in Ewing sarcoma. Nucleic Acids Res 2020; 48:11434-11451. [PMID: 33080033 PMCID: PMC7672457 DOI: 10.1093/nar/gkaa901] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/22/2020] [Accepted: 10/01/2020] [Indexed: 12/11/2022] Open
Abstract
Core regulatory circuitry (CRC)-dependent transcriptional network is critical for developmental tumors in children and adolescents carrying few gene mutations. However, whether and how CRC contributes to transcription regulation in Ewing sarcoma is unknown. Here, we identify and functionally validate a CRC 'trio' constituted by three transcription factors (TFs): KLF15, TCF4 and NKX2-2, in Ewing sarcoma cells. Epigenomic analyses demonstrate that EWS-FLI1, the primary fusion driver for this cancer, directly establishes super-enhancers of each of these three TFs to activate their transcription. In turn, KLF15, TCF4 and NKX2-2 co-bind to their own and each other's super-enhancers and promoters, forming an inter-connected auto-regulatory loop. Functionally, CRC factors contribute significantly to cell proliferation of Ewing sarcoma both in vitro and in vivo. Mechanistically, CRC factors exhibit prominent capacity of co-regulating the epigenome in cooperation with EWS-FLI1, occupying 77.2% of promoters and 55.6% of enhancers genome-wide. Downstream, CRC TFs coordinately regulate gene expression networks in Ewing sarcoma, controlling important signaling pathways for cancer, such as lipid metabolism pathway, PI3K/AKT and MAPK signaling pathways. Together, molecular characterization of the oncogenic CRC model advances our understanding of the biology of Ewing sarcoma. Moreover, CRC-downstream genes and signaling pathways may contain potential therapeutic targets for this malignancy.
Collapse
Affiliation(s)
- Xianping Shi
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease; Affiliated Cancer Hospital of Guangzhou Medical University; Sino-French Hoffmann institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 510120, P.R. China
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Yueyuan Zheng
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Liling Jiang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation; State Key Laboratory of Respiratory Disease; Affiliated Cancer Hospital of Guangzhou Medical University; Sino-French Hoffmann institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 510120, P.R. China
| | - Bo Zhou
- Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Wei Yang
- Departments of Surgery and Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Liyan Li
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Lingwen Ding
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117600, Singapore
| | - Moli Huang
- School of Biology and Basic Medical Sciences, Soochow University, Suzhou 215123, P.R. China
| | - Sigal Gery
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - De-Chen Lin
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - H Phillip Koeffler
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117600, Singapore
- National University Cancer Institute, National University Hospital Singapore, Singapore 119074, Singapore
| |
Collapse
|
109
|
McCulloch KJ, Koenig KM. Krüppel-like factor/specificity protein evolution in the Spiralia and the implications for cephalopod visual system novelties. Proc Biol Sci 2020; 287:20202055. [PMID: 33081641 PMCID: PMC7661307 DOI: 10.1098/rspb.2020.2055] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The cephalopod visual system is an exquisite example of convergence in biological complexity. However, we have little understanding of the genetic and molecular mechanisms underpinning its elaboration. The generation of new genetic material is considered a significant contributor to the evolution of biological novelty. We sought to understand if this mechanism may be contributing to cephalopod-specific visual system novelties. Specifically, we identified duplications in the Krüppel-like factor/specificity protein (KLF/SP) sub-family of C2H2 zinc-finger transcription factors in the squid Doryteuthis pealeii. We cloned and analysed gene expression of the KLF/SP family, including two paralogs of the DpSP6-9 gene. These duplicates showed overlapping expression domains but one paralog showed unique expression in the developing squid lens, suggesting a neofunctionalization of DpSP6-9a. To better understand this neofunctionalization, we performed a thorough phylogenetic analysis of SP6-9 orthologues in the Spiralia. We find multiple duplications and losses of the SP6-9 gene throughout spiralian lineages and at least one cephalopod-specific duplication. This work supports the hypothesis that gene duplication and neofunctionalization contribute to novel traits like the cephalopod image-forming eye and to the diversity found within Spiralia.
