1
|
Li J, Zong H, Zhao X, Liu Y, Zhao S, Li N, Li Z. KLF11/TMEM87B promoted the occurrence of glioma and decreased TMZ sensitivity. Cell Signal 2025; 130:111651. [PMID: 39929351 DOI: 10.1016/j.cellsig.2025.111651] [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: 11/06/2024] [Revised: 01/20/2025] [Accepted: 02/07/2025] [Indexed: 02/25/2025]
Abstract
Whether KLF11 functions as a tumor promoter or inhibitor depends on the type of tumor. Our previous reports revealed the oncogenic role of KLF11 in glioma. In this study, TMEM87B was identified as a downstream gene of KLF11 through ChIP-seq assay, and the binding of KLF11 to the promoter area of TMEM87B was demonstrated using luciferase assay. KLF11 positively regulated the expression of TMEM87B mRNA and protein in glioma cell lines. Furthermore. TMEM87B was highly expressed in glioma samples, which indicated a poor prognosis in glioma patients. The elimination of TMEM87B reduced the proliferation and migration cell viability, along with the formation of tumor spheroids, while increasing TMZ sensitivity, whereas the overexpression of TMEM87B had the opposite effect. Furthermore, both the knockdown of TMEM87B and TMZ treatment could retard tumor growth in xenograft mice, and their combination significantly reduced tumor size and weight. Our findings identified the effects of the KLF11/ TMEM87B axis on glioma progression and TMZ sensitivity, which could provide new targets for glioma therapy.
Collapse
Affiliation(s)
- Jian Li
- Department of Neurosurgery, Changzhi People's Hospital, Changzhi, 046000, Shanxi, China.
| | - Hua Zong
- Department of Neurosurgery, Changzhi People's Hospital, Changzhi, 046000, Shanxi, China
| | - Xiaoli Zhao
- Clinical Laboratory, Changzhi People's Hospital, Changzhi, 046000, Shanxi, China
| | - Yanping Liu
- Department of Neurosurgery, Changzhi People's Hospital, Changzhi, 046000, Shanxi, China
| | - Shaoyun Zhao
- Department of Neurosurgery, Changzhi People's Hospital, Changzhi, 046000, Shanxi, China
| | - Ning Li
- Department of Neurosurgery, Changzhi People's Hospital, Changzhi, 046000, Shanxi, China
| | - Zhuolun Li
- Department of Neurosurgery, Changzhi People's Hospital, Changzhi, 046000, Shanxi, China
| |
Collapse
|
2
|
Li Q, Chen B, Roche LATDL, Gong Z, Wang G, Zhuo R, Wilde RLD, Chen X, Wang W. Ultrasound Genomics Reveals a Signature for Predicting Breast Cancer Prognosis and Therapy Response. Cancer Biother Radiopharm 2025; 40:54-61. [PMID: 39315921 DOI: 10.1089/cbr.2024.0127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2024] Open
Abstract
Background: Breast cancer (BC) in women is the most common malignancy worldwide, but there is still a lack of validated tools to accurately assess patient prognosis and response to available chemotherapy treatment regimens. Method: We collected ultrasound images and transcriptome data of BC from our breast center and public database. Key ultrasound features were then identified by using the support vector machine (SVM) algorithm and correlated with prognostic genes. Long-term survival-related genes were identified through differential expression analysis, and a prognostic evaluation model was established by using Cox regression. In addition, VPS28 from the model was identified as a promising biomarker for BC. Results: Using univariate logistic regression and SVM algorithms, we identified 12 ultrasound features significantly associated with chemotherapy response. Subsequent correlation and differential expression analyses linked 401 genes to these features, from which five key signature genes were derived using Lasso and multivariate Cox regression models. This signature not only facilitates the stratification of patients into risk-specific treatment pathways but also predicts their chemotherapy response, thus supporting personalized medicine in clinical settings. Notably, VPS28, in the signature, emerged as a significant biomarker, strongly associated with poor prognosis, greater tumor invasiveness, and differing expression across demographic groups. Conclusion: In this study, we use ultrasound genomics to reveal a signature that can provide an effective tool for prognostic assessment and predicting chemotherapy response in patients with BC.
Collapse
Affiliation(s)
- Qin Li
- The Fifth Clinical Medical College of Anhui Medical University, Anhui, China
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu City, China
| | - Bin Chen
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu City, China
| | | | - Zimo Gong
- University Hospital for Gynecology, Pius-Hospital, University Medicine Oldenburg, Oldenburg, Germany
| | - Guilin Wang
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Rui Zhuo
- University Hospital for Gynecology, Pius-Hospital, University Medicine Oldenburg, Oldenburg, Germany
| | - Rudy Leon De Wilde
- University Hospital for Gynecology, Pius-Hospital, University Medicine Oldenburg, Oldenburg, Germany
| | - Xiaopeng Chen
- The Fifth Clinical Medical College of Anhui Medical University, Anhui, China
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Wannan Medical College, Wuhu City, China
| | - Wanwan Wang
- Department of Breast and Thyroid Surgery, Xuzhou No.1 People's Hospital, Xuzhou Jiangsu, China
| |
Collapse
|
3
|
Tsoy UA, Sokolnikova PS, Kravchuk EN, Ryazanov PA, Kozyreva AA, Fomicheva YV, Aramisova LS, Karonova TL, Kostareva AA, Grineva E. A Comprehensive Target Panel Allows to Extend the Genetic Spectrum of Neuroendocrine Tumors. Neuroendocrinology 2024:1-21. [PMID: 39536727 DOI: 10.1159/000542223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 09/17/2024] [Indexed: 11/16/2024]
Abstract
INTRODUCTION Neuroendocrine tumors (NETs) frequently have a genetic basis, and the range of genes implicated in NET development continues to expand. Application of targeted gene panels (TGPs) in next-generation sequencing is a central strategy for elucidating novel variants associated with NET development. METHODS In this study, we conducted comprehensive molecular genetic analyses using TGP on a cohort of 93 patients diagnosed with various NETs subtypes, mainly accompanied by various endocrine syndromes: insulinoma (n = 26), pheochromocytoma and paraganglioma (PPGL) (n = 38), parathyroid adenoma (n = 18, including three with insulinoma), and NETs of other locations (n = 14). The TGP encompassed genes linked to diverse NETs and other hereditary endocrine disorders, with subsequent variant classification according to the American College of Medical Genetics and Genomics guidelines. RESULTS Among the identified variants, 20 were found in genes previously linked to specific tumor types, and 10 were found in genes with a limited likelihood and unclear molecular mecanisms of association with observed NETs. Remarkably, 13 variants were discovered in genes not previously associated with the NETs observed in our patients. These genes, such as ABCC8, KCNJ11, KLF11, HABP2, and APC, were implicated in insulinoma; ZNRF3, GNAS, and KCNJ5 were linked with PPGL; parathyroid adenomas were related to variants in SDHB and TP53; while NETs of other locations displayed variants in APC and ABCC8. CONCLUSION Our study demonstrates that utilizing broad TGP in examining patients with various functioning NETs facilitates the identification of new germinal variants in genes that may contribute to the diseases. The verification of revealed findings requires research in vaster sample.
Collapse
Affiliation(s)
- Uliana A Tsoy
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Polina S Sokolnikova
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Ekaterina N Kravchuk
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Pavel A Ryazanov
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Alexandra A Kozyreva
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Yulia V Fomicheva
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Liana S Aramisova
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Tatiana L Karonova
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Anna A Kostareva
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
- Department of Women's and Children's Health, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Elena Grineva
- World-Class Research Centre for Personalized Medicine, Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| |
Collapse
|
4
|
Hou ZH, Tao M, Dong J, Qiu HM, Li F, Bai XY. KLF11 promotes the proliferation of breast cancer cells by inhibiting p53-MDM2 signaling. Cell Signal 2024; 120:111238. [PMID: 38810862 DOI: 10.1016/j.cellsig.2024.111238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/20/2024] [Accepted: 05/26/2024] [Indexed: 05/31/2024]
Abstract
Abnormal Krüppel-like factor 11 (KLF11) expression is frequently found in tumor tissues and is associated with cancer prognosis, but its biological functions and corresponding mechanisms remain elusive. Here, we demonstrated that KLF11 functions as an oncoprotein to promote tumor proliferation in breast cancer cells. Mechanistically, at the transcription level, KLF11 decreased TP53 mRNA expression. Notably, KLF11 also interacted with and stabilized MDM2 through inhibiting MDM2 ubiquitination and subsequent degradation. This increase in MDM2 in turn accelerated the ubiquitin-mediated proteolysis of p53, leading to the reduced expression of p53 and its target genes, including CDKN1A, BAX, and NOXA1. Accordingly, data from animals further confirmed that KLF11 significantly upregulated the growth of breast cancer cells and was inversely correlated with p53 expression. Taken together, our findings reveal a novel mechanism for breast cancer progression in which the function of the tumor suppressor p53 is dramatically weakened.
Collapse
Affiliation(s)
- Zhi-Han Hou
- Chronic Disease Research Center, Medical College, Dalian University, 116622 Dalian, Liaoning, China
| | - Min Tao
- Chronic Disease Research Center, Medical College, Dalian University, 116622 Dalian, Liaoning, China
| | - Jiang Dong
- Chronic Disease Research Center, Medical College, Dalian University, 116622 Dalian, Liaoning, China
| | - Hong-Mei Qiu
- Chronic Disease Research Center, Medical College, Dalian University, 116622 Dalian, Liaoning, China
| | - Fan Li
- Chronic Disease Research Center, Medical College, Dalian University, 116622 Dalian, Liaoning, China
| | - Xiao-Yan Bai
- Chronic Disease Research Center, Medical College, Dalian University, 116622 Dalian, Liaoning, China..
| |
Collapse
|
5
|
Runa F, Ortiz-Soto G, de Barros NR, Kelber JA. Targeting SMAD-Dependent Signaling: Considerations in Epithelial and Mesenchymal Solid Tumors. Pharmaceuticals (Basel) 2024; 17:326. [PMID: 38543112 PMCID: PMC10975212 DOI: 10.3390/ph17030326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 04/01/2024] Open
Abstract
SMADs are the canonical intracellular effector proteins of the TGF-β (transforming growth factor-β). SMADs translocate from plasma membrane receptors to the nucleus regulated by many SMAD-interacting proteins through phosphorylation and other post-translational modifications that govern their nucleocytoplasmic shuttling and subsequent transcriptional activity. The signaling pathway of TGF-β/SMAD exhibits both tumor-suppressing and tumor-promoting phenotypes in epithelial-derived solid tumors. Collectively, the pleiotropic nature of TGF-β/SMAD signaling presents significant challenges for the development of effective cancer therapies. Here, we review preclinical studies that evaluate the efficacy of inhibitors targeting major SMAD-regulating and/or -interacting proteins, particularly enzymes that may play important roles in epithelial or mesenchymal compartments within solid tumors.
Collapse
Affiliation(s)
- Farhana Runa
- Department of Biology, California State University Northridge, Northridge, CA 91330, USA
| | | | | | - Jonathan A Kelber
- Department of Biology, California State University Northridge, Northridge, CA 91330, USA
- Department of Biology, Baylor University, Waco, TX 76706, USA
| |
Collapse
|
6
|
Zhou ZQ, Lv X, Liu SB, Qu HC, Xie QP, Sun LF, Li G. The induction of ferroptosis by KLF11/NCOA4 axis: the inhibitory role in clear cell renal cell carcinoma. Hum Cell 2023; 36:2162-2178. [PMID: 37642832 DOI: 10.1007/s13577-023-00973-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023]
Abstract
Ferroptosis is a form of cell death and has great potential application in the treatment of many cancers, including clear cell renal cell carcinoma (ccRCC). Herein, we identified the essential roles of Krüppel-like factor 11 (KLF11) in suppressing the progression of ccRCC. By analyzing mRNA expression data from the Gene Expression Omnibus (GEO) database, we found that KLF11 was a significantly downregulated gene in ccRCC tissues. The results of subsequent functional assays verified that KLF11 played an antiproliferative role in ccRCC cells and xenograft tumors. Furthermore, gene set enrichment analysis indicated that ferroptosis was involved in ccRCC development, and correlation analysis revealed that KLF11 was positively related to ferroptosis drivers. We also found that KLF11 promoted ferroptosis in ccRCC by downregulating the protein expression of ferritin, system xc (-) cystine/glutamate antiporter (xCT), and glutathione peroxidase 4 (GPX4), acting as the inhibitory factors of ferroptosis and increasing the intracellular levels of lipid reactive oxygen species (ROS). As a transcriptional regulator, KLF11 significantly increased the promoter activity of nuclear receptor coactivator 4 (NCOA4), a gene significantly downregulated in ccRCC and whose low expression is associated with poor survival. The characteristics of ccRCC cells caused by KLF11 overexpression were reversed after NCOA4 silencing. In summary, the present study suggests that KLF11 suppresses the progression of ccRCC by increasing NCOA4 transcription. Therefore, the KLF11/NCOA4 axis may serve as a novel therapeutic target for human ccRCC.
