1
|
Chang FC, James MM, Zhou Y, Ando Y, Zareie HM, Yang J, Zhang M. Human Neural Stem Cell Expansion in Natural Polymer Scaffolds Under Chemically Defined Condition. Adv Biol (Weinh) 2024; 8:e2400224. [PMID: 38963310 DOI: 10.1002/adbi.202400224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/06/2024] [Indexed: 07/05/2024]
Abstract
The maintenance and expansion of human neural stem cells (hNSCs) in 3D tissue scaffolds is a promising strategy in producing cost-effective hNSCs with quality and quantity applicable for clinical applications. A few biopolymers have been extensively used to fabricate 3D scaffolds, including hyaluronic acid, collagen, alginate, and chitosan, due to their bioactive nature and availability. However, these polymers are usually applied in combination with other biomolecules, leading to their responses difficult to ascribe to. Here, scaffolds made of chitosan, alginate, hyaluronic acid, or collagen, are explored for hNSC expansion under xeno-free and chemically defined conditions and compared for hNSC multipotency maintenance. This study shows that the scaffolds made of pure chitosan support the highest adhesion and growth of hNSCs, yielding the most viable cells with NSC marker protein expression. In contrast, the presence of alginate, hyaluronic acid, or collagen induces differentiation toward immature neurons and astrocytes even in the maintenance medium and absence of differentiation factors. The cells in pure chitosan scaffolds preserve the level of transmembrane protein profile similar to that of standard culture. These findings point to the potential of using pure chitosan scaffolds as a base scaffolding material for hNSC expansion in 3D.
Collapse
Affiliation(s)
- Fei-Chien Chang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Matthew Michael James
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Yang Zhou
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Yoshiki Ando
- Materials Department, Medical R&D Center, Corporate R&D Group, KYOCERA Corporation, Yasu, Shiga, 520-2362, Japan
| | - Hadi M Zareie
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Jihui Yang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
2
|
Bahram Sangani N, Koetsier J, Gomes AR, Diogo MM, Fernandes TG, Bouwman FG, Mariman ECM, Ghazvini M, Gribnau J, Curfs LMG, Reutelingsperger CP, Eijssen LMT. Involvement of extracellular vesicle microRNA clusters in developing healthy and Rett syndrome brain organoids. Cell Mol Life Sci 2024; 81:410. [PMID: 39305343 PMCID: PMC11416455 DOI: 10.1007/s00018-024-05409-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/19/2024] [Accepted: 08/10/2024] [Indexed: 09/25/2024]
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder caused by de novo mutations in the MECP2 gene. Although miRNAs in extracellular vesicles (EVs) have been suggested to play an essential role in several neurological conditions, no prior study has utilized brain organoids to profile EV-derived miRNAs during normal and RTT-affected neuronal development. Here we report the spatiotemporal expression pattern of EV-derived miRNAs in region-specific forebrain organoids generated from female hiPSCs with a MeCP2:R255X mutation and the corresponding isogenic control. EV miRNA and protein expression profiles were characterized at day 0, day 13, day 40, and day 75. Several members of the hsa-miR-302/367 cluster were identified as having a time-dependent expression profile with RTT-specific alterations at the latest developmental stage. Moreover, the miRNA species of the chromosome 14 miRNA cluster (C14MC) exhibited strong upregulation in RTT forebrain organoids irrespective of their spatiotemporal location. Together, our results suggest essential roles of the C14MC and hsa-miR-302/367 clusters in EVs during normal and RTT-associated neurodevelopment, displaying promising prospects as biomarkers for monitoring RTT progression.
Collapse
Affiliation(s)
- Nasim Bahram Sangani
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, 6200, MD, The Netherlands
- GKC, Maastricht University Medical Centre, Maastricht, 6229, ER, The Netherlands
| | - Jarno Koetsier
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, 6200, MD, The Netherlands
- GKC, Maastricht University Medical Centre, Maastricht, 6229, ER, The Netherlands
| | - Ana Rita Gomes
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Margarida Diogo
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Tiago G Fernandes
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Freek G Bouwman
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Edwin C M Mariman
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Mehrnaz Ghazvini
- Erasmus MC iPS Facility, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Joost Gribnau
- Erasmus MC iPS Facility, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
- Department of Developmental Biology, Erasmus Medical Center, University Medical Center, Rotterdam, Netherlands
| | - Leopold M G Curfs
- GKC, Maastricht University Medical Centre, Maastricht, 6229, ER, The Netherlands
| | - Chris P Reutelingsperger
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, 6200, MD, The Netherlands.
- GKC, Maastricht University Medical Centre, Maastricht, 6229, ER, The Netherlands.
| | - Lars M T Eijssen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience (MHeNs), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, 6200, MD, The Netherlands
- Department of Bioinformatics-BiGCaT, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Faculty of Health, Medicine and Life Sciences (FHML), Maastricht University, Maastricht, 6200, MD, The Netherlands
| |
Collapse
|
3
|
Saadh MJ, Hussain QM, Alazzawi TS, Fahdil AA, Athab ZH, Yarmukhamedov B, Al-Nuaimi AMA, Alsaikhan F, Farhood B. MicroRNA as Key Players in Hepatocellular Carcinoma: Insights into Their Role in Metastasis. Biochem Genet 2024:10.1007/s10528-024-10897-0. [PMID: 39103713 DOI: 10.1007/s10528-024-10897-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
Liver cancer or hepatocellular carcinoma (HCC) remains the most common cancer in global epidemiology. Both the frequency and fatality of this malignancy have shown an upward trend over recent decades. Liver cancer is a significant concern due to its propensity for both intrahepatic and extrahepatic metastasis. Liver cancer metastasis is a multifaceted process characterized by cell detachment from the bulk tumor, modulation of cellular motility and invasiveness, enhanced proliferation, avoidance of the immune system, and spread either via lymphatic or blood vessels. MicroRNAs (miRNAs) are small non-coding ribonucleic acids (RNAs) playing a crucial function in the intricate mechanisms of tumor metastasis. A number of miRNAs can either increase or reduce metastasis via several mechanisms, such as control of motility, proliferation, attack by the immune system, cancer stem cell properties, altering the microenvironment, and the epithelial-mesenchymal transition (EMT). Besides, two other types of non-coding RNAs, such as long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) can competitively bind to endogenous miRNAs. This competition results in the impaired ability of the miRNAs to inhibit the expression of the specific messenger RNAs (mRNAs) that are targeted. Increasing evidence has shown that the regulatory axis comprising circRNA/lncRNA-miRNA-mRNA is correlated with the regulation of HCC metastasis. This review seeks to present a thorough summary of recent research on miRNAs in HCC, and their roles in the cellular processes of EMT, invasion and migration, as well as the metastasis of malignant cells. Finally, we discuss the function of the lncRNA/circRNA-miRNA-mRNA network as a crucial modulator of carcinogenesis and the regulation of signaling pathways or genes that are relevant to the metastasis of HCC. These findings have the potential to offer valuable insight into the discovery of novel therapeutic approaches for management of liver cancer metastasis.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | | | - Tuqa S Alazzawi
- College of Dentist, National University of Science and Technology, Nasiriyah, Dhi Qar, Iraq
| | - Ali A Fahdil
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Bekhzod Yarmukhamedov
- Department of Public Health and Healthcare management, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, Uzbekistan
| | | | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia.
- School of Pharmacy, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
4
|
Li Y, Fang B. Neural stem cell-derived extracellular vesicles: The light of central nervous system diseases. Biomed Pharmacother 2023; 165:115092. [PMID: 37406512 DOI: 10.1016/j.biopha.2023.115092] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 06/26/2023] [Accepted: 06/26/2023] [Indexed: 07/07/2023] Open
Abstract
Central nervous system (CNS) diseases are the leading cause of death worldwide. By performing compensatory functions and improving the inflammatory microenvironment, the transplantation of neural stem cells (NSCs) can promote functional recovery from brain injury, aging, brain tumours, and other diseases. However, the ability of NSCs to differentiate into neurons is limited, and they are associated with a risk of tumourigenicity. NSC-derived extracellular vesicles (NSC-EVs) can modulate the local microenvironment of the nervous system as well as distant neuronal functions. Thus, cell-free therapy may be a novel remedy for CNS disorders. This article reviews the characteristics, contents, and mechanisms of action of NSC-EVs as well as their roles and application prospects in various CNS diseases.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Bo Fang
- Department of Anesthesiology, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| |
Collapse
|
5
|
Ando Y, Chang FC, James M, Zhou Y, Zhang M. Chitosan Scaffolds as Microcarriers for Dynamic Culture of Human Neural Stem Cells. Pharmaceutics 2023; 15:1957. [PMID: 37514142 PMCID: PMC10384976 DOI: 10.3390/pharmaceutics15071957] [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: 06/12/2023] [Revised: 07/03/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Human neural stem cells (hNSCs) possess remarkable potential for regenerative medicine in the treatment of presently incurable diseases. However, a key challenge lies in producing sufficient quantities of hNSCs, which is necessary for effective treatment. Dynamic culture systems are recognized as a powerful approach to producing large quantities of hNSCs required, where microcarriers play a critical role in supporting cell expansion. Nevertheless, the currently available microcarriers have limitations, including a lack of appropriate surface chemistry to promote cell adhesion, inadequate mechanical properties to protect cells from dynamic forces, and poor suitability for mass production. Here, we present the development of three-dimensional (3D) chitosan scaffolds as microcarriers for hNSC expansion under defined conditions in bioreactors. We demonstrate that chitosan scaffolds with a concentration of 4 wt% (4CS scaffolds) exhibit desirable microstructural characteristics and mechanical properties suited for hNSC expansion. Furthermore, they could also withstand degradation in dynamic conditions. The 4CS scaffold condition yields optimal metabolic activity, cell adhesion, and protein expression, enabling sustained hNSC expansion for up to three weeks in a dynamic culture. Our study introduces an effective microcarrier approach for prolonged expansion of hNSCs, which has the potential for mass production in a three-dimensional setting.
Collapse
Affiliation(s)
- Yoshiki Ando
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
- Materials Department, Medical R&D Center, Corporate R&D Group, KYOCERA Corporation, Yasu 520-2362, Shiga, Japan
| | - Fei-Chien Chang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Matthew James
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Yang Zhou
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
6
|
Elias AE, Nuñez TA, Kun B, Kreiling JA. primiReference: a reference for analysis of primary-microRNA expression in single-nucleus sequencing data. J Genet Genomics 2023; 50:108-121. [PMID: 36371075 PMCID: PMC9974815 DOI: 10.1016/j.jgg.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022]
Abstract
Single-nucleus RNA-sequencing technology has revolutionized understanding of nuanced changes in gene expression between cell types within tissues. Unfortunately, our understanding of regulatory RNAs, such as microRNAs (miRNAs), is limited through both single-cell and single-nucleus techniques due to the short length of miRNAs in the cytoplasm and the incomplete reference of longer primary miRNA (pri-miRNA) transcripts in the nucleus. We build a custom reference to align and count pri-miRNA sequences in single-nucleus data. Using young and aged subventricular zone (SVZ) nuclei, we show differential expression of pri-miRNAs targeting genes involved in neural stem cells (NSC) differentiation in the aged SVZ. Furthermore, using wild-type and 5XFAD mouse model cortex nuclei, to validate the use of primiReference, we find cell-type-specific expression of pri-miRNAs known to be involved in Alzheimer's disease (AD). pri-miRNAs likely contribute to NSC dysregulation with age and AD pathology. primiReference is paramount in capturing a global profile of gene expression and regulation in single-nucleus data and can provide key insights into cell-type-specific expression of pri-miRNAs, paving the way for future studies of regulation and pathway dysregulation. By looking at pri-miRNA abundance and transcriptional differences, regulation of gene expression by miRNAs in disease and aging can be further explored.