Collapse
Affiliation(s)
- Kyle J McCulloch
- Department of Organismic and Evolutionary Biology, Harvard University Cambridge, MA 02138, USA.,John Harvard Distinguished Science Fellows, Harvard University, Cambridge, MA 02138, USA
| | - Kristen M Koenig
- Department of Organismic and Evolutionary Biology, Harvard University Cambridge, MA 02138, USA.,John Harvard Distinguished Science Fellows, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
110
|
Transcription factor Kruppel-like factor 5 positively regulates the expression of AarF domain containing kinase 4. Mol Biol Rep 2020; 47:8419-8427. [PMID: 33033902 DOI: 10.1007/s11033-020-05882-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 09/26/2020] [Accepted: 09/30/2020] [Indexed: 02/04/2023]
Abstract
AarF domain containing kinase 4 (ADCK4) is identified as a candidate gene associated with hereditary nephrotic syndrome (NS). Kruppel-like factor 5 (KLF5) is reported to promote podocyte survival by blocking the ERK/p38 MAPK pathways. Both ADCK4 and KLF5 are involved in the occurrence and development of podocyte disease, but their interaction remains unclear. Firstly, we found that the mRNA levels of ADCK4 and KLF5 decreased in NS patients, and both levels showed an obvious linear relationship. Secondly, we cloned the ADCK4 promoter region and examined its promoter activity in Hela, A549, and HEK 293 cell lines. Deletion analysis showed that the region - 116/- 4 relative to the transcriptional start site (TSS) was the core region of ADCK4 promoter. Thirdly, mutation analysis showed that putative binding sites for KLF5 contributed to the ADCK4 promoter activity. In HEK293 cells, we found that KLF5 upregulated the mRNA and protein levels of ADCK4. Finally, our chromatin immunoprecipitation assay found that KLF5 could bind to the specific region of ADCK4 promoter. These results showed that KLF5 can positively regulate the transcriptional activity of ADCK4.
Collapse
|
111
|
The Krüppel-like factor 15-NFATc1 axis ameliorates podocyte injury: a novel rationale for using glucocorticoids in proteinuria diseases. Clin Sci (Lond) 2020; 134:1305-1318. [PMID: 32478397 DOI: 10.1042/cs20200075] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/07/2020] [Accepted: 05/29/2020] [Indexed: 01/19/2023]
Abstract
Podocyte injury and loss contribute to proteinuria, glomerulosclerosis and eventually kidney failure. Recent studies have demonstrated that the loss of Kruppel-like factor 15 (KLF15) in podocytes increases the susceptibility to injury; however, the mechanism underlying the protective effects on podocyte injury remains incompletely understood. Herein, we showed that KLF15 ameliorates podocyte injury through suppressing NFAT signaling and the salutary effects of the synthetic glucocorticoid dexamethasone in podocyte were partially mediated by the KLF15-NFATc1 axis. We found that KLF15 was significantly reduced in glomerular cells of proteinuric patients and in ADR-, LPS- or HG-treated podocyets in vitro. Overexpression of KLF15 attenuated podocyte apoptosis induced by ADR, LPS or HG and resulted in decreased expression of pro-apoptotic Bax and increased expression of anti-apoptotic Bcl-2. Conversely, the flow cytometry analysis and TUNEl assay demonstrated that loss of KLF15 accelerated podocyte apoptosis and we further found that 11R-VIVIT, a specific NFAT inhibitor, and NFATc1-siRNA rescued KLF15-deficient induced podocyte apoptosis. Meanwhile, Western blot and RT-qPCR showed that the expression of NFATc1 was up-regulated in KLF15 silenced podocytes and reduced in KLF15 overexpressed podocytes. Mechanistically, ChIP analysis showed that KLF15 bound to the NFATc1 promoter region -1984 to -1861base pairs upstream of the transcription start site and the binding amount was decreased after treatment with LPS. The dual-luciferase reporter assay indicated that NFATc1 was a direct target of KLF15. In addition, we found that in vitro treatment with dexamethasone induced a decrease of NFATc1 expression in podocytes and was abrogated by knockdown of KLF15. Hence, our results identify the critical role of the KLF15-NFATc1 axis in podocyte injury and loss, which may be involved in mediating the salutary effects of dexamethasone in podocytes.