Collapse
Affiliation(s)
- Zi-Qi Zhou
- Department of Urology,, Cancer Hospital of China Medical University (Liaoning Cancer Hospital and Institute), No. 44, Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, China
| | - Xi Lv
- Department of Urology,, Cancer Hospital of China Medical University (Liaoning Cancer Hospital and Institute), No. 44, Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, China
| | - Shi-Bo Liu
- Department of Urology,, Cancer Hospital of China Medical University (Liaoning Cancer Hospital and Institute), No. 44, Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, China
| | - Hong-Chen Qu
- Department of Urology,, Cancer Hospital of China Medical University (Liaoning Cancer Hospital and Institute), No. 44, Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, China
| | - Qing-Peng Xie
- Department of Urology,, Cancer Hospital of China Medical University (Liaoning Cancer Hospital and Institute), No. 44, Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, China
| | - Long-Feng Sun
- Department of Geriatric Cardiology, The First Hospital of China Medical University, No. 155, Nanjing North Street, Shenyang, Liaoning Province, China.
| | - Gang Li
- Department of Urology,, Cancer Hospital of China Medical University (Liaoning Cancer Hospital and Institute), No. 44, Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, China.
| |
Collapse
|
7
|
Lin L, Pfender K, Ditsch N, Kuhn C, Rahmeh M, Peng L, Schmoeckel E, Mayr D, Trillsch F, Mahner S, Kessler M, Jeschke U, Hester A. KLF11 is an independent negative prognostic factor for breast cancer from a cohort study and induces proliferation and inhibits apoptosis in vitro. Breast Cancer 2023; 30:758-771. [PMID: 37199905 PMCID: PMC10404175 DOI: 10.1007/s12282-023-01470-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 05/10/2023] [Indexed: 05/19/2023]
Abstract
BACKGROUND The therapy concepts that target several members of krüppel like factor (KLF) family have been achieved in breast cancer (BC). However, the role of KLF11 in BC remains unclear. This study explored the prognostic significance of KLF11 in BC patients and investigated its functional roles in this malignancy. METHODS Immunohistochemistry (IHC) staining of KLF11 in 298 patients' samples was performed to determine the prognostic role of the KLF11. Then the protein level was correlated to clinicopathological characteristics and survival outcomes. Afterward, the function of KLF11 was explored in vitro with siRNA-mediated loss-of-function of cell viability, proliferation, and apoptosis. RESULTS From the cohort study, we found that the expression of KLF11 was positively associated with highly proliferative BC of BC. Furthermore, prognostic analysis demonstrated that KLF11 was an independent negative factor for disease-free survival (DFS) and distant-metastasis-free survival (DMFS) of BC. The KLF11-related prognostic model for DFS and DMFS showed high accuracy in predicting the 3-,5- and 10 -year survival probability of BC patients. Additionally, the knockdown of KLF11 inhibited cell viability and proliferation, as well as induced cell apoptosis in MCF7 and MDA-MB-231 cells, while only inhibited cell viability and induced cell apoptosis in SK-BR-3 cells. CONCLUSIONS Our study indicated that targeting KLF11 is an interesting therapeutic concept and further research could lead to a new therapeutic improvement in BC, especially in highly aggressive molecular subtypes.
Collapse
Affiliation(s)
- Lili Lin
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Kristina Pfender
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Nina Ditsch
- Department of Gynecology and Obstetrics, University Hospital Augsburg, 86156, Augsburg, Germany
| | - Christina Kuhn
- Department of Gynecology and Obstetrics, University Hospital Augsburg, 86156, Augsburg, Germany
| | - Martina Rahmeh
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Lin Peng
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Elisa Schmoeckel
- Department of Pathology, Ludwig-Maximilians University of Munich, 81337, Munich, Germany
| | - Doris Mayr
- Department of Pathology, Ludwig-Maximilians University of Munich, 81337, Munich, Germany
| | - Fabian Trillsch
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Sven Mahner
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Mirjana Kessler
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
- Department of Gynecology and Obstetrics, University Hospital Augsburg, 86156, Augsburg, Germany.
| | - Anna Hester
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| |
Collapse
|
8
|
Giarrizzo M, LaComb JF, Bialkowska AB. The Role of Krüppel-like Factors in Pancreatic Physiology and Pathophysiology. Int J Mol Sci 2023; 24:ijms24108589. [PMID: 37239940 DOI: 10.3390/ijms24108589] [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: 04/13/2023] [Revised: 05/04/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Krüppel-like factors (KLFs) belong to the family of transcription factors with three highly conserved zinc finger domains in the C-terminus. They regulate homeostasis, development, and disease progression in many tissues. It has been shown that KLFs play an essential role in the endocrine and exocrine compartments of the pancreas. They are necessary to maintain glucose homeostasis and have been implicated in the development of diabetes. Furthermore, they can be a vital tool in enabling pancreas regeneration and disease modeling. Finally, the KLF family contains proteins that act as tumor suppressors and oncogenes. A subset of members has a biphasic function, being upregulated in the early stages of oncogenesis and stimulating its progression and downregulated in the late stages to allow for tumor dissemination. Here, we describe KLFs' function in pancreatic physiology and pathophysiology.
Collapse
Affiliation(s)
- Michael Giarrizzo
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794, USA
| | - Joseph F LaComb
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794, USA
| | - Agnieszka B Bialkowska
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|
9
|
Lin HY, Ko CJ, Lo TY, Wu SR, Lan SW, Huang CA, Lin YC, Lin HH, Tu HF, Lee CF, Hsiao PW, Huang HP, Chen MJ, Chang KH, Lee MS. Matriptase-2/NR4A3 axis switches TGF-β action toward suppression of prostate cancer cell invasion, tumor growth, and metastasis. Oncogene 2022; 41:2833-2845. [PMID: 35418692 DOI: 10.1038/s41388-022-02303-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 03/21/2022] [Accepted: 03/29/2022] [Indexed: 11/09/2022]
Abstract
Dysregulation of pericellular proteolysis is strongly implicated in cancer metastasis through alteration of cell invasion and the microenvironment. Matriptase-2 (MT-2) is a membrane-anchored serine protease which can suppress prostate cancer (PCa) cell invasion. In this study, we showed that MT-2 was down-regulated in PCa and could suppress PCa cell motility, tumor growth, and metastasis. Using microarray and biochemical analysis, we found that MT-2 shifted TGF-β action towards its tumor suppressor function by repressing epithelial-to-mesenchymal transition (EMT) and promoting Smad2 phosphorylation and nuclear accumulation to upregulate two TGF-β1 downstream effectors (p21 and PAI-1), culminating in hindrance of PCa cell motility and malignant growth. Mechanistically, MT-2 could dramatically up-regulate the expression of nuclear receptor NR4A3 via iron metabolism in PCa cells. MT-2-induced NR4A3 further coactivated Smad2 to activate p21 and PAI-1 expression. In addition, NR4A3 functioned as a suppressor of PCa and mediated MT-2 signaling to inhibit PCa tumorigenesis and metastasis. These results together indicate that NR4A3 sustains MT-2 signaling to suppress PCa cell invasion, tumor growth, and metastasis, and serves as a contextual factor for the TGF-β/Smad2 signaling pathway in favor of tumor suppression via promoting p21 and PAI-1 expression.
Collapse
Affiliation(s)
- Hsin-Ying Lin
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Chun-Jung Ko
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Tzu-Yu Lo
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Shang-Ru Wu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Shao-Wei Lan
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Chen-An Huang
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Yi-Chin Lin
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Hsin-Hsien Lin
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Hsin-Fang Tu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Cheng-Fan Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Pei-Wen Hsiao
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan, ROC
| | - Hsiang-Po Huang
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Mei-Jou Chen
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, Taipei, Taiwan, ROC
| | - Kai-Hsiung Chang
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan, ROC
| | - Ming-Shyue Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei, Taiwan, ROC.
| |
Collapse
|
10
|
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
|
11
|
Viola L, Londero AP, Bertozzi S, Orsaria M, Marzinotto S, Antoniazzi F, Renda V, Cinel J, Fruscalzo A, Lellé RJ, Mariuzzi L. Prognostic Role of Krüppel-Like Factors 5, 9, and 11 in Endometrial Endometrioid Cancer. Pathol Oncol Res 2020; 26:2265-2272. [PMID: 32451988 DOI: 10.1007/s12253-020-00817-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/28/2020] [Indexed: 12/01/2022]
Abstract
BACKGROUND AND OBJECTIVE Krüppel-like factors (KLFs) are transcription factors with the ability to mediate cross-talk with signaling pathways involved in cell proliferation control, apoptosis, migration, and differentiation. They also appear to influence steroid hormone signaling through transcriptional networks involving steroid hormone receptors and members of the nuclear receptor family of transcription factors. Our study aims to evaluate the potential prognostic role of KLF5, KLF9, and KLF11 in endometrial cancer, and their correlation with hormonal receptor status and cellular proliferation. MATERIALS AND METHODS Retrospective observational study on cases of endometrioid endometrial adenocarcinoma collected in the period January 2000-December 2011 at the University of Udine. Formalin-fixed, paraffin-embedded tissue samples were all submitted to tissue microarray immunohistochemical study. A survival analysis was performed. RESULTS One hundred forty seven patients were included in the study with a mean age at surgery of 65.6 years (±10.2). 80.3% of endometrial malignancies were classified as stage FIGO I (118/147). Radiation therapy and chemotherapy were administered in 62.3% (91/146) and 6.2% (9/145) of patients respectively. Five-year overall survival and disease-free survival resulted 85.4% (95% CI, 79.8-91.4%) and 79.4% (95% CI, 73.0-86.4%) respectively. A high Ki-67, cytoplasmatic KLF5 (HR 4.72, CI.95 1.61-13.89, p < 0.05), and nuclear KLF11 (HR 3.04, CI.95 0.99-9.36, p = 0.053) scores correlated with a shorter overall survival. In addition, a high nuclear KLF11 (HR 2.59, CI.95 1.13-5.95, p < 0.05) score correlated with a shorter disease-free survival. CONCLUSIONS In patients affected by endometrioid endometrial carcinoma, higher staining levels of KLF5 and KLF11 correlated with a poorer prognosis. However, further studies are required in order to better clarify the role of KLFs in the natural history of endometrial cancer.
Collapse
Affiliation(s)
- Luigi Viola
- Radiology Department, University of Campania "Luigi Vanvitelli", Naples, 80138, Italy
| | - Ambrogio P Londero
- Clinic of Obstetrics and Gynecology, DAME, Academic Hospital of Udine, University of Udine, Piazzale Santa Maria della Misericordia, 15, 33100, Udine, Italy. .,Ennergi Research (Non-Profit Organization), Lestizza, 33050, Italy.
| | - Serena Bertozzi
- Ennergi Research (Non-Profit Organization), Lestizza, 33050, Italy.,Breast Unit, DAME, Academic Hospital of Udine, University of Udine, 33100, Udine, Italy
| | - Maria Orsaria
- Institute of Pathology, DAME, Academic Hospital of Udine, University of Udine, Udine, 33100, Italy
| | - Stefania Marzinotto
- Institute of Pathology, DAME, Academic Hospital of Udine, University of Udine, Udine, 33100, Italy
| | - Fulvio Antoniazzi
- Institute of Pathology, DAME, Academic Hospital of Udine, University of Udine, Udine, 33100, Italy
| | - Valentina Renda
- Institute of Pathology, DAME, Academic Hospital of Udine, University of Udine, Udine, 33100, Italy
| | - Jacqueline Cinel
- Clinic of Surgery, Academic Hospital of Udine, University of Udine, 33100, Udine, Italy
| | - Arrigo Fruscalzo
- Clinic of Obstetrics and Gynecology, Christophorus-Kliniken, 48653, Coesfeld, Germany.,Clinic of Obstetrics and Gynecology, University Hospital of Münster, Albert-Schweitzer-Campus 1, Gebäude: A1, 48149, Münster, Germany
| | - Ralph J Lellé
- Clinic of Obstetrics and Gynecology, University Hospital of Münster, Albert-Schweitzer-Campus 1, Gebäude: A1, 48149, Münster, Germany
| | - Laura Mariuzzi
- Institute of Pathology, DAME, Academic Hospital of Udine, University of Udine, Udine, 33100, Italy
| |
Collapse
|
12
|
The Distinct Roles of Transcriptional Factor KLF11 in Normal Cell Growth Regulation and Cancer as a Mediator of TGF-β Signaling Pathway. Int J Mol Sci 2020; 21:ijms21082928. [PMID: 32331236 PMCID: PMC7215894 DOI: 10.3390/ijms21082928] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/14/2022] Open
Abstract
KLF11 (Krüppel-like factor 11) belongs to the family of Sp1/Krüppel-like zinc finger transcription factors that play important roles in a variety of cell types and tissues. KLF11 was initially described as a transforming growth factor-beta (TGF-β) inducible immediate early gene (TIEG). KLF11 promotes the effects of TGF-β on cell growth control by influencing the TGFβ–Smads signaling pathway and regulating the transcription of genes that induce either apoptosis or cell cycle arrest. In carcinogenesis, KLF11 can show diverse effects. Its function as a tumor suppressor gene can be suppressed by phosphorylation of its binding domains via oncogenic pathways. However, KLF 11 can itself also show tumor-promoting effects and seems to have a crucial role in the epithelial–mesenchymal transition process. Here, we review the current knowledge about the function of KLF11 in cell growth regulation. We focus on its transcriptional regulatory function and its influence on the TGF-β signaling pathway. We further discuss its possible role in mediating crosstalk between various signaling pathways in normal cell growth and in carcinogenesis.
