1
|
Wang LJ, Tsai CC, Chao HR, Lee SY, Chen CC, Li SC. MicroRNAs in Umbilical Cord Blood and Development in Full-Term Newborns: A Prospective Study. Biomark Insights 2024; 19:11772719241258017. [PMID: 38863527 PMCID: PMC11165956 DOI: 10.1177/11772719241258017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 05/10/2024] [Indexed: 06/13/2024] Open
Abstract
Background Exploring the epigenetic regulations, such as microRNA, in newborns holds significant promise for enhancing our ability to address and potentially prevent early-life developmental delays. Objectives Hence, this research seeks to investigate if the expression of miRNA in the umbilical cord blood of infants can forecast their developmental outcomes as they grow older. Design and method We enrolled 143 full-term newborns, delivered either via cesarean section (CS) or through natural spontaneous delivery (NSD). We then analyzed the profiles of specific miRNAs (miR-486-5p, miR-126-5p, miR-140-3p, miR-151a-3p, miR-142-5p, and miR-30e-5p) in the umbilical cord blood of these infants. Subsequently, we performed follow-up assessments using Bayley-III scores when the cohort reached 1 year of age. Furthermore, we conducted pathway-enrichment analyses on the target genes associated with these examined miRNAs. Results When comparing newborns delivered via cesarean section (CS) to those born via natural spontaneous delivery (NSD), we observed notable differences. Specifically, newborns through NSD displayed significantly higher ΔCt values for miR-486-5p, alongside lower ΔCt values for miR-126-5p and miR-151a-3p in their cord blood. At 1 year of age, cognitive development was significantly linked to the ΔCt values of miR-140-3p and miR-142-5p, while language development showed a significant association with the ΔCt values of miR-140-3p. Moreover, our pathway enrichment analyses revealed that the target genes of these miRNAs were consistently involved in the pathways related to neurons, such as axon guidance and the neurotrophin signaling pathway. Conclusion In summary, this study represents a pioneering effort in elucidating the potential connections between miRNA levels in cord blood and the health indicators and neurodevelopment of newborns at 1 year of age. Our findings underscore the significance of miRNA levels at birth in influencing mechanisms related to neurodevelopment.
Collapse
Affiliation(s)
- Liang-Jen Wang
- Department of Child and Adolescent Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Ching-Chang Tsai
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - How-Ran Chao
- Department of Environmental Science and Engineering, College of Engineering, National Pingtung University of Science and Technology, Pingtung County, Taiwan
| | - Sheng-Yu Lee
- Department of Psychiatry, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Department of Psychiatry, College of Medicine, Graduate Institute of Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Cheng Chen
- Section of Neonatology, Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
- Department of Early Childhood Care and Education, Cheng-Shiu University, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Sung-Chou Li
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- Department of Dental Technology, Shu-Zen Junior College of Medicine and Management, Kaohsiung, Taiwan
| |
Collapse
|
2
|
Mohammadi P, Nadri S, Abdanipour A, Mortazavi Y. Microchip encapsulation and microRNA-7 overexpression of trabecular meshwork mesenchymal stem/stromal cells improve motor function after spinal cord injury. J Biomed Mater Res A 2023; 111:1482-1494. [PMID: 37042544 DOI: 10.1002/jbm.a.37549] [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: 10/02/2022] [Revised: 03/24/2023] [Accepted: 04/04/2023] [Indexed: 04/13/2023]
Abstract
Manipulation of stem cells and microencapsulation through microfluidic chips has shown more promising results in treating complex conditions, such as spinal cord injury (SCI), than traditional treatments. This study aimed to investigate the potency of neural differentiation and its therapeutic role in SCI animal model of trabecular meshwork mesenchymal stem/stromal cells (TMMSCs) via miR-7 overexpression and microchip-encapsulated. TMMSCs are transduced with miR-7 via a lentiviral vector (TMMSCs-miR-7[+]) and encapsulated in alginate-reduced graphene oxide (alginate-rGO) hydrogel via a microfluidic chip. Neuronal differentiation of transduced cells in hydrogel (3D) and tissue cultures plate (2D) was assessed by expressing specific mRNAs and proteins. Further evaluation is being carried out through 3D and 2D TMMSCs-miR-7(+ and -) transplantation into the rat contusion SCI model. TMMSCs-miR-7(+) encapsulated in the microfluidic chip (miR-7-3D) increased nestin, β-tubulin III, and MAP-2 expression compared with 2D culture. Moreover, miR-7-3D could improve locomotor behavior in contusion SCI rats, decrease cavity size, and increase myelination. Our results revealed that miR-7 and alginate-rGO hydrogel were involved in the neuronal differentiation of TMMSCs in a time-dependent manner. In addition, the microfluidic-encapsulated miR-7 overexpression TMMSCs represented a better survival and integration of the transplanted cells and the repair of SCI. Collectively, the combination of miR-7 overexpression and encapsulation of TMMSCs in hydrogels may represent a promising new treatment for SCI.
