1
|
Park JS, Kang M, Kim HB, Hong H, Lee J, Song Y, Hur Y, Kim S, Kim TK, Lee Y. The capicua-ataxin-1-like complex regulates Notch-driven marginal zone B cell development and sepsis progression. Nat Commun 2024; 15:10579. [PMID: 39632849 PMCID: PMC11618371 DOI: 10.1038/s41467-024-54803-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024] Open
Abstract
Follicular B (FOB) and marginal zone B (MZB) cells are pivotal in humoral immune responses against pathogenic infections. MZB cells can exacerbate endotoxic shock via interleukin-6 secretion. Here we show that the transcriptional repressor capicua (CIC) and its binding partner, ataxin-1-like (ATXN1L), play important roles in FOB and MZB cell development. CIC deficiency reduces the size of both FOB and MZB cell populations, whereas ATXN1L deficiency specifically affects MZB cells. B cell receptor signaling is impaired only in Cic-deficient FOB cells, whereas Notch signaling is disrupted in both Cic-deficient and Atxn1l-deficient MZB cells. Mechanistically, ETV4 de-repression leads to inhibition of Notch1 and Notch2 transcription, thereby inhibiting MZB cell development in B cell-specific Cic-deficient (Cicf/f;Cd19-Cre) and Atxn1l-deficient (Atxn1lf/f;Cd19-Cre) mice. In Cicf/f;Cd19-Cre and Atxn1lf/f; Cd19-Cre mice, humoral immune responses and lipopolysaccharide-induced sepsis progression are attenuated but are restored upon Etv4-deletion. These findings highlight the importance of the CIC-ATXN1L complex in MZB cell development and may provide proof of principle for therapeutic targeting in sepsis.
Collapse
Affiliation(s)
- Jong Seok Park
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, Republic of Korea
| | - Minjung Kang
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, Republic of Korea
| | - Han Bit Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, Republic of Korea
| | - Hyebeen Hong
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, Republic of Korea
| | - Jongeun Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, Republic of Korea
| | - Youngkwon Song
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, Republic of Korea
| | - Yunjung Hur
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, Republic of Korea
| | - Soeun Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, Republic of Korea
| | - Tae-Kyung Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, Republic of Korea
| | - Yoontae Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, Republic of Korea.
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Ghafouri-Fard S, Hussen BM, Eslami S. Identification of miR-125a and miR-106b signature as a potential diagnostic biomarker in breast cancer tissues. Pathol Res Pract 2024; 256:155277. [PMID: 38579577 DOI: 10.1016/j.prp.2024.155277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/19/2024] [Accepted: 03/26/2024] [Indexed: 04/07/2024]
Abstract
MicroRNAs (miRNAs) have essential roles in the etiology of breast cancer and are regarded as possible markers in this malignancy. In order to find new markers for breast cancer, the current study has measured expression level of four miRNAs, namely miR-125a, miR-106b, miR-96 and miR-92a-3p in the paired breast samples. Expression levels of miR-125a and miR-106b were higher in tumoral tissues compared with control tissues (Expression ratios (95% CI) = 4.01 (1.96-8.19) and 3.9 (1.95-7.81); P values = 0.0005 and 0.0003, respectively). miR-106b and miR-125a differentiated between malignant and non-malignant tissues with AUC values of 0.7 and 0.67, respectively. We detected association between expression of miR-106b and clinical stage (P = 0.03), in a way that its expression was the lowest in the advanced stages. Finally, significant relationships were found between miR-96 and miR-125a in both tumoral and non-tumoral specimens (ρ = 0.76 and 0.69, respectively). This nonparametric measure of rank correlation also showed relationship between miR-106b and miR-96 in both sets of samples (ρ = 0.63 and 0.61, respectively). Cumulatively, the assessed miRNAs, particularly miR-125a and miR-106b are putative targets for further expression and functional assays in breast cancer.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Bashdar Mahmud Hussen
- Department of Biomedical Sciences, Cihan University-Erbil, Erbil, Kurdistan Region, Iraq; Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region, Iraq.
| | - Solat Eslami
- Department of Medical Biotechnology, School of Medicine, Alborz University of Medical Sciences, Karaj, Islamic Republic of Iran; Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Islamic Republic of Iran
| |
Collapse
|
3
|
Hsu YC, Huang WC, Kuo CY, Li YS, Cheng SP. Downregulation of cellular retinoic acid binding protein 1 fosters epithelial-mesenchymal transition in thyroid cancer. Mol Carcinog 2023; 62:1935-1946. [PMID: 37642311 DOI: 10.1002/mc.23626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023]
Abstract
Cellular retinoic acid binding protein 1 (CRABP1) participates in the regulation of retinoid signaling. Previous studies showed conflicting results regarding the role of CRABP1 in tumor biology, including protumorigenic and tumor-suppressive effects in different types of cancer. Our bioinformatics analyses suggested that CRABP1 expression was downregulated in thyroid cancer. Ectopic expression of CRABP1 in thyroid cancer cells suppressed migratory and invasive activity without affecting cell growth or cell cycle distribution. In transformed normal thyroid follicular epithelial cells, silencing of CRABP1 expression increased invasiveness. Additionally, CRABP1 overexpression was associated with downregulation of the mesenchymal phenotype. Kinase phosphorylation profiling indicated that CRABP1 overexpression was accompanied by a decrease in phosphorylation of epidermal growth factor (EGF) receptor and downstream phosphorylation of Akt, STAT3, and FAK, which were reversed by exogenous EGF treatment. Immunohistochemical analysis of our tissue microarrays revealed an inverse association between CRABP1 expression and disease stage of differentiated thyroid cancer. Taken together, our results suggest that CRABP1 expression is aberrantly lost in thyroid cancer, and this downregulation promotes the epithelial-mesenchymal transition at least partly through modulating EGF receptor signaling.
Collapse
Affiliation(s)
- Yi-Chiung Hsu
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan City, Taiwan
| | - Wen-Chien Huang
- Department of Surgery, MacKay Memorial Hospital and MacKay Medical College, Taipei, Taiwan
| | - Chi-Yu Kuo
- Department of Surgery, MacKay Memorial Hospital and MacKay Medical College, Taipei, Taiwan
| | - Ying-Syuan Li
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
| | - Shih-Ping Cheng
- Department of Surgery, MacKay Memorial Hospital and MacKay Medical College, Taipei, Taiwan
- Department of Medical Research, MacKay Memorial Hospital, Taipei, Taiwan
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, Taiwan
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
4
|
Gazzaz H, Habchi ME, Feniche ME, Aatik YE, Ouardi AE, Ameur A, Dami A. Diagnostic and Prognostic Value of miR-93 in Prostate Cancer: A Meta-Analysis and Bioinformatics Analysis. IRANIAN JOURNAL OF PUBLIC HEALTH 2023; 52:2260-2271. [PMID: 38106826 PMCID: PMC10719693 DOI: 10.18502/ijph.v52i11.14026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/19/2023] [Indexed: 12/19/2023]
Abstract
Background Accurate and non-invasive diagnostic and prognostic markers are necessary to improve patient outcomes. MicroRNAs have been proposed as relatively non-invasive and pertinent biomarkers. miR-93 has been studied for its potential as a diagnostic and prognostic marker in prostate cancer (PCa), but findings from individual studies are inconsistent. We conducted a meta-analysis of its overall differential expression in 13 PCa studies and a bioinformatics analysis to provide a comprehensive appraisal of its diagnostic and prognostic role. Methods We searched all published papers on miR-93 expression in PCa up to Nov 30, 2022 using PubMed, Science Direct, Web of Science, Cochrane Central Register of Controlled Trials databases. We used RevMan software to Meta-analyze the included literature. A bioinformatics analysis of genes and pathways that might be target to the effect of the mature miR-93-5p was carried out. Results The pooled standardized mean difference (SMD) of miR-93 expression in PCa, its area under the curve (AUC) and hazard ratio (HR) were 1.26, 95% CI [-0.34-2.86], 0.84, 95% CI [0.76 -0.93] and 1.67, 95% CI [0.98, 2.84] respectively. Bioinformatics analysis revealed that mature miR-93-5p may regulate genes such as SMAD1, SMAD7 and MAPK and the PI3K-Akt signaling pathways. Conclusion miR-93 has significant diagnostic and prognostic value in PCa. These findings highlight the potential of miR-93 as a non-invasive biomarker for PCa and may contribute to earlier detection and prognostic assessment. The target genes and signaling pathways regulated by miR-93 may provide insights into the underlying molecular mechanisms of PCa.
Collapse
Affiliation(s)
- Hassane Gazzaz
- Clinical, Metabolic and Molecular Biochemistry Team, Faculty of Medicine and Pharmacy, Mohammed V University,10100 Rabat, Morocco
- Higher Institute of Nursing Professions and Health Techniques of Marrakech, annex of Safi, Morocco
| | - Maha El Habchi
- Research Laboratory of Psychiatry, Medical Psychology and History of Medicine, Faculty of Medicine and Pharmacy, Mohammed V University, 10100 Rabat, Morocco
| | - Mohammed El Feniche
- Laboratory of Biostatistics, Clinical Research and Epidemiology, Faculty of Medicine and Pharmacy, Mohammed V University, 10100 Rabat, Morocco
| | - Yassine El Aatik
- Research Laboratory of Psychiatry, Medical Psychology and History of Medicine, Faculty of Medicine and Pharmacy, Mohammed V University, 10100 Rabat, Morocco
| | - Abdelghani El Ouardi
- Research Laboratory of Psychiatry, Medical Psychology and History of Medicine, Faculty of Medicine and Pharmacy, Mohammed V University, 10100 Rabat, Morocco
| | - Ahmed Ameur
- Department of Urology, Military Hospital Mohammed V, 10045 Rabat, Morocco
| | - Abdellah Dami
- Clinical, Metabolic and Molecular Biochemistry Team, Faculty of Medicine and Pharmacy, Mohammed V University,10100 Rabat, Morocco
- Department of Biochemistry and Toxicology, Military Hospital Mohammed V, 10045 Rabat, Morocco
| |
Collapse
|
5
|
Mokhtari H, Ebrahimi A, Nejati M, Barartabar Z, Damchi M, Khonakdar-Tarsi A, Zahedi M. The effect of encomir-93 mimic transfection on the expression of miR-93 and PSA and androgen receptor in prostate cancer LNcap cell line. Horm Mol Biol Clin Investig 2023; 44:237-241. [PMID: 36995990 DOI: 10.1515/hmbci-2022-0096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/12/2023] [Indexed: 03/31/2023]
Abstract
OBJECTIVES Prostate cancer (PCa) is one of the most common cancers in men with high mortality rate which is a major concern for men's health. However, the molecular mechanisms remain poorly understood. miR-93 is an important oncogene which may have important function in prostate cancer.So, this study aimed to predict that encomir-93 mimic transfection on the expression of miR-93 and PSA and AR in prostate cancer LNcap cell line. METHODS Lymph node carcinoma of the prostate (LNCaP) was cultured and then miR-93 mimics was designed, synthesized and the transfected to LNCaP. The expression level of prostate-specific antigen (PSA) and androgen receptor (AR) was determined via Real-time PCR after treated with 15 pmol of miR-93 mimics. RESULTS miR-93 mimic transfection led to significant increase in PSA and AR expression in comparison with control group (p≤0.05). CONCLUSIONS The miR-93 and its target genes has important role in PCa progression via enhancement in PSA and AR expression. Further research on the function of the miR-93 and its target genes in tumorgenesis and progression PCa could be helpful for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Hossein Mokhtari
- Amol Faculty of Paramedicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ali Ebrahimi
- Department of Biology, Qaemshahr Branch, Islamic Azad University, Qaemshahr, Iran
| | - Mohaddeseh Nejati
- Department of Biology, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Zeinab Barartabar
- Department of Clinical Biochemistry, School of Medicine, Hamedan University of Medical Sciences, Hamedan, Iran
| | - Mehdi Damchi
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Abbas Khonakdar-Tarsi
- Department of Medical Biochemistry and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mahlagha Zahedi
- Department of Pathology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
6
|
Takemon Y, LeBlanc VG, Song J, Chan SY, Lee SD, Trinh DL, Ahmad ST, Brothers WR, Corbett RD, Gagliardi A, Moradian A, Cairncross JG, Yip S, Aparicio SAJR, Chan JA, Hughes CS, Morin GB, Gorski SM, Chittaranjan S, Marra MA. Multi-Omic Analysis of CIC's Functional Networks Reveals Novel Interaction Partners and a Potential Role in Mitotic Fidelity. Cancers (Basel) 2023; 15:2805. [PMID: 37345142 PMCID: PMC10216487 DOI: 10.3390/cancers15102805] [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: 04/07/2023] [Revised: 05/11/2023] [Accepted: 05/15/2023] [Indexed: 06/23/2023] Open
Abstract
CIC encodes a transcriptional repressor and MAPK signalling effector that is inactivated by loss-of-function mutations in several cancer types, consistent with a role as a tumour suppressor. Here, we used bioinformatic, genomic, and proteomic approaches to investigate CIC's interaction networks. We observed both previously identified and novel candidate interactions between CIC and SWI/SNF complex members, as well as novel interactions between CIC and cell cycle regulators and RNA processing factors. We found that CIC loss is associated with an increased frequency of mitotic defects in human cell lines and an in vivo mouse model and with dysregulated expression of mitotic regulators. We also observed aberrant splicing in CIC-deficient cell lines, predominantly at 3' and 5' untranslated regions of genes, including genes involved in MAPK signalling, DNA repair, and cell cycle regulation. Our study thus characterises the complexity of CIC's functional network and describes the effect of its loss on cell cycle regulation, mitotic integrity, and transcriptional splicing, thereby expanding our understanding of CIC's potential roles in cancer. In addition, our work exemplifies how multi-omic, network-based analyses can be used to uncover novel insights into the interconnected functions of pleiotropic genes/proteins across cellular contexts.
Collapse
Affiliation(s)
- Yuka Takemon
- Genome Science and Technology Graduate Program, University of British Columbia, Vancouver, BC V5Z 4S6, Canada;
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.L.); (A.M.); (S.M.G.)
| | - Véronique G. LeBlanc
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.L.); (A.M.); (S.M.G.)
| | - Jungeun Song
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.L.); (A.M.); (S.M.G.)
| | - Susanna Y. Chan
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.L.); (A.M.); (S.M.G.)
| | - Stephen Dongsoo Lee
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.L.); (A.M.); (S.M.G.)
| | - Diane L. Trinh
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.L.); (A.M.); (S.M.G.)
| | - Shiekh Tanveer Ahmad
- Department of Pathology & Laboratory Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - William R. Brothers
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.L.); (A.M.); (S.M.G.)
| | - Richard D. Corbett
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.L.); (A.M.); (S.M.G.)
| | - Alessia Gagliardi
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.L.); (A.M.); (S.M.G.)
| | - Annie Moradian
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.L.); (A.M.); (S.M.G.)
| | - J. Gregory Cairncross
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Stephen Yip
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (S.Y.); (S.A.J.R.A.); (C.S.H.)