Collapse
|
13
|
Cheng L, Shi L, Dai H. Bioinformatics analysis of potential prognostic biomarkers among Krüppel-like transcription Factors (KLFs) in breast cancer. Cancer Biomark 2019; 26:411-420. [PMID: 31640084 DOI: 10.3233/cbm-190199] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Lin Cheng
- Department of Breast Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Breast Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Liang Shi
- Jiangsu Breast Disease Center, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Breast Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Hong Dai
- Department of General Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
14
|
Son B, Jeon J, Lee S, Kim H, Kang H, Youn H, Jo S, Youn B. Radiotherapy in combination with hyperthermia suppresses lung cancer progression via increased NR4A3 and KLF11 expression. Int J Radiat Biol 2019; 95:1696-1707. [PMID: 31498019 DOI: 10.1080/09553002.2019.1665213] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Purpose: Hyperthermia (HT), a clinical treatment involving delivery of heat to tumors, has been used in combination with traditional chemotherapy and radiotherapy to enhance their effects. However, the molecular mechanism underlying the high efficacy of combination therapy is not clear. This study was conducted to identify the molecular mechanism underlying the sensitization of lung cancer to radiotherapy by HT.Materials and methods: Nuclear receptor subfamily 4, group A, member 3 (NR4A3) and Krüppel-like factor 11 (KLF11) expression in non-small-cell lung cancer cells was confirmed by performing real-time quantitative reverse transcription-polymerase chain reaction. Tumor cell proliferation and apoptosis were assessed via a colony-forming assay and Annexin V/propidium iodide staining.Results and conclusions: Expression profile analysis revealed elevated levels of NR4A3 and KLF11 in A549 lung cancer cells after treatment with HT combined with radiation. We also confirmed that NR4A3 and KLF11 induced apoptosis and inhibited cell proliferation by elevating intracellular reactive oxygen species levels. Knockdown of NR4A3 or KLF11 using siRNA led to decreased effects of radiohyperthermia. Finally, the effect of these two factors on lung cancer progression was evaluated by in vivo xenograft studies. Taken together, the results suggest that NR4A3 and KLF11 are critical for increasing the efficacy of radiotherapy in combination with HT.
Collapse
Affiliation(s)
- Beomseok Son
- Department of Integrated Biological Science, Pusan National University, Busan, Republic of Korea
| | - Jaewan Jeon
- Department of Integrated Biological Science, Pusan National University, Busan, Republic of Korea.,Department of Radiation Oncology, Haeundae Paik Hospital, Inje University School of Medicine, Busan, Republic of Korea
| | - Sungmin Lee
- Department of Integrated Biological Science, Pusan National University, Busan, Republic of Korea
| | - Hyunwoo Kim
- Department of Integrated Biological Science, Pusan National University, Busan, Republic of Korea
| | - Hyunkoo Kang
- Department of Integrated Biological Science, Pusan National University, Busan, Republic of Korea
| | - HyeSook Youn
- Department of Integrative Bioscience and Biotechnology, Sejong University, Seoul, Republic of Korea
| | - Sunmi Jo
- Department of Radiation Oncology, Haeundae Paik Hospital, Inje University School of Medicine, Busan, Republic of Korea
| | - BuHyun Youn
- Department of Integrated Biological Science, Pusan National University, Busan, Republic of Korea.,Department of Biological Sciences, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
15
|
Han M, Wang Y, Guo G, Li L, Dou D, Ge X, Lv P, Wang F, Gu Y. microRNA-30d mediated breast cancer invasion, migration, and EMT by targeting KLF11 and activating STAT3 pathway. J Cell Biochem 2018; 119:8138-8145. [PMID: 29923255 DOI: 10.1002/jcb.26767] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 02/02/2018] [Indexed: 12/15/2022]
Abstract
miR-30d has been shown to play pivotal roles in cancer development, and has the potential to act as a diagnostic biomarker and therapeutic target in breast cancer. However, the specific function and molecular mechanism of miR-30d in breast cancer cell growth and metastasis is still unknown. The present study seeks to shed light on the potential contribution of the MiR-30d-KLF-11-STAT3 pathway in breast cancer. The results revealed that miR-30d levels were markedly increased in the breast cancer cell lines BT474, MDA-MB-231, HCC197, and MDA-MB-468 compared with the non-tumor mammary gland MCF10A cell line. Furthermore, the miR-30d mimic increased BT474 and MDA-MB-231 breast cancer cell survival, inhibited apoptosis and increased Bcl-2 expression, whilst inhibited Bax protein levels. miR-30d mimics promote BT474 and MDA-MB-231 cell migration, invasion, and mediate the EMT phenotype. However, miR-30d inhibitors reverse all of the effects of miR-30d mimics on breast cancer cell biology. Also, we observed that KLF-11 is a direct target of miR-30d and KLF-11 and pSTAT3 expression are determined by miR-30d. Finally, the results suggest that miR-30d plays essential roles in breast cancer cells in a manner that is dependent on the levels of KLF-1 and pSTAT3. In summary, miR-30d appears to be a novel diagnostic biomarker and treatment target in breast cancer.
Collapse
Affiliation(s)
- Mingli Han
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,The Key Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yimeng Wang
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Guangcheng Guo
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lin Li
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dongwei Dou
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,The Key Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Ge
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pengwei Lv
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fang Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuanting Gu
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
16
|
Ewing RM, Song J, Gokulrangan G, Bai S, Bowler EH, Bolton R, Skipp P, Wang Y, Wang Z. Multiproteomic and Transcriptomic Analysis of Oncogenic β-Catenin Molecular Networks. J Proteome Res 2018; 17:2216-2225. [PMID: 29747501 DOI: 10.1021/acs.jproteome.8b00180] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The dysregulation of Wnt signaling is a frequent occurrence in many different cancers. Oncogenic mutations of CTNNB1/β-catenin, the key nuclear effector of canonical Wnt signaling, lead to the accumulation and stabilization of β-catenin protein with diverse effects in cancer cells. Although the transcriptional response to Wnt/β-catenin signaling activation has been widely studied, an integrated understanding of the effects of oncogenic β-catenin on molecular networks is lacking. We used affinity-purification mass spectrometry (AP-MS), label-free liquid chromatography-tandem mass spectrometry, and RNA-Seq to compare protein-protein interactions, protein expression, and gene expression in colorectal cancer cells expressing mutant (oncogenic) or wild-type β-catenin. We generate an integrated molecular network and use it to identify novel protein modules that are associated with mutant or wild-type β-catenin. We identify a DNA methyltransferase I associated subnetwork that is enriched in cells with mutant β-catenin and a subnetwork enriched in wild-type cells associated with the CDKN2A tumor suppressor, linking these processes to the transformation of colorectal cancer cells through oncogenic β-catenin signaling. In summary, multiomics analysis of a defined colorectal cancer cell model provides a significantly more comprehensive identification of functional molecular networks associated with oncogenic β-catenin signaling.
Collapse
Affiliation(s)
- Rob M Ewing
- School of Biological Sciences , University of Southampton , Southampton SO17 1BJ , United Kingdom
| | - Jing Song
- School of Medicine , Case Western Reserve University , Cleveland , Ohio 44106 , United States
| | - Giridharan Gokulrangan
- School of Medicine , Case Western Reserve University , Cleveland , Ohio 44106 , United States
| | - Sheldon Bai
- School of Medicine , Case Western Reserve University , Cleveland , Ohio 44106 , United States
| | - Emily H Bowler
- School of Biological Sciences , University of Southampton , Southampton SO17 1BJ , United Kingdom
| | - Rachel Bolton
- School of Biological Sciences , University of Southampton , Southampton SO17 1BJ , United Kingdom
| | - Paul Skipp
- School of Biological Sciences , University of Southampton , Southampton SO17 1BJ , United Kingdom
| | - Yihua Wang
- School of Biological Sciences , University of Southampton , Southampton SO17 1BJ , United Kingdom
| | - Zhenghe Wang
- School of Medicine , Case Western Reserve University , Cleveland , Ohio 44106 , United States
| |
Collapse
|
17
|
Zhao JF, Zhao Q, Hu H, Liao JZ, Lin JS, Xia C, Chang Y, Liu J, Guo AY, He XX. The ASH1-miR-375-YWHAZ Signaling Axis Regulates Tumor Properties in Hepatocellular Carcinoma. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 11:538-553. [PMID: 29858089 PMCID: PMC5944419 DOI: 10.1016/j.omtn.2018.04.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 04/18/2018] [Accepted: 04/18/2018] [Indexed: 12/11/2022]
Abstract
Hepatocellular carcinoma (HCC) is a worldwide malignance, and the underlying mechanisms of this disease are not fully elucidated. In this study, the existence and function of achaete-scute homolog-1 (ASH1)-miR-375-YWHAZ signaling axis in HCC were determined. Our experiments and the Cancer Genome Atlas (TCGA) sequencing data analyses showed that ASH1 and miR-375 were significantly downregulated, whereas YWHAZ was significantly upregulated in HCC. Furthermore, we found that ASH1 positively regulates miR-375, and miR-375 directly downregulates its target YWHAZ. Gain- and loss-of-function study demonstrated ASH1 and miR-375 function as tumor suppressors, whereas YWHAZ acts as an oncogene in HCC. Animal experiment indicated that YWHAZ small interfering RNAs (siRNAs) (si-YWHAZ) delivered by nanoliposomes could suppress the growth of hepatoma xenografts and was well tolerant by nude mice. Further studies revealed that YWHAZ was involved in several protein networks, such as cell autophagy, epithelial-mesenchymal transition (EMT), apoptosis, cell cycle, invasion, and migration. In addition, the patient group with ASH1-high-expression-miR-375-high-expression-YWHAZ-low-expression was correlated with a better clinical prognosis compared with the opposite expression group. In conclusion, we proved the existence of ASH1-miR-375-YWHAZ signaling axis and interpreted its important role in driving HCC tumor progression.
Collapse
Affiliation(s)
- Juan-Feng Zhao
- Institute of Liver Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Hui Ya Hospital of The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qiu Zhao
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hui Hu
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Zhi Liao
- Institute of Liver Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ju-Sheng Lin
- Institute of Liver Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chao Xia
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ying Chang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jing Liu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - An-Yuan Guo
- Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Bioinformatics and Systems Biology, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
| | - Xing-Xing He
- Institute of Liver Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
18
|
Yan X, Xiong X, Chen YG. Feedback regulation of TGF-β signaling. Acta Biochim Biophys Sin (Shanghai) 2018; 50:37-50. [PMID: 29228156 DOI: 10.1093/abbs/gmx129] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Indexed: 12/20/2022] Open
Abstract
Transforming growth factor beta (TGF-β) is a multi-functional polypeptide that plays a critical role in regulating a broad range of cellular functions and physiological processes. Signaling is initiated when TGF-β ligands bind to two types of cell membrane receptors with intrinsic Ser/Thr kinase activity and transmitted by the intracellular Smad proteins, which act as transcription factors to regulate gene expression in the nucleus. Although it is relatively simple and straight-forward, this TGF-β/Smad pathway is regulated by various feedback loops at different levels, including the ligand, the receptor, Smads and transcription, and is thus fine-tuned in terms of signaling robustness, duration, specificity, and plasticity. The precise control gives rise to versatile and context-dependent pathophysiological functions. In this review, we firstly give an overview of TGF-β signaling, and then discuss how each step of TGF-β signaling is finely controlled by distinct modes of feedback mechanisms, involving both protein regulators and miRNAs.
Collapse
Affiliation(s)
- Xiaohua Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Xiangyang Xiong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
19
|
Rontauroli S, Norfo R, Pennucci V, Zini R, Ruberti S, Bianchi E, Salati S, Prudente Z, Rossi C, Rosti V, Guglielmelli P, Barosi G, Vannucchi A, Tagliafico E, Manfredini R. miR-494-3p overexpression promotes megakaryocytopoiesis in primary myelofibrosis hematopoietic stem/progenitor cells by targeting SOCS6. Oncotarget 2017; 8:21380-21397. [PMID: 28423484 PMCID: PMC5400591 DOI: 10.18632/oncotarget.15226] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 01/23/2017] [Indexed: 11/25/2022] Open
Abstract
Primary myelofibrosis (PMF) is a chronic Philadelphia-negative myeloproliferative neoplasm characterized by hematopoietic stem cell-derived clonal myeloproliferation, involving especially the megakaryocyte lineage. To better characterize how the altered expression of microRNAs might contribute to PMF pathogenesis, we have previously performed the integrative analysis of gene and microRNA expression profiles of PMF hematopoietic stem/progenitor cells (HSPCs), which allowed us to identify miR-494-3p as the upregulated microRNA predicted to target the highest number of downregulated mRNAs.To elucidate the role of miR-494-3p in hematopoietic differentiation, in the present study we demonstrated that miR-494-3p enforced expression in normal HSPCs promotes megakaryocytopoiesis. Gene expression profiling upon miR-494-3p overexpression allowed the identification of genes commonly downregulated both after microRNA overexpression and in PMF CD34+ cells. Among them, suppressor of cytokine signaling 6 (SOCS6) was confirmed to be a miR-494-3p target by luciferase assay. Western blot analysis showed reduced level of SOCS6 protein as well as STAT3 activation in miR-494-3p overexpressing cells. Furthermore, transient inhibition of SOCS6 expression in HSPCs demonstrated that SOCS6 silencing stimulates megakaryocytopoiesis, mimicking the phenotypic effects observed upon miR-494-3p overexpression. Finally, to disclose the contribution of miR-494-3p upregulation to PMF pathogenesis, we performed inhibition experiments in PMF HSPCs, which showed that miR-494-3p silencing led to SOCS6 upregulation and impaired megakaryocyte differentiation.Taken together, our results describe for the first time the role of miR-494-3p during normal HSPC differentiation and suggest that its increased expression, and the subsequent downregulation of its target SOCS6, might contribute to the megakaryocyte hyperplasia commonly observed in PMF patients.