Collapse
Affiliation(s)
- Parvin Mohammadi
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Samad Nadri
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
- Department of Medical Nanotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Zanjan Pharmaceutical Nanotechnology Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Alireza Abdanipour
- Department of Anatomy, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Yousef Mortazavi
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
- Cancer Gene Therapy Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
3
|
Ma L, He X, Wu Q. The Molecular Regulatory Mechanism in Multipotency and Differentiation of Wharton's Jelly Stem Cells. Int J Mol Sci 2023; 24:12909. [PMID: 37629090 PMCID: PMC10454700 DOI: 10.3390/ijms241612909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/06/2023] [Accepted: 08/10/2023] [Indexed: 08/27/2023] Open
Abstract
Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) are isolated from Wharton's jelly tissue of umbilical cords. They possess the ability to differentiate into lineage cells of three germ layers. WJ-MSCs have robust proliferative ability and strong immune modulation capacity. They can be easily collected and there are no ethical problems associated with their use. Therefore, WJ-MSCs have great tissue engineering value and clinical application prospects. The identity and functions of WJ-MSCs are regulated by multiple interrelated regulatory mechanisms, including transcriptional regulation and epigenetic modifications. In this article, we summarize the latest research progress on the genetic/epigenetic regulation mechanisms and essential signaling pathways that play crucial roles in pluripotency and differentiation of WJ-MSCs.
Collapse
Affiliation(s)
| | | | - Qiang Wu
- The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| |
Collapse
|
4
|
Thomaidou AC, Goulielmaki M, Tsintarakis A, Zoumpourlis P, Toya M, Christodoulou I, Zoumpourlis V. miRNA-Guided Regulation of Mesenchymal Stem Cells Derived from the Umbilical Cord: Paving the Way for Stem-Cell Based Regeneration and Therapy. Int J Mol Sci 2023; 24:ijms24119189. [PMID: 37298143 DOI: 10.3390/ijms24119189] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/19/2023] [Accepted: 05/21/2023] [Indexed: 06/12/2023] Open
Abstract
The human body is an abundant source of multipotent cells primed with unique properties that can be exploited in a multitude of applications and interventions. Mesenchymal stem cells (MSCs) represent a heterogenous population of undifferentiated cells programmed to self-renew and, depending on their origin, differentiate into distinct lineages. Alongside their proven ability to transmigrate toward inflammation sites, the secretion of various factors that participate in tissue regeneration and their immunoregulatory function render MSCs attractive candidates for use in the cytotherapy of a wide spectrum of diseases and conditions, as well as in different aspects of regenerative medicine. In particular, MSCs that can be found in fetal, perinatal, or neonatal tissues possess additional capabilities, including predominant proliferation potential, increased responsiveness to environmental stimuli, and hypoimmunogenicity. Since microRNA (miRNA)-guided gene regulation governs multiple cellular functions, miRNAs are increasingly being studied in the context of driving the differentiation process of MSCs. In the present review, we explore the mechanisms of miRNA-directed differentiation of MSCs, with a special focus on umbilical cord-derived mesenchymal stem cells (UCMSCs), and we identify the most relevant miRNAs and miRNA sets and signatures. Overall, we discuss the potent exploitations of miRNA-driven multi-lineage differentiation and regulation of UCMSCs in regenerative and therapeutic protocols against a range of diseases and/or injuries that will achieve a meaningful clinical impact through maximizing treatment success rates, while lacking severe adverse events.
