1
|
Qiu W, Zhang S, Yu W, Liu J, Wu H. Non-coding RNAs in hepatocellular carcinoma metastasis: Remarkable indicators and potential oncogenic mechanism. Comput Biol Med 2024; 180:108867. [PMID: 39089114 DOI: 10.1016/j.compbiomed.2024.108867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 06/12/2024] [Accepted: 07/07/2024] [Indexed: 08/03/2024]
Abstract
Non-coding RNAs (ncRNAs), as key regulators involving in intercellular biological processes, are more prominent in many malignancies, especially for hepatocellular carcinoma (HCC). Herein, we conduct a comprehensive review to summarize diverse ncRNAs roles in HCC metastatic mechanism. We focus on four signaling pathways that predominate in HCC metastatic process, including Wnt/β-catenin, HIF-1α, IL-6, and TGF-β pathways. MicroRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs) employed different mechanisms to participate in the regulation of the key genes in these pathways, typical as interaction with DNA to control transcription, with RNA to control translation, and with protein to control stability. Therefore, ncRNAs may become potential biomarkers and therapeutic targets for HCC metastasis.
Collapse
Affiliation(s)
- Wenqi Qiu
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Song Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Yu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jian Liu
- Department of Intensive Care Unit, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huiling Wu
- Department of Plastic and Aesthetic Center, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Wu HH, Leng S, Sergi C, Leng R. How MicroRNAs Command the Battle against Cancer. Int J Mol Sci 2024; 25:5865. [PMID: 38892054 PMCID: PMC11172831 DOI: 10.3390/ijms25115865] [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: 04/24/2024] [Revised: 05/23/2024] [Accepted: 05/26/2024] [Indexed: 06/21/2024] Open
Abstract
MicroRNAs (miRNAs) are small RNA molecules that regulate more than 30% of genes in humans. Recent studies have revealed that miRNAs play a crucial role in tumorigenesis. Large sets of miRNAs in human tumors are under-expressed compared to normal tissues. Furthermore, experiments have shown that interference with miRNA processing enhances tumorigenesis. Multiple studies have documented the causal role of miRNAs in cancer, and miRNA-based anticancer therapies are currently being developed. This review primarily focuses on two key points: (1) miRNAs and their role in human cancer and (2) the regulation of tumor suppressors by miRNAs. The review discusses (a) the regulation of the tumor suppressor p53 by miRNA, (b) the critical role of the miR-144/451 cluster in regulating the Itch-p63-Ago2 pathway, and (c) the regulation of PTEN by miRNAs. Future research and the perspectives of miRNA in cancer are also discussed. Understanding these pathways will open avenues for therapeutic interventions targeting miRNA regulation.
Collapse
Affiliation(s)
- Hong Helena Wu
- 370 Heritage Medical Research Center, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2S2, Canada;
| | - Sarah Leng
- Department of Laboratory Medicine and Pathology (5B4. 09), University of Alberta, Edmonton, AB T6G 2B7, Canada (C.S.)
| | - Consolato Sergi
- Department of Laboratory Medicine and Pathology (5B4. 09), University of Alberta, Edmonton, AB T6G 2B7, Canada (C.S.)