Collapse
Affiliation(s)
- Amy E Elias
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, 02903, USA
| | - Thomas A Nuñez
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, 02903, USA
| | - Bianca Kun
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, 02903, USA
| | - Jill A Kreiling
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, 02903, USA.
| |
Collapse
|
7
|
Hu L, Zhou Y, Yang J, Zhao X, Mao L, Zheng W, Zhao J, Guo M, Chen C, He Z, Xu L. MicroRNA-7 overexpression positively regulates the CD8 + SP cell development via targeting PIK3R1. Exp Cell Res 2021; 407:112824. [PMID: 34516985 DOI: 10.1016/j.yexcr.2021.112824] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 08/22/2021] [Accepted: 09/04/2021] [Indexed: 12/21/2022]
Abstract
microRNA-7 (miR-7), a distinct miRNA family member, has been reported to be involved in the biological functions of immune cells. However, the potential role of miR-7 in the CD8+ T cell development remains to be elucidated. In this study, we estimated the potential effects of miR-7 overexpression in the thymic CD8+ SP cell development using miR-7 overexpression mice. Our results showed that compared with those in control wild type (WT) mice, the volume, weight and total cell numbers of thymus in miR-7 overexpression (OE) mice increased significantly. The absolute cell number of CD8+ SP cells in miR-7 OE mice increased and its ability of activation and proliferation enhanced. Futhermore, we clarified that miR-7 overexpression had an intrinsic promote role in CD8+ SP cell development by adoptive cell transfer assay. Mechanistically, the expression level of PIK3R1, a target of miR-7, decreased significantly in CD8+ SP cells of miR-7 OE mice. Moreover, the expression level of phosphorylated (p)-AKT and p-ERK changed inversely and indicating that miR-7 overexpression impaired the balance of AKE and ERK pathways. In summary, our work reveals an essential role of miR-7 in promoting CD8+ SP cell development through the regulation of PIK3R1 and balance of AKT and ERK pathways.
Collapse
Affiliation(s)
- Lin Hu
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Provincial Education Department, Guizhou, 563000, China; Department of Immunology & Talent Base of Biological Therapy of Guizhou Province, Zunyi Medical University, Guizhou, 563000, China
| | - Ya Zhou
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Provincial Education Department, Guizhou, 563000, China; Department of Medical Physics, Zunyi Medical University, Zunyi, Guizhou, 563003, China
| | - Jing Yang
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Provincial Education Department, Guizhou, 563000, China; Department of Immunology & Talent Base of Biological Therapy of Guizhou Province, Zunyi Medical University, Guizhou, 563000, China
| | - Xu Zhao
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Provincial Education Department, Guizhou, 563000, China; Department of Immunology & Talent Base of Biological Therapy of Guizhou Province, Zunyi Medical University, Guizhou, 563000, China
| | - Ling Mao
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Provincial Education Department, Guizhou, 563000, China; Department of Immunology & Talent Base of Biological Therapy of Guizhou Province, Zunyi Medical University, Guizhou, 563000, China
| | - Wen Zheng
- Department of Laboratory Medicine, Qiannan Medical University for Nationalities, Guizhou 558000, China
| | - Juanjuan Zhao
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Provincial Education Department, Guizhou, 563000, China; Department of Immunology & Talent Base of Biological Therapy of Guizhou Province, Zunyi Medical University, Guizhou, 563000, China
| | - Mengmeng Guo
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Provincial Education Department, Guizhou, 563000, China; Department of Immunology & Talent Base of Biological Therapy of Guizhou Province, Zunyi Medical University, Guizhou, 563000, China
| | - Chao Chen
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Provincial Education Department, Guizhou, 563000, China; Department of Immunology & Talent Base of Biological Therapy of Guizhou Province, Zunyi Medical University, Guizhou, 563000, China
| | - Zhixu He
- Department of Paediatrics, Affiliated Hospital of Zunyi Medical University, Guizhou, 563000, China; Key Laboratory of Adult Stem Cell Transformation Research, Chinese Academy of Medical Sciences, Guizhou, 563000, China
| | - Lin Xu
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Provincial Education Department, Guizhou, 563000, China; Department of Immunology & Talent Base of Biological Therapy of Guizhou Province, Zunyi Medical University, Guizhou, 563000, China.
| |
Collapse
|
8
|
Augenlicht A, Saiselet M, Decaussin-Petrucci M, Andry G, Dumont JE, Maenhaut C. MiR-7-5p inhibits thyroid cell proliferation by targeting the EGFR/MAPK and IRS2/PI3K signaling pathways. Oncotarget 2021; 12:1587-1599. [PMID: 34381564 PMCID: PMC8351599 DOI: 10.18632/oncotarget.28030] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 07/13/2021] [Indexed: 12/14/2022] Open
Abstract
The aberrant expression of miRNAs is often correlated to tumor development. MiR-7-5p is a recently discovered downregulated miRNA in thyroid papillary carcinoma (PTC). The goal of this project was to characterize its functional role in thyroid tumorigenesis and to identify the targeted modulated pathways. MiR-7-5p overexpression following transfection in TPC1 and HT-ori3 cells decreased proliferation of the two thyroid cell lines. Analysis of global transcriptome modifications showed that miR-7-5p inhibits thyroid cell proliferation by modulating the MAPK and PI3K signaling pathways which are both necessary for normal thyroid proliferation and play central roles in PTC tumorigenesis. Several effectors of these pathways are indeed targets of miR-7-5p, among which EGFR and IRS2, two upstream activators. We confirmed the upregulation of IRS2 and EGFR in human PTC and showed the existence of a negative correlation between the decreased expression of miR-7-5p and the increased expression of IRS2 or EGFR. Our results thus support a tumor-suppressor activity of miR-7-5p. The decreased expression of miR-7-5p during PTC tumorigenesis might give the cells a proliferative advantage and delivery of miR-7-5p may represent an innovative approach for therapy.
Collapse
Affiliation(s)
- Alice Augenlicht
- Institute of Interdisciplinary Research, Université libre de Bruxelles, Brussels, Belgium
| | - Manuel Saiselet
- Institute of Interdisciplinary Research, Université libre de Bruxelles, Brussels, Belgium
| | - Myriam Decaussin-Petrucci
- Service d'Anatomie et Cytologie Pathologiques, Centre Hospitalier Lyon Sud, Université Lyon 1, Pierre Benite Cedex 69495, France
| | - Guy Andry
- Surgery Department, J. Bordet Institute, Brussels 1000, Belgium
| | - Jacques E Dumont
- Institute of Interdisciplinary Research, Université libre de Bruxelles, Brussels, Belgium
| | - Carine Maenhaut
- Institute of Interdisciplinary Research, Université libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
9
|
Bahram Sangani N, Gomes AR, Curfs LMG, Reutelingsperger CP. The role of Extracellular Vesicles during CNS development. Prog Neurobiol 2021; 205:102124. [PMID: 34314775 DOI: 10.1016/j.pneurobio.2021.102124] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 04/16/2021] [Accepted: 07/20/2021] [Indexed: 12/21/2022]
Abstract
With a diverse set of neuronal and glial cell populations, Central Nervous System (CNS) has one of the most complex structures in the body. Intercellular communication is therefore highly important to coordinate cell-to-cell interactions. Besides electrical and chemical messengers, CNS cells also benefit from another communication route, what is known as extracellular vesicles, to harmonize their interactions. Extracellular Vesicles (EVs) and their subtype exosomes are membranous particles secreted by cells and contain information packaged in the form of biomolecules such as small fragments of DNA, lipids, miRNAs, mRNAs, and proteins. They are able to efficiently drive changes upon their arrival to recipient cells. EVs actively participate in all stages of CNS development by stimulating neural cell proliferation, differentiation, synaptic formation, and mediating reciprocal interactions between neurons and oligodendrocyte for myelination process. The aim of the present review is to enlighten the presence and contribution of EVs at each CNS developmental milestone.
Collapse
Affiliation(s)
- Nasim Bahram Sangani
- Department of Biochemistry, Maastricht University, Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands; GKC-Rett Expertise Centre, Maastricht University Medical Centre, Maastricht, the Netherlands.
| | - Ana Rita Gomes
- Department of Bioengineering and IBB - Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001, Lisboa, Portugal; Instituto de Medicina Molecular - João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Portugal.
| | - Leopold M G Curfs
- GKC-Rett Expertise Centre, Maastricht University Medical Centre, Maastricht, the Netherlands.
| | - Chris P Reutelingsperger
- Department of Biochemistry, Maastricht University, Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands; GKC-Rett Expertise Centre, Maastricht University Medical Centre, Maastricht, the Netherlands.
| |
Collapse
|
10
|
Yang GH, Lee YB, Kang D, Choi E, Nam Y, Lee KH, You HJ, Kang HJ, An SH, Jeon H. Overcome the barriers of the skin: exosome therapy. Biomater Res 2021; 25:22. [PMID: 34217362 PMCID: PMC8254055 DOI: 10.1186/s40824-021-00224-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/23/2021] [Indexed: 01/06/2023] Open
Abstract
Exosomes are nano-sized cargos with a lipid bilayer structure carrying diverse biomolecules including lipids, proteins, and nucleic acids. These small vesicles are secreted by most types of cells to communicate with each other. Since exosomes circulate through bodily fluids, they can transfer information not only to local cells but also to remote cells. Therefore, exosomes are considered potential biomarkers for various treatments. Recently, studies have shown the efficacy of exosomes in skin defects such as aging, atopic dermatitis, and wounds. Also, exosomes are being studied to be used as ingredients in commercialized skin treatment products. In this review, we discussed the need for exosomes in skin therapy together with the current challenges. Moreover, the functional roles of exosomes in terms of skin treatment and regeneration are overviewed. Finally, we highlighted the major limitations and the future perspective in exosome engineering.