Collapse
Affiliation(s)
- Arsinoe C Thomaidou
- Laboratory of Clinical Virology, Medical School, University of Crete, 71500 Heraklion, Greece
| | - Maria Goulielmaki
- Cancer Immunology and Immunotherapy Center, Cancer Research Center, Saint Savas Cancer Hospital, 11522 Athens, Greece
| | - Antonis Tsintarakis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece
| | - Panagiotis Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece
| | - Marialena Toya
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece
| | - Ioannis Christodoulou
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece
| | - Vassilis Zoumpourlis
- Biomedical Applications Unit, Institute of Chemical Biology, National Hellenic Research Foundation (NHRF), 11635 Athens, Greece
| |
Collapse
|
5
|
miR-34a regulates silent synapse and synaptic plasticity in mature hippocampus. Prog Neurobiol 2023; 222:102404. [PMID: 36642095 DOI: 10.1016/j.pneurobio.2023.102404] [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: 10/18/2022] [Revised: 12/26/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
AMPAR-lacking silent synapses are prevailed and essential for synaptic refinement and synaptic plasticity in developing brains. In mature brain, they are sparse but could be induced under several pathological conditions. How they are regulated molecularly is far from clear. miR-34a is a highly conserved and brain-enriched microRNA with age-dependent upregulated expression profile. Its neuronal function in mature brain remains to be revealed. Here by analyzing synaptic properties of the heterozygous miR-34a knock out mice (34a_ht), we have discovered that mature but not juvenile 34a_ht mice have more silent synapses in the hippocampus accompanied with enhanced synaptic NMDAR but not AMPAR function and increased spine density. As a result, 34a_ht mice display enhanced long-term potentiation (LTP) in the Schaffer collateral synapses and better spatial learning and memory. We further found that Creb1 is a direct target of miR-34a, whose upregulation and activation may mediate the silent synapse increment in 34a_ht mice. Hence, we reveal a novel physiological role of miR-34a in mature brains and provide a molecular mechanism underlying silent synapse regulation.
Collapse
|
6
|
Korotkov A, Sim NS, Luinenburg MJ, Anink JJ, van Scheppingen J, Zimmer TS, Bongaarts A, Broekaart DWM, Mijnsbergen C, Jansen FE, Van Hecke W, Spliet WGM, van Rijen PC, Feucht M, Hainfellner JA, Kršek P, Zamecnik J, Crino PB, Kotulska K, Lagae L, Jansen AC, Kwiatkowski DJ, Jozwiak S, Curatolo P, Mühlebner A, Lee JH, Mills JD, van Vliet EA, Aronica E. MicroRNA-34a activation in tuberous sclerosis complex during early brain development may lead to impaired corticogenesis. Neuropathol Appl Neurobiol 2021; 47:796-811. [PMID: 33942341 PMCID: PMC8519131 DOI: 10.1111/nan.12717] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 01/26/2021] [Accepted: 04/19/2021] [Indexed: 12/14/2022]
Abstract
AIMS Tuberous sclerosis complex (TSC) is a genetic disorder associated with dysregulation of the mechanistic target of rapamycin complex 1 (mTORC1) signalling pathway. Neurodevelopmental disorders, frequently present in TSC, are linked to cortical tubers in the brain. We previously reported microRNA-34a (miR-34a) among the most upregulated miRs in tubers. Here, we characterised miR-34a expression in tubers with the focus on the early brain development and assessed the regulation of mTORC1 pathway and corticogenesis by miR-34a. METHODS We analysed the expression of miR-34a in resected cortical tubers (n = 37) compared with autopsy-derived control tissue (n = 27). The effect of miR-34a overexpression on corticogenesis was assessed in mice at E18. The regulation of the mTORC1 pathway and the expression of the bioinformatically predicted target genes were assessed in primary astrocyte cultures from three patients with TSC and in SH-SY5Y cells following miR-34a transfection. RESULTS The peak of miR-34a overexpression in tubers was observed during infancy, concomitant with the presence of pathological markers, particularly in giant cells and dysmorphic neurons. miR-34a was also strongly expressed in foetal TSC cortex. Overexpression of miR-34a in mouse embryos decreased the percentage of cells migrated to the cortical plate. The transfection of miR-34a mimic in TSC astrocytes negatively regulated mTORC1 and decreased the expression of the target genes RAS related (RRAS) and NOTCH1. CONCLUSIONS MicroRNA-34a is most highly overexpressed in tubers during foetal and early postnatal brain development. miR-34a can negatively regulate mTORC1; however, it may also contribute to abnormal corticogenesis in TSC.