- Division of Anatomical Pathology, Children’s Hospital of Eastern Ontario (CHEO), University of Ottawa, 401 Smyth Road, Ottawa, ON K1H 8L1, Canada
| | - Roger Leng
- 370 Heritage Medical Research Center, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G 2S2, Canada;
| |
Collapse
|
3
|
García-Andrade F, Vigueras-Villaseñor RM, Chávez-Saldaña MD, Rojas-Castañeda JC, Bahena-Ocampo IU, Aréchaga-Ocampo E, Flores-Fortis M, Díaz-Chávez J, Herrera LA, Landero-Huerta DA. Molecular Characterization of Patients with Cryptorchidism: Preliminary Search for an Expression Profile Related to That of Testicular Germ-Cell Tumors. Diagnostics (Basel) 2023; 13:3020. [PMID: 37761387 PMCID: PMC10529510 DOI: 10.3390/diagnostics13183020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/30/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
Cryptorchidism (CO) is a risk factor for the development of testicular germ-cell tumors (TGCT). This is supported by reports showing the persistence of gonocytes in CO patients. These cells are proposed to be related to the development of germ-cell neoplasia in situ (GCNIS), which is considered the precursor stage/lesion of TGCT. Therefore, it is proposed that some patients with CO could express some molecular markers related to TGCT. In this study, we analyzed testicular tissue samples from CO, TGCT, and controls. We determined the expression of POU5F1, PLAP, and KIT by immunohistochemistry and that of the hsa-miR-371-373 cluster, hsa-miR-367, and LATS2, PTEN, and IGFR1 genes by RT-qPCR. We then carried out a bioinformatic analysis to identify other possible candidate genes as tumor biomarkers. We found that 16.7% (2/12) of the CO patients presented increased expression of POU5F1, KIT, PLAP, hsa-miR-371-373, and hsa-miR-367 and decreased expression of LATS2 and IGF1R. Finally, the genes ARID4B, GALNT3, and KPNA6 were identified as other possible candidate tumor biomarkers. This is the first report describing the expression of the hsa-miR-371-373 cluster, hsa-miR-367, LATS2, and IGF1R in the testicular tissues of two CO patients with cells immune-positive to POU5F1, PLAP, and KIT, which is similar to what is observed in TGCT.
Collapse
Affiliation(s)
- Fabiola García-Andrade
- Laboratorio de Biología de la Reproducción, Instituto Nacional de Pediatría, Ciudad de Mexico 04530, Mexico
- Posgrado en Biología Experimental, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Ciudad de Mexico 09310, Mexico
| | | | | | | | - Ivan Uriel Bahena-Ocampo
- Departamento de Ciencias de la Salud, Universidad Autónoma Metropolitana, Unidad Iztapalapa, Ciudad de Mexico 09310, Mexico
| | - Elena Aréchaga-Ocampo
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana, Unidad Cuajimalpa, Ciudad de Mexico 05348, Mexico
| | - Mauricio Flores-Fortis
- Posgrado en Ciencias Naturales e Ingeniería, Universidad Autónoma Metropolitana, Unidad Cuajimalpa, Ciudad de Mexico 05348, Mexico
| | - José Díaz-Chávez
- Unidad de Investigación en Cáncer, Instituto de Investigaciones Biomédicas-Universidad Nacional Autónoma de México, Instituto Nacional de Cancerología, Ciudad de Mexico 14080, Mexico
| | - Luis Alonso Herrera
- Unidad de Investigación en Cáncer, Instituto de Investigaciones Biomédicas-Universidad Nacional Autónoma de México, Instituto Nacional de Cancerología, Ciudad de Mexico 14080, Mexico
- Escuela de Medicina y Ciencias de la Salud-Tecnológico de Monterrey, Ciudad de Mexico 14380, Mexico
| | | |
Collapse
|
4
|
Marroncini G, Anceschi C, Naldi L, Fibbi B, Baldanzi F, Maggi M, Peri A. The V 2 receptor antagonist tolvaptan counteracts proliferation and invasivity in human cancer cells. J Endocrinol Invest 2022; 45:1693-1708. [PMID: 35604542 PMCID: PMC9360171 DOI: 10.1007/s40618-022-01807-5] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 04/20/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE Hyponatremia, the most frequent electrolyte alteration in clinical practice, has been associated with a worse prognosis in cancer patients. On the other hand, a better outcome has been related to serum sodium normalization. In vitro studies have shown that low extracellular sodium promotes