Collapse
Affiliation(s)
- Gi Hoon Yang
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc., 55 Hanyangdaehak-Ro, Ansan-si, Gyeonggi-Do, 15588, South Korea
| | - Yoon Bum Lee
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu, 41061, South Korea
| | - Donggu Kang
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc., 55 Hanyangdaehak-Ro, Ansan-si, Gyeonggi-Do, 15588, South Korea
| | - Eunjeong Choi
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc., 55 Hanyangdaehak-Ro, Ansan-si, Gyeonggi-Do, 15588, South Korea
| | - Yoonju Nam
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc., 55 Hanyangdaehak-Ro, Ansan-si, Gyeonggi-Do, 15588, South Korea
| | - Kyoung Ho Lee
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu, 41061, South Korea
| | - Hi-Jin You
- Department of Plastic Surgery, Korea University Ansan Hospital, 123 Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-Do, 15355, South Korea
| | - Hyo Jin Kang
- Biomedical Research Center, Korea University Ansan Hospital, 123 Jeokgeum-ro, Danwon-gu, Ansan-si, Gyeonggi-Do, 15355, South Korea
| | - Sang Hyun An
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), 80 Cheombok-ro, Dong-gu, Daegu, 41061, South Korea.
| | - Hojun Jeon
- Research Institute of Additive Manufacturing and Regenerative Medicine, Baobab Healthcare Inc., 55 Hanyangdaehak-Ro, Ansan-si, Gyeonggi-Do, 15588, South Korea.
| |
Collapse
|
11
|
Consales C, Butera A, Merla C, Pasquali E, Lopresto V, Pinto R, Pierdomenico M, Mancuso M, Marino C, Benassi B. Exposure of the SH-SY5Y Human Neuroblastoma Cells to 50-Hz Magnetic Field: Comparison Between Two-Dimensional (2D) and Three-Dimensional (3D) In Vitro Cultures. Mol Neurobiol 2020; 58:1634-1649. [PMID: 33230715 PMCID: PMC7932966 DOI: 10.1007/s12035-020-02192-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 10/30/2020] [Indexed: 12/13/2022]
Abstract
We here characterize the response to the extremely low-frequency (ELF) magnetic field (MF, 50 Hz, 1 mT) of SH-SY5Y human neuroblastoma cells, cultured in a three-dimensional (3D) Alvetex® scaffold compared to conventional two-dimensional (2D) monolayers. We proved that the growing phenotype of proliferating SH-SY5Y cells is not affected by the culturing conditions, as morphology, cell cycle distribution, proliferation/differentiation gene expression of 3D-cultures overlap what reported in 2D plates. In response to 72-h exposure to 50-Hz MF, we demonstrated that no proliferation change and apoptosis activation occur in both 2D and 3D cultures. Consistently, no modulation of Ki67, MYCN, CCDN1, and Nestin, of invasiveness and neo-angiogenesis-controlling genes (HIF-1α, VEGF, and PDGF) and of microRNA epigenetic signature (miR-21-5p, miR-222-3p and miR-133b) is driven by ELF exposure. Conversely, intracellular glutathione content and SOD1 expression are exclusively impaired in 3D-culture cells in response to the MF, whereas no change of such redox modulators is observed in SH-SY5Y cells if grown on 2D monolayers. Moreover, ELF-MF synergizes with the differentiating agents to stimulate neuroblastoma differentiation into a dopaminergic (DA) phenotype in the 3D-scaffold culture only, as growth arrest and induction of p21, TH, DAT, and GAP43 are reported in ELF-exposed SH-SY5Y cells exclusively if grown on 3D scaffolds. As overall, our findings prove that 3D culture is a more reliable experimental model for studying SH-SY5Y response to ELF-MF if compared to 2D conventional monolayer, and put the bases for promoting 3D systems in future studies addressing the interaction between electromagnetic fields and biological systems.
Collapse
Affiliation(s)
- Claudia Consales
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Alessio Butera
- Experimental Medicine and Surgery, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Caterina Merla
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Emanuela Pasquali
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Vanni Lopresto
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Rosanna Pinto
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Maria Pierdomenico
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Mariateresa Mancuso
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Carmela Marino
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy
| | - Barbara Benassi
- Division of Health Protection Technologies, ENEA-Casaccia Italian National Agency for New Technologies, Energy and Sustainable Economic Development, Via Anguillarese 301, 00123, Rome, Italy.
| |
Collapse
|
12
|
Chen J, Zhang L, Gan X, Zhang R, He Y, Lv Q, Fu H, Liu X, Miao L. Effects of Retinal Transcription Regulation After GB20 Needling Treatment in Retina With Optic Neuritis. Front Integr Neurosci 2020; 14:568449. [PMID: 33117136 PMCID: PMC7550785 DOI: 10.3389/fnint.2020.568449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/13/2020] [Indexed: 11/13/2022] Open
Abstract
Optic neuritis (ON) is one of the most frequent symptoms of multiple sclerosis (MS) that results in progressive loss of axons and neurons. In clinical trials of Traditional Chinese Medicine, needling at the GB20 acupoint has been widely used for the treatment of ocular diseases, including ON. However, the molecular mechanisms of needling at this site are still unclear. In this study, we generated an experimental autoimmune encephalomyelitis (EAE) mouse model and investigated the effects of needling treatment at the GB20 acupoint on retina with EAE-associated ON. RNA sequencing of the retinal transcriptome revealed that, of the 234 differentially expressed genes induced by ON, 100 genes were upregulated, and 134 genes were downregulated by ON, while needling at the GB20 acupoint specifically reversed the expression of 21 genes compared with control treatment at GV16 acupoint. Among the reversed genes, Nr4a3, Sncg, Uchl1, and Tppp3 were involved in axon development and regeneration and were downregulated by ON, indicating the beneficial effect of needling at GB20. Further gene ontology (GO) enrichment analysis revealed that needling at GB20 affected the molecular process of Circadian rhythm in mouse retina with ON. Our study first reported that needling treatment after ON at the GB20 acupoint regulated gene expression of the retina and reversed the expression of downregulated axon development-related genes. This study also demonstrated that GV16 was a perfect control treatment site for GB20 in animal research. Our study provided a scientific basis for needling treatments at GB20 for ocular diseases.
Collapse
Affiliation(s)
- Jie Chen
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing, China.,School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Li Zhang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Xiulun Gan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Rong Zhang
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Yinjia He
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Qiuyi Lv
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Haonan Fu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaodong Liu
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Linqing Miao
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing, China.,School of Mechatronical Engineering, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
13
|
MicroRNA-7: expression and function in brain physiological and pathological processes. Cell Biosci 2020; 10:77. [PMID: 32537124 PMCID: PMC7288475 DOI: 10.1186/s13578-020-00436-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/23/2020] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs that regulate gene expression at the post-transcriptional level and play critical roles in regulating physiological function, and are becoming worldwide research hot spot in brain development and diseases. However, the exact value of miRNAs in brain physiological and pathological processes remain to be fully elucidated, which is vital for the application of miRNAs as diagnostic, prognostic, and therapeutic biomarkers for brain diseases. MicroRNA-7 (miR-7), as a highly expressed miRNA molecule in the mammalian brain, is well documented to play a critical role in development of various diseases. Importantly, accumulating evidence has shown that miR-7 is involved in a range of developmental and pathological processes of brain. Expressively, miR-7, encoded by three genes located different chromosomes, is dominantly expressed in neurons with sensory or neurosecretory. Moreover, the expression of miR-7 is regulated at three levels including gene transcription, process of primary and precursor sequence and formation of mature sequence. Physiologically, miR-7 principally governs the physiological development of Pituitary gland, Optic nervous system and Cerebral cortex. Pathologically, miR-7 can regulate multiple genes thereby manipulating the process of various brain diseases including neurodegenerative diseases, neuroinflammation, and mental disorders and so on. These emerging studies have shown that miR-7, a representative member of miRNA family, might be a novel intrinsic regulatory molecule involved in the physiological and pathological process of brain. Therefore, in-depth studies on the role of miR-7 in brain physiology and pathology undoubtedly not only provide a light on the roles of miRNAs in brain development and diseases, but also are much helpful for ultimate development of therapeutic strategies against brain diseases. In this review, we provide an overview of current scientific knowledge regarding the expression and function of miR-7 in development and disease of brain and raise many issues involved in the relationship between miR-7 and brain physiological and pathological processes.
Collapse
|
14
|
Pan J, Lee YC, Lee HHC, Sung TC, Jen SH, Ban LK, Su HY, Chen DC, Hsu ST, Higuchi A, Chen H. Culture and differentiation of purified human adipose-derived stem cells by membrane filtration via nylon mesh filters. J Mater Chem B 2020; 8:5204-5214. [PMID: 32490480 DOI: 10.1039/d0tb00947d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Human adipose derived stem cells purified by the membrane migration method through filter membranes coated with vitronectin showed high osteogenic differentiation.
Collapse
|
15
|
Gu J, Shao R, Li M, Yan Q, Hu H. MiR-485-3p modulates neural stem cell differentiation and proliferation via regulating TRIP6 expression. J Cell Mol Med 2019; 24:398-404. [PMID: 31730275 PMCID: PMC6933395 DOI: 10.1111/jcmm.14743] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 08/16/2019] [Accepted: 08/27/2019] [Indexed: 12/12/2022] Open
Abstract
Recent references have showed crucial roles of several miRNAs in neural stem cell differentiation and proliferation. However, the expression and role of miR‐485‐3p remains unknown. In our reference, we indicated that miR‐485‐3p expression was down‐regulated during NSCs differentiation to neural and astrocytes cell. In addition, the TRIP6 expression was up‐regulated during NSCs differentiation to neural and astrocytes cell. We carried out the dual‐luciferase reporter and found that overexpression of miR‐485‐3p decreased the luciferase activity of pmirGLO‐TRIP6‐wt but not the pmirGLO‐TRIP6‐mut. Ectopic expression of miR‐485‐3p decreased the expression of TRIP6 in NSC. Ectopic miR‐485‐3p expression suppressed the cell growth of NSCs and inhibited nestin expression of NSCs. Moreover, elevated expression of miR‐485‐3p decreased the ki‐67 and cyclin D1 expression in NSCs. Furthermore, we indicated that miR‐485‐3p reduced proliferation and induced differentiation of NSCs via targeting TRIP6 expression. These data suggested that a crucial role of miR‐485‐3p in self‐proliferation and differentiation of NSCs. Thus, altering miR‐485‐3p and TRIP6 modulation may be one promising therapy for treating with neurodegenerative and neurogenesis diseases.
Collapse
Affiliation(s)
- Juxian Gu
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, China
| | - Rusheng Shao
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, China
| | - Meng Li
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, China
| | - Qiuyue Yan
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, China
| | - Hongwei Hu
- Department of Pain, Cangzhou Central Hospital, Cangzhou, China
| |
Collapse
|
16
|
Zadehbagheri F, Hosseini E, Bagheri-Hosseinabadi Z, Rekabdarkolaee HM, Sadeghi I. Profiling of miRNAs in serum of children with attention-deficit hyperactivity disorder shows significant alterations. J Psychiatr Res 2019; 109:185-192. [PMID: 30557705 DOI: 10.1016/j.jpsychires.2018.12.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/05/2018] [Accepted: 12/07/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Attention-deficit/hyperactivity disorder (ADHD), a common psychiatric disorder, is identified by abnormal levels of impulsivity, inattention, and hyperactivity. MiRNAs play important roles in neural network development of the brain. Circulating miRNAs (cmiRNAs) are offered as promising noninvasive markers for psychiatric disorders. In this study, the expression level of neurologically relevant miRNAs was evaluated in serum samples of ADHD individuals. METHODS RNA extraction was performed for 60 subjects with ADHD and 60 healthy controls, and the cDNAs were synthesized for all the miRNAs. The expression level of 84 cmiRNAs was then examined in 4 ADHD subjects and 4 controls. The altered expression of 10 cmiRNAs was further evaluated in validation cohort comprising 56 ADHD and 56 control samples by qPCR. The diagnostic power of the miRNAs was determined by use of Receiver-operating characteristic (ROC) analysis. The cmiRNAs target genes were predicted using DIANA mirPath software and gene ontology enrichment analysis was performed using Cytoscape CLUGO. RESULTS Initially, 10 miRNAs showed differential expression in ADHD individuals. Further analysis confirmed four miRNAs (hsa-miR-101-3p, hsa-miR-130a-3p, hsa-miR-138-5p and hsa-miR-195-5p) upregulated and one miRNA (hsa-miR-106b-5p) downregulated. These miRNAs showed significant predictive values for discriminating ADHD individuals. Enrichment analysis highlighted the involvement of the deregulated cmiRNAs in many canonical neurobiological pathways and mechanisms. CONCLUSIONS Our report is the first comprehensive study on the expression profiling of miRNAs in serum of ADHD subjects. These findings suggest a set of cmiRNAs as potential noninvasive biomarkers for ADHD.