Collapse
Affiliation(s)
- Sebastiano Rontauroli
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, Modena, Italy
| | - Ruggiero Norfo
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, Modena, Italy
| | - Valentina Pennucci
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberta Zini
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, Modena, Italy
| | - Samantha Ruberti
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisa Bianchi
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, Modena, Italy
| | - Simona Salati
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, Modena, Italy
| | - Zelia Prudente
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, Modena, Italy
| | - Chiara Rossi
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, Modena, Italy
| | - Vittorio Rosti
- Center for The Study of Myelofibrosis, Biotechnology Research Area, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| | - Paola Guglielmelli
- CRIMM-Center for Research and Innovation for Myeloproliferative Neoplasms, AOU Careggi, and Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Giovanni Barosi
- Center for The Study of Myelofibrosis, Biotechnology Research Area, IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| | - Alessandro Vannucchi
- CRIMM-Center for Research and Innovation for Myeloproliferative Neoplasms, AOU Careggi, and Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Enrico Tagliafico
- Center for Genome Research, University of Modena and Reggio Emilia, Modena, Italy
| | - Rossella Manfredini
- Centre for Regenerative Medicine, Life Sciences Department, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
20
|
Bialkowska AB, Yang VW, Mallipattu SK. Krüppel-like factors in mammalian stem cells and development. Development 2017; 144:737-754. [PMID: 28246209 DOI: 10.1242/dev.145441] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Krüppel-like factors (KLFs) are a family of zinc-finger transcription factors that are found in many species. Recent studies have shown that KLFs play a fundamental role in regulating diverse biological processes such as cell proliferation, differentiation, development and regeneration. Of note, several KLFs are also crucial for maintaining pluripotency and, hence, have been linked to reprogramming and regenerative medicine approaches. Here, we review the crucial functions of KLFs in mammalian embryogenesis, stem cell biology and regeneration, as revealed by studies of animal models. We also highlight how KLFs have been implicated in human diseases and outline potential avenues for future research.
Collapse
Affiliation(s)
- Agnieszka B Bialkowska
- Division of Gastroenterology, Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY 11794-8176, USA
| | - Vincent W Yang
- Division of Gastroenterology, Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY 11794-8176, USA.,Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY 11794-8176, USA
| | - Sandeep K Mallipattu
- Division of Nephrology, Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY 11794-8176, USA
| |
Collapse
|
21
|
Kim CK, He P, Bialkowska AB, Yang VW. SP and KLF Transcription Factors in Digestive Physiology and Diseases. Gastroenterology 2017; 152:1845-1875. [PMID: 28366734 PMCID: PMC5815166 DOI: 10.1053/j.gastro.2017.03.035] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 03/21/2017] [Accepted: 03/24/2017] [Indexed: 12/14/2022]
Abstract
Specificity proteins (SPs) and Krüppel-like factors (KLFs) belong to the family of transcription factors that contain conserved zinc finger domains involved in binding to target DNA sequences. Many of these proteins are expressed in different tissues and have distinct tissue-specific activities and functions. Studies have shown that SPs and KLFs regulate not only physiological processes such as growth, development, differentiation, proliferation, and embryogenesis, but pathogenesis of many diseases, including cancer and inflammatory disorders. Consistently, these proteins have been shown to regulate normal functions and pathobiology in the digestive system. We review recent findings on the tissue- and organ-specific functions of SPs and KLFs in the digestive system including the oral cavity, esophagus, stomach, small and large intestines, pancreas, and liver. We provide a list of agents under development to target these proteins.
Collapse
Affiliation(s)
- Chang-Kyung Kim
- Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY
| | - Ping He
- Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY
| | - Agnieszka B. Bialkowska
- Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY,Corresponding Authors: Vincent W. Yang & Agnieszka B. Bialkowska, Department of Medicine, Stony Brook University School of Medicine, HSC T-16, Rm. 020; Stony Brook, NY, USA. Tel: (631) 444-2066; Fax: (631) 444-3144; ;
| | - Vincent W. Yang
- Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY,Department of Physiology and Biophysics, Stony Brook University School of Medicine, Stony Brook, NY,Corresponding Authors: Vincent W. Yang & Agnieszka B. Bialkowska, Department of Medicine, Stony Brook University School of Medicine, HSC T-16, Rm. 020; Stony Brook, NY, USA. Tel: (631) 444-2066; Fax: (631) 444-3144; ;
| |
Collapse
|
22
|
Yan Q, Zhang W, Wu Y, Wu M, Zhang M, Shi X, Zhao J, Nan Q, Chen Y, Wang L, Cheng T, Li J, Bai Y, Liu S, Wang J. KLF8 promotes tumorigenesis, invasion and metastasis of colorectal cancer cells by transcriptional activation of FHL2. Oncotarget 2016; 6:25402-17. [PMID: 26320172 PMCID: PMC4694840 DOI: 10.18632/oncotarget.4517] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 07/03/2015] [Indexed: 12/23/2022] Open
Abstract
The transcription factor Krüppel-like factor (KLF)8 plays an important role in the formation of several human tumors, including colorectal cancer. We recently identified four-and-a-half LIM protein 2 (FHL2) as a critical inducer of the epithelial-to-mesenchymal transition (EMT) and invasion. However, the molecular mechanism by which KLF8 affects FHL2-mediated tumor proliferation, EMT and metastasis remains unknown. Here, we showed that KLF8 overexpression promoted EMT and metastatic phenotypes. KLF8 expression was stimulated by transforming growth factor (TGF)-β1. Moreover, KLF8 acted as a potential EMT inducer by stimulating vimentin expression and inducing a loss of E-cadherin in stable KLF8-transfected cells. KLF8 overexpression induced a strong increase in FHL2 expression, and a positive correlation between the expression patterns of KLF8 and FHL2 was observed in CRC cells. Promoter reporter and chromatin immunoprecipitation (ChIP) assays demonstrated that KLF8 directly bound to and activated the human FHL2 gene promoter. However, siRNA-mediated repression of FHL2 in KLF8-overexpressing cells reversed the EMT and the proliferative and metastatic phenotypes. In vivo, KLF8 promoted FHL2-mediated proliferation and metastasis via orthotopic implantation. Taken together, this work identified KLF8-induced FHL2 activation as a novel and critical signaling mechanism underlying human breast/colorectal cancer invasion and metastasis.
Collapse
Affiliation(s)
- Qingqing Yan
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenjing Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yao Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Meiyan Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mengnan Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinpeng Shi
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinjun Zhao
- Department of Rheumatism, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qingzhen Nan
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ye Chen
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Long Wang
- Division of Vascular Interventional Radiology, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tianming Cheng
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiachu Li
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yang Bai
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Side Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jide Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
23
|
Backes C, Khaleeq QT, Meese E, Keller A. miEAA: microRNA enrichment analysis and annotation. Nucleic Acids Res 2016; 44:W110-6. [PMID: 27131362 PMCID: PMC4987907 DOI: 10.1093/nar/gkw345] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 04/18/2016] [Indexed: 12/29/2022] Open
Abstract
Similar to the development of gene set enrichment and gene regulatory network analysis tools over a decade ago, microRNA enrichment tools are currently gaining importance. Building on our experience with the gene set analysis toolkit GeneTrail, we implemented the miRNA Enrichment Analysis and Annotation tool (miEAA). MiEAA is a web-based application that offers a variety of commonly applied statistical tests such as over-representation analysis and miRNA set enrichment analysis, which is similar to Gene Set Enrichment Analysis. Besides the different statistical tests, miEAA also provides rich functionality in terms of miRNA categories. Altogether, over 14 000 miRNA sets have been added, including pathways, diseases, organs and target genes. Importantly, our tool can be applied for miRNA precursors as well as mature miRNAs. To make the tool as useful as possible we additionally implemented supporting tools such as converters between different miRBase versions and converters from miRNA names to precursor names. We evaluated the performance of miEAA on two sets of miRNAs that are affected in lung adenocarcinomas and have been detected by array analysis. The web-based application is freely accessible at: http://www.ccb.uni-saarland.de/mieaa_tool/.
Collapse
Affiliation(s)
- Christina Backes
- Chair for Clinical Bioinformatics, Saarland University, Building E 2.1, 66123 Saarbrücken, Germany
| | - Qurratulain T Khaleeq
- Chair for Clinical Bioinformatics, Saarland University, Building E 2.1, 66123 Saarbrücken, Germany
| | - Eckart Meese
- Institute of Human Genetics, Saarland University, Medical School, 66421 Homburg, Germany
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, Building E 2.1, 66123 Saarbrücken, Germany
| |
Collapse
|
24
|
Park SJ, Yang SW, Kim BC. Transforming growth factor-β1 induces cell cycle arrest by activating atypical cyclin-dependent kinase 5 through up-regulation of Smad3-dependent p35 expression in human MCF10A mammary epithelial cells. Biochem Biophys Res Commun 2016; 472:502-7. [PMID: 26966064 DOI: 10.1016/j.bbrc.2016.02.121] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 02/29/2016] [Indexed: 12/22/2022]
Abstract
Cyclin-dependent kinases (Cdks) play important roles in control of cell division. Cdk5 is an atypical member of Cdk family with non-cyclin-like regulatory subunit, p35, but its role in cell cycle progression is still unclear. In the present study, we investigated the role of Cdk5/p35 on transforming growth factor-β1 (TGF-β1)-induced cell cycle arrest. In human MCF10A mammary epithelial cells, TGF-β1 induced cell cycle arrest at G1 phase and increased p27KIP1 expression. Interestingly, pretreatment with roscovitine, an inhibitor of Cdk5, or transfection with small interfering (si) RNAs specific to Cdk5 and p35 significantly attenuated the TGF-β1-induced p27KIP1 expression and cell cycle arrest. TGF-β1 increased Cdk5 activity via up-regulation of p35 gene at transcriptional level, and these effects were abolished by transfection with Smad3 siRNA or infection of adenovirus carrying Smad3 mutant at the C-tail (3SA). Chromatin immunoprecipitation assay further revealed that wild type Smad3, but not mutant Smad3 (3SA), binds to the region of the p35 promoter region (-1000--755) in a TGF-β1-dependent manner. These results for the first time demonstrate a role of Cdk5/p35 in the regulation of cell cycle progression modulated by TGF-β1.
Collapse
Affiliation(s)
- Seong Ji Park
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon 200-701, South Korea
| | - Sun Woo Yang
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon 200-701, South Korea
| | - Byung-Chul Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon 200-701, South Korea.
| |
Collapse
|
25
|
Duncan JW, Zhang X, Wang N, Johnson S, Harris S, Udemgba C, Ou XM, Youdim MB, Stockmeier CA, Wang JM. Binge ethanol exposure increases the Krüppel-like factor 11-monoamine oxidase (MAO) pathway in rats: Examining the use of MAO inhibitors to prevent ethanol-induced brain injury. Neuropharmacology 2016; 105:329-340. [PMID: 26805422 DOI: 10.1016/j.neuropharm.2016.01.024] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 01/14/2016] [Accepted: 01/20/2016] [Indexed: 12/25/2022]
Abstract
Binge drinking induces several neurotoxic consequences including oxidative stress and neurodegeneration. Because of these effects, drugs which prevent ethanol-induced damage to the brain may be clinically beneficial. In this study, we investigated the ethanol-mediated KLF11-MAO cell death cascade in the frontal cortex of Sprague-Dawley rats exposed to a modified Majchowicz 4-day binge ethanol model and control rats. Moreover, MAO inhibitors (MAOIs) were investigated for neuroprotective activity against binge ethanol. Binge ethanol-treated rats demonstrated a significant increase in KLF11, both MAO isoforms, protein oxidation and caspase-3, as well as a reduction in BDNF expression in the frontal cortex compared to control rats. MAOIs prevented these binge ethanol-induced changes, suggesting a neuroprotective benefit. Neither binge ethanol nor MAOI treatment significantly affected protein expression levels of the oxidative stress enzymes, SOD2 or catalase. Furthermore, ethanol-induced antinociception was enhanced following exposure to the 4-day ethanol binge. These results demonstrate that the KLF11-MAO pathway is activated by binge ethanol exposure and MAOIs are neuroprotective by preventing the binge ethanol-induced changes associated with this cell death cascade. This study supports KLF11-MAO as a mechanism of ethanol-induced neurotoxicity and cell death that could be targeted with MAOI drug therapy to alleviate alcohol-related brain injury. Further examination of MAOIs to reduce alcohol use disorder-related brain injury could provide pivotal insight to future pharmacotherapeutic opportunities.
Collapse
Affiliation(s)
- Jeremy W Duncan
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216, USA; Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Xiao Zhang
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216, USA; Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Niping Wang
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Shakevia Johnson
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Sharonda Harris
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Chinelo Udemgba
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Xiao-Ming Ou
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Moussa B Youdim
- Technion-Rappaport Family Faculty of Medicine, Eve Topf Center of Excellence, For Neurodegenerative Diseases Research, Haifa, 31096, Israel
| | - Craig A Stockmeier
- Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Jun Ming Wang
- Program in Neuroscience, University of Mississippi Medical Center, Jackson, MS 39216, USA; Department of Psychiatry and Human Behavior, University of Mississippi Medical Center, Jackson, MS 39216, USA; Department of Pathology, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| |
Collapse
|
26
|
Baines A, Martin P, Rorie C. Current and Emerging Targeting Strategies for Treatment of Pancreatic Cancer. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 144:277-320. [DOI: 10.1016/bs.pmbts.2016.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
27
|
Sokolova V, Fiorino A, Zoni E, Crippa E, Reid JF, Gariboldi M, Pierotti MA. The Effects of miR-20a on p21: Two Mechanisms Blocking Growth Arrest in TGF-β-Responsive Colon Carcinoma. J Cell Physiol 2015; 230:3105-14. [PMID: 26012475 DOI: 10.1002/jcp.25051] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 05/19/2015] [Indexed: 12/18/2022]
Abstract
Loss of response to TGF-β is a central event in the genesis of colorectal cancer (CRC), a disease that, in the majority cases, is refractory to growth inhibition induced by this cytokine. However, inactivating mutations at receptors and transducers from the TGF-β cascade occur only in approximately half of CRCs, suggesting the involvement of additional mechanisms altering the response to the cytokine. We have recently described the amplification of the 13q31 locus, where the miR-17-92 cluster maps, associated with overexpression of its members. In this study, we address the potential role of miR-20a, from the miR-17-92 cluster, in the suppression of TGF-β cytostatic response in CRC. Using the poorly tumorigenic and TGF-β-sensitive FET cell line that expresses low miR-20a levels, we first confirmed that miR-20a downmodulated CDKN1A expression, both at mRNA and protein level, through direct binding to its 3'-UTR. We demonstrated that miR-20a significantly diminished cell response to TGF-β by preventing its delay of G1/S transition and promoting progression into cell cycle. Moreover, besides modulating CDKN1A, miR-20a blocked TGF-β-induced transactivation of its promoter without affecting the post-receptor activation of Smad3/4 effectors directly. Finally, miR-20a abrogated the TGF-β-mediated c-Myc repression, a direct inhibitor of the CDKN1A promoter activation, most likely by reducing the expression of specific MYC-regulating genes from the Smad/E2F-based core repressor complex. Our experiments indicate that miR-20a interferes with the colonic epithelium homeostasis by disrupting the regulation of Myc/p21 by TGF-β, which is essential for its malignant transformation.