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada
| | - Samuel A. J. R. Aparicio
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (S.Y.); (S.A.J.R.A.); (C.S.H.)
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada
| | - Jennifer A. Chan
- Department of Pathology & Laboratory Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB T2N 4Z6, Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Christopher S. Hughes
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (S.Y.); (S.A.J.R.A.); (C.S.H.)
| | - Gregg B. Morin
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.L.); (A.M.); (S.M.G.)
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Sharon M. Gorski
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.L.); (A.M.); (S.M.G.)
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Suganthi Chittaranjan
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.L.); (A.M.); (S.M.G.)
| | - Marco A. Marra
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada; (V.G.L.); (A.M.); (S.M.G.)
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| |
Collapse
|
7
|
Liu Y, Yang C, Chen S, Liu W, Liang J, He S, Hui J. Cancer-derived exosomal miR-375 targets DIP2C and promotes osteoblastic metastasis and prostate cancer progression by regulating the Wnt signaling pathway. Cancer Gene Ther 2023; 30:437-449. [PMID: 36434177 DOI: 10.1038/s41417-022-00563-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 10/23/2022] [Accepted: 11/08/2022] [Indexed: 11/27/2022]
Abstract
Bone metastasis is the most common complication responsible for most deaths in the advanced stages of prostate cancer (PCa). However, the exact mechanism of bone metastasis in PCa remains unelucidated. Herein, we explored the function and potential underlying mechanism of exosomal miR-375 in bone metastasis and tumor progression in PCa. This study revealed that miR-375 expression was markedly upregulated in advanced PCa with bone metastasis and metastatic PCa cell lines. Moreover, miR-375 showed high expression in PCa-derived exosomes and could be delivered to human mesenchymal stem cells (hMSCs) via exosomes. Mechanistically, miR-375 directly targeted DIP2C and upregulated the Wnt signaling pathway, thereby promoting osteoblastic differentiation in hMSCs. Furthermore, miR-375 promoted the proliferation, invasion, and migration of PCa cells in vitro and enhanced tumor progression and osteoblastic metastasis in vivo. Notably, the expression of miR-375, TCF-1, LEF-1, and β-catenin in was higher in PCa tissues with bone metastasis than in PCa tissues without bone metastasis and showed a continuous increase, whereas DIP2C, cyclin D1, and Axin2 showed an opposite expression pattern. In conclusion, our study suggests that cancer-derived exosomal miR-375 targets DIP2C, activates the Wnt signaling pathway, and promotes osteoblastic metastasis and PCa progression.
Collapse
Affiliation(s)
- Ying Liu
- Department of Oncology, Affiliated Huadu Hospital, Southern Medical University (People's Hospital of Huadu District), Guangzhou, 510810, Guangdong, China
| | - Changmou Yang
- Department of Urology, Shenshan Central Hospital, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Shanwei, 516600, Guangdong, China.,Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Shisheng Chen
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.,Department of Urology, Dongguan Tungwah Hospital, Dongguan, 523110, Guangdong, China
| | - Weihao Liu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Jingyi Liang
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Shuhua He
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| | - Jialiang Hui
- Department of Transplantation, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
8
|
Park J, Park GY, Lee J, Park J, Kim S, Kim E, Park SY, Yoon JH, Lee Y. ERK phosphorylation disrupts the intramolecular interaction of capicua to promote cytoplasmic translocation of capicua and tumor growth. Front Mol Biosci 2022; 9:1030725. [PMID: 36619173 PMCID: PMC9814488 DOI: 10.3389/fmolb.2022.1030725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Activation of receptor tyrosine kinase signaling inactivates capicua (CIC), a transcriptional repressor that functions as a tumor suppressor, via degradation and/or cytoplasmic translocation. Although CIC is known to be inactivated by phosphorylation, the mechanisms underlying the cytoplasmic translocation of CIC remain poorly understood. Therefore, we aimed to evaluate the roles of extracellular signal-regulated kinase (ERK), p90RSK, and c-SRC in the epidermal growth factor receptor (EGFR) activation-induced cytoplasmic translocation of CIC and further investigated the molecular basis for this process. We found that nuclear ERK induced the cytoplasmic translocation of CIC-S. We identified 12 serine and threonine (S/T) residues within CIC, including S173 and S301 residues that are phosphorylated by p90RSK, which contribute to the cytoplasmic translocation of CIC-S when phosphorylated. The amino-terminal (CIC-S-N) and carboxyl-terminal (CIC-S-C) regions of CIC-S were found to interact with each other to promote their nuclear localization. EGF treatment disrupted the interaction between CIC-S-N and CIC-S-C and induced their cytoplasmic translocation. Alanine substitution for the 12 S/T residues blocked the cytoplasmic translocation of CIC-S and consequently enhanced the tumor suppressor activity of CIC-S. Our study demonstrates that ERK-mediated disruption of intramolecular interaction of CIC is critical for the cytoplasmic translocation of CIC, and suggests that the nuclear retention of CIC may represent a strategy for cancer therapy.
Collapse
Affiliation(s)
- Jongmin Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, South Korea
| | - Guk-Yeol Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, South Korea
| | - Jongeun Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, South Korea
| | - Joonyoung Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, South Korea
| | - Soeun Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, South Korea
| | - Eunjeong Kim
- Department of Biology, College of Natural Sciences, Kyungpook National University, Daegu, South Korea
| | - Seung-Yeol Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, South Korea
| | - Jong Hyuk Yoon
- Neurodegenerative Diseases Research Group, Korea Brain Research Institute, Daegu, South Korea
| | - Yoontae Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, South Korea,Institute of Convergence Science, Yonsei University, Seoul, South Korea,*Correspondence: Yoontae Lee,
| |
Collapse
|
9
|
Gupta N, Song H, Wu W, Ponce RK, Lin YK, Kim JW, Small EJ, Feng FY, Huang FW, Okimoto RA. The CIC-ERF co-deletion underlies fusion-independent activation of ETS family member, ETV1, to drive prostate cancer progression. eLife 2022; 11:e77072. [PMID: 36383412 PMCID: PMC9668335 DOI: 10.7554/elife.77072] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 10/16/2022] [Indexed: 11/13/2022] Open
Abstract
Human prostate cancer can result from chromosomal rearrangements that lead to aberrant ETS gene expression. The mechanisms that lead to fusion-independent ETS factor upregulation and prostate oncogenesis remain relatively unknown. Here, we show that two neighboring transcription factors, Capicua (CIC) and ETS2 repressor factor (ERF), which are co-deleted in human prostate tumors can drive prostate oncogenesis. Concurrent CIC and ERF loss commonly occur through focal genomic deletions at chromosome 19q13.2. Mechanistically, CIC and ERF co-bind the proximal regulatory element and mutually repress the ETS transcription factor, ETV1. Targeting ETV1 in CIC and ERF-deficient prostate cancer limits tumor growth. Thus, we have uncovered a fusion-independent mode of ETS transcriptional activation defined by concurrent loss of CIC and ERF.
Collapse
Affiliation(s)
- Nehal Gupta
- Department of Medicine, University of CaliforniaSan FranciscoUnited States
| | - Hanbing Song
- Department of Medicine, University of CaliforniaSan FranciscoUnited States
| | - Wei Wu
- Department of Medicine, University of CaliforniaSan FranciscoUnited States
| | - Rovingaile K Ponce
- Department of Medicine, University of CaliforniaSan FranciscoUnited States
| | - Yone K Lin
- Department of Medicine, University of CaliforniaSan FranciscoUnited States
| | - Ji Won Kim
- Department of Medicine, University of CaliforniaSan FranciscoUnited States
| | - Eric J Small
- Department of Medicine, University of CaliforniaSan FranciscoUnited States
- Helen Diller Family Comprehensive Cancer Center, University of CaliforniaSan FranciscoUnited States
| | - Felix Y Feng
- Department of Medicine, University of CaliforniaSan FranciscoUnited States
- Helen Diller Family Comprehensive Cancer Center, University of CaliforniaSan FranciscoUnited States
- Department of Radiation Oncology, University of CaliforniaSan FranciscoUnited States
| | - Franklin W Huang
- Department of Medicine, University of CaliforniaSan FranciscoUnited States
- Helen Diller Family Comprehensive Cancer Center, University of CaliforniaSan FranciscoUnited States
| | - Ross A Okimoto
- Department of Medicine, University of CaliforniaSan FranciscoUnited States
- Helen Diller Family Comprehensive Cancer Center, University of CaliforniaSan FranciscoUnited States
| |
Collapse
|
10
|
Schaefer M, Nabih A, Spies D, Hermes V, Bodak M, Wischnewski H, Stalder P, Ngondo RP, Liechti LA, Sajic T, Aebersold R, Gatfield D, Ciaudo C. Global and precise identification of functional
miRNA
targets in
mESCs
by integrative analysis. EMBO Rep 2022; 23:e54762. [PMID: 35899551 PMCID: PMC9442311 DOI: 10.15252/embr.202254762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 12/03/2022] Open
Abstract
MicroRNA (miRNA) loaded Argonaute (AGO) complexes regulate gene expression via direct base pairing with their mRNA targets. Previous works suggest that up to 60% of mammalian transcripts might be subject to miRNA‐mediated regulation, but it remains largely unknown which fraction of these interactions are functional in a specific cellular context. Here, we integrate transcriptome data from a set of miRNA‐depleted mouse embryonic stem cell (mESC) lines with published miRNA interaction predictions and AGO‐binding profiles. Using this integrative approach, combined with molecular validation data, we present evidence that < 10% of expressed genes are functionally and directly regulated by miRNAs in mESCs. In addition, analyses of the stem cell‐specific miR‐290‐295 cluster target genes identify TFAP4 as an important transcription factor for early development. The extensive datasets developed in this study will support the development of improved predictive models for miRNA‐mRNA functional interactions.
Collapse
Affiliation(s)
- Moritz Schaefer
- Swiss Federal Institute of Technology Zurich IMHS, Chair of RNAi and Genome Integrity Zurich Switzerland
- Life Science Zurich Graduate School University of Zürich Zurich Switzerland
| | - Amena Nabih
- Swiss Federal Institute of Technology Zurich IMHS, Chair of RNAi and Genome Integrity Zurich Switzerland
- Life Science Zurich Graduate School University of Zürich Zurich Switzerland
| | - Daniel Spies
- Swiss Federal Institute of Technology Zurich IMHS, Chair of RNAi and Genome Integrity Zurich Switzerland
- Life Science Zurich Graduate School University of Zürich Zurich Switzerland
| | - Victoria Hermes
- Swiss Federal Institute of Technology Zurich IMHS, Chair of RNAi and Genome Integrity Zurich Switzerland
| | - Maxime Bodak
- Swiss Federal Institute of Technology Zurich IMHS, Chair of RNAi and Genome Integrity Zurich Switzerland
- Life Science Zurich Graduate School University of Zürich Zurich Switzerland
| | - Harry Wischnewski
- Swiss Federal Institute of Technology Zurich IMHS, Chair of RNAi and Genome Integrity Zurich Switzerland
| | - Patrick Stalder
- Swiss Federal Institute of Technology Zurich IMHS, Chair of RNAi and Genome Integrity Zurich Switzerland
- Life Science Zurich Graduate School University of Zürich Zurich Switzerland
| | - Richard Patryk Ngondo
- Swiss Federal Institute of Technology Zurich IMHS, Chair of RNAi and Genome Integrity Zurich Switzerland
| | - Luz Angelica Liechti
- Center for Integrative Genomics (CIG) University of Lausanne Lausanne Switzerland
| | - Tatjana Sajic
- Swiss Federal Institute of Technology Zurich, IMSB Zürich Switzerland
| | - Ruedi Aebersold
- Swiss Federal Institute of Technology Zurich, IMSB Zürich Switzerland
| | - David Gatfield
- Center for Integrative Genomics (CIG) University of Lausanne Lausanne Switzerland
| | - Constance Ciaudo
- Swiss Federal Institute of Technology Zurich IMHS, Chair of RNAi and Genome Integrity Zurich Switzerland
| |
Collapse
|
11
|
Gan J, Liu S, Zhang Y, He L, Bai L, Liao R, Zhao J, Guo M, Jiang W, Li J, Li Q, Mu G, Wu Y, Wang X, Zhang X, Zhou D, Lv H, Wang Z, Zhang Y, Qian C, Feng M, Chen H, Meng Q, Huang X. MicroRNA-375 is a therapeutic target for castration-resistant prostate cancer through the PTPN4/STAT3 axis. Exp Mol Med 2022; 54:1290-1305. [PMID: 36042375 PMCID: PMC9440249 DOI: 10.1038/s12276-022-00837-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 05/31/2022] [Accepted: 06/27/2022] [Indexed: 04/08/2023] Open
Abstract
The functional role of microRNA-375 (miR-375) in the development of prostate cancer (PCa) remains controversial. Previously, we found that plasma exosomal miR-375 is significantly elevated in castration-resistant PCa (CRPC) patients compared with castration-sensitive PCa patients. Here, we aimed to determine how miR-375 modulates CRPC progression and thereafter to evaluate the therapeutic potential of human umbilical cord mesenchymal stem cell (hucMSC)-derived exosomes loaded with miR-375 antisense oligonucleotides (e-375i). We used miRNA in situ hybridization technique to evaluate miR-375 expression in PCa tissues, gain- and loss-of-function experiments to determine miR-375 function, and bioinformatic methods, dual-luciferase reporter assay, qPCR, IHC and western blotting to determine and validate the target as well as the effects of miR-375 at the molecular level. Then, e-375i complexes were assessed for their antagonizing effects against miR-375. We found that the expression of miR-375 was elevated in PCa tissues and cancer exosomes, correlating with the Gleason score. Forced expression of miR-375 enhanced the expression of EMT markers and AR but suppressed apoptosis markers, leading to enhanced proliferation, migration, invasion, and enzalutamide resistance and decreased apoptosis of PCa cells. These effects could be reversed by miR-375 silencing. Mechanistically, miR-375 directly interfered with the expression of phosphatase nonreceptor type 4 (PTPN4), which in turn stabilized phosphorylated STAT3. Application of e-375i could inhibit miR-375, upregulate PTPN4 and downregulate p-STAT3, eventually repressing the growth of PCa. Collectively, we identified a novel miR-375 target, PTPN4, that functions upstream of STAT3, and targeting miR-375 may be an alternative therapeutic for PCa, especially for CRPC with high AR levels.