cancer cells proliferation and invasiveness. Tolvaptan, a selective vasopressin receptor type 2 (V2) antagonist, has been effectively used in the last decade for the treatment of hyponatremia secondary to the Syndrome of Inappropriate Antidiuresis. A few in vitro data suggested a direct role of tolvaptan in counteracting cancer progression, so far. The aim of this study was to evaluate the effect and the mechanism of action of tolvaptan in cell lines from different tumours [i.e. colon cancer (HCT-8), hepatocarcinoma (HepG2), neuroblastoma (SK-N-AS)]. METHODS AND RESULTS First, we showed that these cell lines express the V2 receptor. Tolvaptan significantly reduced cell proliferation with an IC50 in the micromolar range. Accordingly, reduced levels of cAMP, of the catalytic α subunit of PKA, and a reduced pAKT/AKT ratio were observed. Tolvaptan effectively inhibited cell cycle progression, whereas it induced apoptotis. Furthermore, it reduced cell invasiveness. In particular, anchorage-independent growth and the activity of collagenases type IV were blunted in the three cell lines. Accordingly, tolvaptan counteracted the RhoA/ROCK1-2 pathway, which has a pivotal role in regulating cell movement. CONCLUSIONS Overall, these findings indicate that tolvaptan effectively inhibits tumour progression in vitro. Further studies should clarify whether the V2 receptor might be considered a possible target in anti-cancer strategies in the future.
Collapse
Affiliation(s)
- G Marroncini
- Pituitary Diseases and Sodium Alterations Unit, AOU Careggi, 50139, Florence, Italy
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, AOU Careggi, Viale Pieraccini, 6, 50139, Florence, Italy
| | - C Anceschi
- Pituitary Diseases and Sodium Alterations Unit, AOU Careggi, 50139, Florence, Italy
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, AOU Careggi, Viale Pieraccini, 6, 50139, Florence, Italy
| | - L Naldi
- Pituitary Diseases and Sodium Alterations Unit, AOU Careggi, 50139, Florence, Italy
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, AOU Careggi, Viale Pieraccini, 6, 50139, Florence, Italy
| | - B Fibbi
- Pituitary Diseases and Sodium Alterations Unit, AOU Careggi, 50139, Florence, Italy
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, AOU Careggi, Viale Pieraccini, 6, 50139, Florence, Italy
| | - F Baldanzi
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, AOU Careggi, Viale Pieraccini, 6, 50139, Florence, Italy
| | - M Maggi
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, AOU Careggi, Viale Pieraccini, 6, 50139, Florence, Italy
| | - A Peri
- Pituitary Diseases and Sodium Alterations Unit, AOU Careggi, 50139, Florence, Italy.
- Endocrinology, Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, AOU Careggi, Viale Pieraccini, 6, 50139, Florence, Italy.
| |
Collapse
|
5
|
Pavani KC, Meese T, Pascottini OB, Guan X, Lin X, Peelman L, Hamacher J, Van Nieuwerburgh F, Deforce D, Boel A, Heindryckx B, Tilleman K, Van Soom A, Gadella BM, Hendrix A, Smits K. Hatching is modulated by microRNA-378a-3p derived from extracellular vesicles secreted by blastocysts. Proc Natl Acad Sci U S A 2022; 119:e2122708119. [PMID: 35298333 PMCID: PMC8944274 DOI: 10.1073/pnas.2122708119] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 02/04/2022] [Indexed: 12/17/2022] Open
Abstract
SignificanceHatching from the zona pellucida is a prerequisite for embryo implantation and is less likely to occur in vitro for reasons unknown. Extracellular vesicles (EVs) are secreted by the embryo into the culture medium. Yet the role that embryonic EVs and their cargo microRNAs (miRNAs) play in blastocyst hatching has not been elucidated, partially due to the difficulties of isolating them from low amounts of culture medium. Here, we optimized EV-miRNA isolation from medium conditioned by individually cultured bovine embryos and subsequently showed that miR-378a-3p, which was up-regulated in EVs secreted by blastocysts, plays a crucial role in promoting blastocyst hatching. This demonstrates the regulatory effect of miR-378-3p on hatching, which is an established embryo quality parameter linked with implantation.