Collapse
Affiliation(s)
- Viktorija Sokolova
- Molecular Genetics of Cancer, Fondazione Istituto FIRC di Oncologia Molecolare, Milano, Italy.,Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Antonio Fiorino
- Department of Predictive and Preventive Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Eugenio Zoni
- Molecular Genetics of Cancer, Fondazione Istituto FIRC di Oncologia Molecolare, Milano, Italy.,Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elisabetta Crippa
- Molecular Genetics of Cancer, Fondazione Istituto FIRC di Oncologia Molecolare, Milano, Italy.,Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - James F Reid
- Molecular Genetics of Cancer, Fondazione Istituto FIRC di Oncologia Molecolare, Milano, Italy.,Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Manuela Gariboldi
- Molecular Genetics of Cancer, Fondazione Istituto FIRC di Oncologia Molecolare, Milano, Italy.,Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Marco A Pierotti
- Scientific Directorate, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
28
|
Mathison A, Escande C, Calvo E, Seo S, White T, Salmonson A, Faubion WA, Buttar N, Iovanna J, Lomberk G, Chini EN, Urrutia R. Phenotypic Characterization of Mice Carrying Homozygous Deletion of KLF11, a Gene in Which Mutations Cause Human Neonatal and MODY VII Diabetes. Endocrinology 2015; 156:3581-95. [PMID: 26248217 PMCID: PMC4588811 DOI: 10.1210/en.2015-1145] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We have previously shown that amino acid changes in the human Kruppel-Like Factor (KLF) 11 protein is associated with the development of maturity onset diabetes of the young VII, whereas complete inactivation of this pathway by the -331 human insulin mutation causes neonatal diabetes mellitus. Here, we report that Klf11-/- mice have decreased circulating insulin levels, alterations in the control of blood glucose and body weight, as well as serum dyslipidemia, but do not develop diabetes. Functional assays using ex vivo liver tissue sections demonstrate that Klf11-/- mice display increased insulin sensitivity. Genome-wide experiments validated by pathway-specific quantitative PCR arrays reveal that the Klf11-/- phenotype associates to alterations in the regulation of gene networks involved in lipid metabolism, in particular those regulated by peroxisome proliferator-activated receptor-γ. Combined, these results demonstrate that the major phenotype given by the whole-body deletion of Klf11 in mouse is not diabetes but increased insulin sensitivity, likely due to altered transcriptional regulation in target tissues. The absence of diabetes in the Klf11-/- mouse either indicates an interspecies difference for the role of this transcription factor in metabolic homeostasis between mouse and humans, or potentially highlights the fact that other molecular factors can compensate for its absence. Nevertheless, the data of this study, gathered at the whole-organism level, further support a role for KLF11 in metabolic processes like insulin sensitivity, which regulation is critical in several forms of diabetes.
Collapse
Affiliation(s)
- Angela Mathison
- Laboratory of Epigenetics and Chromatin Dynamics (A.M., A.S., W.A.F., N.B., G.L., R.U.), Gastroenterology Research Unit, Departments of Biochemistry and Molecular Biology, Biophysics, Medicine, Epigenomics Translation Program Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota 55905; Metabolic Diseases and Aging Laboratory (C.E.), Institut Pasteur Montevideo, Montevideo 11400, Uruguay; Department of Anesthesia and Robert and Arlene Kogod Center on Aging (C.E., T.W., E.N.C.), Mayo Clinic, Rochester, Minnesota 55905; Endocrinology and Nephrology (E.C.), Centre Hospitalier Universitaire de Québec Research Center and Laval University, Québec, Québec, G1V 4G2, Canada; Lieber Institute for Brain Development (S.S.), Baltimore, Maryland 21205; and Centre de Recherche en Cancérologie de Marseille (J.I.), INSERM U1068, Centre Nationale de la Recherche Scientifique Unité Mixte de Recherche 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, 13288, France
| | - Carlos Escande
- Laboratory of Epigenetics and Chromatin Dynamics (A.M., A.S., W.A.F., N.B., G.L., R.U.), Gastroenterology Research Unit, Departments of Biochemistry and Molecular Biology, Biophysics, Medicine, Epigenomics Translation Program Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota 55905; Metabolic Diseases and Aging Laboratory (C.E.), Institut Pasteur Montevideo, Montevideo 11400, Uruguay; Department of Anesthesia and Robert and Arlene Kogod Center on Aging (C.E., T.W., E.N.C.), Mayo Clinic, Rochester, Minnesota 55905; Endocrinology and Nephrology (E.C.), Centre Hospitalier Universitaire de Québec Research Center and Laval University, Québec, Québec, G1V 4G2, Canada; Lieber Institute for Brain Development (S.S.), Baltimore, Maryland 21205; and Centre de Recherche en Cancérologie de Marseille (J.I.), INSERM U1068, Centre Nationale de la Recherche Scientifique Unité Mixte de Recherche 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, 13288, France
| | - Ezequiel Calvo
- Laboratory of Epigenetics and Chromatin Dynamics (A.M., A.S., W.A.F., N.B., G.L., R.U.), Gastroenterology Research Unit, Departments of Biochemistry and Molecular Biology, Biophysics, Medicine, Epigenomics Translation Program Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota 55905; Metabolic Diseases and Aging Laboratory (C.E.), Institut Pasteur Montevideo, Montevideo 11400, Uruguay; Department of Anesthesia and Robert and Arlene Kogod Center on Aging (C.E., T.W., E.N.C.), Mayo Clinic, Rochester, Minnesota 55905; Endocrinology and Nephrology (E.C.), Centre Hospitalier Universitaire de Québec Research Center and Laval University, Québec, Québec, G1V 4G2, Canada; Lieber Institute for Brain Development (S.S.), Baltimore, Maryland 21205; and Centre de Recherche en Cancérologie de Marseille (J.I.), INSERM U1068, Centre Nationale de la Recherche Scientifique Unité Mixte de Recherche 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, 13288, France
| | - Seungmae Seo
- Laboratory of Epigenetics and Chromatin Dynamics (A.M., A.S., W.A.F., N.B., G.L., R.U.), Gastroenterology Research Unit, Departments of Biochemistry and Molecular Biology, Biophysics, Medicine, Epigenomics Translation Program Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota 55905; Metabolic Diseases and Aging Laboratory (C.E.), Institut Pasteur Montevideo, Montevideo 11400, Uruguay; Department of Anesthesia and Robert and Arlene Kogod Center on Aging (C.E., T.W., E.N.C.), Mayo Clinic, Rochester, Minnesota 55905; Endocrinology and Nephrology (E.C.), Centre Hospitalier Universitaire de Québec Research Center and Laval University, Québec, Québec, G1V 4G2, Canada; Lieber Institute for Brain Development (S.S.), Baltimore, Maryland 21205; and Centre de Recherche en Cancérologie de Marseille (J.I.), INSERM U1068, Centre Nationale de la Recherche Scientifique Unité Mixte de Recherche 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, 13288, France
| | - Thomas White
- Laboratory of Epigenetics and Chromatin Dynamics (A.M., A.S., W.A.F., N.B., G.L., R.U.), Gastroenterology Research Unit, Departments of Biochemistry and Molecular Biology, Biophysics, Medicine, Epigenomics Translation Program Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota 55905; Metabolic Diseases and Aging Laboratory (C.E.), Institut Pasteur Montevideo, Montevideo 11400, Uruguay; Department of Anesthesia and Robert and Arlene Kogod Center on Aging (C.E., T.W., E.N.C.), Mayo Clinic, Rochester, Minnesota 55905; Endocrinology and Nephrology (E.C.), Centre Hospitalier Universitaire de Québec Research Center and Laval University, Québec, Québec, G1V 4G2, Canada; Lieber Institute for Brain Development (S.S.), Baltimore, Maryland 21205; and Centre de Recherche en Cancérologie de Marseille (J.I.), INSERM U1068, Centre Nationale de la Recherche Scientifique Unité Mixte de Recherche 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, 13288, France
| | - Ann Salmonson
- Laboratory of Epigenetics and Chromatin Dynamics (A.M., A.S., W.A.F., N.B., G.L., R.U.), Gastroenterology Research Unit, Departments of Biochemistry and Molecular Biology, Biophysics, Medicine, Epigenomics Translation Program Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota 55905; Metabolic Diseases and Aging Laboratory (C.E.), Institut Pasteur Montevideo, Montevideo 11400, Uruguay; Department of Anesthesia and Robert and Arlene Kogod Center on Aging (C.E., T.W., E.N.C.), Mayo Clinic, Rochester, Minnesota 55905; Endocrinology and Nephrology (E.C.), Centre Hospitalier Universitaire de Québec Research Center and Laval University, Québec, Québec, G1V 4G2, Canada; Lieber Institute for Brain Development (S.S.), Baltimore, Maryland 21205; and Centre de Recherche en Cancérologie de Marseille (J.I.), INSERM U1068, Centre Nationale de la Recherche Scientifique Unité Mixte de Recherche 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, 13288, France
| | - William A Faubion
- Laboratory of Epigenetics and Chromatin Dynamics (A.M., A.S., W.A.F., N.B., G.L., R.U.), Gastroenterology Research Unit, Departments of Biochemistry and Molecular Biology, Biophysics, Medicine, Epigenomics Translation Program Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota 55905; Metabolic Diseases and Aging Laboratory (C.E.), Institut Pasteur Montevideo, Montevideo 11400, Uruguay; Department of Anesthesia and Robert and Arlene Kogod Center on Aging (C.E., T.W., E.N.C.), Mayo Clinic, Rochester, Minnesota 55905; Endocrinology and Nephrology (E.C.), Centre Hospitalier Universitaire de Québec Research Center and Laval University, Québec, Québec, G1V 4G2, Canada; Lieber Institute for Brain Development (S.S.), Baltimore, Maryland 21205; and Centre de Recherche en Cancérologie de Marseille (J.I.), INSERM U1068, Centre Nationale de la Recherche Scientifique Unité Mixte de Recherche 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, 13288, France
| | - Navtej Buttar
- Laboratory of Epigenetics and Chromatin Dynamics (A.M., A.S., W.A.F., N.B., G.L., R.U.), Gastroenterology Research Unit, Departments of Biochemistry and Molecular Biology, Biophysics, Medicine, Epigenomics Translation Program Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota 55905; Metabolic Diseases and Aging Laboratory (C.E.), Institut Pasteur Montevideo, Montevideo 11400, Uruguay; Department of Anesthesia and Robert and Arlene Kogod Center on Aging (C.E., T.W., E.N.C.), Mayo Clinic, Rochester, Minnesota 55905; Endocrinology and Nephrology (E.C.), Centre Hospitalier Universitaire de Québec Research Center and Laval University, Québec, Québec, G1V 4G2, Canada; Lieber Institute for Brain Development (S.S.), Baltimore, Maryland 21205; and Centre de Recherche en Cancérologie de Marseille (J.I.), INSERM U1068, Centre Nationale de la Recherche Scientifique Unité Mixte de Recherche 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, 13288, France
| | - Juan Iovanna
- Laboratory of Epigenetics and Chromatin Dynamics (A.M., A.S., W.A.F., N.B., G.L., R.U.), Gastroenterology Research Unit, Departments of Biochemistry and Molecular Biology, Biophysics, Medicine, Epigenomics Translation Program Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota 55905; Metabolic Diseases and Aging Laboratory (C.E.), Institut Pasteur Montevideo, Montevideo 11400, Uruguay; Department of Anesthesia and Robert and Arlene Kogod Center on Aging (C.E., T.W., E.N.C.), Mayo Clinic, Rochester, Minnesota 55905; Endocrinology and Nephrology (E.C.), Centre Hospitalier Universitaire de Québec Research Center and Laval University, Québec, Québec, G1V 4G2, Canada; Lieber Institute for Brain Development (S.S.), Baltimore, Maryland 21205; and Centre de Recherche en Cancérologie de Marseille (J.I.), INSERM U1068, Centre Nationale de la Recherche Scientifique Unité Mixte de Recherche 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, 13288, France
| | - Gwen Lomberk
- Laboratory of Epigenetics and Chromatin Dynamics (A.M., A.S., W.A.F., N.B., G.L., R.U.), Gastroenterology Research Unit, Departments of Biochemistry and Molecular Biology, Biophysics, Medicine, Epigenomics Translation Program Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota 55905; Metabolic Diseases and Aging Laboratory (C.E.), Institut Pasteur Montevideo, Montevideo 11400, Uruguay; Department of Anesthesia and Robert and Arlene Kogod Center on Aging (C.E., T.W., E.N.C.), Mayo Clinic, Rochester, Minnesota 55905; Endocrinology and Nephrology (E.C.), Centre Hospitalier Universitaire de Québec Research Center and Laval University, Québec, Québec, G1V 4G2, Canada; Lieber Institute for Brain Development (S.S.), Baltimore, Maryland 21205; and Centre de Recherche en Cancérologie de Marseille (J.I.), INSERM U1068, Centre Nationale de la Recherche Scientifique Unité Mixte de Recherche 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, 13288, France
| | - Eduardo N Chini