Collapse
Affiliation(s)
- Anatoly Korotkov
- Department of (Neuro) PathologyAmsterdam UMCUniversity of AmsterdamAmsterdam NeuroscienceAmsterdamThe Netherlands
| | - Nam Suk Sim
- Graduate School of Medical Science and EngineeringKorea Advanced Institute of Science and TechnologyDaejeonRepublic of Korea
| | - Mark J. Luinenburg
- Department of (Neuro) PathologyAmsterdam UMCUniversity of AmsterdamAmsterdam NeuroscienceAmsterdamThe Netherlands
| | - Jasper J. Anink
- Department of (Neuro) PathologyAmsterdam UMCUniversity of AmsterdamAmsterdam NeuroscienceAmsterdamThe Netherlands
| | - Jackelien van Scheppingen
- Department of (Neuro) PathologyAmsterdam UMCUniversity of AmsterdamAmsterdam NeuroscienceAmsterdamThe Netherlands
- Department of NeuroimmunologyNetherlands Institute for NeuroscienceAmsterdamThe Netherlands
| | - Till S. Zimmer
- Department of (Neuro) PathologyAmsterdam UMCUniversity of AmsterdamAmsterdam NeuroscienceAmsterdamThe Netherlands
| | - Anika Bongaarts
- Department of (Neuro) PathologyAmsterdam UMCUniversity of AmsterdamAmsterdam NeuroscienceAmsterdamThe Netherlands
| | - Diede W. M. Broekaart
- Department of (Neuro) PathologyAmsterdam UMCUniversity of AmsterdamAmsterdam NeuroscienceAmsterdamThe Netherlands
| | - Caroline Mijnsbergen
- Department of (Neuro) PathologyAmsterdam UMCUniversity of AmsterdamAmsterdam NeuroscienceAmsterdamThe Netherlands
| | - Floor E. Jansen
- Department of Paediatric NeurologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Wim Van Hecke
- Department of PathologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Wim G. M. Spliet
- Department of PathologyUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Peter C. van Rijen
- University Medical CenterBrain CentreRudolf Magnus Institute for NeuroscienceUtrechtThe Netherlands
| | - Martha Feucht
- Department of PediatricsMedical University ViennaViennaAustria
| | | | - Pavel Kršek
- Department of Pediatric Neurology2nd Faculty of Medicine and Motol University HospitalPragueCzech Republic
| | - Josef Zamecnik
- Department of Pathology and Molecular Medicine2nd Faculty of Medicine and Motol University HospitalPragueCzech Republic
| | - Peter B. Crino
- Department of NeurologyUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Katarzyna Kotulska
- Department of Neurology and EpileptologyThe Children's Memorial Health InstituteWarsawPoland
| | - Lieven Lagae
- Department of Development and Regeneration‐Section Pediatric NeurologyUniversity Hospitals KU LeuvenLeuvenBelgium
| | - Anna C. Jansen
- Pediatric Neurology UnitUniversitair Ziekenhuis BrusselBrusselsBelgium
| | | | - Sergiusz Jozwiak
- Department of Neurology and EpileptologyThe Children's Memorial Health InstituteWarsawPoland
- Department of Child NeurologyMedical University of WarsawWarsawPoland
| | - Paolo Curatolo
- Child Neurology and Psychiatry UnitSystems Medicine DepartmentTor Vergata UniversityRomeItaly
| | - Angelika Mühlebner
- Department of (Neuro) PathologyAmsterdam UMCUniversity of AmsterdamAmsterdam NeuroscienceAmsterdamThe Netherlands
| | - Jeong H. Lee
- Graduate School of Medical Science and EngineeringKorea Advanced Institute of Science and TechnologyDaejeonRepublic of Korea
- SoVarGen, IncDaejeonRepublic of Korea
| | - James D. Mills
- Department of (Neuro) PathologyAmsterdam UMCUniversity of AmsterdamAmsterdam NeuroscienceAmsterdamThe Netherlands
- Department of Clinical and Experimental EpilepsyUniversity College LondonLondonUK
- Chalfont Centre for EpilepsyChalfont St PeterUK
| | - Erwin A. van Vliet
- Department of (Neuro) PathologyAmsterdam UMCUniversity of AmsterdamAmsterdam NeuroscienceAmsterdamThe Netherlands
- Center for NeuroscienceSwammerdam Institute for Life SciencesUniversity of AmsterdamAmsterdamThe Netherlands
| | - Eleonora Aronica
- Department of (Neuro) PathologyAmsterdam UMCUniversity of AmsterdamAmsterdam NeuroscienceAmsterdamThe Netherlands
- Stichting Epilepsie Instellingen NederlandHeemstedeThe Netherlands
| |
Collapse
|
7
|