Collapse
|
112
|
Yu T, Gong Y, Liu Y, Xia L, Zhao C, Liu L, Xie M, Wu Z, Zhao D, Qiu W, Wang Y, Zhang J, Ji M. KLF6 Acetylation Promotes Sublytic C5b-9-Induced Production of MCP-1 and RANTES in Experimental Mesangial Proliferative Glomerulonephritis. Int J Biol Sci 2020; 16:2340-2356. [PMID: 32760202 PMCID: PMC7378648 DOI: 10.7150/ijbs.46573] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/08/2020] [Indexed: 12/13/2022] Open
Abstract
Rat Thy-1 nephritis (Thy-1N) is an experimental mesangial proliferative glomerulonephritis (MsPGN) for studying human MsPGN. Although sublytic C5b-9 complex formation on glomerular mesangial cells (GMCs) and renal MCP-1 and RANTES production in rats with Thy-1N have been proved, the role and mechanism of MCP-1 or RANTES synthesis in GMCs induced by sublytic C5b-9 are poorly elucidated. In this study, we first found the expression of transcription factor (KLF6), co-activator (KAT7) and chemokines (MCP-1 and RANTES) was all up-regulated both in renal tissue of Thy-1N rats (in vivo) and in sublytic C5b-9-induced GMCs (in vitro). Further in vitro experiments revealed that KLF6 bound to MCP-1 promoter (-297 to -123 nt) and RANTES promoter (-343 to -191 nt), leading to MCP-1 and RANTES gene transcription. Meanwhile, KAT7 also bound to the same region of MCP-1 and RANTES promoter in a KLF6-dependent manner, and KLF6 was acetylated by KAT7 at lysine residue 100, which finally promoted MCP-1 and RANTES expression. Moreover, our in vivo experiments discovered that knockdown of renal KAT7 or KLF6 gene obviously reduced MCP-1 and RANTES production, GMCs proliferation, ECM accumulation, and proteinuria secretion in Thy-1N rats. Collectively, our study indicates that sublytic C5b-9-induced MCP-1 and RANTES synthesis is associated with KAT7-mediated KLF6 acetylation and elevated KLF6 transcriptional activity, which might provide a new insight into the pathogenesis of rat Thy-1N and human MsPGN.
Collapse
Affiliation(s)
- Tianyi Yu
- Department of Immunology, Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yajuan Gong
- Department of Immunology, Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yu Liu
- Department of Immunology, Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Lu Xia
- Department of Immunology, Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Chenhui Zhao
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Longfei Liu
- Department of Immunology, Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Mengxiao Xie
- Department of Immunology, Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Zhijiao Wu
- Department of Immunology, Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Dan Zhao
- Department of Immunology, Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Wen Qiu
- Department of Immunology, Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yingwei Wang
- Department of Immunology, Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Jing Zhang
- Department of Immunology, Key Laboratory of Immunological Environment and Disease, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Key Laboratory of Antibody Technology of Ministry of Health, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Mingde Ji
- Department of Laboratory Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| |
Collapse
|
113
|
Li Y, Tu S, Zeng Y, Zhang C, Deng T, Luo W, Lian L, Chen L, Xiong X, Yan X. KLF2 inhibits TGF-β-mediated cancer cell motility in hepatocellular carcinoma. Acta Biochim Biophys Sin (Shanghai) 2020; 52:485-494. [PMID: 32318691 DOI: 10.1093/abbs/gmaa024] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 10/17/2019] [Accepted: 12/26/2019] [Indexed: 12/16/2022] Open
Abstract
Feedback regulation plays a pivotal role in determining the intensity and duration of TGF-β signaling and subsequently affecting the pathophysiological roles of TGF-β, including those in liver malignancy. KLF2, a member of the Krüppel-like factor (KLF) family transcription factors, has been implicated in impeding hepatocellular carcinoma (HCC) development. However, the underlying molecular mechanisms are not fully understood. In the present study, we found that TGF-β stimulates the expression of KLF2 gene in several HCC cell lines. KLF2 protein is able to inhibit TGF-β/Smad signaling in HCC cells as assessed by luciferase reporter assay. Further studies indicated that KLF2 inhibits the transcriptional activity of Smad2/3 and Smad4 and ameliorates TGF-β-induced target gene expression, therefore creating a novel negative feedback loop in TGF-β signaling. Functionally, stably expression of KLF2 in HCCLM3 cells attenuated TGF-β-induced cancer cell motility in wound-healing and transwell assays by interfering with TGF-β-mediated upregulation of MMP2. Together, our results revealed that KLF2 protein has a tumor-suppressive function in HCC through a negative feedback loop over TGF-β signaling.