- Laboratory of Epigenetics and Chromatin Dynamics (A.M., A.S., W.A.F., N.B., G.L., R.U.), Gastroenterology Research Unit, Departments of Biochemistry and Molecular Biology, Biophysics, Medicine, Epigenomics Translation Program Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota 55905; Metabolic Diseases and Aging Laboratory (C.E.), Institut Pasteur Montevideo, Montevideo 11400, Uruguay; Department of Anesthesia and Robert and Arlene Kogod Center on Aging (C.E., T.W., E.N.C.), Mayo Clinic, Rochester, Minnesota 55905; Endocrinology and Nephrology (E.C.), Centre Hospitalier Universitaire de Québec Research Center and Laval University, Québec, Québec, G1V 4G2, Canada; Lieber Institute for Brain Development (S.S.), Baltimore, Maryland 21205; and Centre de Recherche en Cancérologie de Marseille (J.I.), INSERM U1068, Centre Nationale de la Recherche Scientifique Unité Mixte de Recherche 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, 13288, France
| | - Raul Urrutia
- Laboratory of Epigenetics and Chromatin Dynamics (A.M., A.S., W.A.F., N.B., G.L., R.U.), Gastroenterology Research Unit, Departments of Biochemistry and Molecular Biology, Biophysics, Medicine, Epigenomics Translation Program Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota 55905; Metabolic Diseases and Aging Laboratory (C.E.), Institut Pasteur Montevideo, Montevideo 11400, Uruguay; Department of Anesthesia and Robert and Arlene Kogod Center on Aging (C.E., T.W., E.N.C.), Mayo Clinic, Rochester, Minnesota 55905; Endocrinology and Nephrology (E.C.), Centre Hospitalier Universitaire de Québec Research Center and Laval University, Québec, Québec, G1V 4G2, Canada; Lieber Institute for Brain Development (S.S.), Baltimore, Maryland 21205; and Centre de Recherche en Cancérologie de Marseille (J.I.), INSERM U1068, Centre Nationale de la Recherche Scientifique Unité Mixte de Recherche 7258, Aix-Marseille Université and Institut Paoli-Calmettes, Parc Scientifique et Technologique de Luminy, Marseille, 13288, France
| |
Collapse
|
29
|
Harris S, Johnson S, Duncan JW, Udemgba C, Meyer JH, Albert PR, Lomberk G, Urrutia R, Ou XM, Stockmeier CA, Wang JM. Evidence revealing deregulation of the KLF11-MAO A pathway in association with chronic stress and depressive disorders. Neuropsychopharmacology 2015; 40:1373-82. [PMID: 25502632 PMCID: PMC4397395 DOI: 10.1038/npp.2014.321] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 10/08/2014] [Accepted: 10/23/2014] [Indexed: 12/18/2022]
Abstract
The biochemical pathways underlying major depressive disorder (MDD) and chronic stress are not well understood. However, it has been reported that monoamine oxidase A (MAO A, a major neurotransmitter-degrading enzyme) is significantly increased in the brains of human subjects affected with MDD and rats exposed to chronic social defeat (CSD) stress, which is used to model depression. In the current study, we compared the protein levels of a MAO A-transcriptional activator, Kruppel-like factor 11 (KLF11 , also recognized as transforming growth factor-beta-inducible early gene 2) between the brains of 18 human subjects with MDD and 18 control subjects. We found that, indeed, the expression of KLF11 is increased by 36% (p<0.02) in the postmortem prefrontal cortex of human subjects with MDD compared with controls. We also observed a positive correlation between KLF11 levels and those of its target gene, MAO A, both in association with MDD. KLF11 protein expression was also increased by 44% (p<0.02) in the frontal cortex of KLF11 wild-type mice (Klf11(+/+)) vs Klf11(-/-) when both exposed to CSD stress. In contrast, locomotor activities, central box duration and sucrose preference were significantly reduced in the stressed Klf11(+/+) mice, suggesting that Klf11(+/+) mice are more severely affected by the stress model compared with Klf11(-/-) mice. These results serve to assign an important role of KLF11 in upregulating MAO A in MDD and chronic social stress, suggesting that inhibition of the pathways regulated by this transcription factor may aid in the therapeutics of neuropsychiatric illnesses. Thus, the new knowledge derived from the current study extends our understanding of transcriptional mechanisms that are operational in the pathophysiology of common human diseases and thus bears significant biomedical relevance.
Collapse
Affiliation(s)
- Sharonda Harris
- Department of Psychiatry and Human Behavior, Jackson, MS, USA
| | | | - Jeremy W Duncan
- Department of Psychiatry and Human Behavior, Jackson, MS, USA,Program in Neuroscience, Jackson, MS, USA
| | - Chinelo Udemgba
- Department of Psychiatry and Human Behavior, Jackson, MS, USA
| | - Jeffrey H Meyer
- Centre for Addiction and Mental Health and Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Paul R Albert
- Ottawa Hospital Research Institute (Neuroscience), Ottawa, Ontario, Canada
| | - Gwen Lomberk
- Epigenetics and Chromatin Dynamics Laboratory, GI Research Unit, Mayo Clinic, Rochester, MN, USA
| | - Raul Urrutia
- Epigenetics and Chromatin Dynamics Laboratory, GI Research Unit, Mayo Clinic, Rochester, MN, USA
| | - Xiao-Ming Ou
- Department of Psychiatry and Human Behavior, Jackson, MS, USA
| | - Craig A Stockmeier
- Department of Psychiatry and Human Behavior, Jackson, MS, USA,Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
| | - Jun Ming Wang
- Department of Psychiatry and Human Behavior, Jackson, MS, USA,Program in Neuroscience, Jackson, MS, USA,Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA,Department of Pathology, University of Mississippi Medical Center, 2500 N. State Street, Jackson, MS 39216, USA, Tel: +1-601-984-4644, Fax: +1-601-984-5899, E-mail:
| |
Collapse
|
30
|
Tu X, Zheng X, Li H, Cao Z, Chang H, Luan S, Zhu J, Chen J, Zang Y, Zhang J. MicroRNA-30 Protects Against Carbon Tetrachloride-induced Liver Fibrosis by Attenuating Transforming Growth Factor Beta Signaling in Hepatic Stellate Cells. Toxicol Sci 2015; 146:157-69. [PMID: 25912033 DOI: 10.1093/toxsci/kfv081] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Transforming growth factor beta (TGF-β) is crucial for transdifferentiation of hepatic stellate cells (HSCs) and the blunting of TGF-β signaling in HSCs can effectively prevent liver fibrosis. Krüppel-like factor 11 (KLF11) is an early response transcription factor that potentiates TGF-β/Smad signaling by suppressing the transcription of inhibitory Smad7. Using a mouse model of carbon tetrachloride (CCl4)-induced liver fibrosis, we observed significant upregulation of KLF11 in the activated HSCs during liver fibrogenesis. Meanwhile, the downregulation of miR-30 was observed in the HSCs isolated from fibrotic liver. Adenovirus-mediated ectopic expression of miR-30 was under the control of smooth muscle α-actin promoter, showing that the increase in miR-30 in HSC greatly reduced CCl4-induced liver fibrosis. Subsequent investigations showed that miR-30 suppressed KLF11 expression in HSC and led to a significant upregulation of Smad7 in vivo. Mechanistic studies further confirmed that KLF11 was the direct target of miR-30, and revealed that miR-30 blunted the profibrogenic TGF-β signaling in HSC by suppressing KLF11 expression and thus enhanced the negative feedback loop of TGF-β signaling imposed by Smad7. Finally, we demonstrated that miR-30 facilitated the reversal of activated HSC to a quiescent state as indicated by the inhibition of proliferation and migration, the loss of activation markers, and the gain of quiescent HSC markers. In conclusion, our results define miR-30 as a crucial suppressor of TGF-β signaling in HSCs activation and provide useful insights into the mechanisms underlying liver fibrosis.
Collapse
Affiliation(s)
- Xiaolong Tu
- *State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University and Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Xiuxiu Zheng
- *State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University and Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Huanan Li
- *State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University and Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Zhipeng Cao
- *State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University and Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Hanwen Chang
- *State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University and Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Shaoyuan Luan
- *State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University and Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Jie Zhu
- *State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University and Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Jiangning Chen
- *State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University and Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Yuhui Zang
- *State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University and Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China *State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University and Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| | - Junfeng Zhang
- *State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University and Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China *State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University and Jiangsu Engineering Research Center for microRNA Biology and Biotechnology, Nanjing University, Nanjing 210093, People's Republic of China
| |
Collapse
|
31
|
Liu WJ, Lo CF, Kou GH, Leu JH, Lai YJ, Chang LK, Chang YS. The promoter of the white spot syndrome virus immediate-early gene WSSV108 is activated by the cellular KLF transcription factor. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2015; 49:7-18. [PMID: 25445906 DOI: 10.1016/j.dci.2014.10.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 10/24/2014] [Accepted: 10/25/2014] [Indexed: 06/04/2023]
Abstract
A series of deletion and mutation assays of the white spot syndrome virus (WSSV) immediate-early gene WSSV108 promoter showed that a Krüppel-like factor (KLF) binding site located from -504 to -495 (relative to the transcription start site) is important for the overall level of WSSV108 promoter activity. Electrophoretic mobility shift assays further showed that overexpressed recombinant Penaeus monodon KLF (rPmKLF) formed a specific protein-DNA complex with the (32)P-labeled KLF binding site of the WSSV108 promoter, and that higher levels of Litopenaeus vannamei KLF (LvKLF) were expressed in WSSV-infected shrimp. A transactivation assay indicated that the WSSV108 promoter was strongly activated by rPmKLF in a dose-dependent manner. Lastly, we found that specific silencing of LvKLF expression in vivo by dsRNA injection dramatically reduced both WSSV108 expression and WSSV replication. We conclude that shrimp KLF is important for WSSV genome replication and gene expression, and that it binds to the WSSV108 promoter to enhance the expression of this immediate-early gene.
Collapse
Affiliation(s)
- Wang-Jing Liu
- Department of Earth and Life Science, College of Science, University of Taipei, Taipei 100, Taiwan
| | - Chu-Fang Lo
- Institute of Bioinformatics and Biosignal Transduction, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan 701, Taiwan
| | - Guang-Hsiung Kou
- Department of Life Science, College of Life Science, National Taiwan University, Taipei 106, Taiwan
| | - Jiann-Horng Leu
- Institute of Marine Biology, College of Life Sciences, National Taiwan Ocean University, Keelung 202, Taiwan
| | - Ying-Jang Lai
- Department of Food Science, College of Science and Engineering, National Quemoy University, Kinmen 892, Taiwan
| | - Li-Kwan Chang
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei 106, Taiwan
| | - Yun-Shiang Chang
- Department of Molecular Biotechnology, College of Biotechnology and Bioresources, Da-Yeh University, Changhua 515, Taiwan.
| |
Collapse
|
32
|
Human cancer: Is it linked to dysfunctional lipid metabolism? Biochim Biophys Acta Gen Subj 2015; 1850:352-64. [DOI: 10.1016/j.bbagen.2014.11.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 10/27/2014] [Accepted: 11/03/2014] [Indexed: 11/23/2022]
|
33
|
Abstract
Krüppel-like factors (KLFs) comprise a highly conserved family of zinc finger transcription factors, that are involved in a plethora of cellular processes, ranging from proliferation and apoptosis to differentiation, migration and pluripotency. During the last few years, evidence on their role and deregulation in different human cancers has been emerging. This review will discuss current knowledge on Krüppel-like transcription in the epithelial-mesenchymal transition (EMT), invasion and metastasis, with a focus on epithelial cancer biology and the extensive interface with pluripotency. Furthermore, as KLFs are able to mediate different outcomes, important influences of the cellular and microenvironmental context will be highlighted. Finally, we attempt to integrate diverse findings on KLF functions in EMT and stem cell biology to ft in the current model of cellular plasticity as a tool for successful metastatic dissemination.
Collapse
|
34
|
Abstract
Krüppel-like factors (KLFs) are a family of DNA-binding transcriptional regulators with diverse and essential functions in a multitude of cellular processes, including proliferation, differentiation, migration, inflammation and pluripotency. In this Review, we discuss the roles and regulation of the 17 known KLFs in various cancer-relevant processes. Importantly, the functions of KLFs are context dependent, with some KLFs having different roles in normal cells and cancer, during cancer development and progression and in different cancer types. We also identify key questions for the field that are likely to lead to important new translational research and discoveries in cancer biology.