Collapse
Affiliation(s)
- Fatemeh Zadehbagheri
- Department of Internal Medicine, Yasuj University of Medical Sciences, Yasuj, Iran; Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Ebrahim Hosseini
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Zahra Bagheri-Hosseinabadi
- Department of Clinical Biochemistry, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | | | - Iman Sadeghi
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran; CEINGE-biotecnologie Avanzate, Naples, Italy.
| |
Collapse
|
17
|
Magalhães L, Quintana LG, Lopes DCF, Vidal AF, Pereira AL, D'Araujo Pinto LC, de Jesus Viana Pinheiro J, Khayat AS, Goulart LR, Burbano R, de Assumpção PP, Ribeiro-Dos-Santos Â. APC gene is modulated by hsa-miR-135b-5p in both diffuse and intestinal gastric cancer subtypes. BMC Cancer 2018; 18:1055. [PMID: 30376837 PMCID: PMC6208123 DOI: 10.1186/s12885-018-4980-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 10/21/2018] [Indexed: 12/20/2022] Open
Abstract
Background Several genetic and epigenetic alterations are related to the development and progression of Gastric Cancer (GC), one of those being the deregulated microRNA (miRNA) expression profile. miRNAs are small noncoding RNAs that negatively regulate the expression of thousands of genes, including oncogenes and tumor suppressor genes. Our group identified, in previous studies, some miRNAs that are differentially expressed in GC when compared to the gastric mucosa without cancer, including hsa-miR-29c and hsa-miR-135b. The aim of the study was to modulate the expression of the miRNAs hsa-miR-29c-5p and hsa-miR-135b-5p and evaluate the expression of their target genes in 2D and 3D cell cultures. Methods hsa-miR-29c-5p and hsa-miR-135b-5p expression profiles were modulated by transfecting mimics and antimiRs, respectively, in 2D and 3D cell cultures. The expression of the proteins coded by the genes CDC42, DNMT3A (target genes of hsa-miR-29c-5p) and APC (target gene of hsa-miR-135b-5p) were measured by Western Blot. Results Results showed that mimics and antimiRs transfection significantly altered the expression of both miRNAs, increasing the expression of hsa-miR-29c-5p and reducing the expression of hsa-miR-135b-5p, especially in the 3D culture of the cell lines. When analyzing the proteins expression, we observed that AGP01 and AGP03 cell lines transfected with mimics had a reduction in the levels of CDC42 and DNMT3A and all three cell lines transfected with antimiRs had an increase in the expression of the protein APC. Conclusion We concluded that three-dimensional culture can be a more representative in vitro model that resembles better the in vivo reality. Our results also showed that hsa-miR-29c-5p is an important regulator of CDC42 and DNMT3A genes in the intestinal subtype gastric cancer and hsa-miR-135b-5p regulates the APC gene in both intestinal and diffuse subtypes of GC. Dysregulation in their expression, and consequently in their respectively signaling pathways, shows how these miRNAs can influence the carcinogenesis of different histological subtypes of gastric cancer. Electronic supplementary material The online version of this article (10.1186/s12885-018-4980-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Leandro Magalhães
- Laboratório de Genética Humana e Médica, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Luciana Gonçalves Quintana
- Núcleo de Pesquisas em Oncologia, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | - Dielly Catrina Favacho Lopes
- Laboratório de Neuropatologia Experimental, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | - Amanda Ferreira Vidal
- Laboratório de Genética Humana e Médica, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Adenilson Leão Pereira
- Laboratório de Genética Humana e Médica, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Lara Carolina D'Araujo Pinto
- Laboratório de Cultivo Celular, Faculdade de Odontologia, Instituto de Ciências da Saúde, Universidade Federal do Pará, Belém, Brazil
| | - João de Jesus Viana Pinheiro
- Laboratório de Cultivo Celular, Faculdade de Odontologia, Instituto de Ciências da Saúde, Universidade Federal do Pará, Belém, Brazil
| | - André Salim Khayat
- Núcleo de Pesquisas em Oncologia, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil.,Laboratório de Citogenética Humana, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Luiz Ricardo Goulart
- Laboratório de Nanobiotecnologia, Instituto de Genética e Bioquímica, Universidade Federal de Uberlândia, Uberlândia, Brazil
| | - Rommel Burbano
- Núcleo de Pesquisas em Oncologia, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil.,Laboratório de Citogenética Humana, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Paulo Pimentel de Assumpção
- Núcleo de Pesquisas em Oncologia, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil
| | - Ândrea Ribeiro-Dos-Santos
- Laboratório de Genética Humana e Médica, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil. .,Núcleo de Pesquisas em Oncologia, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, Brazil.
| |
Collapse
|
18
|
Vieira MS, Santos AK, Vasconcellos R, Goulart VAM, Parreira RC, Kihara AH, Ulrich H, Resende RR. Neural stem cell differentiation into mature neurons: Mechanisms of regulation and biotechnological applications. Biotechnol Adv 2018; 36:1946-1970. [PMID: 30077716 DOI: 10.1016/j.biotechadv.2018.08.002] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 07/31/2018] [Accepted: 08/01/2018] [Indexed: 02/07/2023]
Abstract
The abilities of stem cells to self-renew and form different mature cells expand the possibilities of applications in cell-based therapies such as tissue recomposition in regenerative medicine, drug screening, and treatment of neurodegenerative diseases. In addition to stem cells found in the embryo, various adult organs and tissues have niches of stem cells in an undifferentiated state. In the central nervous system of adult mammals, neurogenesis occurs in two regions: the subventricular zone and the dentate gyrus in the hippocampus. The generation of the different neural lines originates in adult neural stem cells that can self-renew or differentiate into astrocytes, oligodendrocytes, or neurons in response to specific stimuli. The regulation of the fate of neural stem cells is a finely controlled process relying on a complex regulatory network that extends from the epigenetic to the translational level and involves extracellular matrix components. Thus, a better understanding of the mechanisms underlying how the process of neurogenesis is induced, regulated, and maintained will provide elues for development of novel for strategies for neurodegenerative therapies. In this review, we focus on describing the mechanisms underlying the regulation of the neuronal differentiation process by transcription factors, microRNAs, and extracellular matrix components.
Collapse
Affiliation(s)
- Mariana S Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinopólis, MG, Brazil
| | - Anderson K Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Rebecca Vasconcellos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinopólis, MG, Brazil
| | - Vânia A M Goulart
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ricardo C Parreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinopólis, MG, Brazil
| | - Alexandre H Kihara
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, SP, Brazil.
| | - Rodrigo R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil; Instituto Nanocell, Divinopólis, MG, Brazil.
| |
Collapse
|
19
|
Li WY, Zhang WT, Cheng YX, Liu YC, Zhai FG, Sun P, Li HT, Deng LX, Zhu XF, Wang Y. Inhibition of KLF7-Targeting MicroRNA 146b Promotes Sciatic Nerve Regeneration. Neurosci Bull 2018; 34:419-437. [PMID: 29356943 DOI: 10.1007/s12264-018-0206-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 10/28/2017] [Indexed: 12/12/2022] Open
Abstract
A previous study has indicated that Krüppel-like factor 7 (KLF7), a transcription factor that stimulates Schwann cell (SC) proliferation and axonal regeneration after peripheral nerve injury, is a promising therapeutic transcription factor in nerve injury. We aimed to identify whether inhibition of microRNA-146b (miR-146b) affected SC proliferation, migration, and myelinated axon regeneration following sciatic nerve injury by regulating its direct target KLF7. SCs were transfected with miRNA lentivirus, miRNA inhibitor lentivirus, or KLF7 siRNA lentivirus in vitro. The expression of miR146b and KLF7, as well as SC proliferation and migration, were subsequently evaluated. In vivo, an acellular nerve allograft (ANA) followed by injection of GFP control vector or a lentiviral vector encoding an miR-146b inhibitor was used to assess the repair potential in a model of sciatic nerve gap. miR-146b directly targeted KLF7 by binding to the 3'-UTR, suppressing KLF7. Up-regulation of miR-146b and KLF7 knockdown significantly reduced the proliferation and migration of SCs, whereas silencing miR-146b resulted in increased proliferation and migration. KLF7 protein was localized in SCs in which miR-146b was expressed in vivo. Similarly, 4 weeks after the ANA, anti-miR-146b increased KLF7 and its target gene nerve growth factor cascade, promoting axonal outgrowth. Closer analysis revealed improved nerve conduction and sciatic function index score, and enhanced expression of neurofilaments, P0 (anti-peripheral myelin), and myelinated axon regeneration. Our findings provide new insight into the regulation of KLF7 by miR-146b during peripheral nerve regeneration and suggest a potential therapeutic strategy for peripheral nerve injury.
Collapse
Affiliation(s)
- Wen-Yuan Li
- Department of Anatomy, Mudanjiang College of Medicine, Mudanjiang, 157011, China
| | - Wei-Ting Zhang
- The Affiliated Hongqi Hospital, Mudanjiang College of Medicine, Mudanjiang, 157011, China
| | - Yong-Xia Cheng
- Department of Pathology, Mudanjiang College of Medicine, Mudanjiang, 157011, China
| | - Yan-Cui Liu
- Department of Anatomy, Mudanjiang College of Medicine, Mudanjiang, 157011, China
| | - Feng-Guo Zhai
- Department of Pharmacy, Mudanjiang College of Medicine, Mudanjiang, 157011, China
| | - Ping Sun
- Department of Anatomy, Mudanjiang College of Medicine, Mudanjiang, 157011, China
| | - Hui-Ting Li
- The Affiliated Hongqi Hospital, Mudanjiang College of Medicine, Mudanjiang, 157011, China
| | - Ling-Xiao Deng
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Xiao-Feng Zhu
- Department of Anatomy, Mudanjiang College of Medicine, Mudanjiang, 157011, China.
| | - Ying Wang
- Department of Anatomy, Mudanjiang College of Medicine, Mudanjiang, 157011, China.