Bazrgar M, Khodabakhsh P, Prudencio M, Mohagheghi F, Ahmadiani A. The role of microRNA-34 family in Alzheimer's disease: A potential molecular link between neurodegeneration and metabolic disorders. Pharmacol Res 2021; 172:105805. [PMID: 34371173 DOI: 10.1016/j.phrs.2021.105805] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/27/2021] [Accepted: 08/05/2021] [Indexed: 02/09/2023]
Abstract
Growing evidence indicates that overexpression of the microRNA-34 (miR-34) family in the brain may play a crucial role in Alzheimer's disease (AD) pathogenesis by targeting and downregulating genes associated with neuronal survival, synapse formation and plasticity, Aβ clearance, mitochondrial function, antioxidant defense system, and energy metabolism. Additionally, elevated levels of the miR-34 family in the liver and pancreas promote the development of metabolic syndromes (MetS), such as diabetes and obesity. Importantly, MetS represent a well-documented risk factor for sporadic AD. This review focuses on the recent findings regarding the role of the miR-34 family in the pathogenesis of AD and MetS, and proposes miR-34 as a potential molecular link between both disorders. A comprehensive understanding of the functional roles of miR-34 family in the molecular and cellular pathogenesis of AD brains may lead to the discovery of a breakthrough treatment strategy for this disease.
Collapse
Affiliation(s)
- Maryam Bazrgar
- Neuroscience Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Pariya Khodabakhsh
- Department of Pharmacology, Shahid Beheshti University of Medical Science, Tehran, Iran
| | | | - Fatemeh Mohagheghi
- Institute of Experimental Hematology, Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - Abolhassan Ahmadiani
- Neuroscience Research Center, Shahid Beheshti University of Medical Science, Tehran, Iran.
| |
Collapse
|
8
|
Early Life Irradiation-Induced Hypoplasia and Impairment of Neurogenesis in the Dentate Gyrus and Adult Depression Are Mediated by MicroRNA- 34a-5p/T-Cell Intracytoplasmic Antigen-1 Pathway. Cells 2021; 10:cells10092476. [PMID: 34572124 PMCID: PMC8466295 DOI: 10.3390/cells10092476] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/10/2021] [Accepted: 09/15/2021] [Indexed: 02/05/2023] Open
Abstract
Early life radiation exposure causes abnormal brain development, leading to adult depression. However, few studies have been conducted to explore pre- or post-natal irradiation-induced depression-related neuropathological changes. Relevant molecular mechanisms are also poorly understood. We induced adult depression by irradiation of mice at postnatal day 3 (P3) to reveal hippocampal neuropathological changes and investigate their molecular mechanism, focusing on MicroRNA (miR) and its target mRNA and protein. P3 mice were irradiated by γ-rays with 5Gy, and euthanized at 1, 7 and 120 days after irradiation. A behavioral test was conducted before the animals were euthanized at 120 days after irradiation. The animal brains were used for different studies including immunohistochemistry, CAP-miRSeq, Real-Time Quantitative Reverse Transcription PCR (qRT-PCR) and western blotting. The interaction of miR-34a-5p and its target T-cell intracytoplasmic antigen-1 (Tia1) was confirmed by luciferase reporter assay. Overexpression of Tia1 in a neural stem cell (NSC) model was used to further validate findings from the mouse model. Irradiation with 5 Gy at P3 induced depression in adult mice. Animal hippocampal pathological changes included hypoplasia of the infrapyramidal blade of the stratum granulosum, aberrant and impaired cell division, and neurogenesis in the dentate gyrus. At the molecular level, upregulation of miR-34a-5p and downregulation of Tia1 mRNA were observed in both animal and neural stem cell models. The luciferase reporter assay and gene transfection studies further confirmed a direct interaction between miR-43a-5p and Tia1. Our results indicate that the early life γ-radiation-activated miR-43a-5p/Tia1 pathway is involved in the pathogenesis of adult depression. This novel finding may provide a new therapeutic target by inhibiting the miR-43a-5p/Tia1 pathway to prevent radiation-induced pathogenesis of depression.