Collapse
Affiliation(s)
- Junqing Gan
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Shan Liu
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Yu Zhang
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Liangzi He
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Lu Bai
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Ran Liao
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Juan Zhao
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Madi Guo
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Wei Jiang
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Jiade Li
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Qi Li
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Guannan Mu
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Yangjiazi Wu
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Xinling Wang
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Xingli Zhang
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Dan Zhou
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Huimin Lv
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Zhengfeng Wang
- Department of Neurosurgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yanqiao Zhang
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Cheng Qian
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - MeiYan Feng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Hui Chen
- Department of Urologic Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Qingwei Meng
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Xiaoyi Huang
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China.
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, Heilongjiang, 150081, China.
| |
Collapse
|
12
|
Martinucci B, Cucielo MS, Minatel BC, Cury SS, Caxali GH, Aal MCE, Felisbino SL, Pinhal D, Carvalho RF, Delella FK. Fibronectin Modulates the Expression of miRNAs in Prostate Cancer Cell Lines. Front Vet Sci 2022; 9:879997. [PMID: 35898539 PMCID: PMC9310065 DOI: 10.3389/fvets.2022.879997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 06/08/2022] [Indexed: 01/10/2023] Open
Abstract
Prostate cancer (PCa) is a significant cause of cancer-related deaths among men and companion animals, such as dogs. However, despite its high mortality and incidence rates, the molecular mechanisms underlying this disease remain to be fully elucidated. Among the many factors involved in prostate carcinogenesis, the extracellular matrix (ECM) plays a crucial role. This ECM in the prostate is composed mainly of collagen fibers, reticular fibers, elastic fibers, proteoglycans and glycoproteins, such as fibronectin. Fibronectin is a glycoprotein whose dysregulation has been implicated in the development of multiple types of cancer, and it has been associated with cell migration, invasion, and metastasis. Furthermore, our research group has previously shown that fibronectin induces transcriptional changes by modulating the expression of protein coding genes in LNCaP cells. However, potential changes at the post-transcriptional level are still not well understood. This study investigated the impact of exposure to fibronectin on the expression of a key class of regulatory RNAs, the microRNAs (miRNAs), in prostate cancer cell lines LNCaP and PC-3. Five mammalian miRNAs (miR-21, miR-29b, miR-125b, miR-221, and miR-222) were differentially expressed after fibronectin exposure in prostate cell lines. The expression profile of hundreds of mRNAs predicted to be targeted by these miRNAs was analyzed using publicly available RNA-Sequencing data (GSE64025, GSE68645, GSE29155). Also, protein-protein interaction networks and enrichment analysis were performed to gain insights into miRNA biological functions. Altogether, these functional analyzes revealed that fibronectin exposure impacts the expression of miRNAs potentially involved in PCa causing changes in critical signaling pathways such as PI3K-AKT, and response to cell division, death, proliferation, and migration. The relationship here demonstrated between fibronectin exposure and altered miRNA expression improves the comprehension of PCa in both men and other animals, such as dogs, which naturally develop prostate cancer.
Collapse
Affiliation(s)
- Bruno Martinucci
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - Maira Smaniotto Cucielo
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - Brenda Carvalho Minatel
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - Sarah Santiloni Cury
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - Gabriel Henrique Caxali
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - Mirian Carolini Esgoti Aal
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - Sergio Luis Felisbino
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - Danillo Pinhal
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - Robson Francisco Carvalho
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
| | - Flávia Karina Delella
- Department of Structural and Functional Biology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, Brazil
- *Correspondence: Flávia Karina Delella
| |
Collapse
|
13
|
Zheng J, Chen G, Li T, He X, Luo Y, Yang K. Isoflurane Promotes Cell Proliferation, Invasion, and Migration by Regulating BACH1 and miR-375 in Prostate Cancer Cells In Vitro. Int J Toxicol 2022; 41:212-224. [PMID: 35532539 DOI: 10.1177/10915818221084906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The aim of this study was to investigate the mechanism of isoflurane in proliferation, invasion, and migration in prostate cancer (PC) cells in vitro by regulating BACH1 and miR-375. The effect of different concentrations of isoflurane (0%, 0.5%, 1%, and 2%) on PC cell proliferation (PC3 and 22RV1) was measured. After PC cells and normal human prostate stromal immortalized WPMY-1 cells were treated with isoflurane, BACH1 and miR-375 expression was measured. Subsequently, PC3 and 22RV1 cells underwent gain- and loss-of-function assays with or without 4-h 2% isoflurane pretreatment. The levels of miR-375, BACH1, and PTEN were assessed. The binding of BACH1 to miR-375 promoter was detected by ChIP assay. Dual-luciferase reporter assay detected the targeting relationship of miR-375 with BACH1 and PTEN. Isoflurane promoted PC3 and 22RV1 cell proliferation. In addition, isoflurane elevated the levels of BACH1 and miR-375 in a dosage-dependent manner in PC cells. Transfection with miR-375 inhibitor or sh-BACH1 repressed PC cell proliferation, invasion, and migration, while exposure to 2% isoflurane for 4 h before transfection counteracted the inhibitory effects of sh-BACH1 or miR-375 inhibitor on PC cells. PTEN expression was suppressed after 2% isoflurane treatment, but the transfection with miR-375 inhibitor partly abrogated this suppressive effect in PC cells. Moreover, BACH1 bound to miR-375 and miR-375 negatively targeted PTEN. miR-375 mimic could partially reverse the inhibitory effects of sh-BACH1 on the proliferation, invasion, and migration of isoflurane-treated PC cells. Isoflurane facilitated PC cell proliferation, migration, and invasion by activating BACH1 to upregulate miR-375.
Collapse
Affiliation(s)
- Jue Zheng
- Department of Urology, 87803Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, P.R. China
| | - Guiheng Chen
- Department of Urology, 87803Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, P.R. China
| | - Tieqiu Li
- Department of Urology, 87803Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, P.R. China
| | - Xiang He
- Department of Urology, 87803Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, P.R. China
| | - Yuanman Luo
- Department of Urology, 87803Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, P.R. China
| | - Ke Yang
- Department of Urology, 87803Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, P.R. China
| |
Collapse
|
14
|
Nhieu J, Lin YL, Wei LN. CRABP1 in Non-Canonical Activities of Retinoic Acid in Health and Diseases. Nutrients 2022; 14:nu14071528. [PMID: 35406141 PMCID: PMC9003107 DOI: 10.3390/nu14071528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/01/2022] [Accepted: 04/03/2022] [Indexed: 12/30/2022] Open
Abstract
In this review, we discuss the emerging role of Cellular Retinoic Acid Binding Protein 1 (CRABP1) as a mediator of non-canonical activities of retinoic acid (RA) and relevance to human diseases. We first discuss the role of CRABP1 in regulating MAPK activities and its implication in stem cell proliferation, cancers, adipocyte health, and neuro-immune regulation. We then discuss an additional role of CRABP1 in regulating CaMKII activities, and its implication in heart and motor neuron diseases. Through molecular and genetic studies of Crabp1 knockout (CKO) mouse and culture models, it is established that CRABP1 forms complexes with specific signaling molecules to function as RA-regulated signalsomes in a cell context-dependent manner. Gene expression data and CRABP1 gene single nucleotide polymorphisms (SNPs) of human cancer, neurodegeneration, and immune disease patients implicate the potential association of abnormality in CRABP1 with human diseases. Finally, therapeutic strategies for managing certain human diseases by targeting CRABP1 are discussed.
Collapse
Affiliation(s)
| | | | - Li-Na Wei
- Correspondence: ; Tel.: +1-612-6259-402
| |
Collapse
|
15
|
Sienkiewicz K, Yang C, Paschal BM, Ratan A. Genomic analyses of the metastasis-derived prostate cancer cell lines LNCaP, VCaP, and PC3-AR. Prostate 2022; 82:442-451. [PMID: 34951700 PMCID: PMC8792310 DOI: 10.1002/pros.24290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/11/2021] [Accepted: 12/07/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND The lymph node metastasis-derived LNCaP, the bone metastasis-derived PC3 (skull), and VCaP (vertebral) cell lines are widely used as preclinical models of human prostate cancer (CaP) and have been described in more than 19,000 publications. Here, we report on short-read whole-genome sequencing and genomic analyses of LNCaP, VCaP, and PC3 cells stably transduced with WT AR (PC3-AR). METHODS LNCaP, VCaP, and PC3-AR cell lines were sequenced to an average depth of more than 30-fold using Illumina short-read sequencing. Using various computational methods, we identified and compared the single-nucleotide variants, copy-number profiles, and the structural variants observed in the three cell lines. RESULTS LNCaP cells are composed of multiple subpopulations, which results in nonintegral copy number states and a high mutational load when the data is analyzed in bulk. All three cell lines contain pathogenic mutations and homozygous deletions in genes involved in DNA mismatch repair, along with deleterious mutations in cell-cycle, Wnt signaling, and other critical cellular processes. PC3-AR cells have a truncating mutation in TP53 and do not express the p53 protein. The VCaP cells contain a homozygous gain-of-function mutation in TP53 (p.R248W) that promotes cancer invasion, metastasis, and progression and has also been observed in prostate adenocarcinomas. In addition, we detect the signatures of chromothripsis of the q arms of chromosome 5 in both PC3-AR and VCaP cells, strengthening the association of TP53 inactivation with chromothripsis reported in other systems. CONCLUSIONS Our work provides a resource for genetic, genomic, and biological studies employing these commonly-used prostate cancer cell lines.
Collapse
Affiliation(s)
| | - Chunsong Yang
- Center for Cell Signaling, University of Virginia, Virginia, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Virginia, USA
| | - Bryce M. Paschal
- Center for Cell Signaling, University of Virginia, Virginia, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Virginia, USA
| | - Aakrosh Ratan
- Center for Public Health Genomics, University of Virginia, Virginia, USA
- Department of Public Health Sciences, University of Virginia, Virginia, USA
| |
Collapse
|
16
|
Hong H, Lee J, Park GY, Kim S, Park J, Park JS, Song Y, Lee S, Kim TJ, Lee YJ, Roh TY, Kwok SK, Kim SW, Tan Q, Lee Y. Postnatal regulation of B-1a cell development and survival by the CIC-PER2-BHLHE41 axis. Cell Rep 2022; 38:110386. [PMID: 35172136 DOI: 10.1016/j.celrep.2022.110386] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/23/2021] [Accepted: 01/24/2022] [Indexed: 02/07/2023] Open
Abstract
B-1 cell development mainly occurs via fetal and neonatal hematopoiesis and is suppressed in adult bone marrow hematopoiesis. However, little is known about the factors inhibiting B-1 cell development at the adult stage. We report that capicua (CIC) suppresses postnatal B-1a cell development and survival. CIC levels are high in B-1a cells and gradually increase in transitional B-1a (TrB-1a) cells with age. B-cell-specific Cic-null mice exhibit expansion of the B-1a cell population and a gradual increase in TrB-1a cell frequency with age but attenuated B-2 cell development. CIC deficiency enhances B cell receptor (BCR) signaling in transitional B cells and B-1a cell viability. Mechanistically, CIC-deficiency-mediated Per2 derepression upregulates Bhlhe41 levels by inhibiting CRY-mediated transcriptional repression for Bhlhe41, consequently promoting B-1a cell formation in Cic-null mice. Taken together, CIC is a key transcription factor that limits the B-1a cell population at the adult stage and balances B-1 versus B-2 cell formation.
Collapse
Affiliation(s)
- Hyebeen Hong
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, Republic of Korea
| | - Jongeun Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, Republic of Korea
| | - Guk-Yeol Park
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, Republic of Korea
| | - Soeun Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, Republic of Korea
| | - Jiho Park
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, Republic of Korea
| | - Jong Seok Park
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, Republic of Korea
| | - Youngkwon Song
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, Republic of Korea
| | - Sujin Lee
- Department of Immunology, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Tae Jin Kim
- Department of Immunology, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - You Jeong Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, Republic of Korea
| | - Tae-Young Roh
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, Republic of Korea
| | - Seung-Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sung Won Kim
- Department of Otolaryngology-Head and Neck Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Qiumin Tan
- Department of Cell Biology, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Yoontae Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk 37673, Republic of Korea.
| |
Collapse
|
17
|
Slabáková E, Kahounová Z, Procházková J, Souček K. Regulation of Neuroendocrine-like Differentiation in Prostate Cancer by Non-Coding RNAs. Noncoding RNA 2021; 7:ncrna7040075. [PMID: 34940756 PMCID: PMC8704250 DOI: 10.3390/ncrna7040075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/18/2021] [Accepted: 11/29/2021] [Indexed: 12/21/2022] Open
Abstract
Neuroendocrine prostate cancer (NEPC) represents a variant of prostate cancer that occurs in response to treatment resistance or, to a much lesser extent, de novo. Unravelling the molecular mechanisms behind transdifferentiation of cancer cells to neuroendocrine-like cancer cells is essential for development of new treatment opportunities. This review focuses on summarizing the role of small molecules, predominantly microRNAs, in this phenomenon. A published literature search was performed to identify microRNAs, which are reported and experimentally validated to modulate neuroendocrine markers and/or regulators and to affect the complex neuroendocrine phenotype. Next, available patients’ expression datasets were surveyed to identify deregulated microRNAs, and their effect on NEPC and prostate cancer progression is summarized. Finally, possibilities of miRNA detection and quantification in body fluids of prostate cancer patients and their possible use as liquid biopsy in prostate cancer monitoring are discussed. All the addressed clinical and experimental contexts point to an association of NEPC with upregulation of miR-375 and downregulation of miR-34a and miR-19b-3p. Together, this review provides an overview of different roles of non-coding RNAs in the emergence of neuroendocrine prostate cancer.