| |
Collapse
|
20
|
Zhou N, Hao S, Huang Z, Wang W, Yan P, Zhou W, Zhu Q, Liu X. MiR-7 inhibited peripheral nerve injury repair by affecting neural stem cells migration and proliferation through cdc42. Mol Pain 2018; 14:1744806918766793. [PMID: 29663842 PMCID: PMC5912295 DOI: 10.1177/1744806918766793] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 01/29/2018] [Accepted: 02/05/2018] [Indexed: 01/30/2023] Open
Abstract
Objective Neural stem cells play an important role in the recovery and regeneration of peripheral nerve injury, and the microRNA-7 (miR-7) regulates differentiation of neural stem cells. This study aimed to explore the role of miR-7 in neural stem cells homing and proliferation and its influence on peripheral nerve injury repair. Methods The mice model of peripheral nerve injury was created by segmental sciatic nerve defect (sciatic nerve injury), and neural stem cells treatment was performed with a gelatin hydrogel conduit containing neural stem cells inserted into the sciatic nerve injury mice. The Sciatic Function Index was used to quantify sciatic nerve functional recovery in the mice. The messenger RNA and protein expression were detected by reverse transcription polymerase chain reaction and Western blot, respectively. Luciferase reporter assay was used to confirm the binding between miR-7 and the 3'UTR of cell division cycle protein 42 (cdc42). The neural stem cells migration and proliferation were analyzed by transwell assay and a Cell-LightTM EdU DNA Cell Proliferation kit, respectively. Results Neural stem cells treatment significantly promoted nerve repair in sciatic nerve injury mice. MiR-7 expression was decreased in sciatic nerve injury mice with neural stem cells treatment, and miR-7 mimic transfected into neural stem cells suppressed migration and proliferation, while miR-7 inhibitor promoted migration and proliferation. The expression level and effect of cdc42 on neural stem cells migration and proliferation were opposite to miR-7, and the luciferase reporter assay proved that cdc42 was a target of miR-7. Using co-transfection into neural stem cells, we found pcDNA3.1-cdc42 and si-cdc42 could reverse respectively the role of miR-7 mimic and miR-7 inhibitor on neural stem cells migration and proliferation. In addition, miR-7 mimic-transfected neural stem cells could abolish the protective role of neural stem cells on peripheral nerve injury. Conclusion MiR-7 inhibited peripheral nerve injury repair by affecting neural stem cells migration and proliferation through cdc42.
Collapse
Affiliation(s)
- Nan Zhou
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuang Hao
- Department of Cardiac Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zongqiang Huang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weiwei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Penghui Yan
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wei Zhou
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qihang Zhu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaokang Liu
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
21
|
OIP5, a target of miR-15b-5p, regulates hepatocellular carcinoma growth and metastasis through the AKT/mTORC1 and β-catenin signaling pathways. Oncotarget 2017; 8:18129-18144. [PMID: 28184024 PMCID: PMC5392313 DOI: 10.18632/oncotarget.15185] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 01/16/2017] [Indexed: 12/12/2022] Open
Abstract
Opa interacting protein 5 (OIP5) is upregulated in some types of human cancers, but the biological implications of its upregulation have not yet been clarified in human hepatocellular carcinoma (HCC). In this study, the signaling pathway downstream of OIP5 was analyzed by proteome kinase profiling. A putative microRNA targeting OIP5 was identified using a miRNA PCR array. Tumorigenicity and metastatic ability were examined in an orthotopic animal model. OIP5 expression was strongly detected in early and advanced tumors via gene expression profiling and immunohistochemical staining analyses. Cells with knockdown of OIP5 via target shRNA exhibited reduced hepatic mass formation and metastatic tumor nodules in an orthotopic mouse model. OIP5-induced AKT activation was mediated by both mTORC2 and p38/PTEN activation. AKT activation was linked to mTORC1 and GSK-3β/β-catenin signaling, which are primarily associated with tumor cell growth and metastasis, respectively. miR-15b-5p, which targets OIP5, efficiently inhibited OIP5-mediated mTORC1 and GSK-3β/β-catenin signaling. These findings suggest that OIP5 may be involved in HCC growth and metastasis and that miR-15b-5p inhibits OIP5-mediated oncogenic signaling in HCC.
Collapse
|
22
|
Piwecka M, Glažar P, Hernandez-Miranda LR, Memczak S, Wolf SA, Rybak-Wolf A, Filipchyk A, Klironomos F, Cerda Jara CA, Fenske P, Trimbuch T, Zywitza V, Plass M, Schreyer L, Ayoub S, Kocks C, Kühn R, Rosenmund C, Birchmeier C, Rajewsky N. Loss of a mammalian circular RNA locus causes miRNA deregulation and affects brain function. Science 2017; 357:science.aam8526. [DOI: 10.1126/science.aam8526] [Citation(s) in RCA: 713] [Impact Index Per Article: 101.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 07/26/2017] [Indexed: 12/29/2022]
|
23
|
Zhang N, Lyu Y, Pan X, Xu L, Xuan A, He X, Huang W, Long D. miR‑146b‑5p promotes the neural conversion of pluripotent stem cells by targeting Smad4. Int J Mol Med 2017; 40:814-824. [PMID: 28713933 PMCID: PMC5548013 DOI: 10.3892/ijmm.2017.3064] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/30/2017] [Indexed: 01/06/2023] Open
Abstract
Pluripotent stem cells (PSCs) are regarded as potential sources that provide specific neural cells for cell therapy in some nervous system diseases. However, the mechanisms underlying the neural differentiation of PSCs remain largely unknown. MicroRNAs (miRNAs or miRs) are a class of small non-protein-coding RNAs that act as critical regulatory molecules in many cellular processes. In this study, we found that miR-146b-5p expression was markedly increased following the neural induction of mouse embryonic stem cells (ESCs) or induced PSCs (iPSCs). In this study, to further identify the role of miR-146b-5p, we generated stable miR-146b-5p- overexpressing ESC and iPSC cell lines, and induced the differentiation of these cells by the adherent monolayer culture method. In the miR-146b-5p-overexpressing ESC- or iPSC- derived cultures, RT-qPCR analysis revealed that the mRNA expression levels of neuroectoderm markers, such as Sox1, Nestin and Pax6, were markedly increased, and flow cytometric analysis verified that the number of Nestin-positive cells was higher in the miR-146b-5p-overexpressing compared with the control cells. Mechanistically, the miR-146b-5p-overexpressing ESCs or iPSCs exhibited a significant reduction in Oct4 expression, which may be an explanation for these cells having a tendency to differentiate towards the neural lineage. Moreover, we confirmed that miR-146b-5p directly targeted Smad4 and negatively regulated the transforming growth factor (TGF)-β signaling pathway, which contributed to the neural commitment of PSCs. Collectively, our findings uncover the essential role of miR-146b-5p in the neural conversion of PSCs.
Collapse
Affiliation(s)
- Nianping Zhang
- Department of Human Anatomy, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Ying Lyu
- Department of Human Anatomy, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Xuebing Pan
- Department of Human Anatomy, College of Health Sciences of Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Liping Xu
- Department of Human Anatomy, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Aiguo Xuan
- Department of Human Anatomy, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Xiaosong He
- Department of Human Anatomy, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Wandan Huang
- Department of Human Anatomy, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| | - Dahong Long
- Department of Human Anatomy, Guangzhou Medical University, Guangzhou, Guangdong 511436, P.R. China
| |
Collapse
|
24
|
MicroRNA-Directed Neuronal Reprogramming as a Therapeutic Strategy for Neurological Diseases. Mol Neurobiol 2017; 55:4428-4436. [PMID: 28664454 DOI: 10.1007/s12035-017-0671-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 06/20/2017] [Indexed: 01/22/2023]
Abstract
The loss of neurons due to injury and disease results in a wide spectrum of highly disabling neurological and neurodegenerative conditions, given the apparent limited capacity of endogenous repair of the adult central nervous system (CNS). Therefore, it is important to develop technologies that can promote de novo neural stem cell and neuron generation. Current insights in CNS development and cellular reprogramming have provided the knowledge to finely modulate lineage-restricted transcription factors and microRNAs (miRNA) to elicit correct neurogenesis. Here, we discuss the current knowledge on the direct reprogramming of somatic non-neuronal cells into neural stem cells or subtype specific neurons in vitro and in vivo focusing on miRNA driven reprogramming. miRNA can allow rapid and efficient direct phenotype conversion by modulating gene networks active during development, which promote global shifts in the epigenetic landscape pivoting cell fate decisions. Furthermore, we critically present state-of-the-art and recent advances on miRNA therapeutics that can be applied to the diseased CNS. Together, the advances in our understanding of miRNA role in CNS development and disease, recent progress in miRNA-based therapeutic strategies, and innovative drug delivery methods create novel perspectives for meaningful therapies for neurodegenerative disorders.
Collapse
|
25
|
Murphy AR, Laslett A, O'Brien CM, Cameron NR. Scaffolds for 3D in vitro culture of neural lineage cells. Acta Biomater 2017; 54:1-20. [PMID: 28259835 DOI: 10.1016/j.actbio.2017.02.046] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 02/28/2017] [Accepted: 02/28/2017] [Indexed: 12/22/2022]
Abstract
Understanding how neurodegenerative disorders develop is not only a key challenge for researchers but also for the wider society, given the rapidly aging populations in developed countries. Advances in this field require new tools with which to recreate neural tissue in vitro and produce realistic disease models. This in turn requires robust and reliable systems for performing 3D in vitro culture of neural lineage cells. This review provides a state of the art update on three-dimensional culture systems for in vitro development of neural tissue, employing a wide range of scaffold types including hydrogels, solid porous polymers, fibrous materials and decellularised tissues as well as microfluidic devices and lab-on-a-chip systems. To provide some context with in vivo development of the central nervous system (CNS), we also provide a brief overview of the neural stem cell niche, neural development and neural differentiation in vitro. We conclude with a discussion of future directions for this exciting and important field of biomaterials research. STATEMENT OF SIGNIFICANCE Neurodegenerative diseases, including dementia, Parkinson's and Alzheimer's diseases and motor neuron diseases, are a major societal challenge for aging populations. Understanding these conditions and developing therapies against them will require the development of new physical models of healthy and diseased neural tissue. Cellular models resembling neural tissue can be cultured in the laboratory with the help of 3D scaffolds - materials that allow the organization of neural cells into tissue-like structures. This review presents recent work on the development of different types of scaffolds for the 3D culture of neural lineage cells and the generation of functioning neural-like tissue. These in vitro culture systems are enabling the development of new approaches for modelling and tackling diseases of the brain and CNS.