Collapse
Affiliation(s)
- Krishna Chaitanya Pavani
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, University of Ghent, B-9820 Merelbeke, Belgium
- Department for Reproductive Medicine, Ghent University Hospital, 9000 Gent, Belgium
| | - Tim Meese
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Ghent University, B-9000 Ghent, Belgium
| | - Osvaldo Bogado Pascottini
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, University of Ghent, B-9820 Merelbeke, Belgium
- Department of Veterinary Sciences, Gamete Research Center, University of Antwerp, 2610 Antwerp, Belgium
| | - XueFeng Guan
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, B-9000 Ghent, Belgium
| | - Xiaoyuan Lin
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, B-9000 Ghent, Belgium
| | - Luc Peelman
- Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, B-9000 Ghent, Belgium
| | - Joachim Hamacher
- Institute of Crop Science and Resource Conservation, Plant Pathology, Rheinische Friedrich-Wilhelms-University of Bonn, D-53115 Bonn, Germany
| | - Filip Van Nieuwerburgh
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Ghent University, B-9000 Ghent, Belgium
| | - Dieter Deforce
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Ghent University, B-9000 Ghent, Belgium
| | - Annekatrien Boel
- Ghent-Fertility and Stem Cell Team, Department for Reproductive Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Björn Heindryckx
- Ghent-Fertility and Stem Cell Team, Department for Reproductive Medicine, Ghent University Hospital, 9000 Ghent, Belgium
| | - Kelly Tilleman
- Department for Reproductive Medicine, Ghent University Hospital, 9000 Gent, Belgium
| | - Ann Van Soom
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, University of Ghent, B-9820 Merelbeke, Belgium
| | - Bart M. Gadella
- Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CM Utrecht, The Netherlands
| | - An Hendrix
- Laboratory of Experimental Cancer Research, Department of Human Structure and Repair, Ghent University, B-9000 Ghent, Belgium
- Cancer Research Institute Ghent, B-9000 Ghent, Belgium
| | - Katrien Smits
- Department of Reproduction, Obstetrics and Herd Health, Faculty of Veterinary Medicine, University of Ghent, B-9820 Merelbeke, Belgium
| |
Collapse
|
6
|
Zuo J, Zhang Z, Li M, Yang Y, Zheng B, Wang P, Huang C, Zhou S. The crosstalk between reactive oxygen species and noncoding RNAs: from cancer code to drug role. Mol Cancer 2022; 21:30. [PMID: 35081965 PMCID: PMC8790843 DOI: 10.1186/s12943-021-01488-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/26/2021] [Indexed: 02/08/2023] Open
Abstract
Oxidative stress (OS), characterized by the excessive accumulation of reactive oxygen species (ROS), is an emerging hallmark of cancer. Tumorigenesis and development driven by ROS require an aberrant redox homeostasis, that activates onco-signaling and avoids ROS-induced programmed death by orchestrating antioxidant systems. These processes are revealed to closely associate with noncoding RNAs (ncRNAs). On the basis of the available evidence, ncRNAs have been widely identified as multifarious modulators with the involvement of several key redox sensing pathways, such as NF-κB and Nrf2 signaling, therefore potentially becoming effective targets for cancer therapy. Furthermore, the vast majority of ncRNAs with property of easy detected in fluid samples (e.g., blood and urine) facilitate clinicians to monitor redox homeostasis, indicating a novel method for cancer diagnosis. Herein, focusing on carcinoma initiation, metastasis and chemoradiotherapy resistance, we aimed to discuss the ncRNAs-ROS network involved in cancer progression, and the potential clinical application as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Jing Zuo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China
| | - Maomao Li
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, People's Republic of China
| | - Yun Yang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, People's Republic of China
| | - Bohao Zheng
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, People's Republic of China
| | - Ping Wang
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, People's Republic of China.