Collapse
Affiliation(s)
- Yining Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Shuo Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Yi Zeng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Cheng Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Tian Deng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Weicheng Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Lingyan Lian
- The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Ling Chen
- The Health Department of the First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - Xiangyang Xiong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Xiaohua Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
- Institute of Biomedical Sciences, Nanchang University, Nanchang 330006, China
| |
Collapse
|
114
|
Liang X, Xu W. miR-181a-5p regulates the proliferation and apoptosis of glomerular mesangial cells by targeting KLF6. Exp Ther Med 2020; 20:1121-1128. [PMID: 32742352 DOI: 10.3892/etm.2020.8780] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 12/12/2019] [Indexed: 12/22/2022] Open
Abstract
Diabetic nephropathy (DN) is a chronic loss of kidney function that frequently occurs in patients with diabetes mellitus and is characterized by abnormal glomerular mesangial cell (GMC) proliferation and apoptosis. By using microarray analysis, microRNA (miR)-181a-5p has previously been identified to be dysregulated in DN. The present study aimed to determine the underlying molecular mechanisms and function of miR-181a-5p in GMCs under DN conditions. First, reverse transcription-quantitative PCR was performed to detect miR-181a-5p and kruppel-like factor 6 (KLF6) expression in GMCs following high-glucose treatment. Subsequently, MTT and flow cytometric assays were performed in order to determine the effect of miR-181a-5p and KLF6 on high-glucose-driven GMC proliferation and apoptosis. After confirming that KLF6 was a target gene of miR-181a-5p via a bioinformatics analysis and luciferase reporter assay, the mRNA and protein expression levels of associated factors in different treatment groups were measured. The results demonstrated that miR-181a-5p was significantly downregulated, while KLF6 was significantly upregulated in GMCs following treatment with high glucose. Furthermore, overexpression of miR-181a led to suppression of cell proliferation and promoted apoptosis of GMCs induced by high glucose, while these effects were inhibited by co-transfection with KLF6. Finally, miR-181-5p was demonstrated to inhibit the expression of KLF6, Bcl-2, Wnt1 and β-catenin, while increasing the expression levels of Bax and caspase-3. In conclusion, the expression levels of miR-181a-5p were downregulated in GMCs following treatment with high glucose and overexpression of miR-181a-5p may inhibit GMC proliferation and promote apoptosis, at least partially through targeting KLF6 via the Wnt/β-catenin signaling pathway. Overall, the results of the present study suggest that miR-181a-5p may have a crucial role in the occurrence and development of DN and may be a valuable diagnostic marker and therapeutic target for DN.
Collapse
Affiliation(s)
- Xinyue Liang
- Department of Geriatrics, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, P.R. China
| | - Wen Xu
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200127, P.R. China
| |
Collapse
|
115
|
Zhao J, Su Y, Jiao J, Wang Z, Fang X, He X, Zhang X, Liu Z, Xu X. Identification of lncRNA and mRNA Biomarkers in Osteoarthritic Degenerative Meniscus by Weighted Gene Coexpression Network and Competing Endogenous RNA Network Analysis. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2123787. [PMID: 32685450 PMCID: PMC7341399 DOI: 10.1155/2020/2123787] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/26/2020] [Accepted: 04/15/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) play a crucial role in varieties of biological processes. This study is aimed at investigating meniscal degeneration-specific lncRNAs and mRNAs and their related networks in knee osteoarthritis (KOA). METHODS The dataset GSE98918, which included 24 meniscus samples and related clinical data, was downloaded from the Gene Expression Omnibus database. The differentially expressed lncRNAs and mRNAs in the meniscus between KOA and control groups were identified. Based on the enriched differentially expressed lncRNAs and mRNAs, we constructed the coexpression network using WGCNA (weighted correlation network analysis) and identified the critical module related to KOA. For mRNAs in the key module, gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were carried out using the DAVID database. A competing endogenous RNA network (ceRNA) based on the screened mRNAs, lncRNAs, and related miRNAs was constructed to reveal presumptive biomarkers further. Finally, the hub lncRNAs and mRNAs were screened, and the diagnostic value was evaluated using a receiver operating characteristic (ROC) curve. Hub mRNAs were validated using the dataset GSE113825. RESULTS We screened 208 significantly differentially expressed lncRNAs and mRNAs in menisci between the KOA and non-KOA samples, which were enriched in sixteen modules using WGCNA, especially the green module. Coexpression network based on the enriched differentially expressed lncRNAs and mRNAs in the green module uncovered 5 lncRNAs and 56 mRNAs. The lncRNA-miRNA-mRNA ceRNA network revealed that lnc-HLA-DQA1-5, lnc-RP11-127H5.1.1-1, lnc-RTN2-1, IGFBP4 (insulin-like growth factor binding protein 4), and KLF2 (Kruppel-like factor 2) were significantly correlated with the meniscus degeneration of KOA. ROC curve analysis revealed that these hub lncRNAs and mRNAs showed excellent diagnostic value for KOA. CONCLUSIONS These hub lncRNAs and mRNAs were potential prognostic biomarkers for the meniscus degeneration of KOA. Further studies are required to validate these new biomarkers and better understand the pathological process of the meniscus degeneration of KOA.