Collapse
Affiliation(s)
- Ashley R Murphy
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, VIC 3800, Australia
| | - Andrew Laslett
- CSIRO Manufacturing, Bag 10, Clayton South MDC, VIC 3168, Australia; Australian Regenerative Medicine Institute, Science, Technology, Research and Innovation Precinct (STRIP), Monash University, Clayton Campus, Wellington Road, Clayton, VIC 3800, Australia
| | - Carmel M O'Brien
- CSIRO Manufacturing, Bag 10, Clayton South MDC, VIC 3168, Australia; Australian Regenerative Medicine Institute, Science, Technology, Research and Innovation Precinct (STRIP), Monash University, Clayton Campus, Wellington Road, Clayton, VIC 3800, Australia
| | - Neil R Cameron
- Department of Materials Science and Engineering, Monash University, 22 Alliance Lane, Clayton, VIC 3800, Australia.
| |
Collapse
|
26
|
miR-18a counteracts AKT and ERK activation to inhibit the proliferation of pancreatic progenitor cells. Sci Rep 2017; 7:45002. [PMID: 28332553 PMCID: PMC5362961 DOI: 10.1038/srep45002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 02/17/2017] [Indexed: 12/19/2022] Open
Abstract
Activation of endogenous stem/progenitor cells to repair injured tissues is an ideal option for disease treatment. However, adult pancreatic progenitor cells remain in a quiescent state in vivo. Thus, it is difficult to stimulate proliferation and differentiation in these progenitor cells, and the cause remains elusive. miR-17-92 cluster miRNAs are highly conserved in mammals and are expressed in multiple tissue stem/progenitor cells, but their role in pancreatic progenitor cells are less well known. In the present study, we demonstrate that miR-18a, but not the other members of the miR-17-92 gene cluster, inhibits the proliferation of pancreatic progenitor cells in vitro and ex vivo. miR-18a inhibits proliferation of adult pancreatic progenitor cells through arresting the cell cycle at G1 stage, indicating that miR-18a plays a role in keeping the adult pancreatic progenitor cells in quiescence. miR-18a inhibits pancreatic progenitor proliferation by targeting the gene expressions of connective tissue growth factor (CTGF), neural precursor cell expressed, developmentally down-regulated 9 (Nedd9), and cyclin dependent kinase 19 (CDK19), as well as by suppressing activation of the proliferation-related signaling pathways phosphatidylinositol 3-kinase–protein kinase B (PI3K/AKT) and extracellular signal-regulated kinase (ERK).
Collapse
|
27
|
Casselli T, Qureshi H, Peterson E, Perley D, Blake E, Jokinen B, Abbas A, Nechaev S, Watt JA, Dhasarathy A, Brissette CA. MicroRNA and mRNA Transcriptome Profiling in Primary Human Astrocytes Infected with Borrelia burgdorferi. PLoS One 2017; 12:e0170961. [PMID: 28135303 PMCID: PMC5279786 DOI: 10.1371/journal.pone.0170961] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 01/14/2017] [Indexed: 02/07/2023] Open
Abstract
Lyme disease is caused by infection with the bacterium Borrelia burgdorferi (Bb), which is transmitted to humans by deer ticks. The infection manifests usually as a rash and minor systemic symptoms; however, the bacteria can spread to other tissues, causing joint pain, carditis, and neurological symptoms. Lyme neuroborreliosis presents itself in several ways, such as Bell's palsy, meningitis, and encephalitis. The molecular basis for neuroborreliosis is poorly understood. Analysis of the changes in the expression levels of messenger RNAs and non-coding RNAs, including microRNAs, following Bb infection could therefore provide vital information on the pathogenesis and clinical symptoms of neuroborreliosis. To this end, we used cultured primary human astrocytes, key responders to CNS infection and important components of the blood-brain barrier, as a model system to study RNA and microRNA changes in the CNS caused by Bb. Using whole transcriptome RNA-seq, we found significant changes in 38 microRNAs and 275 mRNAs at 24 and 48 hours following Bb infection. Several of the RNA changes affect pathways involved in immune response, development, chromatin assembly (including histones) and cell adhesion. Further, several of the microRNA predicted target mRNAs were also differentially regulated. Overall, our results indicate that exposure to Bb causes significant changes to the transcriptome and microRNA profile of astrocytes, which has implications in the pathogenesis, and hence potential treatment strategies to combat this disease.
Collapse
Affiliation(s)
- Timothy Casselli
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Humaira Qureshi
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Elizabeth Peterson
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Danielle Perley
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Emily Blake
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Bradley Jokinen
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Ata Abbas
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Sergei Nechaev
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - John A. Watt
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Archana Dhasarathy
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| | - Catherine A. Brissette
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States of America
| |
Collapse
|
28
|
Vasconcellos R, Alvarenga ÉC, Parreira RC, Lima SS, Resende RR. Exploring the cell signalling in hepatocyte differentiation. Cell Signal 2016; 28:1773-88. [DOI: 10.1016/j.cellsig.2016.08.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/18/2016] [Accepted: 08/18/2016] [Indexed: 02/08/2023]
|
29
|
Chu J, Tu Y, Chen J, Tan D, Liu X, Pi R. Effects of melatonin and its analogues on neural stem cells. Mol Cell Endocrinol 2016; 420:169-79. [PMID: 26499395 DOI: 10.1016/j.mce.2015.10.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Revised: 09/27/2015] [Accepted: 10/18/2015] [Indexed: 12/30/2022]
Abstract
Neural stem cells (NSCs) are multipotent cells which are capable of self-replication and differentiation into neurons, astrocytes or oligodendrocytes in the central nervous system (CNS). NSCs are found in two main regions in the adult brain: the subgranular zone (SGZ) in the hippocampal dentate gyrus (DG) and the subventricular zone (SVZ). The recent discovery of NSCs in the adult mammalian brain has fostered a plethora of translational and preclinical studies to investigate novel approaches for the therapy of neurodegenerative diseases. Melatonin is the major secretory product synthesized and secreted by the pineal gland and shows both a wide distribution within phylogenetically distant organisms from bacteria to humans and a great functional versatility. Recently, accumulated experimental evidence showed that melatonin plays an important role in NSCs, including its proliferation, differentiation and survival, which are modulated by many factors including MAPK/ERK signaling pathway, histone acetylation, neurotrophic factors, transcription factors, and apoptotic genes. The purpose of this review is to summarize the beneficial effects of melatonin on NSCs and further to discuss the potential usage of melatonin and its derivatives or analogues in the treatment of CNS neurodegenerative diseases.
Collapse
Affiliation(s)
- Jiaqi Chu
- Department of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510080, China; International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of Guangdong, Guangzhou 510006, China; National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yalin Tu
- Department of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510080, China; International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of Guangdong, Guangzhou 510006, China; National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jingkao Chen
- Department of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510080, China; International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of Guangdong, Guangzhou 510006, China; National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-Sen University, Guangzhou 510080, China
| | - Dunxian Tan
- Department of Cellular and Structural Biology, The University of Texas, Health Science Center at San Antonio, 7703 Floyd Curl, San Antonio, TX 78229, USA
| | - Xingguo Liu
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, China
| | - Rongbiao Pi
- Department of Pharmacology & Toxicology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510080, China; International Joint Laboratory (SYSU-PolyU HK) of Novel Anti-Dementia Drugs of Guangdong, Guangzhou 510006, China; National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-Sen University, Guangzhou 510080, China; Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, 74 Zhongshan 2nd Road, Guangzhou 510080, China.
| |
Collapse
|
30
|
Stevanato L, Thanabalasundaram L, Vysokov N, Sinden JD. Investigation of Content, Stoichiometry and Transfer of miRNA from Human Neural Stem Cell Line Derived Exosomes. PLoS One 2016; 11:e0146353. [PMID: 26752061 PMCID: PMC4713432 DOI: 10.1371/journal.pone.0146353] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 12/16/2015] [Indexed: 01/05/2023] Open
Abstract
Exosomes are small (30–100 nm) membrane vesicles secreted by a variety of cell types and only recently have emerged as a new avenue for cell-to-cell communication. They are natural shuttles of RNA and protein cargo, making them attractive as potential therapeutic delivery vehicles. MicroRNAs (miRNAs) are short non-coding RNAs which regulate biological processes and can be found in exosomes. Here we characterized the miRNA contents of exosomes derived from human neural stem cells (hNSCs). Our investigated hNSC line is a clonal, conditionally immortalized cell line, compliant with good manufacturing practice (GMP), and in clinical trials for stroke and critical limb ischemia in the UK (clinicaltrials.gov: NCT01151124, NCT02117635, and NCT01916369). By using next generation sequencing (NGS) technology we identified the presence of a variety of miRNAs in both exosomal and cellular preparations. Many of these miRNAs were enriched in exosomes indicating that cells specifically sort them for extracellular release. Although exosomes have been proven to contain miRNAs, the copy number quantification per exosome of a given miRNA remains unclear. Herein we quantified by real-time PCR a highly shuttled exosomal miRNA subtype (hsa-miR-1246) in order to assess its stoichiometry per exosome. Furthermore, we utilized an in vitro system to confirm its functional transfer by measuring the reduction in luciferase expression using a 3’ untranslated region dual luciferase reporter assay. In summary, NGS analysis allowed the identification of a unique set of hNSC derived exosomal miRNAs. Stoichiometry and functional transfer analysis of one of the most abundant identified miRNA, hsa-miR-1246, were measured to support biological relevance of exosomal miRNA delivery.
Collapse
Affiliation(s)
- Lara Stevanato
- Stem Cell Discovery, ReNeuron, Guildford, United Kingdom
- * E-mail:
| | | | - Nickolai Vysokov
- Stem Cell Discovery, ReNeuron, Guildford, United Kingdom
- Wolfson CARD, Kings College London, Guys Campus, London, United Kingdom
| | - John D. Sinden
- Stem Cell Discovery, ReNeuron, Guildford, United Kingdom
| |
Collapse
|
31
|
Petersen GF, Hilbert BJ, Trope GD, Kalle WHJ, Strappe PM. Direct Conversion of Equine Adipose-Derived Stem Cells into Induced Neuronal Cells Is Enhanced in Three-Dimensional Culture. Cell Reprogram 2015; 17:419-26. [PMID: 26579833 DOI: 10.1089/cell.2015.0046] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The ability to culture neurons from horses may allow further investigation into equine neurological disorders. In this study, we demonstrate the generation of induced neuronal cells from equine adipose-derived stem cells (EADSCs) using a combination of lentiviral vector expression of the neuronal transcription factors Brn2, Ascl1, Myt1l (BAM) and NeuroD1 and a defined chemical induction medium, with βIII-tubulin-positive induced neuronal cells displaying a distinct neuronal morphology of rounded and compact cell bodies, extensive neurite outgrowth, and branching of processes. Furthermore, we investigated the effects of dimensionality on neuronal transdifferentiation, comparing conventional two-dimensional (2D) monolayer culture against three-dimensional (3D) culture on a porous polystyrene scaffold. Neuronal transdifferentiation was enhanced in 3D culture, with evenly distributed cells located on the surface and throughout the scaffold. Transdifferentiation efficiency was increased in 3D culture, with an increase in mean percent conversion of more than 100% compared to 2D culture. Additionally, induced neuronal cells were shown to transit through a Nestin-positive precursor state, with MAP2 and Synapsin 2 expression significantly increased in 3D culture. These findings will help to increase our understanding of equine neuropathogenesis, with prospective roles in disease modeling, drug screening, and cellular replacement for treatment of equine neurological disorders.