Collapse
|
18
|
Mei Y, Li K, Zhang Z, Li M, Yang H, Wang H, Huang X, Li X, Shi S, Yang H. miR-33b-3p Acts as a Tumor Suppressor by Targeting DOCK4 in Prostate Cancer. Front Oncol 2021; 11:740452. [PMID: 34804930 PMCID: PMC8595470 DOI: 10.3389/fonc.2021.740452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/18/2021] [Indexed: 11/28/2022] Open
Abstract
Despite that androgen-deprivation therapy results in long-lasting responses, the disease inevitably progresses to metastatic castration-resistant prostate cancer. In this study, we identified miR-33b-3p as a tumor suppressor in prostate cancer. miR-33b-3p was significantly reduced in prostate cancer tissues, and the low expression of miR-33b-3p was correlated with poor overall survival of prostate cancer patients. Overexpression of miR-33b-3p inhibited both migration and invasion of highly metastatic prostate cancer cells whereas inhibition of miR-33b-3p promoted those processes in lowly metastatic cells. The in vivo results demonstrate that miR-33b-3p suppresses metastasis of tail vein inoculated prostate cancer cells to lung and lymph nodes in mice. DOCK4 was validated as the direct target of miR-33b-3p. miR-33b-3p decreased the expression of DOCK4 and restoration of DOCK4 could rescue miR-33b-3p inhibition on cell migration and invasion. Moreover, downregulation of miR-33b-3p was induced by bortezomib, the clinically used proteasome inhibitor, and overexpression of miR-33b-3p enhanced the insufficient inhibition of bortezomib on migration and invasion as well as metastasis of prostate cancer cells. In summary, our findings demonstrate that miR-33b-3p suppresses metastasis by targeting DOCK4 in prostate cancer. Our results suggest that enhancing miR-33b-3p expression may provide a promising therapeutic strategy for overcoming that proteasome inhibitor’s poor efficacy against metastatic prostate cancer.
Collapse
Affiliation(s)
- Yu Mei
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Kai Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Zhicheng Zhang
- Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mengmeng Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Hong Yang
- Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hui Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xuemei Huang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Xinyuan Li
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Shuhua Shi
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Huanjie Yang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| |
Collapse
|
19
|
Lee SD, Song J, LeBlanc VG, Marra MA. Integrative multi-omic analysis reveals neurodevelopmental gene dysregulation in CIC-knockout and IDH1 mutant cells. J Pathol 2021; 256:297-309. [PMID: 34767259 PMCID: PMC9305137 DOI: 10.1002/path.5835] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 10/22/2021] [Accepted: 11/09/2021] [Indexed: 11/18/2022]
Abstract
Capicua (CIC)'s transcriptional repressor function is implicated in neurodevelopment and in oligodendroglioma (ODG) aetiology. However, CIC's role in these contexts remains obscure, primarily from our currently limited knowledge regarding its biological functions. Moreover, CIC mutations in ODG invariably co‐occur with a neomorphic IDH1/2 mutation, yet the functional relationship between these two genetic events is unknown. Here, we analysed models derived from an E6/E7/hTERT‐immortalized (i.e. p53‐ and RB‐deficient) normal human astrocyte cell line. To examine the consequences of CIC loss, we compared transcriptomic and epigenomic profiles between CIC wild‐type and knockout cell lines, with and without mutant IDH1 expression. Our analyses revealed dysregulation of neurodevelopmental genes in association with CIC loss. CIC ChIP‐seq was also performed to expand upon the currently limited ensemble of known CIC target genes. Among the newly identified direct CIC target genes were EPHA2 and ID1, whose functions are linked to neurodevelopment and the tumourigenicity of in vivo glioma tumour models. NFIA, a known mediator of gliogenesis, was discovered to be uniquely overexpressed in CIC‐knockout cells expressing mutant IDH1‐R132H protein. These results identify neurodevelopment and specific genes within this context as candidate targets through which CIC alterations may contribute to the progression of IDH‐mutant gliomas. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Stephen D Lee
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, Canada
| | - Jungeun Song
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, Canada
| | | | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| |
Collapse
|
20
|
Kachris S, Papadaki C, Rounis K, Tsitoura E, Kokkinaki C, Nikolaou C, Sourvinos G, Mavroudis D. Circulating miRNAs as Potential Biomarkers in Prostate Cancer Patients Undergoing Radiotherapy. Cancer Manag Res 2021; 13:8257-8271. [PMID: 34754245 PMCID: PMC8572024 DOI: 10.2147/cmar.s325246] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/21/2021] [Indexed: 12/21/2022] Open
Abstract
Introduction Disease recurrence is a major concern in patients with localized prostate cancer (PCa) following treatment with radiotherapy (RT), and few studies have evaluated the clinical relevance of microRNAs (miRNAs) prior and post-RT. Purpose We aimed to investigate the significance of miRNAs in the outcomes of prostate cancer patients undergoing radiotherapy and to identify the related pathways through bioinformatics analysis. Materials and Methods The expression levels of miR-21, miR-106b, miR-141 and miR-375 involved in the response to radiotherapy were assessed by RT-qPCR in the serum of PCa patients (n=56) prior- and post-RT. Results Low expression levels of miR-106b prior-RT were associated with extracapsular extension and seminal vesicles invasion by the tumor (p=0.031 and 0.044, respectively). In the high-risk subgroup (n=47), post-RT expression levels of miR-21 were higher in patients with biochemical relapse (BR) compared to non-relapse (p=0.043). Also, in the salvage treatment subgroup (post-operative BR; n=20), post-RT expression levels of miR-21 and miR-106b were higher in patients with BR compared to non-relapse (p=0.043 and p=0.032, respectively). In the whole group of patients, high expression levels of miR-21 prior-RT and of miR-106b post-RT were associated with significantly shorter overall survival (OS; p=0.049 and p=0.050, respectively). No associations were observed among miR-141 and miR-375 expression levels with clinicopathological features or treatment outcome. Bioinformatics analysis revealed significant enrichment in DNA damage response pathways. Conclusion Circulating miRNAs prior or post-RT may hold prognostic implications in patients with PCa.
Collapse
Affiliation(s)
- Stefanos Kachris
- Department of Radiation Oncology, University General Hospital, Heraklion, Crete, Greece
| | - Chara Papadaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Konstantinos Rounis
- Department of Medical Oncology, University General Hospital, Heraklion, Crete, Greece
| | - Eliza Tsitoura
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Chrysanthi Kokkinaki
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Christoforos Nikolaou
- Department of Biology, University of Crete, Heraklion, Crete, Greece.,Institute of Molecular Biology and Biotechnology (IMBB), Foundation of Research and Technology (FORTH), Heraklion, Crete, Greece.,Institute of Bioinnovation, Biomedical Science Research Center "Alexander Fleming", Athens, Greece
| | - George Sourvinos
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion, Crete, Greece
| | - Dimitrios Mavroudis
- Laboratory of Translational Oncology, School of Medicine, University of Crete, Heraklion, Crete, Greece.,Department of Medical Oncology, University General Hospital, Heraklion, Crete, Greece
| |
Collapse
|
21
|
Li K, Shao S, Ji T, Liu M, Wang L, Pang Y, Chen M, Xu S, Zhang K, Wang Q, Zhuang Z, Wei L, Zhang Y, Chen Y, Wang Y, Zhang J, Chen K, Lian H, Zhong C. Capicua Regulates Dendritic Morphogenesis Through Ets in Hippocampal Neurons in vitro. Front Neuroanat 2021; 15:669310. [PMID: 34385910 PMCID: PMC8353115 DOI: 10.3389/fnana.2021.669310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/28/2021] [Indexed: 11/25/2022] Open
Abstract
Capicua (Cic), a transcriptional repressor frequently mutated in brain cancer oligodendroglioma, is highly expressed in adult neurons. However, its function in the dendritic growth of neurons in the hippocampus remains poorly understood. Here, we confirmed that Cic was expressed in hippocampal neurons during the main period of dendritogenesis, suggesting that Cic has a function in dendrite growth. Loss-of-function and gain-of function assays indicated that Cic plays a central role in the inhibition of dendritic morphogenesis and dendritic spines in vitro. Further studies showed that overexpression of Cic reduced the expression of Ets in HT22 cells, while in vitro knockdown of Cic in hippocampal neurons significantly elevated the expression of Ets. These results suggest that Cic may negatively control dendrite growth through Ets, which was confirmed by ShRNA knockdown of either Etv4 or Etv5 abolishing the phenotype of Cic knockdown in cultured neurons. Taken together, our results suggest that Cic inhibits dendritic morphogenesis and the growth of dendritic spines through Ets.
Collapse
Affiliation(s)
- Keqin Li
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Shuai Shao
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tongjie Ji
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Min Liu
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lufeng Wang
- Department of Neurology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ying Pang
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Mu Chen
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Siyi Xu
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kuiming Zhang
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qi Wang
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhongwei Zhuang
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Liang Wei
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanfei Zhang
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanlin Chen
- Department of Neurosurgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yang Wang
- Department of Emergency, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Zhang
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kui Chen
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hao Lian
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chunlong Zhong
- Department of Neurosurgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
22
|
Ma H, Li X, Yu S, Hu Y, Yin M, Zhu F, Xu L, Wang T, Wang H, Li H, Zhao B, Huang Y. Deletion of the miR-25/93/106b cluster induces glomerular deposition of immune complexes and renal fibrosis in mice. J Cell Mol Med 2021; 25:7922-7934. [PMID: 34197043 PMCID: PMC8358857 DOI: 10.1111/jcmm.16721] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/20/2021] [Accepted: 05/22/2021] [Indexed: 01/17/2023] Open
Abstract
IgA nephropathy (IgAN), the most common form of primary glomerulonephritis, is caused by immune system dysfunction and affects only the kidneys. miRNA was involved in IgAN, in which their roles are still unknown. Herein, we found increased glomerular medulla size, proteinuria, kidney artery resistance, kidney fibrosis and immune complex deposition in 5‐month miR‐25/93/106b cluster knockout (miR‐TKO) mice. In vitro, the inhibition of miR‐25 cluster could promote cell proliferation and increase fibrosis‐related protein and transferrin receptor (TFRC) expression in human renal glomerular mesangial cell (HRMC). Luciferase assay revealed that inhibition of miR‐93/106b cluster could upregulate Ccnd1 expression through direct binding with the 3’UTR of Ccnd1. Conversely, inhibition of Ccnd1 expression prevented miR‐93/106b‐induced effect in HRMC. These findings suggested that miR‐25 cluster played an important role in the progression of IgAN, which provided new insights into the pathogenesis and treatment of IgAN.
Collapse
Affiliation(s)
- Hongchuang Ma
- Department of Cell Biology, Jinan University, Guangzhou, China.,Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin, China
| | - Xiang Li
- Nephrosis Precision Medicine Innovation Center, University of Beihua School of Medicine, Beihua University, Jilin, China
| | - Shanshan Yu
- Nephrosis Precision Medicine Innovation Center, University of Beihua School of Medicine, Beihua University, Jilin, China
| | - Yanling Hu
- Nephrosis Precision Medicine Innovation Center, University of Beihua School of Medicine, Beihua University, Jilin, China
| | | | - Fubin Zhu
- Nephrosis Precision Medicine Innovation Center, University of Beihua School of Medicine, Beihua University, Jilin, China
| | - Licheng Xu
- Department of Cell Biology, Jinan University, Guangzhou, China
| | - Tianhe Wang
- Department of Cell Biology, Jinan University, Guangzhou, China
| | - Huiyan Wang
- Jilin Collaborative Innovation Center for Antibody Engineering, Jilin Medical University, Jilin, China
| | - Hongzhi Li
- Nephrosis Precision Medicine Innovation Center, University of Beihua School of Medicine, Beihua University, Jilin, China
| | - Binghai Zhao
- Nephrosis Precision Medicine Innovation Center, University of Beihua School of Medicine, Beihua University, Jilin, China
| | - Yadong Huang
- Department of Cell Biology, Jinan University, Guangzhou, China
| |
Collapse
|
23
|
Liu Y, Wang Q, Wen J, Wu Y, Man C. MiR-375: A novel multifunctional regulator. Life Sci 2021; 275:119323. [PMID: 33744323 DOI: 10.1016/j.lfs.2021.119323] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/20/2021] [Accepted: 02/27/2021] [Indexed: 01/23/2023]
Abstract
MiR-375, a primitively described beta cell-specific miRNA, is confirmed to function as multi-functional regulator in diverse typical cellular pathways according to the follow-up researches. Based on the existing studies, miR-375 can regulate many functional genes and ectopic expressions of miR-375 are usually associated with pathological changes, and its expression regulation mechanism is mainly related to promoter methylation or circRNA. In this review, the regulatory functions of miR-375 in immunity, such as its relevance with macrophages, T helper cells and autoimmune diseases were briefly discussed. Also, the functions of miR-375 involved in inflammation, development and virus replication were reviewed. Finally, the mechanisms and application prospects of miR-375 in cancers were analyzed. Studies show that the application of miR-375 as therapeutic target and biomarker has a broad developing space in future. We hope this paper can provide reference for its further study.
Collapse
Affiliation(s)
- Yang Liu
- College of Life Science and Technology, Harbin Normal University, Harbin 150001, PR China
| | - Qiuyuan Wang
- College of Life Science and Technology, Harbin Normal University, Harbin 150001, PR China
| | - Jie Wen
- College of Life Science and Technology, Harbin Normal University, Harbin 150001, PR China
| | - Yiru Wu
- College of Life Science and Technology, Harbin Normal University, Harbin 150001, PR China
| | - Chaolai Man
- College of Life Science and Technology, Harbin Normal University, Harbin 150001, PR China.
| |
Collapse
|
24
|
Li Z, Li LX, Diao YJ, Wang J, Ye Y, Hao XK. Identification of Urinary Exosomal miRNAs for the Non-Invasive Diagnosis of Prostate Cancer. Cancer Manag Res 2021; 13:25-35. [PMID: 33442291 PMCID: PMC7797303 DOI: 10.2147/cmar.s272140] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 12/15/2020] [Indexed: 12/13/2022] Open
Abstract
Background Novel and non-invasive biomarkers with higher sensitivity and specificity for the diagnosis of prostate cancer (PCa) is urgently needed. In this study, we used next-generation sequencing (NGS) to characterize the genome-wide exosomal miRNA expression profiling in urine specimens and explored the diagnostic potential of urinary exosomal miRNAs for PCa. Methods Urinary exosomal microRNA expression profiling was performed by next-generation sequencing (NGS) and then validated by quantitative real-time PCR. Results Significant downregulation of urinary exosomal miR-375 was observed in PCa patients compared with healthy controls, while the expression levels of urinary exosomal miR-451a, miR-486-3p and miR-486-5p were found to be significantly up-regulated in the PCa patients. Furthermore, the expression level of urinary exosomal miR-375 showed a significant correlation with the clinical T-stage and bone metastasis of patients with PCa (P<0.05). Receiver operator characteristic curve demonstrated that the urinary exosomal miR-375, miR-451a, miR-486-3p and miR-486-5p levels can be used to differentiate PCa patients from healthy controls, with area under the curves (AUCs) of 0.788, 0.757, 0.704 and 0.796, respectively. The urinary exosomal miR-375 was found to be superior in discriminating between localized and metastatic PCa with an AUC of 0.806. Moreover, PCa patients can be distinguished from patients with benign prostatic hyperplasia by using a panel combining urinary exosomal miR-375 and miR-451a with an AUC of 0.726. Conclusion These findings demonstrate that the urinary exosomal miRNAs can serve as novel and non-invasive biomarkers for diagnosing and predicting the progression of PCa.