Collapse
Affiliation(s)
- Jun Zhao
- Department of Orthopaedics, Heilongjiang University of Chinese Medicine, Heping Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Yu Su
- Harbin Fifth Hospital, Jiankang Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Jianfei Jiao
- Harbin Fifth Hospital, Jiankang Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Zhengchun Wang
- Department of Orthopaedics, Heilongjiang University of Chinese Medicine, Heping Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Xiangchun Fang
- Department of Orthopaedics, Heilongjiang University of Chinese Medicine, Heping Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Xuefeng He
- Department of Orthopaedics, Heilongjiang University of Chinese Medicine, Heping Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Xiaofeng Zhang
- Department of Orthopaedics, Heilongjiang University of Chinese Medicine, Heping Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Zhao Liu
- Department of Orthopaedics, Heilongjiang University of Chinese Medicine, Heping Road, Xiangfang District, Harbin, Heilongjiang, China
| | - Xilin Xu
- Department of Orthopaedics, Heilongjiang University of Chinese Medicine, Heping Road, Xiangfang District, Harbin, Heilongjiang, China
| |
Collapse
|
116
|
Piskacek M, Havelka M, Jendruchova K, Knight A, Keegan LP. The evolution of the 9aaTAD domain in Sp2 proteins: inactivation with valines and intron reservoirs. Cell Mol Life Sci 2020; 77:1793-1810. [PMID: 31375868 PMCID: PMC11105055 DOI: 10.1007/s00018-019-03251-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/10/2019] [Accepted: 07/24/2019] [Indexed: 12/28/2022]
Abstract
The universal nine-amino-acid transactivation domains (9aaTADs) have been identified in numerous transcription activators. Here, we identified the conserved 9aaTAD motif in all nine members of the specificity protein (SP) family. Previously, the Sp1 transcription factor has been defined as a glutamine-rich activator. We showed by amino acid substitutions that the glutamine residues are completely dispensable for 9aaTAD function and are not conserved in the SP family. We described the origin and evolutionary history of 9aaTADs. The 9aaTADs of the ancestral Sp2 gene became inactivated in early chordates. We next discovered that an accumulation of valines in 9aaTADs inactivated their transactivation function and enabled their strict conservation during evolution. Subsequently, in chordates, Sp2 has duplicated and created new paralogs, Sp1, Sp3, and Sp4 (the SP1-4 clade). During chordate evolution, the dormancy of the Sp2 activation domain lasted over 100 million years. The dormant but still intact ancestral Sp2 activation domains allowed diversification of the SP1-4 clade into activators and repressors. By valine substitution in the 9aaTADs, Sp1 and Sp3 regained their original activator function found in ancestral lower metazoan sea sponges. Therefore, the vertebrate SP1-4 clade could include both repressors and activators. Furthermore, we identified secondary 9aaTADs in Sp2 introns present from fish to primates, including humans. In the gibbon genome, introns containing 9aaTADs were used as exons, which turned the Sp2 gene into an activator. Similarly, we identified introns containing 9aaTADs used conditionally as exons in the (SP family-unrelated) transcription factor SREBP1, suggesting that the intron-9aaTAD reservoir is a general phenomenon.
Collapse
Affiliation(s)
- Martin Piskacek
- Department of Pathological Physiology, Faculty of Medicine, Laboratory of Cancer Biology and Genetics, Masaryk University Brno, Kamenice 5, Brno, 625 00, Czech Republic.
| | - Marek Havelka
- Department of Pathological Physiology, Faculty of Medicine, Laboratory of Cancer Biology and Genetics, Masaryk University Brno, Kamenice 5, Brno, 625 00, Czech Republic
| | - Kristina Jendruchova
- Department of Pathological Physiology, Faculty of Medicine, Laboratory of Cancer Biology and Genetics, Masaryk University Brno, Kamenice 5, Brno, 625 00, Czech Republic
| | - Andrea Knight
- Department of Pathological Physiology, Faculty of Medicine, Gamma Delta T Cell Laboratory, Masaryk University Brno, Kamenice 5, Brno, 625 00, Czech Republic.
| | - Liam P Keegan
- CEITEC, Masaryk University, Kamenice 753/5, Pavilion A35, Brno, 62 500, Czech Republic.