Collapse
Affiliation(s)
- Gayle F Petersen
- 1 School of Biomedical Sciences, Charles Sturt University , Wagga Wagga, New South Wales, Australia
| | - Bryan J Hilbert
- 2 School of Animal and Veterinary Sciences, Charles Sturt University , Wagga Wagga, New South Wales, Australia
| | - Gareth D Trope
- 2 School of Animal and Veterinary Sciences, Charles Sturt University , Wagga Wagga, New South Wales, Australia
| | - Wouter H J Kalle
- 1 School of Biomedical Sciences, Charles Sturt University , Wagga Wagga, New South Wales, Australia
| | - Padraig M Strappe
- 1 School of Biomedical Sciences, Charles Sturt University , Wagga Wagga, New South Wales, Australia
| |
Collapse
|
32
|
Horsham JL, Ganda C, Kalinowski FC, Brown RAM, Epis MR, Leedman PJ. MicroRNA-7: A miRNA with expanding roles in development and disease. Int J Biochem Cell Biol 2015; 69:215-24. [PMID: 26546742 DOI: 10.1016/j.biocel.2015.11.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 11/01/2015] [Accepted: 11/02/2015] [Indexed: 12/15/2022]
Abstract
MicroRNAs (miRNAs) are a family of short, non-coding RNA molecules (∼22nt) involved in post-transcriptional control of gene expression. They act via base-pairing with mRNA transcripts that harbour target sequences, resulting in accelerated mRNA decay and/or translational attenuation. Given miRNAs mediate the expression of molecules involved in many aspects of normal cell development and functioning, it is not surprising that aberrant miRNA expression is closely associated with many human diseases. Their pivotal role in driving a range of normal cellular physiology as well as pathological processes has established miRNAs as potential therapeutics, as well as potential diagnostic and prognostic tools in human health. MicroRNA-7 (miR-7) is a highly conserved miRNA which displays restricted spatiotemporal expression during development and in maturity. In humans and mice, mature miR-7 is generated from three different genes, illustrating unexpected redundancy and also the importance of this miRNA in regulating key cellular processes. In this review we examine the expanding role of miR-7 in the context of health, with emphasis on organ differentiation and development, as well as in various mammalian diseases, particularly of the brain, heart, endocrine pancreas and skin, as well as in cancer. The more we learn about miR-7, the more we realise the complexity of its regulation and potential functional application both from a biomarker and therapeutic perspective.
Collapse
Affiliation(s)
- Jessica L Horsham
- Laboratory for Cancer Medicine, Harry Perkins Institute of Medical Research, The University of Western Australia Centre for Medical Research, Perth, WA 6000, Australia; School of Medicine and Pharmacology, University of Western Australia, Nedlands, WA 6009, Australia
| | - Clarissa Ganda
- Laboratory for Cancer Medicine, Harry Perkins Institute of Medical Research, The University of Western Australia Centre for Medical Research, Perth, WA 6000, Australia
| | - Felicity C Kalinowski
- Laboratory for Cancer Medicine, Harry Perkins Institute of Medical Research, The University of Western Australia Centre for Medical Research, Perth, WA 6000, Australia
| | - Rikki A M Brown
- Laboratory for Cancer Medicine, Harry Perkins Institute of Medical Research, The University of Western Australia Centre for Medical Research, Perth, WA 6000, Australia
| | - Michael R Epis
- Laboratory for Cancer Medicine, Harry Perkins Institute of Medical Research, The University of Western Australia Centre for Medical Research, Perth, WA 6000, Australia
| | - Peter J Leedman
- Laboratory for Cancer Medicine, Harry Perkins Institute of Medical Research, The University of Western Australia Centre for Medical Research, Perth, WA 6000, Australia; School of Medicine and Pharmacology, University of Western Australia, Nedlands, WA 6009, Australia.
| |
Collapse
|
33
|
MicroRNA delivery for regenerative medicine. Adv Drug Deliv Rev 2015; 88:108-22. [PMID: 26024978 DOI: 10.1016/j.addr.2015.05.014] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 05/13/2015] [Accepted: 05/21/2015] [Indexed: 12/26/2022]
Abstract
MicroRNA (miRNA) directs post-transcriptional regulation of a network of genes by targeting mRNA. Although relatively recent in development, many miRNAs direct differentiation of various stem cells including induced pluripotent stem cells (iPSCs), a major player in regenerative medicine. An effective and safe delivery of miRNA holds the key to translating miRNA technologies. Both viral and nonviral delivery systems have seen success in miRNA delivery, and each approach possesses advantages and disadvantages. A number of studies have demonstrated success in augmenting osteogenesis, improving cardiogenesis, and reducing fibrosis among many other tissue engineering applications. A scaffold-based approach with the possibility of local and sustained delivery of miRNA is particularly attractive since the physical cues provided by the scaffold may synergize with the biochemical cues induced by miRNA therapy. Herein, we first briefly cover the application of miRNA to direct stem cell fate via replacement and inhibition therapies, followed by the discussion of the promising viral and nonviral delivery systems. Next we present the unique advantages of a scaffold-based delivery in achieving lineage-specific differentiation and tissue development.
Collapse
|
34
|
Li X, Li Z, Yang G, Pan Z. MicroRNA-338-3p suppresses tumor growth of esophageal squamous cell carcinoma in vitro and in vivo. Mol Med Rep 2015; 12:3951-3957. [PMID: 26004521 DOI: 10.3892/mmr.2015.3820] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 04/30/2015] [Indexed: 11/06/2022] Open
Abstract
Accumulating evidence has shown that microRNAs (miRNAs) are aberrantly expressed in human esophageal squamous cell carcinoma (ESCC) and are crucial in tumorigenesis, among which miR‑338‑3p has been examined to be downregulated in patients with ESCC. However, the role of miR‑338‑3p in ESCC remains to be elucidated. In the present study, the role of miR‑338‑3p on the growth and survival of an ESCC cell line was determined with several in vitro approaches and in nude mouse models. It was determined that miR‑338‑3p expression was frequently downregulated in ESCC tissue compared with corresponding adjacent non‑tumor tissue, and that its expression was significantly correlated with tumor stage and metastasis. Overexpression of miR‑338‑3p in ESCC cells suppressed cell proliferation, colony formation, migration and invasion, and induced cell arrest at the G0/G1 stage and cell apoptosis in vitro. In addition, it was demonstrated that overexpression of miR‑338‑3p significantly suppresses tumor growth of xenograft tumors in mice (P<0.05). These findings revealed that miR‑338‑3p may act as a tumor suppressor in ESCC, and its dysregulation may be involved in the initiation and development of human ESCC. In addition, it was suggested that miR‑338‑3p may be a potential therapeutic agent for treatment of ESCC.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Anesthesiology, The Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| | - Zhihong Li
- Department of Thoracic Surgery, The First Hospital, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Guiyun Yang
- Department of Anesthesiology, The Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| | - Zhenxiang Pan
- Department of Anesthesiology, The Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
35
|
Zhang L, Li Z, Gai F, Wang Y. MicroRNA-137 suppresses tumor growth in epithelial ovarian cancer in vitro and in vivo. Mol Med Rep 2015; 12:3107-14. [PMID: 25955305 DOI: 10.3892/mmr.2015.3756] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 04/08/2015] [Indexed: 11/05/2022] Open
Abstract
Epithelial ovarian cancer (EOC) remains a major gynecological problem, with a poor 5-year-survival rate due to distant metastases. The identification of microRNAs (miRNAs) may provide a novel avenue for diagnostic and treatment regimens for EOC. Several miRNAs have been reported to be involved in the progression of EOC, among which miRNA (miR)-137 has been observed to be downregulated in the ovarian tissues of patients with EOC. However, the functions of miR-137 in EOC cell apoptosis, migration and invasion remain to be elucidated. In the present study, the expression of miR-137 was measured in clinical ovarian cancer specimens and cell lines using reverse transcription-quantitative polymerase chain reaction. The role of miR-137 in the growth and survival of the SKOV3 human ovarian cancer cell line was determined using several in vitro approaches and in nude mouse models. The results demonstrated that the expression of miR-137 was downregulated in the ovarian cancer specimens and cell lines. It was also observed that enforced expression of miR-137 in the EOC cell lines decreased cell proliferation, clonogenicity, migration and invasion, and induced G1 arrest and cell apoptosis in vitro. Notably, the enforced expression of miR-137 suppressed tumor growth in the nude mice models. These findings suggested that miR-137 may act as a tumor suppressor and be used as a potential therapeutic agent for the treatment of EOC.
Collapse
Affiliation(s)
- Liyu Zhang
- Department of Obstetrics and Gynecology, The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Zhihong Li
- Department of Obstetrics and Gynecology, The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Fengchun Gai
- Department of Infectious Diseases, The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| | - Yanping Wang
- Department of Obstetrics and Gynecology, The Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, Jilin 130117, P.R. China
| |
Collapse
|
36
|
Stevanato L, Hicks C, Sinden JD. Differentiation of a Human Neural Stem Cell Line on Three Dimensional Cultures, Analysis of MicroRNA and Putative Target Genes. J Vis Exp 2015:52410. [PMID: 25938519 PMCID: PMC4541566 DOI: 10.3791/52410] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Neural stem cells (NSCs) are capable of self-renewal and differentiation into neurons, astrocytes and oligodendrocytes under specific local microenvironments. In here, we present a set of methods used for three dimensional (3D) differentiation and miRNA analysis of a clonal human neural stem cell (hNSC) line, currently in clinical trials for stroke disability (NCT01151124 and NCT02117635, Clinicaltrials.gov). HNSCs were derived from an ethical approved first trimester human fetal cortex and conditionally immortalized using retroviral integration of a single copy of the c-mycER(TAM)construct. We describe how to measure axon process outgrowth of hNSCs differentiated on 3D scaffolds and how to quantify associated changes in miRNA expression using PCR array. Furthermore we exemplify computational analysis with the aim of selecting miRNA putative targets. SOX5 and NR4A3 were identified as suitable miRNA putative target of selected significantly down-regulated miRNAs in differentiated hNSC. MiRNA target validation was performed on SOX5 and NR4A3 3'UTRs by dual reporter plasmid transfection and dual luciferase assay.
Collapse
|
37
|
Arslan SY, Yu Y, Burdette JE, Pavone ME, Hope TJ, Woodruff TK, Kim JJ. Novel three dimensional human endocervix cultures respond to 28-day hormone treatment. Endocrinology 2015; 156:1602-9. [PMID: 25635622 PMCID: PMC4399320 DOI: 10.1210/en.2014-1840] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The endocervix has both anatomical and biological functions that participate in the delicate balance between tolerance necessary for conception and protection from pathogens. Our goal was to develop a robust 3-dimensional (3D) endocervix model that was a reliable representation of the in vivo tissues and to identify the physiological responses to changing levels of steroid hormones during a 28-day time period. Human endocervical cells were grown on polystyrene scaffolds, and the morphologic and hormonal responses of cultured cells were assessed in response to fluctuating levels of estradiol (E2) or progesterone (P4). Morphologically, the 3D cultures were composed of a mixed population of cells, including epithelial and stromal cells. Treatment with E2 and P4 (d 28) increased cell growth and proliferation as compared with no treatment control. Cells expressed estrogen receptor and P4 receptor and produced both neutral and acidic mucins, including Mucin 16. In addition, a 45-plex Luminex assay identified numerous factors secreted and regulated by hormones. Specifically, IL-1β and leukemia inhibitory factor significantly decreased in the presence of E2 and P4 as compared with the no hormone control at day 26. Cotreatment with RU486 (mifepristone) attenuated the inhibition of IL-1β and leukemia inhibitory factor secretion. In summary, a robust, novel 3D endocervical culture was developed, and physiologic responses to the menstrual cycle mimic of E2 and P4 levels for a period of 28 days were identified.