Collapse
Affiliation(s)
- Zhuo Li
- Department of Laboratory Medicine, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, People's Republic of China.,The National Engineering Research Center for Miniaturized Detection Systems, College of Life Science, Northwest University, Xi'an, Shaanxi 710069, People's Republic of China.,Department of Laboratory Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| | - La-Xiu Li
- Department of Laboratory Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China.,Department of Laboratory Medicine, Xi'an Fourth Hospital, Xi'an, Shaanxi 710004, People's Republic of China
| | - Yan-Jun Diao
- Department of Laboratory Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| | - Juan Wang
- Department of Laboratory Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| | - Yun Ye
- Department of Laboratory Medicine, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710077, People's Republic of China.,Department of Laboratory Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China
| | - Xiao-Ke Hao
- The National Engineering Research Center for Miniaturized Detection Systems, College of Life Science, Northwest University, Xi'an, Shaanxi 710069, People's Republic of China.,Department of Laboratory Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, Shaanxi 710032, People's Republic of China.,School of Medicine, Northwest University, Xi'an, Shaanxi 710069, People's Republic of China
| |
Collapse
|
25
|
MicroRNAs as Biomarkers in Canine Osteosarcoma: A New Future? Vet Sci 2020; 7:vetsci7040146. [PMID: 33008041 PMCID: PMC7711435 DOI: 10.3390/vetsci7040146] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 09/24/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022] Open
Abstract
Sarcomas are frequent in dogs and canine species are excellent animal models for studying the human counterpart. However, osteosarcomas are a rare form of sarcoma with high death rates in humans and dogs. miRNAs are small endogenous RNAs that regulate gene expression post-transcriptionally. The discovery of miRNAs could give a contribute in the diagnosis and prognosis of different types of tumors in animal species, as already in humans. The differentiated expression of miRNAs is a frequent finding in cancers and is related to their pathogenesis in many cases. Most canine and human sarcomas show similar miRNA aberrations. Lower levels of miR-1 and miR-133b in canine osteosarcoma tissues were found to increase tumorigenesis through a higher expression of their target genes MET and MCL1. The overexpression of miR-9 promotes a metastatic phenotype in canine osteosarcomas and its capacity as a prognostic biomarker for the disease is currently being evaluated. MicroRNAs at the 14q32 locus could be used as prognostic biomarkers, since their decreased expression has been associated with poor prognosis in canine and human osteosarcomas. Furthermore, a decreased expression of miR-34a in osteosarcoma tumour cells has been associated with shorter disease-free survival times and its reintroduction as a synthetic prodrug shows good potential as a novel therapeutic target to fight the disease. Circulating miR-214 and miR-126 are significantly increased in a broad-spectrum cancer and have the ability to successfully predict the prognosis of dogs. However, further studies are needed to make the use of miRNAs as biomarkers a common practice.
Collapse
|
26
|
Capicua in Human Cancer. Trends Cancer 2020; 7:77-86. [PMID: 32978089 DOI: 10.1016/j.trecan.2020.08.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/27/2020] [Accepted: 08/28/2020] [Indexed: 12/11/2022]
Abstract
Capicua (CIC) is a highly conserved transcriptional repressor that is differentially regulated through mitogen-activated protein kinase (MAPK) signaling or genetic alteration across human cancer. CIC contributes to tumor progression and metastasis through direct transcriptional control of effector target genes. Recent findings indicate that CIC dysregulation is mechanistically linked and restricted to specific cancer subtypes, yet convergence on key downstream transcriptional nodes are critical for CIC-regulated oncogenesis across these cancers. In this review, we focus on how differential regulation of CIC through functional and genetic mechanisms contributes to subtype-specific cancer phenotypes and we propose new therapeutic strategies to effectively target CIC-altered cancers.
Collapse
|
27
|
Pudova EA, Krasnov GS, Nyushko KM, Kobelyatskaya AA, Savvateeva MV, Poloznikov AA, Dolotkazin DR, Klimina KM, Guvatova ZG, Simanovsky SA, Gladysh NS, Tokarev AT, Melnikova NV, Dmitriev AA, Alekseev BY, Kaprin AD, Kiseleva MV, Snezhkina AV, Kudryavtseva AV. miRNAs expression signature potentially associated with lymphatic dissemination in locally advanced prostate cancer. BMC Med Genomics 2020; 13:129. [PMID: 32948204 PMCID: PMC7500008 DOI: 10.1186/s12920-020-00788-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 08/31/2020] [Indexed: 02/08/2023] Open
Abstract
Background Prostate cancer is one of the most common and socially significant cancers among men. The aim of our study was to reveal changes in miRNA expression profiles associated with lymphatic dissemination in prostate cancer and to identify the most prominent miRNAs as potential prognostic markers for future studies. Methods High-throughput miRNA sequencing was performed for 44 prostate cancer specimens taken from Russian patients, with and without lymphatic dissemination (N1 – 20 samples; N0 – 24 samples). Results We found at least 18 microRNAs with differential expression between N0 and N1 sample groups: miR-182-5p, miR-183-5p, miR-96-5p, miR-25-3p, miR-93-5p, miR-7-5p, miR-615-3p, miR-10b, miR-1248 (N1-miRs; elevated expression in N1 cohort; p < 0.05); miR-1271-5p, miR-184, miR-222-3p, miR-221-5p, miR-221-3p, miR-455-3p, miR-143-5p, miR-181c-3p and miR-455-5p (N0-miRs; elevated expression in N0; p < 0.05). The expression levels of N1-miRs were highly correlated between each other (the same is applied for N0-miRs) and the expression levels of N0-miRs and N1-miRs were anti-correlated. The tumor samples can be divided into two groups depending on the expression ratio between N0-miRs and N1-miRs. Conclusions We found the miRNA expression signature associated with lymphatic dissemination, in particular on the Russian patient cohort. Many of these miRNAs are well-known players in either oncogenic transformation or tumor suppression. Further experimental studies with extended sampling are required to validate these results.
Collapse
Affiliation(s)
- Elena A Pudova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
| | - George S Krasnov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Kirill M Nyushko
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | | | - Maria V Savvateeva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Andrey A Poloznikov
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Daniyar R Dolotkazin
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Kseniya M Klimina
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Zulfiya G Guvatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Sergey A Simanovsky
- A. N. Severtsov Institute of Ecology and Evolution, Russian Academy of Sciences, Moscow, Russia
| | | | | | - Nataliya V Melnikova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Alexey A Dmitriev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Boris Y Alekseev
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Andrey D Kaprin
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Marina V Kiseleva
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | | | - Anna V Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
28
|
Cao X, Wolf A, Kim SE, Cabrera RM, Wlodarczyk BJ, Zhu H, Parker M, Lin Y, Steele JW, Han X, Ramaekers VT, Steinfeld R, Finnell RH, Lei Y. CIC de novo loss of function variants contribute to cerebral folate deficiency by downregulating FOLR1 expression. J Med Genet 2020; 58:484-494. [PMID: 32820034 PMCID: PMC7895856 DOI: 10.1136/jmedgenet-2020-106987] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/27/2020] [Accepted: 06/05/2020] [Indexed: 01/09/2023]
Abstract
Background Cerebral folate deficiency (CFD) syndrome is characterised by a low concentration of 5-methyltetrahydrofolate in cerebrospinal fluid, while folate levels in plasma and red blood cells are in the low normal range. Mutations in several folate pathway genes, including FOLR1 (folate receptor alpha, FRα), DHFR (dihydrofolate reductase) and PCFT (proton coupled folate transporter) have been previously identified in patients with CFD. Methods In an effort to identify causal mutations for CFD, we performed whole exome sequencing analysis on eight CFD trios and identified eight de novo mutations in seven trios. Results Notably, we found a de novo stop gain mutation in the capicua (CIC) gene. Using 48 sporadic CFD samples as a validation cohort, we identified three additional rare variants in CIC that are putatively deleterious mutations. Functional analysis indicates that CIC binds to an octameric sequence in the promoter regions of folate transport genes: FOLR1, PCFT and reduced folate carrier (Slc19A1; RFC1). The CIC nonsense variant (p.R353X) downregulated FOLR1 expression in HeLa cells as well as in the induced pluripotent stem cell (iPSCs) derived from the original CFD proband. Folate binding assay demonstrated that the p.R353X variant decreased cellular binding of folic acid in cells. Conclusion This study indicates that CIC loss of function variants can contribute to the genetic aetiology of CFD through regulating FOLR1 expression. Our study described the first mutations in a non-folate pathway gene that can contribute to the aetiology of CFD.
Collapse
Affiliation(s)
- Xuanye Cao
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Annika Wolf
- Department of Pediatric Neurology, University Medical Center Göttingen, Gottingen, Niedersachsen, Germany
| | - Sung-Eun Kim
- Department of Pediatrics, University of Texas at Austin, Austin, Texas, USA
| | - Robert M Cabrera
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Bogdan J Wlodarczyk
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Huiping Zhu
- Department of Nutritional Sciences, University of Texas at Austin Dell Medical School, Austin, Texas, USA
| | - Margaret Parker
- Department of Pediatrics, University of Texas at Austin, Austin, Texas, USA
| | - Ying Lin
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - John W Steele
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA.,Institute for Cell and Molecular Biology, The University of Texas at Austin, Austin, Texas, USA
| | - Xiao Han
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Vincent Th Ramaekers
- Department of Pediatric Neurology, University Hospital Center Liège, Liège, Belgium
| | - Robert Steinfeld
- Department of Pediatric Neurology, University Medical Center Göttingen, Gottingen, Niedersachsen, Germany .,University Children's Hospital Zurich, Zurich, Switzerland
| | - Richard H Finnell
- Department of Pediatrics, University of Texas at Austin, Austin, Texas, USA .,Center for Precision Environmental Health, Departments of Molecular and Cellular Biology and Medicine, Houston, Texas, USA.,Departments of Molecular and Human Genetics and Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Yunping Lei
- Center for Precision Environmental Health, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
29
|
MicroRNAs as Guardians of the Prostate: Those Who Stand before Cancer. What Do We Really Know about the Role of microRNAs in Prostate Biology? Int J Mol Sci 2020; 21:ijms21134796. [PMID: 32645914 PMCID: PMC7370012 DOI: 10.3390/ijms21134796] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 06/29/2020] [Accepted: 07/02/2020] [Indexed: 12/17/2022] Open
Abstract
Prostate cancer is the second leading cause of cancer-related deaths of men in the Western world. Despite recent advancement in genomics, transcriptomics and proteomics to understand prostate cancer biology and disease progression, castration resistant metastatic prostate cancer remains a major clinical challenge and often becomes incurable. MicroRNAs (miRNAs), about 22-nucleotide-long non-coding RNAs, are a group of regulatory molecules that mainly work through post-transcriptional gene silencing via translational repression. Expression analysis studies have revealed that miRNAs are aberrantly expressed in cancers and have been recognized as regulators of prostate cancer progression. In this critical review, we provide an analysis of reported miRNA functions and conflicting studies as they relate to expression levels of specific miRNAs and prostate cancer progression; oncogenic and/or tumor suppressor roles; androgen receptor signaling; epithelial plasticity; and the current status of diagnostic and therapeutic applications. This review focuses on select miRNAs, highly expressed in normal and cancer tissue, to emphasize the current obstacles faced in utilizing miRNA data for significant impacts on prostate cancer therapeutics.
Collapse
|
30
|
Ashrafizadeh M, Najafi M, Mohammadinejad R, Farkhondeh T, Samarghandian S. Flaming the fight against cancer cells: the role of microRNA-93. Cancer Cell Int 2020; 20:277. [PMID: 32612456 PMCID: PMC7325196 DOI: 10.1186/s12935-020-01349-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 06/15/2020] [Indexed: 12/14/2022] Open
Abstract
There have been attempts to develop novel anti-tumor drugs in cancer therapy. Although satisfying results have been observed at a consequence of application of chemotherapeutic agents, the cancer cells are capable of making resistance into these agents. This has forced scientists into genetic manipulation as genetic alterations are responsible for generation of a high number of cancer cells. MicroRNAs (miRs) are endogenous, short non-coding RNAs that affect target genes at the post-transcriptional level. Increasing evidence reveals the potential role of miRs in regulation of biological processes including angiogenesis, metabolism, cell proliferation, cell division, and cell differentiation. Abnormal expression of miRs is associated with development of a number of pathologic events, particularly cancer. MiR-93 plays a significant role in both physiological and pathological mechanisms. At the present review, we show how this miR dually affects the proliferation and invasion of cancer cells. Besides, we elucidate the oncogenesis or oncosuppressor function of miR-93.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Tahereh Farkhondeh
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Saeed Samarghandian
- Healthy Ageing Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
31
|
Hübner M, Moellhoff N, Effinger D, Hinske CL, Hirschberger S, Wu T, Müller MB, Strauß G, Kreth FW, Kreth S. MicroRNA-93 acts as an "anti-inflammatory tumor suppressor" in glioblastoma. Neurooncol Adv 2020; 2:vdaa047. [PMID: 32642700 PMCID: PMC7282490 DOI: 10.1093/noajnl/vdaa047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background Inflammation is an important driver of malignant glioma disease. Inflammatory mediators are not only produced by immune cells in the tumor microenvironment, but also by glioblastoma (GBM) cells themselves creating a mutually reinforcing loop. We here aimed at identifying an “anti-inflammatory switch” that allows to dampen inflammation in GBM. Methods We used human GBM specimens, primary cultures, and cell lines. The response of GBM cells toward inflammatory stimuli was tested by incubation with supernatant of stimulated human immune cells. Expression levels were measured by whole transcriptome microarrays and qRT-PCR, and protein was quantified by LUMINEX and SDS-PAGE. MicroRNA binding to 3′UTRs was analyzed by luciferase assays. Proliferation rates were determined by flow cytometry, and invasion and angiogenesis were studied using migration and endothelial tube formation assays. Results We demonstrated GBM cells to secrete high amounts of proinflammatory mediators in an inflammatory microenvironment. We found miR-93 as a potential “anti-inflammatory tumor suppressor” dramatically downregulated in GBM. Concordantly, cytokine secretion dropped after miR-93 re-expression. Transfection of miR-93 in GBM cells led to down-regulation of hubs of the inflammatory networks, namely, HIF-1α and MAP3K2 as well as IL-6, G-CSF, IL-8, LIF, IL-1β, COX2, and CXCL5. We showed only COX2 and CXCL5 to be indirectly regulated by miR-93 while all other genes are true targets. Phenotypically, re-expression of miR-93 in GBM cells substantially suppressed proliferation, migration, and angiogenesis. Conclusions Alleviating GBM-derived inflammation by re-expression of miR-93 may be a powerful tool to mitigate these tumors’ aggressiveness and holds promise for new clinical approaches.