| |
Collapse
|
117
|
Zhao H, Chen B, Li Z, Wang B, Li L. Long Noncoding RNA DANCR Suppressed Lipopolysaccharide-Induced Septic Acute Kidney Injury by Regulating miR-214 in HK-2 Cells. Med Sci Monit 2020; 26:e921822. [PMID: 32222722 PMCID: PMC7139187 DOI: 10.12659/msm.921822] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Acute kidney injury (AKI) is one of the most important causes of death in sepsis patients. Here, we first measured the level of DANCR (differentiation antagonizing nonprotein coding RNA) expression in AKI, and the potential mechanism was further elucidated. MATERIAL AND METHODS We used qRT-PCR to examine the level of DANCR in patient blood serum samples and in HK-2 cells. In addition, DANCR overexpression was established using lentiviral transfection in HK-2 cells. Subsequently, Cell Counting Kit-8 (CCK-8) assay and flow cytometry were applied to evaluate the role of DANCR in HK-2 cells treated with lipopolysaccharide (LPS). Enzyme linked immunosorbent assay (ELISA), western blot and recovery experiments were performed to elucidate the further mechanism. RESULTS We found that DANCR was decreased in the serum of AKI patients and HK-2 cells treated with LPS. Additionally, DANCR promoted cell viability and suppressed cell apoptosis and cytokine production in LPS-treated HK-2 cells. Bioinformatics analysis showed that DANCR served as a sponge for miR-214. Furthermore, DANCR inhibited the expression of Krüppel-like factor 6 (KLF6). CONCLUSIONS Our study suggests that AKI development could be alleviated by sponging miR-214 and regulating KLF6 expression, which provides a novel potential mechanism involved in the diagnosis and treatment of sepsis-induced AKI patients.
Collapse
Affiliation(s)
- Huajie Zhao
- Department of Intensive Care Unit, The Second Hospital of Tianjin Medical University, Tianjin, China (mainland)
| | - Bing Chen
- Department of Intensive Care Unit, The Second Hospital of Tianjin Medical University, Tianjin, China (mainland)
| | - Zhenyu Li
- Department of Intensive Care Unit, The Second Hospital of Tianjin Medical University, Tianjin, China (mainland)
| | - Bin Wang
- Department of Intensive Care Unit, The Second Hospital of Tianjin Medical University, Tianjin, China (mainland)
| | - Liyu Li
- Department of Intensive Care Unit, The Second Hospital of Tianjin Medical University, Tianjin, China (mainland)
| |
Collapse
|
118
|
Wang Z, Qiu X, Zhang H, Li W. KLF5 influences cell biological function and chemotherapy sensitivity through the JNK signaling pathway in anaplastic thyroid carcinoma. J Biochem Mol Toxicol 2020; 34:e22469. [PMID: 32173973 DOI: 10.1002/jbt.22469] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 01/03/2020] [Accepted: 01/31/2020] [Indexed: 01/15/2023]
Abstract
We aimed to investigate the effects of Krüppel-like factor 5 (KLF5) on cell biological function and chemotherapy sensitivity of anaplastic thyroid carcinoma (ATC) and explore the underlying mechanism. In this study, we found that KLF5 was expressed higher in ATC cells than that in normal thyroid cells. Knockdown of KLF5 inhibited proliferation, induced apoptosis and restrained invasion and migration abilities of ATC cells. KLF5 overexpression promoted proliferation and inhibited apoptosis of ATC cells in response to doxorubicin (Dox), whereas KLF5 knockdown increased the sensitivity of ATC cells to Dox. Multidrug resistance gene 1/permeability glycoprotein and ATP-binding cassette superfamily G member 2 were heightened in ATC cells with KLF5 overexpression, but the opposite results were found in sh-KLF5-treated cells. Phosphorylation (p)-c-Jun N-terminal kinase (JNK) was upregulated in KLF5 overexpression cells, whereas it was downregulated in the KLF5 knockdown treatment group. Furthermore, KLF5 knockdown inhibited ATC growth and enhanced the Dox sensitivity of ATC by inactivating the JNK signaling pathway. Taken together, our findings concluded that KLF5 knockdown can remarkably inhibit the proliferation, invasion, and migration and induce apoptosis of ATC cells, and increase the chemotherapy sensitivity of ATC, all of which probably through inhibiting the JNK signaling pathway.
Collapse
Affiliation(s)
- Zheng Wang
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Department of Breast and Thyroid Surgery, Nanyang Central Hospital, Nanyang, China
| | - Xinguang Qiu
- Department of Thyroid Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hao Zhang
- Department of Breast and Thyroid Surgery, Nanyang Central Hospital, Nanyang, China
| | - Weihan Li
- Department of Breast and Thyroid Surgery, Nanyang Central Hospital, Nanyang, China
| |
Collapse
|
119
|
Yuedi D, Houbao L, Pinxiang L, Hui W, Min T, Dexiang Z. KLF2 induces the senescence of pancreatic cancer cells by cooperating with FOXO4 to upregulate p21. Exp Cell Res 2020; 388:111784. [PMID: 31866399 DOI: 10.1016/j.yexcr.2019.111784] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 02/02/2023]
Abstract
Pancreatic cancer is one of the most common malignancies in the world. Senescence is frequently observed in the progression of pancreatic cancer. In a previous study, we showed that KLF2 inhibited the growth and migration of pancreatic cancer. However, the mechanisms are not fully understood. In this study, we showed that overexpression of KLF2 induced the senescence of pancreatic cancer cells and inhibited tumorigenesis, and knockdown of KLF2 inhibited senescence and p21 expression. In the molecular mechanism study, KLF2 was found to interact with FOXO4 and cooperated with FOXO4 to induce the expression of p21. Downregulation of p21 and FOXO4 impaired the induction of senescence by KLF2. Overall, this study revealed the functions and mechanisms of KLF2 in senescence and provided a novel explanation for the suppressive roles of KLF2 in pancreatic cancer.