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, People's Republic of China.
| | - Shengtao Zhou
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE and State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, People's Republic of China.
| |
Collapse
|
7
|
Alvarado-Flores F, Kaneko-Tarui T, Beyer W, Katz J, Chu T, Catalano P, Sadovsky Y, Hivert MF, O’Tierney-Ginn P. Placental miR-3940-3p Is Associated With Maternal Insulin Resistance in Late Pregnancy. J Clin Endocrinol Metab 2021; 106:3526-3535. [PMID: 34333643 PMCID: PMC8787748 DOI: 10.1210/clinem/dgab571] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Indexed: 02/07/2023]
Abstract
CONTEXT An increase in maternal insulin resistance (IR) during pregnancy is essential for normal fetal growth. The mechanisms underlying this adaptation are poorly understood. Placental factors are believed to instigate and maintain these changes, as IR decreases shortly after delivery. Methylation of placental gene loci that are common targets for miRNAs are associated with maternal IR. OBJECTIVE We hypothesized that placental miRNAs targeting methylated loci are associated with maternal IR during late pregnancy. METHODS We collected placentas from 132 elective cesarean sections and fasting blood samples at delivery to estimate maternal homeostasis model assessment of insulin resistance (HOMA-IR). Placental miRNA expression was measured via whole genome small-RNA sequencing in a subset of 40 placentas selected by maternal pre-gravid body mass index (BMI) and neonatal adiposity. Five miRNAs correlated with maternal HOMA-IR and previously identified as targeting methylated genes were selected for validation in all 132 placenta samples via RT-qPCR. Multiple regression adjusted for relevant clinical variables. RESULTS Median maternal age was 27.5 years, with median pre-pregnancy BMI of 24.7 kg/m2, and median HOMA-IR of 2.9. Among the 5 selected miRNA, maternal HOMA-IR correlated with the placental expression of miRNA-371b-3p (r = 0.25; P = 0.008) and miRNA-3940-3p (r = 0.32; P = 0.0004) across the 132 individuals. After adjustment for confounding variables, placental miRNA-3940-3p expression remained significantly associated with HOMA-IR (β = 0.16; P = 0.03). CONCLUSION Placental miRNA-3940-3p was associated with maternal IR at delivery. This placental miRNA may have an autocrine or paracrine effect-regulating placental genes involved in modulating maternal IR.
Collapse
Affiliation(s)
| | - Tomoko Kaneko-Tarui
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - William Beyer
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Jacqueline Katz
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Tianjiao Chu
- Magee Womens Research Institute, Pittsburgh, PA 15213, USA
| | - Patrick Catalano
- Mother Infant Research Institute, Tufts Medical Center, Boston, MA 02111, USA
| | - Yoel Sadovsky
- Magee Womens Research Institute, Pittsburgh, PA 15213, USA
| | - Marie-France Hivert
- Department of Population Medicine, Harvard Medical School; Harvard Pilgrim Health Care Institute, Boston, MA 02115, USA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | | |
Collapse
|
8
|
Zhao L, Yang Q, Liu J. Clinical Value Evaluation of microRNA-324-3p and Other Available Biomarkers in Patients With HBV Infection-Related Hepatocellular Carcinoma. Open Forum Infect Dis 2021; 8:ofab108. [PMID: 34189151 PMCID: PMC8232384 DOI: 10.1093/ofid/ofab108] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
Background Patients with hepatitis B virus (HBV) infection are at high risk of hepatocellular carcinoma (HCC). This study aimed to evaluate the expression of microRNA-324-3p (miR-324-3p) in HBV-related HCC and explore the clinical significance of serum miR-324-3p and other available biomarkers in the diagnosis and prognosis of HBV-related HCC. Methods Expression of miR-324-3p in HBV infection–related cells and patients was estimated using quantitative real-time polymerase chain reaction (PCR). Receiver operating characteristic (ROC) curves were constructed to evaluate the diagnostic performance of serum miR-324-3p, alpha-fetoprotein (AFP), and protein induced by vitamin K absence/antagonist II (PIVKA-II) in the differentiation of HBV-related HCC from healthy controls and chronic hepatitis B patients (CHB). The relationship between serum miR-324-3p and patients’ clinical features was assessed using the chi-square test, and the value of miR-324-3p to predict overall survival prognosis was evaluated using Kaplan-Meier methods and Cox regression assay in patients with HBV-related HCC. Results HBV-related HCC cells had significantly increased miR-324-3p compared with normal and HBV-unrelated HCC cells, and serum miR-324-3p in HCC patients with HBV infection was also higher than that in healthy controls and CHB. Serum miR-324-3p had relatively high diagnostic accuracy for the screening of HCC cases with HBV infection, and the combination of miR-324-3p, AFP, and PIVKA-II showed improved diagnostic performance. Additionally, high-serum miR-324-2p in HBV-related HCC patients was associated with cirrhosis, tumor size, clinical stage, and poor overall survival prognosis. Conclusions High-serum miR-324-3p may be involved in the progression of HBV-related hepatitis to HCC and may serve as a candidate biomarker for the diagnosis and prognosis of HBV-related HCC.