Collapse
Affiliation(s)
- Max Hübner
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.,Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Nicholas Moellhoff
- Division of Hand, Plastic and Aesthetic Surgery, University Hospital, LMU Munich, Munich, Germany
| | - David Effinger
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.,Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | | | - Simon Hirschberger
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.,Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Tingting Wu
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Martin Bernhard Müller
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.,Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Gabriele Strauß
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.,Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | | | - Simone Kreth
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.,Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
32
|
Valera VA, Parra-Medina R, Walter BA, Pinto P, Merino MJ. microRNA Expression Profiling in Young Prostate Cancer Patients. J Cancer 2020; 11:4106-4114. [PMID: 32368293 PMCID: PMC7196262 DOI: 10.7150/jca.37842] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 03/27/2020] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) are small, non-coding RNA molecules with multiple roles in many biological processes. Few studies have shown the molecular characteristics in younger prostate cancer (PCa) patients. In this study, we performed miRNA profiling in young PCa (EO-PCa) cases compared with PCa arising in older men (LO-PCa). Experimental Design: Formalin-fixed, paraffin embedded tissue was used. miRNA was extracted for PCR array and NanoString methods. Relative miRNAs expression levels were obtained by comparing young vs older men, and young PCa tumor samples vs normal epithelium. Results: miRNA profiling showed a different expression pattern in PCa arising in younger men, and young PCa tumoral and its normal counterpart. Nine miRNAs (hsa-miR-140-5p, hsa-miR-146a, hsa-miR-29b, hsa-miR-9, hsa-miR-124-3p, hsa-let-7f-5p, hsa-miR-184, hsa-miR-373, hsa-miR-146b-5p) showed differences in the expression compared to LO-PCa. Fourteen miRNAs were significantly up-regulated (miR-1973, miR-663a, miR-575, miR-93-5p, miR-630, miR-600, miR-494, miR-150-5p, miR-137, miR-25-3p, miR-375, miR-489, miR-888-5p, miR-142-3p), while 9 were found down-regulated (miR-21-5p, miR-363-3p, miR-205-5p, miR-548ai, miR-3195, 145-5p, miR-143-3p, miR-222-3p, miR-221-3p) comparing young PCa tumoral tissue compared to normal counterpart. The higher expression of miR-600 and miR-137 were associated with high Gleason score, extraprostatic extension and lymphatic invasion. Conclusion: These results suggest that PCa in younger patients has a different expression profile compared to normal tissue and PCa arising in older man. Differentially expressed miRNAs provide insights of molecular mechanisms involve in this PCa subtype.
Collapse
Affiliation(s)
- Vladimir A Valera
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health. Bethesda MD
| | - Rafael Parra-Medina
- Translational Surgical Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda MD.,Faculty of Natural Science and Mathematics, Universidad del Rosario, Bogotá, Colombia
| | - Beatriz A Walter
- Translational Surgical Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda MD
| | - Peter Pinto
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health. Bethesda MD
| | - Maria J Merino
- Translational Surgical Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda MD
| |
Collapse
|
33
|
Lee Y. Regulation and function of capicua in mammals. Exp Mol Med 2020; 52:531-537. [PMID: 32238859 PMCID: PMC7210929 DOI: 10.1038/s12276-020-0411-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/27/2020] [Accepted: 03/03/2020] [Indexed: 12/20/2022] Open
Abstract
Capicua (CIC) is an evolutionarily conserved transcription factor. CIC contains a high-mobility group (HMG) box that recognizes specific DNA sequences to regulate the expression of various target genes. CIC was originally identified in Drosophila melanogaster as a transcriptional repressor that suppresses the receptor tyrosine kinase signaling pathway. This molecule controls normal organ growth and tissue patterning as well as embryogenesis in Drosophila. Recent studies have also demonstrated its extensive functions in mammals. For example, CIC regulates several developmental and physiological processes, including lung development, abdominal wall closure during embryogenesis, brain development and function, neural stem cell homeostasis, T cell differentiation, and enterohepatic circulation of bile acids. CIC is also associated with the progression of various types of cancer and neurodegeneration in spinocerebellar ataxia type-1, systemic autoimmunity, and liver injury. In this review, I provide a broad overview of our current understanding of the regulation and functions of CIC in mammals and discuss future research directions.
Collapse
Affiliation(s)
- Yoontae Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Republic of Korea.
- Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
34
|
Wong D, Yip S. Making heads or tails - the emergence of capicua (CIC) as an important multifunctional tumour suppressor. J Pathol 2020; 250:532-540. [PMID: 32073140 DOI: 10.1002/path.5400] [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: 01/10/2020] [Revised: 01/31/2020] [Accepted: 02/12/2020] [Indexed: 12/24/2022]
Abstract
Capicua, encoded by the gene CIC, is an evolutionarily conserved high-mobility group-box transcription factor downstream of the receptor tyrosine kinase and mitogen-activated protein kinase pathways. It was initially discovered and studied in Drosophila. Recurrent mutations in CIC were first identified in oligodendroglioma, a subtype of low-grade glioma. Subsequent studies have identified CIC aberrations in multiple types of cancer and have established CIC as a potent tumour suppressor involved in regulating pathways related to cell growth and proliferation, invasion and treatment resistance. The most well-known and studied targets of mammalian CIC are the oncogenic E-Twenty Six transcription factors ETV1/4/5, which have been found to be elevated in cancers with CIC aberrations. Here, we review the role of CIC in normal mammalian development, oncogenesis and tumour progression, and the functional interactors that mediate them. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Derek Wong
- Molecular Oncology, British Columbia Cancer Research Center, Vancouver, British Columbia, Canada.,Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Stephen Yip
- Molecular Oncology, British Columbia Cancer Research Center, Vancouver, British Columbia, Canada.,Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
35
|
Yoe J, Kim D, Kim S, Lee Y. Capicua restricts cancer stem cell-like properties in breast cancer cells. Oncogene 2020; 39:3489-3506. [PMID: 32108163 DOI: 10.1038/s41388-020-1230-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 02/14/2020] [Accepted: 02/17/2020] [Indexed: 12/27/2022]
Abstract
Cancer stem cells (CSCs) play a central role in cancer initiation, progression, therapeutic resistance, and recurrence in patients. Here we present Capicua (CIC), a developmental transcriptional repressor, as a suppressor of CSC properties in breast cancer cells. CIC deficiency critically enhances CSC self-renewal and multiple CSC subpopulations of breast cancer cells without altering their growth rate or invasiveness. Loss of CIC relieves repression of ETV4 and ETV5 expression, consequently promoting self-renewal capability, EpCAM+/CD44+/CD24low/- expression, and ALDH activity. In xenograft models, CIC deficiency significantly increases CSC frequency and drives tumor initiation through derepression of ETV4. Consistent with the experimental data, the CD44high/CD24low CSC-like feature is inversely correlated with CIC levels in breast cancer patients. We also identify SOX2 as a downstream target gene of CIC that partly promotes CSC properties. Taken together, our study demonstrates that CIC suppresses breast cancer formation via restricting cancer stemness and proposes CIC as a potential regulator of stem cell maintenance.
Collapse
Affiliation(s)
- Jeehyun Yoe
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Donghyo Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Sanguk Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, Republic of Korea.,Division of Integrative Bioscience and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, Republic of Korea
| | - Yoontae Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, Republic of Korea. .,Division of Integrative Bioscience and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, Gyeongbuk, 37673, Republic of Korea.
| |
Collapse
|
36
|
Lee JS, Kim E, Lee J, Kim D, Kim H, Kim CJ, Kim S, Jeong D, Lee Y. Capicua suppresses colorectal cancer progression via repression of ETV4 expression. Cancer Cell Int 2020; 20:42. [PMID: 32042269 PMCID: PMC7003492 DOI: 10.1186/s12935-020-1111-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 01/17/2020] [Indexed: 02/08/2023] Open
Abstract
Background Although major driver gene mutations have been identified, the complex molecular heterogeneity of colorectal cancer (CRC) remains unclear. Capicua (CIC) functions as a tumor suppressor in various types of cancers; however, its role in CRC progression has not been examined. Methods Databases for gene expression profile in CRC patient samples were used to evaluate the association of the levels of CIC and Polyoma enhancer activator 3 (PEA3) group genes (ETS translocation variant 1 (ETV1), ETV4, and ETV5), the best-characterized CIC targets in terms of CIC functions, with clinicopathological features of CRC. CIC and ETV4 protein levels were also examined in CRC patient tissue samples. Gain- and loss-of function experiments in cell lines and mouse xenograft models were performed to investigate regulatory functions of CIC and ETV4 in CRC cell growth and invasion. qRT-PCR and western blot analyses were performed to verify the CIC regulation of ETV4 expression in CRC cells. Rescue experiments were conducted using siRNA against ETV4 and CIC-deficient CRC cell lines. Results CIC expression was decreased in the tissue samples of CRC patients. Cell invasion, migration, and proliferation were enhanced in CIC-deficient CRC cells and suppressed in CIC-overexpressing cells. Among PEA3 group genes, ETV4 levels were most dramatically upregulated and inversely correlated with the CIC levels in CRC patient samples. Furthermore, derepression of ETV4 was more prominent in CIC-deficient CRC cells, when compared with that observed for ETV1 and ETV5. The enhanced cell proliferative and invasive capabilities in CIC-deficient CRC cells were completely recovered by knockdown of ETV4. Conclusion Collectively, the CIC-ETV4 axis is not only a key module that controls CRC progression but also a novel therapeutic and/or diagnostic target for CRC.
Collapse
Affiliation(s)
- Jeon-Soo Lee
- 1Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk Republic of Korea
| | - Eunjeong Kim
- 1Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk Republic of Korea
| | - Jongeun Lee
- 1Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk Republic of Korea
| | - Donghyo Kim
- 1Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk Republic of Korea
| | - Hyeongjoo Kim
- 3Soonchunhyang Medical Science Research Institute, Soonchunhyang University, Cheonan, Chungnam Republic of Korea
| | - Chang-Jin Kim
- 4Department of Pathology, College of Medicine, Soonchunhyang University, Room 601, 31 Soonchunhyang 6gil, Dongnam-gu, Cheonan, Chungnam 31151 Republic of Korea
| | - Sanguk Kim
- 1Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk Republic of Korea.,2Division of Integrative Bioscience and Biotechnology, Pohang University of Science and Technology, Pohang, Gyeongbuk Republic of Korea
| | - Dongjun Jeong
- 3Soonchunhyang Medical Science Research Institute, Soonchunhyang University, Cheonan, Chungnam Republic of Korea.,4Department of Pathology, College of Medicine, Soonchunhyang University, Room 601, 31 Soonchunhyang 6gil, Dongnam-gu, Cheonan, Chungnam 31151 Republic of Korea
| | - Yoontae Lee
- 1Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk Republic of Korea.,2Division of Integrative Bioscience and Biotechnology, Pohang University of Science and Technology, Pohang, Gyeongbuk Republic of Korea.,POSTECH Biotech Center, Room 388, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeongbuk 37673 Republic of Korea
| |
Collapse
|
37
|
Stefely JA, Zhang Y, Freiberger EC, Kwiecien NW, Thomas HE, Davis AM, Lowry ND, Vincent CE, Shishkova E, Clark NA, Medvedovic M, Coon JJ, Pagliarini DJ, Mercer CA. Mass spectrometry proteomics reveals a function for mammalian CALCOCO1 in MTOR-regulated selective autophagy. Autophagy 2020; 16:2219-2237. [PMID: 31971854 DOI: 10.1080/15548627.2020.1719746] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Macroautophagy/autophagy is suppressed by MTOR (mechanistic target of rapamycin kinase) and is an anticancer target under active investigation. Yet, MTOR-regulated autophagy remains incompletely mapped. We used proteomic profiling to identify proteins in the MTOR-autophagy axis. Wild-type (WT) mouse cell lines and cell lines lacking individual autophagy genes (Atg5 or Ulk1/Ulk2) were treated with an MTOR inhibitor to induce autophagy and cultured in media with either glucose or galactose. Mass spectrometry proteome profiling revealed an elevation of known autophagy proteins and candidates for new autophagy components, including CALCOCO1 (calcium binding and coiled-coil domain protein 1). We show that CALCOCO1 physically interacts with MAP1LC3C, a key protein in the machinery of autophagy. Genetic deletion of CALCOCO1 disrupted autophagy of the endoplasmic reticulum (reticulophagy). Together, these results reveal a role for CALCOCO1 in MTOR-regulated selective autophagy. More generally, the resource generated by this work provides a foundation for establishing links between the MTOR-autophagy axis and proteins not previously linked to this pathway. Abbreviations: ATG: autophagy-related; CALCOCO1: calcium binding and coiled-coil domain protein 1; CALCOCO2/NDP52: calcium binding and coiled-coil domain protein 2; CLIR: MAP1LC3C-interacting region; CQ: chloroquine; KO: knockout; LIR: MAP1LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MEF: mouse embryonic fibroblast; MLN: MLN0128 ATP-competitive MTOR kinase inhibitor; MTOR: mechanistic target of rapamycin kinase; reticulophagy: selective autophagy of the endoplasmic reticulum; TAX1BP1/CALCOCO3: TAX1 binding protein 1; ULK: unc 51-like autophagy activating kinase; WT: wild-type.