Collapse
Affiliation(s)
- Dai Yuedi
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Soochow, 215006, China; Department of Medical Oncology, Fudan University Shanghai Cancer Center, Minhang Branch, Shanghai, 200240, China.
| | - Liu Houbao
- General Surgery Department, Zhongshan Hospital, General Surgery Institute, Fudan University, 180 Fenglin Rd., Shanghai, 200032, China.
| | - Lu Pinxiang
- General Surgery Department, Zhongshan-Xuhui Hospital Affiliated to Fudan University, Shanghai, 200031, China.
| | - Wang Hui
- General Surgery Department, Zhongshan-Xuhui Hospital Affiliated to Fudan University, Shanghai, 200031, China.
| | - Tao Min
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Soochow, 215006, China.
| | - Zhang Dexiang
- General Surgery Department, Zhongshan-Xuhui Hospital Affiliated to Fudan University, Shanghai, 200031, China.
| |
Collapse
|
120
|
Gu YY, Liu XS, Huang XR, Yu XQ, Lan HY. Diverse Role of TGF-β in Kidney Disease. Front Cell Dev Biol 2020; 8:123. [PMID: 32258028 PMCID: PMC7093020 DOI: 10.3389/fcell.2020.00123] [Citation(s) in RCA: 171] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 02/12/2020] [Indexed: 12/13/2022] Open
Abstract
Inflammation and fibrosis are two pathological features of chronic kidney disease (CKD). Transforming growth factor-β (TGF-β) has been long considered as a key mediator of renal fibrosis. In addition, TGF-β also acts as a potent anti-inflammatory cytokine that negatively regulates renal inflammation. Thus, blockade of TGF-β inhibits renal fibrosis while promoting inflammation, revealing a diverse role for TGF-β in CKD. It is now well documented that TGF-β1 activates its downstream signaling molecules such as Smad3 and Smad3-dependent non-coding RNAs to transcriptionally and differentially regulate renal inflammation and fibrosis, which is negatively regulated by Smad7. Therefore, treatments by rebalancing Smad3/Smad7 signaling or by specifically targeting Smad3-dependent non-coding RNAs that regulate renal fibrosis or inflammation could be a better therapeutic approach. In this review, the paradoxical functions and underlying mechanisms by which TGF-β1 regulates in renal inflammation and fibrosis are discussed and novel therapeutic strategies for kidney disease by targeting downstream TGF-β/Smad signaling and transcriptomes are highlighted.
Collapse
Affiliation(s)
- Yue-Yu Gu
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xu-Sheng Liu
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Department of Nephrology, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiao-Ru Huang
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Guangdong-Hong Kong Joint Laboratory for Immunity and Genetics of Chronic Kidney Disease, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Xue-Qing Yu
- Guangdong-Hong Kong Joint Laboratory for Immunity and Genetics of Chronic Kidney Disease, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Guangdong-Hong Kong Joint Laboratory for Immunity and Genetics of Chronic Kidney Disease, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| |
Collapse
|
121
|
Altered Expression of Long Noncoding and Messenger RNAs in Diabetic Nephropathy following Treatment with Rosiglitazone. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1360843. [PMID: 32025515 PMCID: PMC6983290 DOI: 10.1155/2020/1360843] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 11/09/2019] [Indexed: 02/07/2023]
Abstract
Diabetic nephropathy (DN) is characterized by metabolic disorder and inflammation. However, the regulatory effects that long noncoding RNAs (lncRNAs) have on the pathogenesis of DN and on the efficacy of rosiglitazone treatment have yet to be clearly defined. Herein, we performed unbiased RNA sequencing to characterize the transcriptomic profiles in db/db diabetic mouse model with or without rosiglitazone treatment that served to improve the phenotypes of DN. Moreover, RNA-seq profiling revealed that the development of DN caused an upregulation in the expression of 1176 mRNAs and a downregulation in the expression of 1010 mRNAs compared to controls, with the expression of 251 mRNAs being returned to normal following treatment with rosiglitazone. Further, 88 upregulated and 68 downregulated lncRNAs were identified in db/db mice compared to controls, 10 of which had their normal expression restored following treatment with rosiglitazone. Bioinformatic analysis revealed that the primary pathways involved in the pathogenesis of DN, and subsequently in the therapeutic effects of PPARγ, are related to inflammatory and metabolic processes. From bioinformatics analysis, lncRNA-AI838599 emerged as a novel molecular mechanism for rosiglitazone treatment in DN through TNFα-NFκb pathway. These findings may indicate a new molecular regulatory approach for the development of DN therapeutic agents.