Collapse
Affiliation(s)
- Li Zhao
- Department of Infectious Diseases, Yidu Central Hospital of Weifang, Qingzhou, Shandong, China
| | - Qian Yang
- Department of Infectious Diseases, Yidu Central Hospital of Weifang, Qingzhou, Shandong, China
| | - Jianbo Liu
- Public Health Division, Yidu Central Hospital of Weifang, Qingzhou, Shandong, China
| |
Collapse
|
9
|
Ghafouri-Fard S, Honarmand Tamizkar K, Hussen BM, Taheri M. MicroRNA signature in liver cancer. Pathol Res Pract 2021; 219:153369. [PMID: 33626406 DOI: 10.1016/j.prp.2021.153369] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/31/2021] [Accepted: 02/02/2021] [Indexed: 12/24/2022]
Abstract
Liver cancer is the 7th utmost frequent neoplasm and the 4th principal source of cancer deaths. This malignancy is linked with several environmental and lifestyle-related factors emphasizing the role of epigenetics in its pathogenesis. MicroRNAs (miRNAs) have been regarded as potent epigenetic mechanisms partaking in the pathogenesis of liver cancer. Dysregulation of miRNAs has been related with poor outcome of patients with liver cancer. In the current manuscript, we provide a concise review of the results of recent studies about the role of miRNAs in the progression of liver cancer and their diagnostic and prognostic utility.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Bashdar Mahmud Hussen
- Pharmacognosy Department, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Weldy CS, Syed SA, Amsallem M, Hu DQ, Ji X, Punn R, Taylor A, Navarre B, Reddy S. Circulating whole genome miRNA expression corresponds to progressive right ventricle enlargement and systolic dysfunction in adults with tetralogy of Fallot. PLoS One 2020; 15:e0241476. [PMID: 33175850 PMCID: PMC7657553 DOI: 10.1371/journal.pone.0241476] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 10/15/2020] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION The adult congenital heart disease population with repaired tetralogy of Fallot (TOF) is subject to chronic volume and pressure loading leading to a 40% probability of right ventricular (RV) failure by the 3rd decade of life. We sought to identify a non-invasive signature of adverse RV remodeling using peripheral blood microRNA (miRNA) profiling to better understand the mechanisms of RV failure. METHODS Demographic, clinical data, and blood samples were collected from adults with repaired TOF (N = 20). RNA was isolated from the buffy coat of peripheral blood and whole genome miRNA expression was profiled using Agilent's global miRNA microarray platform. Fold change, pathway analysis, and unbiased hierarchical clustering of miRNA expression was performed and correlated to RV size and function assessed by echocardiography performed at or near the time of blood collection. RESULTS MiRNA expression was profiled in the following groups: 1. normal RV size (N = 4), 2. mild/moderate RV enlargement (N = 11) and 3. severe RV enlargement (N = 5). 267 miRNAs were downregulated, and 66 were upregulated across the three groups (fold change >2.0, FDR corrected p<0.05) as RV enlargement increased and systolic function decreased. qPCR validation of a subset of these miRNAs identified increasing expression of miRNA 28-3p, 433-3p, and 371b-3p to be associated with increasing RV size and decreasing RV systolic function. Unbiased hierarchical clustering of all patients based on miRNA expression demonstrates three distinct patient clusters that largely coincide with progressive RV enlargement. Pathway analysis of dysregulated miRNAs demonstrates up and downregulation of cell cycle pathways, extracellular matrix proteins and fatty acid synthesis. HIF 1α signaling was downregulated while p53 signaling was predicted to be upregulated. CONCLUSION Adults with TOF have a distinct miRNA profile with progressive RV enlargement and dysfunction implicating cell cycle dysregulation and upregulation in extracellular matrix and fatty acid metabolism. These data suggest peripheral blood miRNA can provide insight into the mechanisms of RV failure and can potentially be used for monitoring disease progression and to develop RV specific therapeutics to prevent RV failure in TOF.