Collapse
|
9
|
Ataei A, Poorebrahim M, Rajabpour A, Rizvanov A, Shahriar Arab S. Topological Analysis of Regulatory Networks Reveals Functionally Key Genes and miRNAs Involved in the Differentiation of Mesenchymal Stem Cells. IRANIAN JOURNAL OF BIOTECHNOLOGY 2021; 19:e2565. [PMID: 34179189 PMCID: PMC8217530 DOI: 10.30498/ijb.2021.2565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Background The details of molecular mechanisms underlying the differentiation of Mesenchymal Stem Cells (MSCs) into specific lineages are not well understood. Objectives We aimed to construct the interactome network and topology analysis of bone marrow mesenchymal stem cell of CAGE data. Applying the enrichment results, we wanted to introduce the common genes and hub-microRNA and hub-genes of these giant network. Materials and Methods In this study, we constructed gene regulatory networks for each non-mesenchymal cell lineage according to their gene expression profiles obtained from FANTOM5 database. The putative interactions of TF-gene and protein-protein were determined using TRED, STRING, HPRD and GeneMANIA servers. In parallel, a regulatory network including corresponding miRNAs and total differentially expressed genes (DEGs) was constructed for each cell lineage. Results The results indicated that analysis of networks' topology can significantly distinguish the hub regulatory genes and miRNAs involved in the differentiation of MSCs. The functional annotation of identified hub genes and miRNAs revealed that several signal transduction pathways i.e. AKT, WNT and TGFβ and cell proliferation related pathways play a pivotal role in the regulation of MSCs differentiation. We also classified cell lineages into two groups based on their predicted miRNA profiles. Conclusions In conclusion, we found a number of hub genes and miRNAs which seem to have key regulatory functions during differentiation of MSCs. Our results also introduce a number of new regulatory genes and miRNAs which can be considered as the new candidates for genetic manipulation of MSCs in vitro.
Collapse
Affiliation(s)
- Atousa Ataei
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Equal contribution
| | - Mansour Poorebrahim
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, University of Medical Sciences, Tehran, Iran.,Equal contribution
| | - Azam Rajabpour
- Cellular and Molecular Research Center, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Albert Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Seyed Shahriar Arab
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
10
|
MicroRNA-34a: the bad guy in age-related vascular diseases. Cell Mol Life Sci 2021; 78:7355-7378. [PMID: 34698884 PMCID: PMC8629897 DOI: 10.1007/s00018-021-03979-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/08/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022]
Abstract
The age-related vasculature alteration is the prominent risk factor for vascular diseases (VD), namely, atherosclerosis, abdominal aortic aneurysm, vascular calcification (VC) and pulmonary arterial hypertension (PAH). The chronic sterile low-grade inflammation state, alias inflammaging, characterizes elderly people and participates in VD development. MicroRNA34-a (miR-34a) is emerging as an important mediator of inflammaging and VD. miR-34a increases with aging in vessels and induces senescence and the acquisition of the senescence-associated secretory phenotype (SASP) in vascular smooth muscle (VSMCs) and endothelial (ECs) cells. Similarly, other VD risk factors, including dyslipidemia, hyperglycemia and hypertension, modify miR-34a expression to promote vascular senescence and inflammation. miR-34a upregulation causes endothelial dysfunction by affecting ECs nitric oxide bioavailability, adhesion molecules expression and inflammatory cells recruitment. miR-34a-induced senescence facilitates VSMCs osteoblastic switch and VC development in hyperphosphatemia conditions. Conversely, atherogenic and hypoxic stimuli downregulate miR-34a levels and promote VSMCs proliferation and migration during atherosclerosis and PAH. MiR34a genetic ablation or miR-34a inhibition by anti-miR-34a molecules in different experimental models of VD reduce vascular inflammation, senescence and apoptosis through sirtuin 1 Notch1, and B-cell lymphoma 2 modulation. Notably, pleiotropic drugs, like statins, liraglutide and metformin, affect miR-34a expression. Finally, human studies report that miR-34a levels associate to atherosclerosis and diabetes and correlate with inflammatory factors during aging. Herein, we comprehensively review the current knowledge about miR-34a-dependent molecular and cellular mechanisms activated by VD risk factors and highlight the diagnostic and therapeutic potential of modulating its expression in order to reduce inflammaging and VD burn and extend healthy lifespan.