Collapse
Affiliation(s)
- Sevim Yildiz Arslan
- Division of Reproductive Science in Medicine (S.Y.A., Y.Y., M.E.P., T.K.W., J.J.K.), Department of Obstetrics and Gynecology, and Department of Cell and Molecular Biology (T.J.H.), Northwestern University, Chicago, Illinois 60611; Department of Medicinal Chemistry and Pharmacognosy (J.E.B.), University of Illinois at Chicago, Chicago, Illinois 60607; and Division of Reproductive Endocrinology and Infertility (M.E.P.), Department of Obstetrics and Gynecology, Northwestern University, Chicago, Illinois 60611
| | | | | | | | | | | | | |
Collapse
|
38
|
Wen C, Liu X, Ma H, Zhang W, Li H. miR‑338‑3p suppresses tumor growth of ovarian epithelial carcinoma by targeting Runx2. Int J Oncol 2015; 46:2277-85. [PMID: 25776272 DOI: 10.3892/ijo.2015.2929] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 03/02/2015] [Indexed: 11/06/2022] Open
Abstract
miR‑338‑3p, a recently discovered miRNA, has been shown to play important roles in tumorigenesis and metastasis in various cancers. However, the exact roles and mechanisms of miR‑338‑3p remain unknown in human ovarian epithelial carcinoma (EOC). The relationship between miR‑338‑3p expression pattern and clinicopathological features of patients with EOC were determined by real-time quantitative RT-PCR. Furthermore, the role of miR‑338‑3p and possible molecular mechanisms in EOC was investigated by several in vitro approaches and in a nude mouse model. We first showed that the expression of miR‑338‑3p was significantly downregulated in EOC tissues compared to those in adjacent normal tissues, and the value was negatively related to advanced FIGO stage, high histological grading and lymph node metastasis (P<0.01). An in vitro analysis revealed that the overexpression of miR‑338‑3p in EOC cells significantly inhibited cell proliferation, colony formation, migration and invasion, inducing cell apoptosis and enhancing caspase-3, -8, and -9 activities. Bioinformatic analysis and dual luciferase assays identified Runx2 as a direct target of miR‑338‑3p. We also found that enforced expression of miR‑338‑3p markedly inhibited the in vivo tumorigenicity in a nude mouse xenograft model system. Furthermore, overexpression of miR‑338‑3p inhibited phosphorylation of PI3K and AKT, which contributed to suppression of ovarian cancer cell growth. These findings revealed that miR‑338‑3p may act as a tumor suppressor that blocks the growth of human ovarian epithelial carcinoma through PI3K/AKT signaling pathways by targeting Runx2.
Collapse
Affiliation(s)
- Chunyan Wen
- Department of Pathology, China-Japan Union Hospital of Jilin University, Nanguan District, Changchun 13033, P.R. China
| | - Xiaojun Liu
- Department of Obstetrics and Gynecology, China-Japan Union Hospital of Jilin University, Nanguan District, Changchun 13033, P.R. China
| | - Hongxi Ma
- Department of Pathology, The First Hospital of Jilin University, Changchun 130021, P.R. China
| | - Wenjie Zhang
- Department of Pathology, China-Japan Union Hospital of Jilin University, Nanguan District, Changchun 13033, P.R. China
| | - Haifeng Li
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun 130021, P.R. China
| |
Collapse
|
39
|
Castro-Vega LJ, Letouzé E, Burnichon N, Buffet A, Disderot PH, Khalifa E, Loriot C, Elarouci N, Morin A, Menara M, Lepoutre-Lussey C, Badoual C, Sibony M, Dousset B, Libé R, Zinzindohoue F, Plouin PF, Bertherat J, Amar L, de Reyniès A, Favier J, Gimenez-Roqueplo AP. Multi-omics analysis defines core genomic alterations in pheochromocytomas and paragangliomas. Nat Commun 2015; 6:6044. [PMID: 25625332 DOI: 10.1038/ncomms7044] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 12/05/2014] [Indexed: 01/07/2023] Open
Abstract
Pheochromocytomas and paragangliomas (PCCs/PGLs) are neural crest-derived tumours with a very strong genetic component. Here we report the first integrated genomic examination of a large collection of PCC/PGL. SNP array analysis reveals distinct copy-number patterns associated with genetic background. Whole-exome sequencing shows a low mutation rate of 0.3 mutations per megabase, with few recurrent somatic mutations in genes not previously associated with PCC/PGL. DNA methylation arrays and miRNA sequencing identify DNA methylation changes and miRNA expression clusters strongly associated with messenger RNA expression profiling. Overexpression of the miRNA cluster 182/96/183 is specific in SDHB-mutated tumours and induces malignant traits, whereas silencing of the imprinted DLK1-MEG3 miRNA cluster appears as a potential driver in a subgroup of sporadic tumours. Altogether, the complete genomic landscape of PCC/PGL is mainly driven by distinct germline and/or somatic mutations in susceptibility genes and reveals different molecular entities, characterized by a set of unique genomic alterations.
Collapse
Affiliation(s)
- Luis Jaime Castro-Vega
- 1] INSERM, UMR970, Paris-Cardiovascular Research Center, F-75015 Paris, France [2] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, F-75006 Paris, France
| | - Eric Letouzé
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre Le Cancer, 75013 Paris, France
| | - Nelly Burnichon
- 1] INSERM, UMR970, Paris-Cardiovascular Research Center, F-75015 Paris, France [2] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, F-75006 Paris, France [3] Department of Genetics, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | - Alexandre Buffet
- 1] INSERM, UMR970, Paris-Cardiovascular Research Center, F-75015 Paris, France [2] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, F-75006 Paris, France [3] Department of Genetics, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | - Pierre-Hélie Disderot
- 1] INSERM, UMR970, Paris-Cardiovascular Research Center, F-75015 Paris, France [2] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, F-75006 Paris, France
| | - Emmanuel Khalifa
- 1] INSERM, UMR970, Paris-Cardiovascular Research Center, F-75015 Paris, France [2] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, F-75006 Paris, France [3] Department of Genetics, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | - Céline Loriot
- 1] INSERM, UMR970, Paris-Cardiovascular Research Center, F-75015 Paris, France [2] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, F-75006 Paris, France
| | - Nabila Elarouci
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre Le Cancer, 75013 Paris, France
| | - Aurélie Morin
- 1] INSERM, UMR970, Paris-Cardiovascular Research Center, F-75015 Paris, France [2] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, F-75006 Paris, France
| | - Mélanie Menara
- 1] INSERM, UMR970, Paris-Cardiovascular Research Center, F-75015 Paris, France [2] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, F-75006 Paris, France
| | - Charlotte Lepoutre-Lussey
- 1] INSERM, UMR970, Paris-Cardiovascular Research Center, F-75015 Paris, France [2] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, F-75006 Paris, France [3] Hypertension Unit, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | - Cécile Badoual
- 1] INSERM, UMR970, Paris-Cardiovascular Research Center, F-75015 Paris, France [2] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, F-75006 Paris, France [3] Department of Pathology, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | - Mathilde Sibony
- 1] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, F-75006 Paris, France [2] Department of Pathology, Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, F-75006 Paris, France
| | - Bertrand Dousset
- 1] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, F-75006 Paris, France [2] Department of Digestive and Endocrine Surgery, Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, F-75006 Paris, France [3] INSERM, U1016, Institut Cochin, F-75006 Paris, France [4] CNRS UMR8104, F-75006 Paris, France
| | - Rossella Libé
- 1] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, F-75006 Paris, France [2] INSERM, U1016, Institut Cochin, F-75006 Paris, France [3] CNRS UMR8104, F-75006 Paris, France [4] Department of Endocrinology, Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, F-75006 Paris, France [5] Rare Adrenal Cancer Network COMETE, F-75006 Paris, France
| | - Franck Zinzindohoue
- 1] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, F-75006 Paris, France [2] Department of Surgery, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | - Pierre François Plouin
- 1] INSERM, UMR970, Paris-Cardiovascular Research Center, F-75015 Paris, France [2] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, F-75006 Paris, France [3] Hypertension Unit, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, F-75015 Paris, France [4] Rare Adrenal Cancer Network COMETE, F-75006 Paris, France
| | - Jérôme Bertherat
- 1] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, F-75006 Paris, France [2] INSERM, U1016, Institut Cochin, F-75006 Paris, France [3] CNRS UMR8104, F-75006 Paris, France [4] Department of Endocrinology, Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, F-75006 Paris, France [5] Rare Adrenal Cancer Network COMETE, F-75006 Paris, France
| | - Laurence Amar
- 1] INSERM, UMR970, Paris-Cardiovascular Research Center, F-75015 Paris, France [2] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, F-75006 Paris, France [3] Hypertension Unit, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, F-75015 Paris, France
| | - Aurélien de Reyniès
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre Le Cancer, 75013 Paris, France
| | - Judith Favier
- 1] INSERM, UMR970, Paris-Cardiovascular Research Center, F-75015 Paris, France [2] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, F-75006 Paris, France
| | - Anne-Paule Gimenez-Roqueplo
- 1] INSERM, UMR970, Paris-Cardiovascular Research Center, F-75015 Paris, France [2] Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, F-75006 Paris, France [3] Department of Genetics, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, F-75015 Paris, France [4] Rare Adrenal Cancer Network COMETE, F-75006 Paris, France
| |
Collapse
|
40
|
Silveyra P, Chroneos ZC, DiAngelo SL, Thomas NJ, Noutsios GT, Tsotakos N, Howrylak JA, Umstead TM, Floros J. Knockdown of Drosha in human alveolar type II cells alters expression of SP-A in culture: a pilot study. Exp Lung Res 2014; 40:354-66. [PMID: 25058539 DOI: 10.3109/01902148.2014.929757] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Human surfactant protein A (SP-A) plays an important role in surfactant metabolism and lung innate immunity. SP-A is synthesized and secreted by alveolar type II (ATII) cells, one of the two cell types of the distal lung epithelium (ATII and ATI). We have shown that miRNA interactions with sequence polymorphisms on the SP-A mRNA 3'UTRs mediate differential expression of SP-A1 and SP-A2 gene variants in vitro. In the present study, we describe a physiologically relevant model to study miRNA regulation of SP-A in human ATII. For these studies, we purified and cultured human ATII on an air-liquid interface matrix (A/L) or plastic wells without matrix (P). Gene expression analyses confirmed that cells cultured in A/L maintained the ATII phenotype for over 5 days, whereas P-cultured cells differentiated to ATI. When we transfected ATII with siRNAs to inhibit the expression of Drosha, a critical effector of miRNA maturation, the levels of SP-A mRNA and protein increased in a time dependent manner. We next characterized cultured ATII and ATI by studying expression of 1,066 human miRNAs using miRNA PCR arrays. We detected expression of >300 miRNAs with 24 miRNAs differentially expressed in ATII versus ATI, 12 of which predicted to bind SP-A 3'UTRs, indicating that these may be implicated in SP-A downregulation in ATI. Thus, miRNAs not only affect SP-A expression, but also may contribute to the maintenance of the ATII cell phenotype and/or the trans-differentiation of ATII to ATI cells, and may represent new molecular markers that distinguish ATII and ATI.
Collapse
Affiliation(s)
- Patricia Silveyra
- 1Center for Host Defense, Inflammation, and Lung Disease (CHILD) Research, Department of Pediatrics, The Pennsylvania State University College of Medicine , Hershey, Pennsylvania , USA
| | | | | | | | | | | | | | | | | |
Collapse
|