Collapse
|
122
|
Yu MY, Kim JE, Lee S, Choi JW, Kim YC, Han SS, Lee H, Cha RH, Lee JP, Lee JW, Kim DK, Kim YS, Yang SH. Krüppel-like factor 15 is a key suppressor of podocyte fibrosis under rotational force-driven pressure. Exp Cell Res 2020; 386:111706. [PMID: 31697927 DOI: 10.1016/j.yexcr.2019.111706] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 10/28/2019] [Accepted: 10/30/2019] [Indexed: 11/16/2022]
Abstract
Krüppel-like factor 15 (KLF15) is a well-known transcription factor associated with podocyte injury and fibrosis. Recently, hypertensive nephropathy was discovered to be closely related to podocyte injury and fibrosis. However, methods to stimulate hypertension in vitro are lacking. Here, we constructed an in vitro model mimicking hypertension using a rotational force device to identify the role of KLF15 in fibrosis due to mechanically induced hypertensive injury. First, we found that KLF15 expression was decreased in patients with hypertensive nephropathy. Then, an in vitro study of hypertension due to rotational force was conducted, and an increase in fibrosis markers and decrease in KLF15 levels were determined after application of 4 mmHg pressure in primary cultured human podocytes. KLF15 and tight junction protein levels increased with retinoic acid treatment. siRNA-mediated inhibition of KLF15 exacerbated pressure-induced fibrosis injury, and KLF15 expression after treatment with angiotensin II was similar to that observed after treatment with the blood pressure modeling device. Furthermore, the reduced KLF15 levels after mechanical pressure application were restored after the administration of an antihypertensive drug. KLF15 expression was also low in vivo. We confirmed the protective role of KLF15 in fibrosis using a mechanically induced in vitro model of hypertensive injury.
Collapse
Affiliation(s)
- Mi-Yeon Yu
- Department of Internal Medicine, Hanyang University Guri Hospital, Republic of Korea
| | - Ji Eun Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Saram Lee
- Seoul National University Hospital Biomedical Research Institute, Seoul, Republic of Korea
| | - Jin Woo Choi
- Interdisciplinary Program in Bioengineering Major, Graduate School, Seoul National University, Seoul, Republic of Korea
| | - Yong Chul Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Seung Seok Han
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hajeong Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea; Kidney Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ran Hui Cha
- Department of Internal Medicine, National Medical Center, Seoul, Republic of Korea
| | - Jung Pyo Lee
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, Republic of Korea
| | - Jae Wook Lee
- Nephrology Clinic, National Cancer Center, Goyang, Gyeonggi-do, Republic of Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea; Kidney Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yon Su Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea; Kidney Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seung Hee Yang
- Seoul National University Hospital Biomedical Research Institute, Seoul, Republic of Korea; Kidney Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
123
|
Wang L, Lin W, Chen J. Krüppel-like Factor 15: A Potential Therapeutic Target For Kidney Disease. Int J Biol Sci 2019; 15:1955-1961. [PMID: 31523196 PMCID: PMC6743293 DOI: 10.7150/ijbs.34838] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/28/2019] [Indexed: 12/26/2022] Open
Abstract
Krüppel-like factor 15 (KLF15) is a zinc-finger transcription factor highly expressed in the glomeruli and interstitial cells of kidneys. An increasing number of studies have demonstrated a critical role for KLF15 in the kidney, involving tubular physiology, podocyte injury, renal fibrosis, and mesangial pathology. In this review, we discuss recent advances and update our overview of the functions of KLF15 in kidney biology, hoping to provide new perspectives on the progression and therapy of Chronic Kidney Disease (CKD). A better understanding of KLF15 with respect to its diverse roles in specific cells or diseases will be beneficial in pursuing novel therapeutic targets and moving forward to precision medicine.
Collapse
Affiliation(s)
- Lefeng Wang
- The Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Weiqiang Lin
- The Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Jianghua Chen
- The Kidney Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|