Collapse
Affiliation(s)
- Marie-Pier Tetreault
- Department of Medicine, Gastroenterology Division, University of Pennsylvania Perelman School of Medicine, 913 Biomedical Research Building II/III, 421 Curie Boulevard, Philadelphia PA 19104-6144, USA
| | | | | |
Collapse
|
35
|
Udemgba C, Johnson S, Stockmeier CA, Luo J, Albert PR, Wang J, May WL, Rajkowska G, Harris S, Sittman DB, Ou XM. The expression of KLF11 (TIEG2), a monoamine oxidase B transcriptional activator in the prefrontal cortex of human alcohol dependence. Alcohol Clin Exp Res 2013; 38:144-51. [PMID: 23915421 DOI: 10.1111/acer.12229] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Accepted: 05/22/2013] [Indexed: 12/24/2022]
Abstract
BACKGROUND The biochemical pathways underlying alcohol abuse and dependence are not well understood, although brain cell loss and neurotoxicity have been reported in subjects with alcohol dependence. Monoamine oxidase B (MAO B; an enzyme that catabolizes neurotransmitters such as dopamine) is consistently increased in this psychiatric illness. MAO B has been implicated in the pathogenesis of alcohol dependence and alcohol-induced brain neurotoxicity. Recently, the cell growth inhibitor protein, Kruppel-like factor 11 (KLF11), has been reported to be an MAO transcriptional activator. KLF11 is also known as TIEG2 (transforming growth factor-beta-inducible early gene 2) and mediates apoptotic cell death. This study investigates the protein expression of KLF11 and its relationship with MAO B using human postmortem prefrontal cortex from subjects with alcohol dependence. METHODS Twelve subjects with alcohol dependence and the respective psychiatrically normal control subjects were investigated. Expression of KLF11 and MAO B proteins in the prefrontal cortex was measured by Western blot analysis. Correlation studies involving KLF11 and MAO B protein expression were performed. Localization of KLF11 in the human prefrontal cortex was also determined by immunohistochemistry. RESULTS Levels of KLF11 protein were significantly increased by 44% (p < 0.03) in the postmortem prefrontal cortex of subjects with alcohol dependence as compared to age- and gender-matched, psychiatrically normal control subjects. Furthermore, KLF11 levels were significantly and positively correlated with both the increased MAO B protein levels and blood alcohol content in alcohol-dependent subjects. In addition, KLF11 protein expression was visualized in both neuronal and glial cells. CONCLUSIONS This novel study shows the important role of KLF11, an MAO transcriptional activator, in human alcohol dependence. It further supports that the KLF11-MAO B cell death cascade may contribute to chronic alcohol-induced brain damage. This argues a case for KLF11-MAO B inhibition as a novel therapeutic strategy that may impact this highly prevalent illness.
Collapse
Affiliation(s)
- Chinelo Udemgba
- Department of Psychiatry and Human Behavior , University of Mississippi Medical Center, Jackson, Mississippi
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Glineur C, Gross B, Neve B, Rommens C, Chew GT, Martin-Nizard F, Rodríguez-Pascual F, Lamas S, Watts GF, Staels B. Fenofibrate Inhibits Endothelin-1 Expression by Peroxisome Proliferator–Activated Receptor α–Dependent and Independent Mechanisms in Human Endothelial Cells. Arterioscler Thromb Vasc Biol 2013; 33:621-8. [DOI: 10.1161/atvbaha.112.300665] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Objective—
Dyslipidemia contributes to endothelial dysfunction in type 2 diabetes mellitus. Fenofibrate (FF), a ligand of the peroxisome proliferator–activated receptor-α (PPARα), has beneficial effects on microvascular complications. FF may act on the endothelium by regulating vasoactive factors, including endothelin-1 (ET-1). In vitro, FF decreases ET-1 expression in human microvascular endothelial cells. We investigated the molecular mechanisms involved in the effect of FF treatment on plasma levels of ET-1 in type 2 diabetes mellitus patients.
Methods and Results—
FF impaired the capacity of transforming growth factor-β to induce
ET-1
gene expression. PPARα activation by FF increased expression of the transcriptional repressor Krüppel-like factor 11 and its binding to the
ET-1
gene promoter. Knockdown of Krüppel-like factor 11 expression potentiated basal and transforming growth factor-β–stimulated ET-1 expression, suggesting that Krüppel-like factor 11 downregulates ET-1 expression. FF, in a PPARα-independent manner, and insulin enhanced glycogen synthase kinase-3β phosphorylation thus reducing glycogen synthase kinase-3 activity that contributes to the FF-mediated reduction of
ET-1
gene expression. In type 2 diabetes mellitus, improvement of flow-mediated dilatation of the brachial artery by FF was associated with a decrease in plasma ET-1.
Conclusion—
FF decreases ET-1 expression by a PPARα-dependent mechanism, via transcriptional induction of the Krüppel-like factor 11 repressor and by PPARα-independent actions via inhibition of glycogen synthase kinase-3 activity.
Collapse
Affiliation(s)
- Corine Glineur
- From the Université Lille Nord de France, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Inserm, U1011, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Université Droit Santé Lille, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Institut Pasteur de Lille, Lille, France (C.G., B.G., B.N., C.R., F.M.-N., B.S.); CNRS, UMR8090, Institut of Biologie, Université Droit Santé Lille, Lille, France (B.N.); School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia,
| | - Barbara Gross
- From the Université Lille Nord de France, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Inserm, U1011, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Université Droit Santé Lille, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Institut Pasteur de Lille, Lille, France (C.G., B.G., B.N., C.R., F.M.-N., B.S.); CNRS, UMR8090, Institut of Biologie, Université Droit Santé Lille, Lille, France (B.N.); School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia,
| | - Bernadette Neve
- From the Université Lille Nord de France, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Inserm, U1011, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Université Droit Santé Lille, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Institut Pasteur de Lille, Lille, France (C.G., B.G., B.N., C.R., F.M.-N., B.S.); CNRS, UMR8090, Institut of Biologie, Université Droit Santé Lille, Lille, France (B.N.); School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia,
| | - Corinne Rommens
- From the Université Lille Nord de France, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Inserm, U1011, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Université Droit Santé Lille, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Institut Pasteur de Lille, Lille, France (C.G., B.G., B.N., C.R., F.M.-N., B.S.); CNRS, UMR8090, Institut of Biologie, Université Droit Santé Lille, Lille, France (B.N.); School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia,
| | - Gerard T. Chew
- From the Université Lille Nord de France, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Inserm, U1011, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Université Droit Santé Lille, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Institut Pasteur de Lille, Lille, France (C.G., B.G., B.N., C.R., F.M.-N., B.S.); CNRS, UMR8090, Institut of Biologie, Université Droit Santé Lille, Lille, France (B.N.); School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia,
| | - Françoise Martin-Nizard
- From the Université Lille Nord de France, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Inserm, U1011, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Université Droit Santé Lille, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Institut Pasteur de Lille, Lille, France (C.G., B.G., B.N., C.R., F.M.-N., B.S.); CNRS, UMR8090, Institut of Biologie, Université Droit Santé Lille, Lille, France (B.N.); School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia,
| | - Fernando Rodríguez-Pascual
- From the Université Lille Nord de France, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Inserm, U1011, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Université Droit Santé Lille, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Institut Pasteur de Lille, Lille, France (C.G., B.G., B.N., C.R., F.M.-N., B.S.); CNRS, UMR8090, Institut of Biologie, Université Droit Santé Lille, Lille, France (B.N.); School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia,
| | - Santiago Lamas
- From the Université Lille Nord de France, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Inserm, U1011, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Université Droit Santé Lille, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Institut Pasteur de Lille, Lille, France (C.G., B.G., B.N., C.R., F.M.-N., B.S.); CNRS, UMR8090, Institut of Biologie, Université Droit Santé Lille, Lille, France (B.N.); School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia,
| | - Gerald F. Watts
- From the Université Lille Nord de France, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Inserm, U1011, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Université Droit Santé Lille, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Institut Pasteur de Lille, Lille, France (C.G., B.G., B.N., C.R., F.M.-N., B.S.); CNRS, UMR8090, Institut of Biologie, Université Droit Santé Lille, Lille, France (B.N.); School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia,
| | - Bart Staels
- From the Université Lille Nord de France, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Inserm, U1011, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Université Droit Santé Lille, Lille, France (C.G., B.G., C.R., F.M.-N., B.S.); Institut Pasteur de Lille, Lille, France (C.G., B.G., B.N., C.R., F.M.-N., B.S.); CNRS, UMR8090, Institut of Biologie, Université Droit Santé Lille, Lille, France (B.N.); School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia,
| |
Collapse
|
37
|
Yin KJ, Fan Y, Hamblin M, Zhang J, Zhu T, Li S, Hawse JR, Subramaniam M, Song CZ, Urrutia R, Lin JD, Chen YE. KLF11 mediates PPARγ cerebrovascular protection in ischaemic stroke. ACTA ACUST UNITED AC 2013; 136:1274-87. [PMID: 23408111 DOI: 10.1093/brain/awt002] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is emerging as a major regulator in neurological diseases. However, the role of (PPARγ) and its co-regulators in cerebrovascular endothelial dysfunction after stroke is unclear. Here, we have demonstrated that (PPARγ) activation by pioglitazone significantly inhibited both oxygen-glucose deprivation-induced cerebral vascular endothelial cell death and middle cerebral artery occlusion-triggered cerebrovascular damage. Consistent with this finding, selective (PPARγ) genetic deletion in vascular endothelial cells resulted in increased cerebrovascular permeability and brain infarction in mice after focal ischaemia. Moreover, we screened for (PPARγ) co-regulators using a genome-wide and high-throughput co-activation system and revealed KLF11 as a novel (PPARγ) co-regulator, which interacted with (PPARγ) and regulated its function in mouse cerebral vascular endothelial cell cultures. Interestingly, KLF11 was also found as a direct transcriptional target of (PPARγ). Furthermore, KLF11 genetic deficiency effectively abolished pioglitazone cytoprotection in mouse cerebral vascular endothelial cell cultures after oxygen-glucose deprivation, as well as pioglitazone-mediated cerebrovascular protection in a mouse middle cerebral artery occlusion model. Mechanistically, we demonstrated that KLF11 enhanced (PPARγ) transcriptional suppression of the pro-apoptotic microRNA-15a (miR-15a) gene, resulting in endothelial protection in cerebral vascular endothelial cell cultures and cerebral microvasculature after ischaemic stimuli. Taken together, our data demonstrate that recruitment of KLF11 as a novel (PPARγ) co-regulator plays a critical role in the cerebrovascular protection after ischaemic insults. It is anticipated that elucidating the coordinated actions of KLF11 and (PPARγ) will provide new insights into understanding the molecular mechanisms underlying (PPARγ) function in the cerebral vasculature and help to develop a novel therapeutic strategy for the treatment of stroke.
Collapse
Affiliation(s)
- Ke-Jie Yin
- Cardiovascular Centre, Department of Internal Medicine, University of Michigan Medical Centre, NCRC Bld 26, Rm 361S, 2800 Plymouth Rd, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Lomberk G, Mathison AJ, Grzenda A, Seo S, DeMars CJ, Rizvi S, Bonilla-Velez J, Calvo E, Fernandez-Zapico ME, Iovanna J, Buttar NS, Urrutia R. Sequence-specific recruitment of heterochromatin protein 1 via interaction with Krüppel-like factor 11, a human transcription factor involved in tumor suppression and metabolic diseases. J Biol Chem 2012; 287:13026-39. [PMID: 22318730 PMCID: PMC3339955 DOI: 10.1074/jbc.m112.342634] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 02/06/2012] [Indexed: 12/17/2022] Open
Abstract
Heterochromatin protein 1 (HP1) proteins are "gatekeepers" of epigenetic gene silencing that is mediated by lysine 9 of histone H3 methylation (H3K9me). Current knowledge supports a paradigm whereby HP1 proteins achieve repression by binding to H3K9me marks and interacting to H3K9 histone methyltransferases (HMTs), such as SUV39H1, which methylate this residue on adjacent nucleosomes thereby compacting chromatin and silencing gene expression. However, the mechanism underlying the recruitment of this epigenetic regulator to target gene promoters remains poorly characterized. In the current study, we reveal for the first time a mechanism whereby HP1 is recruited to promoters by a well characterized Krüppel-like transcription factor (KLF), in a sequence-specific manner, to mediate complex biological phenomena. A PXVXL HP1-interacting domain identified at position 487-491 of KLF11 mediates the binding of HP1α and KLF11 in vitro and in cultured cells. KLF11 also recruits HP1α and its histone methyltransferase, SUV39H1, to promoters to limit KLF11-mediated gene activation. Indeed, a KLF11ΔHP1 mutant derepresses KLF11-regulated cancer genes, by inhibiting HP1-SUV39H1 recruitment, decreasing H3K9me3, while increasing activation-associated marks. Biologically, impairment of KLF11-mediated HP1-HMT recruitment abolishes tumor suppression, providing direct evidence that HP1-HMTs act in a sequence-specific manner to achieve this function rather than its well characterized binding to methylated chromatin without intermediary. Collectively, these studies reveal a novel role for HP1 as a cofactor in tumor suppression, expand our mechanistic understanding of a KLF associated to human disease, and outline cellular and biochemical mechanisms underlying this phenomenon, increasing the specificity of targeting HP1-HMT complexes to gene promoters.