Collapse
Affiliation(s)
- Chad S. Weldy
- Division of Cardiology, Department of Medicine, Stanford University, Stanford, California, United States of America
| | - Saad Ali Syed
- Stanford University School of Medicine, Stanford, California, United States of America
| | - Myriam Amsallem
- Division of Cardiology, Department of Medicine, Stanford University, Stanford, California, United States of America
- Division of Cardiology, Department of Pediatrics, Stanford University, Stanford, California, United States of America
| | - Dong-Qing Hu
- Division of Cardiology, Department of Pediatrics, Stanford University, Stanford, California, United States of America
| | - Xuhuai Ji
- Human Immune Monitoring Center and Functional Genomics Facility, Stanford University, Stanford, California, United States of America
| | - Rajesh Punn
- Division of Cardiology, Department of Pediatrics, Stanford University, Stanford, California, United States of America
| | - Anne Taylor
- Division of Cardiology, Department of Pediatrics, Stanford University, Stanford, California, United States of America
| | - Brittany Navarre
- Division of Cardiology, Department of Pediatrics, Stanford University, Stanford, California, United States of America
| | - Sushma Reddy
- Division of Cardiology, Department of Pediatrics, Stanford University, Stanford, California, United States of America
| |
Collapse
|
11
|
Ghafouri-Fard S, Abak A, Shoorei H, Mohaqiq M, Majidpoor J, Sayad A, Taheri M. Regulatory role of microRNAs on PTEN signaling. Biomed Pharmacother 2020; 133:110986. [PMID: 33166764 DOI: 10.1016/j.biopha.2020.110986] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 10/28/2020] [Accepted: 11/03/2020] [Indexed: 12/13/2022] Open
Abstract
Phosphatase and tensin homolog (PTEN) gene encodes a tumor suppressor protein which is altered in several malignancies. This protein is a negative regulator of the PI3K/AKT signaling. Several transcription factors regulate the expression of PTEN in positive or negative directions. Moreover, numerous microRNAs (miRNAs) have functional interactions with PTEN and inhibit its expression. Suppression of PTEN can attenuate the response of cancer cells to chemotherapeutic agents. Based on the critical role of this tumor suppressor gene, the identification of negative regulators of its expression has practical significance particularly in the prevention and management of cancer. Meanwhile, the interaction between miRNAs and PTEN has functional consequences in non-malignant disorders including myocardial infarction, osteoporosis, cerebral ischemic stroke, and recurrent abortion. In the present review, we describe the role of miRNAs in the regulation of expression and activity of PTEN.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atefe Abak
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mahdi Mohaqiq
- Wake Forest Institute for Regenerative Medicine, School of Medicine, Wake Forest University, Winston-Salem, NC, USA
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Arezou Sayad
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|