Collapse
Affiliation(s)
- Jonathan A Stefely
- Morgridge Institute for Research , Madison, WI, USA.,Medical Scientist Training Program, School of Medicine and Public Health, University of Wisconsin-Madison , Madison, WI, USA.,Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati , Cincinnati, OH, USA
| | - Yu Zhang
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati , Cincinnati, OH, USA
| | - Elyse C Freiberger
- Department of Chemistry, University ofWisconsin-Madison , Madison, WI, USA.,Department of Biomolecular Chemistry, University ofWisconsin-Madison , Madison, WI, USA.,Genome Center of Wisconsin , Madison, WI, USA.,Department of Biochemistry, University ofWisconsin-Madison , Madison, Madison, WI, USA
| | - Nicholas W Kwiecien
- Department of Chemistry, University ofWisconsin-Madison , Madison, WI, USA.,Department of Biomolecular Chemistry, University ofWisconsin-Madison , Madison, WI, USA.,Genome Center of Wisconsin , Madison, WI, USA.,Department of Biochemistry, University ofWisconsin-Madison , Madison, Madison, WI, USA
| | - Hala Elnakat Thomas
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati , Cincinnati, OH, USA
| | - Alexander M Davis
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati , Cincinnati, OH, USA
| | - Nathaniel D Lowry
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati , Cincinnati, OH, USA
| | - Catherine E Vincent
- Genome Center of Wisconsin , Madison, WI, USA.,Department of Chemistry, Hartwick College , Oneonta, NY, USA
| | | | - Nicholas A Clark
- Division of Biostatistics and Bioinformatics, Department of Environmental Health, University of Cincinnati , Cincinnati, OH, USA
| | - Mario Medvedovic
- Division of Biostatistics and Bioinformatics, Department of Environmental Health, University of Cincinnati , Cincinnati, OH, USA
| | - Joshua J Coon
- Morgridge Institute for Research , Madison, WI, USA.,Department of Chemistry, University ofWisconsin-Madison , Madison, WI, USA.,Department of Biomolecular Chemistry, University ofWisconsin-Madison , Madison, WI, USA.,Genome Center of Wisconsin , Madison, WI, USA
| | - David J Pagliarini
- Morgridge Institute for Research , Madison, WI, USA.,Department of Biochemistry, University ofWisconsin-Madison , Madison, Madison, WI, USA
| | - Carol A Mercer
- Division of Hematology/Oncology, Department of Internal Medicine, University of Cincinnati , Cincinnati, OH, USA
| |
Collapse
|
38
|
Liu J, Wang P, Zhang P, Zhang X, Du H, Liu Q, Huang B, Qian C, Zhang S, Zhu W, Yang X, Xiao Y, Liu Z, Luo D. An integrative bioinformatics analysis identified miR-375 as a candidate key regulator of malignant breast cancer. J Appl Genet 2019; 60:335-346. [PMID: 31372832 DOI: 10.1007/s13353-019-00507-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 06/30/2019] [Accepted: 07/09/2019] [Indexed: 12/27/2022]
Abstract
MicroRNAs (miRNAs) are key regulators that play important biological roles in carcinogenesis and are promising biomarkers for cancer diagnosis and therapy. hsa-miR-375-3p (miR-375) has been suggested to serve as a tumor suppressor or oncogene in various tumor types; however, its specific expression and potential regulatory role in malignant breast cancer remain unclear. In this study, the results from noncoding RNA microarray analysis indicated that the miR-375 expression level is significantly decreased in malignant basal-like breast cancer compared with luminal-like breast cancer. A total of 1895 co-downregulated and 1645 co-upregulated genes were identified in miR-375 mimic-transfected basal-like breast cancer cell lines. Predicted miR-375 targets were obtained from the online databases TargetScan and DIANA-microT-CDS. Combined KEGG enrichment analysis for coregulated genes and predicted miR-375 targets provided information and revealed differences in potential dynamic signaling pathways regulated by miR-375 and also indicated specific regulatory pathways, such as RNA transport and processing, in basal-like breast cancer. Additionally, gene expression microarray analysis accompanied by UALCAN analysis was performed to screen upregulated genes in the basal-like subtype. Four potential key genes, including LDHB, CPNE8, QKI, and EIF5A2, were identified as candidate target genes of miR-375. Therefore, the present study demonstrated that miR-375 may be a potential key regulator and provide a promising direction for diagnostic and therapeutic developments for malignant breast cancer.
Collapse
Affiliation(s)
- Jiaxuan Liu
- Queen Mary School, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Ping Wang
- Queen Mary School, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Ping Zhang
- Department of Pathology, The Affiliated Infectious Diseases Hospital, Nanchang University, Nanchang, 330002, Jiangxi, China
| | - Xinyu Zhang
- Queen Mary School, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Hang Du
- Queen Mary School, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Qiang Liu
- National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Bo Huang
- Department of Pathology, The Affiliated Infectious Diseases Hospital, Nanchang University, Nanchang, 330002, Jiangxi, China
| | - Caiyun Qian
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Shuhua Zhang
- Jiangxi Cardiovascular Research Institute, Jiangxi Provincial People's Hospital, Nanchang, 330006, Jiangxi, China
| | - Weifeng Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xiaohong Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yingqun Xiao
- Department of Pathology, The Affiliated Infectious Diseases Hospital, Nanchang University, Nanchang, 330002, Jiangxi, China.
| | - Zhuoqi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi, China.
| | - Daya Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi, China.
- Jiangxi Province Key Laboratory of Tumor Pathogens and Molecular Pathology, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
39
|
Pan X, Shen HB. Inferring Disease-Associated MicroRNAs Using Semi-supervised Multi-Label Graph Convolutional Networks. iScience 2019; 20:265-277. [PMID: 31605942 PMCID: PMC6817654 DOI: 10.1016/j.isci.2019.09.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/05/2019] [Accepted: 09/11/2019] [Indexed: 01/22/2023] Open
Abstract
MicroRNAs (miRNAs) play crucial roles in biological processes involved in diseases. The associations between diseases and protein-coding genes (PCGs) have been well investigated, and miRNAs interact with PCGs to trigger them to be functional. We present a computational method, DimiG, to infer miRNA-associated diseases using a semi-supervised Graph Convolutional Network model (GCN). DimiG uses a multi-label framework to integrate PCG-PCG interactions, PCG-miRNA interactions, PCG-disease associations, and tissue expression profiles. DimiG is trained on disease-PCG associations and an interaction network using a GCN, which is further used to score associations between diseases and miRNAs. We evaluate DimiG on a benchmark set from verified disease-miRNA associations. Our results demonstrate that DimiG outperforms the best unsupervised method and is comparable to two supervised methods. Three case studies of prostate cancer, lung cancer, and inflammatory bowel disease further demonstrate the efficacy of DimiG, where top miRNAs predicted by DimiG are supported by literature.
Collapse
Affiliation(s)
- Xiaoyong Pan
- Institute of Image Processing and Pattern Recognition, Shanghai Jiao Tong University, and Key Laboratory of System Control and Information Processing, Ministry of Education of China, 200240 Shanghai, China; Department of Medical informatics, Erasmus Medical Center, 3015 CE Rotterdam, the Netherlands.
| | - Hong-Bin Shen
- Institute of Image Processing and Pattern Recognition, Shanghai Jiao Tong University, and Key Laboratory of System Control and Information Processing, Ministry of Education of China, 200240 Shanghai, China.
| |
Collapse
|
40
|
Park S, Park J, Kim E, Lee Y. The Capicua/ETS Translocation Variant 5 Axis Regulates Liver-Resident Memory CD8 + T-Cell Development and the Pathogenesis of Liver Injury. Hepatology 2019; 70:358-371. [PMID: 30810242 DOI: 10.1002/hep.30594] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 02/22/2019] [Indexed: 12/28/2022]
Abstract
Liver-resident memory T (liver TRM ) cells exert protective immune responses following liver infection by malaria parasites. However, how these TRM cells are developed and what the consequence is if they are not properly maintained remain poorly understood. Here, we show that the transcriptional repressor, Capicua (CIC), controls liver CD8+ TRM cell development to maintain normal liver function. Cic-deficient mice have a greater number of liver CD8+ TRM cells and liver injury phenotypes accompanied by increased levels of proinflammatory cytokine genes in liver tissues. Excessive formation of CD69+ CD8+ TRM -like cells was also observed in mice with acetaminophen-induced liver injury (AILI). Moreover, expansion of liver CD8+ TRM cell population and liver injury phenotypes in T-cell-specific Cic null mice were rescued by codeletion of ETS translocation variant [Etv]5 alleles, indicating that Etv5 is a CIC target gene responsible for regulation of CD8+ TRM cell development and liver function. We also discovered that ETV5 directly regulates expression of Hobit, a master transcription factor for TRM cell development, in CD8+ T cells. Conclusion: Our findings suggest the CIC-ETV5 axis as a key molecular module that controls CD8+ TRM cell development, indicating a pathogenic role for CD8+ TRM cells in liver injury.
Collapse
Affiliation(s)
- Sungjun Park
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, Republic of Korea
| | - Jiho Park
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, Republic of Korea
| | - Eunjeong Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, Republic of Korea
| | - Yoontae Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, Republic of Korea.,Division of Integrative Bioscience and Biotechnology, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, Republic of Korea
| |
Collapse
|
41
|
Liu H, Wu Z, Zhou H, Cai W, Li X, Hu J, Gao L, Feng T, Wang L, Peng X, Qi M, Liu L, Han B. The SOX4/miR-17-92/RB1 Axis Promotes Prostate Cancer Progression. Neoplasia 2019; 21:765-776. [PMID: 31238254 PMCID: PMC6593351 DOI: 10.1016/j.neo.2019.05.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 05/20/2019] [Accepted: 05/23/2019] [Indexed: 12/24/2022] Open
Abstract
Although androgen-deprivation treatment (ADT) is the main treatment for advanced prostate cancer (PCa), it eventually fails. This failure invariably leads to castration-resistant prostate cancer (CRPC) and the development of the neuroendocrine (NE) phenotype. The molecular basis for PCa progression remains unclear. Previously, we and others have demonstrated that the sex-determining region Y-box 4 (SOX4) gene, a critical developmental transcription factor, is overexpressed and associated with poor prognosis in PCa patients. In this study, we show that SOX4 expression is associated with PCa progression and the development of the NE phenotype in androgen deprivation conditions. High-throughput microRNA profiling and bioinformatics analyses suggest that SOX4 may target the miR-17-92 cluster. SOX4 transcriptionally upregulates miR-17-92 cluster expression in PCa cells. SOX4-induced PCa cell proliferation, migration, and invasion are also mediated by miR-17-92 cluster members. Furthermore, RB1 is a target gene of miR-17-92 cluster. We found that SOX4 downregulates RB1 protein expression by upregulating the miR-17-92 expression. In addition, SOX4-knockdown restrains NE phenotype and PCa cell proliferation. Clinically, the overexpression of miR-17-92 members is shown to be positively correlated with SOX4 expression in PCa patients, whereas RB1 expression is negatively correlated with SOX4 expression in patients with the aggressive PCa phenotype. Collectively, we propose a novel model of a SOX4/miR-17-92/RB1 axis that may exist to promote PCa progression.
Collapse
Affiliation(s)
- Hui Liu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University, School of Basic Medical Sciences, Jinan, 250012, China
| | - Zhen Wu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University, School of Basic Medical Sciences, Jinan, 250012, China
| | - Haibin Zhou
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University, School of Basic Medical Sciences, Jinan, 250012, China
| | - Wenjie Cai
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University, School of Basic Medical Sciences, Jinan, 250012, China
| | - Xinjun Li
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University, School of Basic Medical Sciences, Jinan, 250012, China
| | - Jing Hu
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University, School of Basic Medical Sciences, Jinan, 250012, China
| | - Lin Gao
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University, School of Basic Medical Sciences, Jinan, 250012, China
| | - Tingting Feng
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University, School of Basic Medical Sciences, Jinan, 250012, China
| | - Lin Wang
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University, School of Basic Medical Sciences, Jinan, 250012, China
| | - Xijia Peng
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University, School of Basic Medical Sciences, Jinan, 250012, China
| | - Mei Qi
- Department of Pathology, Shandong University Qilu Hospital, Jinan, 250012, China
| | - Long Liu
- Department of Pathology, Shandong University Qilu Hospital, Jinan, 250012, China
| | - Bo Han
- The Key Laboratory of Experimental Teratology, Ministry of Education and Department of Pathology, Shandong University, School of Basic Medical Sciences, Jinan, 250012, China; Department of Pathology, Shandong University Qilu Hospital, Jinan, 250012, China.
| |
Collapse
|
42
|
He S, Shi J, Mao J, Luo X, Liu W, Liu R, Yang F. The expression of miR-375 in prostate cancer: A study based on GEO, TCGA data and bioinformatics analysis. Pathol Res Pract 2019; 215:152375. [DOI: 10.1016/j.prp.2019.03.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/04/2019] [Accepted: 03/02/2019] [Indexed: 12/15/2022]
|
43
|
Wang YN, Liu SY, Wang L, Han LY. Long noncoding RNA PCA3 contributes to the progression of choriocarcinoma by acting as a ceRNA against miR-106b. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2019; 12:1609-1617. [PMID: 31933978 PMCID: PMC6947099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 12/13/2018] [Indexed: 06/10/2023]
Abstract
BACKGROUND Choriocarcinoma is the most aggressive gestational trophoblastic disease, with massive local trophoblast invasion and vascular percolation, resulting in multiple organ metastases. Recent evidence has shown that long noncoding RNAs (lncRNAs) play an important role in tumor progression. This study aimed to investigate the expression and role of lncRNA PCA3 in the progression of choriocarcinoma. METHODS First, the expression of lncRNA PCA3 in choriocarcinoma cells was detected using quantitative real-time PCR (qRT-PCR). Then functional assays such as cell proliferation assay, wound healing assay, and invasion assay were conducted to determine the role of PCA3. In addition, the specific molecular mechanism was studied using western blot, luciferase assay, and rescue experiment. RESULTS We demonstrated that the expression of PCA3 is significantly higher in choriocarcinoma cells in contrast to normal human chorionic trophoblast cells. Furthermore, PCA3 could promote cell proliferation, migration and invasion in gestational choriocarcinoma cells and facilitated epithelial to mesenchymal transition (EMT) in vitro. In addition, PAC3 could directly bind to miR-106b and effectively liberate the expression of its endogenous target matrix metallopeptidase 2 (MMP2). CONCLUSION Our results suggest that PCA3 contributes to the progression of choriocarcinoma by acting as a ceRNA against miR-106b.