Collapse
Affiliation(s)
- Gwen Lomberk
- From the Department of Biochemistry and Molecular Biology, Laboratory of Epigenetics and Chromatin Dynamics, GIH Division, and
| | - Angela J. Mathison
- From the Department of Biochemistry and Molecular Biology, Laboratory of Epigenetics and Chromatin Dynamics, GIH Division, and
| | - Adrienne Grzenda
- From the Department of Biochemistry and Molecular Biology, Laboratory of Epigenetics and Chromatin Dynamics, GIH Division, and
| | - Seungmae Seo
- From the Department of Biochemistry and Molecular Biology, Laboratory of Epigenetics and Chromatin Dynamics, GIH Division, and
| | - Cathrine J. DeMars
- From the Department of Biochemistry and Molecular Biology, Laboratory of Epigenetics and Chromatin Dynamics, GIH Division, and
| | - Sumera Rizvi
- From the Department of Biochemistry and Molecular Biology, Laboratory of Epigenetics and Chromatin Dynamics, GIH Division, and
| | - Juliana Bonilla-Velez
- From the Department of Biochemistry and Molecular Biology, Laboratory of Epigenetics and Chromatin Dynamics, GIH Division, and
| | - Ezequiel Calvo
- the Molecular Endocrinology and Oncology Research Center, CHUL Research Center, Quebec 61V 462, Canada, and
| | | | - Juan Iovanna
- INSERM U.624, Parc Scientifique et Technologique de Luminy, Marseille F-13288, France
| | - Navtej S. Buttar
- From the Department of Biochemistry and Molecular Biology, Laboratory of Epigenetics and Chromatin Dynamics, GIH Division, and
| | - Raul Urrutia
- From the Department of Biochemistry and Molecular Biology, Laboratory of Epigenetics and Chromatin Dynamics, GIH Division, and
| |
Collapse
|
39
|
Klf10 and Klf11 as mediators of TGF-beta superfamily signaling. Cell Tissue Res 2011; 347:65-72. [PMID: 21574058 DOI: 10.1007/s00441-011-1186-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 04/27/2011] [Indexed: 12/19/2022]
Abstract
Klf10 and Klf11 belong to the family of Sp1/Krüppel-like zinc finger transcription factors that play important roles in a variety of cell types and tissues. Although Klf10 and Klf11 were initially introduced as transforming growth factor-beta (TGF-beta)-inducible genes, several studies have described their upregulation by a plethora of growth factors, cytokines and hormones. Here, we review the current knowledge of the inductive cues for Klf10 and Klf11 and focus on their transcriptional regulation by members of the TGF-beta superfamily. We further summarize their involvement in the regulation of the TGF-beta signaling pathway and discuss their possible role as molecules mediating crosstalk between various signaling pathways. Finally, we provide an overview of the pro-apoptotic and anti-proliferative functions of Klf10 and Klf11.
Collapse
|
40
|
Abstract
The Krüppel-like factor (KLF) family of transcription factors regulates diverse biological processes that include proliferation, differentiation, growth, development, survival, and responses to external stress. Seventeen mammalian KLFs have been identified, and numerous studies have been published that describe their basic biology and contribution to human diseases. KLF proteins have received much attention because of their involvement in the development and homeostasis of numerous organ systems. KLFs are critical regulators of physiological systems that include the cardiovascular, digestive, respiratory, hematological, and immune systems and are involved in disorders such as obesity, cardiovascular disease, cancer, and inflammatory conditions. Furthermore, KLFs play an important role in reprogramming somatic cells into induced pluripotent stem (iPS) cells and maintaining the pluripotent state of embryonic stem cells. As research on KLF proteins progresses, additional KLF functions and associations with disease are likely to be discovered. Here, we review the current knowledge of KLF proteins and describe common attributes of their biochemical and physiological functions and their pathophysiological roles.
Collapse
Affiliation(s)
- Beth B McConnell
- Departments of Medicine and of Hematology and Medical Oncology, Emory University School of Medicine,Atlanta, Georgia 30322, USA
| | | |
Collapse
|
41
|
Fibroblast and prostate tumor cell cross-talk: fibroblast differentiation, TGF-β, and extracellular matrix down-regulation. Exp Cell Res 2010; 316:3207-26. [PMID: 20727350 DOI: 10.1016/j.yexcr.2010.08.005] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Revised: 08/12/2010] [Accepted: 08/12/2010] [Indexed: 01/27/2023]
Abstract
Growth and survival of tumors at a site of metastasis involve interactions with stromal cells in the surrounding environment. Stromal cells aid tumor cell growth by producing cytokines as well as by modifying the environment surrounding the tumor through modulation of the extracellular matrix (ECM). Small leucine-rich proteoglycans (SLRPs) are biologically active components of the ECM which can be altered in the stroma surrounding tumors. The influence tumor cells have on stromal cells has been well elucidated. However, little is understood about the effect metastatic cancer cells have on the cell biology and behavior of the local stromal cells. Our data reveal a significant down-regulation in the expression of ECM components such as collagens I, II, III, and IV, and the SLRPs, decorin, biglycan, lumican, and fibromodulin in stromal cells when grown in the presence of two metastatic prostate cancer cell lines PC3 and DU145. Interestingly, TGF-β down-regulation was observed in stromal cells, as well as actin depolymerization and increased vimentin and α5β1 integrin expression. MT1-MMP expression was upregulated and localized in stromal cell protrusions which extended into the ECM. Moreover, enhanced stromal cell migration was observed after cross-talk with metastatic prostate tumor cells. Xenografting metastatic prostate cancer cells together with "activated" stromal cells led to increased tumorigenicity of the prostate cancer cells. Our findings suggest that metastatic prostate cancer cells create a metastatic niche by altering the phenotype of local stromal cells, leading to changes in the ECM.
Collapse
|
42
|
König A, Linhart T, Schlengemann K, Reutlinger K, Wegele J, Adler G, Singh G, Hofmann L, Kunsch S, Büch T, Schäfer E, Gress TM, Fernandez-Zapico ME, Ellenrieder V. NFAT-induced histone acetylation relay switch promotes c-Myc-dependent growth in pancreatic cancer cells. Gastroenterology 2010; 138:1189-99.e1-2. [PMID: 19900447 PMCID: PMC2895621 DOI: 10.1053/j.gastro.2009.10.045] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2009] [Revised: 09/28/2009] [Accepted: 10/29/2009] [Indexed: 01/12/2023]
Abstract
BACKGROUND & AIMS Induction of immediate early transcription factors (ITF) represents the first transcriptional program controlling mitogen-stimulated cell cycle progression in cancer. Here, we examined the transcriptional mechanisms regulating the ITF protein c-Myc and its role in pancreatic cancer growth in vitro and in vivo. METHODS Expression of ITF proteins was examined by reverse-transcription polymerase chain reaction and immunoblotting, and its implications in cell cycle progression and growth was determined by flow cytometry and [(3)H]-thymidine incorporation. Intracellular Ca(2+) concentrations, calcineurin activity, and cellular nuclear factor of activated T cells (NFAT) distribution were analyzed. Transcription factor complex formations and promoter regulation were examined by immunoprecipitations, reporter gene assays, and chromatin immunoprecipitation. Using a combination of RNA interference knockdown technology and xenograft models, we analyzed the significance for pancreatic cancer tumor growth. RESULTS Serum promotes pancreatic cancer growth through induction of the proproliferative NFAT/c-Myc axis. Mechanistically, serum increases intracellular Ca(2+) concentrations and activates the calcineurin/NFAT pathway to induce c-Myc transcription. NFAT binds to a serum responsive element within the proximal promoter, initiates p300-dependent histone acetylation, and creates a local chromatin structure permissive for the inducible recruitment of Ets-like gene (ELK)-1, a protein required for maximal activation of the c-Myc promoter. The functional significance of this novel pathway was emphasized by impaired c-Myc expression, G1 arrest, and reduced tumor growth upon NFAT depletion in vitro and in vivo. CONCLUSIONS Our study uncovers a novel mechanism regulating cell growth and identifies the NFAT/ELK complex as modulators of early stages of mitogen-stimulated proliferation in pancreatic cancer cells.
Collapse
Affiliation(s)
- Alexander König
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Germany
| | - Thomas Linhart
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Germany
| | - Katrin Schlengemann
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Germany
| | - Kristina Reutlinger
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Germany
| | - Jessica Wegele
- Department of Gastroenterology, University of Ulm, Germany
| | - Guido Adler
- Department of Gastroenterology, University of Ulm, Germany
| | - Garima Singh
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Germany
| | - Leonie Hofmann
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Germany
| | - Steffen Kunsch
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Germany
| | - Thomas Büch
- Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilian University, Munich, Germany
| | - Eva Schäfer
- Walther-Straub-Institute of Pharmacology and Toxicology, Ludwig-Maximilian University, Munich, Germany
| | - Thomas M. Gress
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Germany
| | | | - Volker Ellenrieder
- Department of Gastroenterology and Endocrinology, Philipps-University of Marburg, Germany, Corresponding author: Volker Ellenrieder Dept. of Gastroenterology and Endocrinology, Philipps-University of Marburg, Baldingerstrasse, 35043 Marburg, Germany Phone: 0049-6421-5866460
| |
Collapse
|
43
|
Westfall SD, Sachdev S, Das P, Hearne LB, Hannink M, Roberts RM, Ezashi T. Identification of oxygen-sensitive transcriptional programs in human embryonic stem cells. Stem Cells Dev 2008; 17:869-81. [PMID: 18811242 DOI: 10.1089/scd.2007.0240] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
To realize the full potential of human embryonic stem cells (hESCs), it is important to develop culture conditions that maintain hESCs in a pluripotent, undifferentiated state. A low O(2) atmosphere (approximately 4% O(2)), for example, prevents spontaneous differentiation and supports self-renewal of hESCs. To identify genes whose expression is sensitive to O(2) conditions, microarray analysis was performed on RNA from hESCs that had been maintained under either 4% or 20% O(2). Of 149 genes differentially expressed, 42 were up-regulated and 107 down-regulated under 20% O(2). Several of the down-regulated genes are most likely under the control of hypoxia-inducing factors and include genes encoding enzymes involved in carbohydrate catabolism and cellular redox state. Although genes associated with pluripotency, including OCT4, SOX2, and NANOG were generally unaffected, some genes controlled by these transcription factors, including LEFTY2, showed lowered expression under 20% O(2), while a few genes implicated in lineage specification were up-regulated. Although the differences between O(2) conditions were generally subtle, they were observed in two different hESC lines and at different passage numbers. The data are consistent with the hypothesis that 4% O(2) favors the molecular mechanisms required for the maintenance of pluripotency.
Collapse
Affiliation(s)
- Suzanne D Westfall
- Department of Animal Sciences, University of Missouri, Columbia, Missouri 65211-7310, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Ma L, Hanson RL, Que LN, Mack JL, Franks PW, Infante AM, Kobes S, Bogardus C, Baier LJ. Association analysis of Krüppel-like factor 11 variants with type 2 diabetes in Pima Indians. J Clin Endocrinol Metab 2008; 93:3644-9. [PMID: 18593768 PMCID: PMC2729234 DOI: 10.1210/jc.2008-0546] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Krüppel-like factor 11 (KLF11) is a transcription factor of the zinc finger domain family that has been shown to regulate expression of the insulin gene. An initial study reported that a KLF11 variant predicting a Q62R was associated with type 2 diabetes (T2D) in French Caucasians; however, subsequent studies have failed to identify an association between this variant and T2D in subjects from a similar Northern-European ancestry. OBJECTIVE We sought to determine whether the Q62R or other variants within KLF11 were associated with T2D in Pima Indians, a population with an extremely high prevalence of this disease. DESIGN, SETTING, AND SUBJECTS KLF11 was sequenced in 24 Pima Indians to identify potentially novel variants. There were 18 variants genotyped in a family-based sample of 1337 Pima Indians to analyze the linkage disequilibrium pattern of this gene and identify representative variants. Four representative variants were further genotyped in a population-based sample of 3501 full-heritage Pima Indians for association analyses. Among these subjects, 413 had undergone metabolic studies when they were nondiabetic to measure traits that predict T2D. RESULTS Neither the Q62R nor any other common variant in KLF11 was associated with T2D in the Pima population. In addition, no variant was associated with insulin secretion or insulin-stimulated glucose disposal rate. CONCLUSIONS Common variation in KLF11 variation does not appear to influence the population-based risk for developing T2D among full-heritage Pima Indians. Thus, KLF11 is unlikely to play a major role in the etiology of T2D among this Native American population.
Collapse
Affiliation(s)
- Lijun Ma
- Diabetes Molecular Genetics Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes, Digestive and Kidney Diseases, NIH, Phoenix, Arizona 85004, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
|
46
|
Mallette FA, Gaumont-Leclerc MF, Huot G, Ferbeyre G. Myc Down-regulation as a Mechanism to Activate the Rb Pathway in STAT5A-induced Senescence. J Biol Chem 2007; 282:34938-44. [PMID: 17913706 DOI: 10.1074/jbc.m707074200] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Senescence is a general antiproliferative program that avoids the expansion of cells bearing oncogenic mutations. We found that constitutively active STAT5A (ca-STAT5A) can induce a p53- and Rb-dependent cellular senescence response. However, ca-STAT5A did not induce p21 and p16(INK4a), which are responsible for inhibiting cyclin-dependent protein kinases and engaging the Rb pathway during the senescence response to oncogenic ras. Intriguingly, ca-STAT5A led to a down-regulation of Myc and Myc targets, including CDK4, a negative regulator of Rb. The down-regulation of Myc was in part proteasome-dependent and correlated with its localization to promyelocytic leukemia bodies, which were found to be highly abundant during STAT5-induced senescence. Introduction of CDK4 or Myc bypassed STAT5A-induced senescence in cells in which p53 was also inactivated. These results uncover a novel mechanism to engage the Rb pathway in oncogene-induced senescence and indicate the existence of oncogene-specific pathways that regulate senescence.
Collapse
Affiliation(s)
- Frédérick A Mallette
- Département de Biochimie, Université de Montréal, Montréal, Québec H3C 3J7, Canada
| | | | | | | |
Collapse
|