Collapse
Affiliation(s)
- Yi-Nan Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University Changchun 130041, Jilin Province, China
| | - Shu-Yan Liu
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University Changchun 130041, Jilin Province, China
| | - Ling Wang
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University Changchun 130041, Jilin Province, China
| | - Li-Ying Han
- Department of Obstetrics and Gynecology, The Second Hospital of Jilin University Changchun 130041, Jilin Province, China
| |
Collapse
|
44
|
Wu Y, Hu L, Qin Z, Wang X. MicroRNA‑302a upregulation mediates chemo‑resistance in prostate cancer cells. Mol Med Rep 2019; 19:4433-4440. [PMID: 30942454 DOI: 10.3892/mmr.2019.10098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 12/28/2018] [Indexed: 11/05/2022] Open
Abstract
MicroRNAs (miRNAs) are post‑transcriptional regulators that mediate the initiation and progression of human cancer. Growing evidence suggests that deregulation of miRNA expression levels underlies chemo‑resistance. To investigate whether miRNA‑302a (miR‑302a) is involved in mediating chemo‑resistance to paclitaxel in prostate cancer, a series of in vitro analyses were performed in paclitaxel‑resistant prostate cancer PC‑3PR cells and non‑resistant prostate cancer PC‑3 cells. It was demonstrated that the expression of miR‑302a was upregulated in PC‑3PR cells. Notably, ectopic expression of miR‑302a also increased resistance to paclitaxel in wild‑type PC‑3 cells. By contrast, silencing of miR‑302a in PC‑3PR cells sensitized the cells to paclitaxel. Gene and protein expression analyses suggested that the miR‑302a target gene breast cancer resistance protein (BCRP) may mediate chemo‑resistance to paclitaxel in PC‑3PR cells. In conclusion, the data suggested that elevated miR‑302a levels, in part, mediate sensitivity to paclitaxel in prostate cancer through the aberrant regulation of its downstream targets, AOF2, BCRP and permeability glycoprotein 1. These data have implications for the development of novel therapeutics in prostate cancer that may improve sensitivity to chemotherapeutics.
Collapse
Affiliation(s)
- Yuqi Wu
- Department of Urology, Carson International Cancer Centre, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Centre, Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| | - Li Hu
- Department of Urology, Carson International Cancer Centre, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Centre, Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| | - Zizhen Qin
- Department of Urology, Carson International Cancer Centre, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Centre, Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| | - Xiangwei Wang
- Department of Urology, Carson International Cancer Centre, Shenzhen University General Hospital and Shenzhen University Clinical Medical Academy Centre, Shenzhen University, Shenzhen, Guangdong 518000, P.R. China
| |
Collapse
|
45
|
Pockar S, Globocnik Petrovic M, Peterlin B, Vidovic Valentincic N. MiRNA as biomarker for uveitis - A systematic review of the literature. Gene 2019; 696:162-175. [PMID: 30763668 DOI: 10.1016/j.gene.2019.02.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/13/2019] [Accepted: 02/01/2019] [Indexed: 12/20/2022]
Abstract
AIM A systematic review of miRNA profiling studies in uveitis. METHODS Literature search strategy - Pubmed central central database, using miRNA/microRNA and intraocular inflammation/uveitis as keywords. RESULTS We found twenty publications regarding the experimental and clinical use of miRNA in uveitis, published between 2011 and 2018. CONCLUSION The publications regarding the role of miRNA in uveitis are very scarce, but provide some valuable information about the potential new mechanisms in uveitis. Some of the identified miRNAs in different uveitis entities could serve as a biomarker of intraocular inflammation. Possible candidate miRNAs could be let-7e, miRNA-1, miR-9-3, miR-20a-5p, miR-23a, mir-29a-3p, miR-140-5p, miR-143, miR-146a and miR-146a-5p, miR-155, miR-182 and miR-182-5p, miR-196a2, miR-205, miR-223-3p, miR-301a. MiR-146a, miR-146a-5p, miR-155, miR-182, miR-223-3p, have been found to be possibly associated with uveitis disease in both, human and animal species.
Collapse
Affiliation(s)
- Sasa Pockar
- Eye Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Mojca Globocnik Petrovic
- Eye Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia; Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Borut Peterlin
- Clinical Institute of Medical Genetics, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Natasa Vidovic Valentincic
- Eye Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia; Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia.
| |
Collapse
|
46
|
Yang Y, Jia B, Zhao X, Wang Y, Ye W. miR-93-5p may be an important oncogene in prostate cancer by bioinformatics analysis. J Cell Biochem 2018; 120:10463-10483. [PMID: 30582208 DOI: 10.1002/jcb.28332] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 11/29/2018] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Prostate adenocarcinoma is one of the most prevalent causes of cancer-related deaths in males worldwide. However, the underlying mechanisms remain poorly understood. Hence, it is important to identify specific and effective therapeutic targets, to be able to determine appropriate therapy and management. So, this study aimed to predict that miR-93-5p is an important oncogene in prostate cancer by bioinformatics analysis. METHODS In this study, initially we identified differentially expressed genes (DEGs) and differently expressed miRNAs (DEMs) in the The Cancer Genome Atlas (TCGA) database, performed Gene Ontology (GO) and pathway enrichment analysis, then investigated the relationship between DEGs and DEMs, and finally through consulting the literature and retrieving the database, we found that miR-93-5p may play a major role in prostate cancer, so we predicted the expression and survival of miR-93-5p and its isomers by bioinformatics analysis, meanwhile, evaluated the function of miR-93-5p in vitro. RESULTS In total, 104 DEMs were differently expressed between prostate cancer and normal samples, including 56 downregulated ones and 48 upregulated ones; miR-93-5p (upregulated) was identified as a good biomarker. And 1904 DEGs were retrieved, including 794 downregulated ones and 1110 upregulated ones. We also obtained 1254 DEGs of the DEMs. In GO and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, the significantly enriched pathways involved pathway in focal adhesion, vascular smooth muscle contraction, and regulation of actin cytoskeleton. By the KEGG pathway, we got 16 target genes and 92 pathways of the miR-93-5p in prostate cancer. We also found that the miR-93-5p and its isomers can express in prostate cancer, and which with a high expression had a poor overall survival and a significant difference recurrence rate within 5 years. Further in vitro verification results demonstrated that the low expression of miR-93-5p can inhibit cell proliferation, migration, invasion, change cell cycle, and promote early apoptosis of PC-3 cells. CONCLUSION The miR-93-5p and its target genes were used to define important molecular targets that could serve as a prognostic and predictive marker in the treatment of prostate cancer. Further research on the function of the miR-93-5p and its target genes in the KEGG pathway could provide references for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Yuemei Yang
- Department of R&D Technology Center, Beijing Zhicheng Biomedical Technology Co, Ltd, Beijing, China
| | - Binghan Jia
- Department of R&D Technology Center, Beijing Zhicheng Biomedical Technology Co, Ltd, Beijing, China
| | - Xiaoling Zhao
- Department of R&D Technology Center, Beijing Zhicheng Biomedical Technology Co, Ltd, Beijing, China
| | - Yao Wang
- Department of R&D Technology Center, Beijing Zhicheng Biomedical Technology Co, Ltd, Beijing, China
| | - Weiliang Ye
- Department of R&D Technology Center, Beijing Zhicheng Biomedical Technology Co, Ltd, Beijing, China
| |
Collapse
|
47
|
Yang K, Gao ZY, Li TQ, Song W, Xiao W, Zheng J, Chen H, Chen GH, Zou HY. Anti-tumor activity and the mechanism of a green tea (Camellia sinensis) polysaccharide on prostate cancer. Int J Biol Macromol 2018; 122:95-103. [PMID: 30342140 DOI: 10.1016/j.ijbiomac.2018.10.101] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/22/2018] [Accepted: 10/14/2018] [Indexed: 02/01/2023]
Abstract
In this study, a homogeneous polysaccharide (GTP), with a molecular weight of 7.0 × 104 Da, was isolated from Green tea, which was only composed of glucose. The antitumor effects of GTP on prostate cancer (PC) cell line along with the possible mechanism was examined. First, we investigate the potential role of microRNA-93 (miR-93) in PC progression. Our results showed that miR-93 was significantly upregulated in human PC tissues and several PC cell lines, and its overexpression was correlated with poor survival in PC patients. Furthermore, functional analysis showed that miR-93 overexpression promoted the migration, invasion and proliferation of PC-3 cells transfected with miR-93 mimics, while its knockdown displayed an opposite result in DU145 cells following miR-93 inhibitor transfection. Additionally, in vivo tumorigenic studies on nude mice confirmed that miR-93 mimic treatment accelerated the growth of PC-3 xenograft tumors. As expected, GTP (25, 50 and 100 μg/ml) inhibited growth of PC-3 cells via inducing apoptosis, which was achieved by elevation of bax/bcl-2 ratio and caspae-3 protein expression, as well as a decrease of miR-93. Thus, miR-93 may be a potential therapeutic target by GTP for PC therapy.
Collapse
Affiliation(s)
- Ke Yang
- Department of Urology, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha 410005, P.R. China
| | - Zhi-Yong Gao
- Department of Urology, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha 410005, P.R. China
| | - Tie-Qiu Li
- Department of Urology, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha 410005, P.R. China
| | - Wei Song
- Department of Urology, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha 410005, P.R. China
| | - Wei Xiao
- Department of Urology, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha 410005, P.R. China
| | - Jue Zheng
- Department of Urology, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha 410005, P.R. China
| | - Hao Chen
- Department of Urology, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha 410005, P.R. China
| | - Gui-Heng Chen
- Department of Urology, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha 410005, P.R. China
| | - Hao-Yu Zou
- Department of Surgery, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha 410005, P.R. China.
| |
Collapse
|
48
|
Liu JJ, Zhang X, Wu XH. miR-93 Promotes the Growth and Invasion of Prostate Cancer by Upregulating Its Target Genes TGFBR2, ITGB8, and LATS2. MOLECULAR THERAPY-ONCOLYTICS 2018; 11:14-19. [PMID: 30294667 PMCID: PMC6169426 DOI: 10.1016/j.omto.2018.08.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 08/10/2018] [Indexed: 01/07/2023]
Abstract
This study aimed to evaluate the effects of miR-93 on the growth and invasiveness of prostate cancer (PC) cells (PCCs). Real-time PCR was carried out to detect the expression of miR-93 in the PC tissues and cell lines. The adjacent normal tissues served as controls. For in vitro experiments, methyl thiazolyl tetrazolium, clone formation, tumor cell invasion assays, and western blot analysis (WBA) were performed to confirm the variations in the proliferation and invasiveness of PCCs, prior and subsequent to transfection with an miR-93 antisense oligonucleotide (ASO), which blocks miR-93 binding to its target. Furthermore, the effect of miR-93 on the proliferation of PCCs was examined. Finally, the expression levels of the target genes of miR-93 were determined by WBA. miR-93 expression was higher in PC tissues than in the adjacent normal tissues, and a reduction in the miR-93 level remarkably inhibited the proliferation and invasiveness of PCCs. Moreover, miR-93 enhanced the expression of its target genes TGFΒR2, ITGB8, and LATS2. The results of this study suggest that miR-93 may promote the proliferation and invasion of PCCs by upregulating its target genes TGFBR2, ITGB8, and LATS2.
Collapse
Affiliation(s)
- Jia-Ji Liu
- Department of Urology, Yongchuan Hospital of Chongqing Medical University, Chongqing 402160, China
| | - Xuan Zhang
- Department of Urology, Yongchuan Hospital of Chongqing Medical University, Chongqing 402160, China
| | - Xiao-Hou Wu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400015, China
| |
Collapse
|
49
|
Transcriptomic analysis of CIC and ATXN1L reveal a functional relationship exploited by cancer. Oncogene 2018; 38:273-290. [PMID: 30093628 DOI: 10.1038/s41388-018-0427-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 06/01/2018] [Accepted: 07/02/2018] [Indexed: 12/24/2022]
Abstract
Aberrations in Capicua (CIC) have recently been implicated as a negative prognostic factor in a multitude of cancer types through activation of the MAPK signalling cascade and derepression of oncogenic ETS transcription factors. The Ataxin-family protein ATXN1L has previously been reported to interact with CIC in developmental and disease contexts to facilitate the repression of CIC target genes. To further investigate this relationship, we performed functional in vitro studies utilizing ATXN1LKO and CICKO human cell lines and characterized a reciprocal functional relationship between CIC and ATXN1L. Transcriptomic interrogation of the CIC-ATXN1-ATXN1L axis in low-grade glioma, prostate adenocarcinoma and stomach adenocarcinoma TCGA cohorts revealed context-dependent convergence of gene sets and pathways related to mitotic cell cycle and division. This study highlights the CIC-ATXN1-ATXN1L axis as a more potent regulator of the cell cycle than previously appreciated.
Collapse
|
50
|
Guarnieri AL, Towers CG, Drasin DJ, Oliphant MUJ, Andrysik Z, Hotz TJ, Vartuli RL, Linklater ES, Pandey A, Khanal S, Espinosa JM, Ford HL. The miR-106b-25 cluster mediates breast tumor initiation through activation of NOTCH1 via direct repression of NEDD4L. Oncogene 2018; 37:3879-3893. [PMID: 29662198 PMCID: PMC6043359 DOI: 10.1038/s41388-018-0239-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 02/01/2018] [Accepted: 03/03/2018] [Indexed: 01/07/2023]
Abstract
Tumor-initiating cells (TIC) represent a subset of tumor cells with increased self-renewal capability. TICs display resistance to frontline cancer treatment and retain the ability to repopulate a tumor after therapy, leading to cancer relapse. NOTCH signaling has been identified as an important driver of the TIC population, yet mechanisms governing regulation of this pathway in cancer remain to be fully elucidated. Here we identify a novel mechanism of NOTCH regulation and TIC induction in breast cancer via the miR-106b-25 miRNA cluster. We show that the miR-106b-25 cluster upregulates NOTCH1 in multiple breast cancer cell lines, representing both estrogen receptor (ER+) and triple negative breast cancer (TNBC) through direct repression of the E3 ubiquitin ligase, NEDD4L. We further show that upregulation of NOTCH1 is necessary for TIC induction downstream of miR-106b-25 in both ER + and TNBC breast cancer cells, and that re-expression of NEDD4L is sufficient to reverse miR106b-25-mediated NOTCH1 upregulation and TIC induction. Importantly, we demonstrate a significant positive correlation between miR-106b-25 and NOTCH1 protein, yet a significant inverse correlation between miR-106b-25 and NEDD4L mRNA in human breast cancer, suggesting a critical role for the miR106b-25/NEDD4L/NOTCH1 axis in the disease. Further, we show for the first time that NEDD4L expression alone is significantly associated with a better relapse-free prognosis for breast cancer patients. These data expand our knowledge of the mechanisms underlying NOTCH activation and TIC induction in breast cancer, and may provide new avenues for the development of therapies targeting this resistant subset of tumor cells.
Collapse
Affiliation(s)
- A L Guarnieri
- Program in Molecular Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - C G Towers
- Program in Molecular Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - D J Drasin
- Program in Molecular Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - M U J Oliphant
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Integrated Physiology Program, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Z Andrysik
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - T J Hotz
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - R L Vartuli
- Program in Molecular Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - E S Linklater
- Program in Molecular Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - A Pandey
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - S Khanal
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - J M Espinosa
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Linda Crnic Institute for Down Syndrome, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Molecular, Cellular and Developmental Biology, University of Colorado at Boulder, Boulder, CO, 80309, USA
| | - H L Ford
- Program in Molecular Biology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
- Integrated Physiology Program, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|