Collapse
|
11
|
Shariati A, Nemati R, Sadeghipour Y, Yaghoubi Y, Baghbani R, Javidi K, Zamani M, Hassanzadeh A. Mesenchymal stromal cells (MSCs) for neurodegenerative disease: A promising frontier. Eur J Cell Biol 2020; 99:151097. [PMID: 32800276 DOI: 10.1016/j.ejcb.2020.151097] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative disorders are a variety of diseases including Alzheimer's (AD), Parkinson's (PD), and Huntington's diseases (HD), multiple sclerosis (MS) and amyotrophic lateral sclerosis (ALS) along with some other less common diseases generally described by the advanced deterioration of central or peripheral nervous system, structurally or functionally. In the last two decades, mesenchymal stromal cells (MSCs) due to their unique assets encompassing self-renewal, multipotency and accessibility in association with low ethical concern open new frontiers in the context of neurodegenerative diseases therapy. Interestingly, MSCs can be differentiated into endodermal and ectodermal lineages (e.g., neurons, oligodendrocyte, and astrocyte), and thus could be employed to advance cell-based therapeutic strategy. Additionally, as inflammation ordinarily ensues as a local response provoked by microglia in the neurodegenerative diseases, MSCs therapy because of their pronounced immunomodulatory properties is noticed as a rational approach for their treatment. Recently, varied types of studies have been mostly carried out in vitro and rodent models using MSCs upon their procurement from various sources and expansion. The promising results of the studies in rodent models have motivated researchers to design and perform several clinical trials, with a speedily rising number. In the current review, we aim to deliver a brief overview of MSCs sources, expansion strategies, and their immunosuppressive characteristics and discuss credible functional mechanisms exerted by MSCs to treat neurodegenerative disorders, covering AD, PD, ALS, MS, and HD.
Collapse
Affiliation(s)
- Ali Shariati
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Reza Nemati
- Department of Medical Emergencies, School of Allied Medical Sciences, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Yasin Sadeghipour
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Yoda Yaghoubi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Reza Baghbani
- Department of Medical Emergencies, School of Allied Medical Sciences, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Kamran Javidi
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.
| | - Majid Zamani
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.
| | - Ali Hassanzadeh
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran; Cell Therapy and Regenerative Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Menezes ESB, Badial PR, El Debaky H, Husna AU, Ugur MR, Kaya A, Topper E, Bulla C, Grant KE, Bolden-Tiller O, Moura AA, Memili E. Sperm miR-15a and miR-29b are associated with bull fertility. Andrologia 2019; 52:e13412. [PMID: 31671225 DOI: 10.1111/and.13412] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/26/2019] [Accepted: 08/06/2019] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs modulate male fertility by regulating gene expression. In this study, dynamics of sperm miR-15a, miR-29b and miR-34a from high fertility (HF) and low fertility (LF) bulls using RT-qPCR were evaluated. Bioinformatic tools were employed to ascertain genes of interest of the sperm miRNAs. The expression levels of p53, BCL2, BAX and DNMT1 in bull spermatozoa were determined by immunoblotting. MicroRNA levels of miR-15a and miR-29 were higher in LF sires when compared with those present in HF bulls. Expression levels of miR-34a did not differ between the two groups. We found an inverse correlation between miR-15a and bull fertility. MiR29-b was also negatively associated with fertility scores. BCL2 and DNMT1 were higher in HF bulls while BAX was higher in the LF group. Our data showed a positive correlation between BCL2 and bull fertility. In addition, DNMT1 was positively associated with bull fertility. Furthermore, levels of BAX were negatively linked with bull fertility scores. Identification of miRNAs found in the spermatozoa of sires with different in vivo fertility helps understand the alterations in the fertilising capacity from cattle and other mammals. These potential biomarkers can be used in reproductive biotechnology as fertility markers to assess semen quality and predict male fertility.
Collapse
Affiliation(s)
- Erika S B Menezes
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, USA.,Department of Animal Sciences, Federal University of Ceara, Fortaleza, Brazil
| | - Peres Ramos Badial
- Department of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, USA
| | - Hazem El Debaky
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, USA.,National Research Center, Cairo, Egypt
| | - Asma Ul Husna
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, USA.,Department of Zoology, Pir Mehr Ali Shah Arid Agriculture University, Rawalpindi, Pakistan
| | - Muhammet Rasit Ugur
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, USA
| | - Abdullah Kaya
- URUS Group LP, Madison, WI, USA.,Department of Reproduction and Artificial Insemination, Selcuk University, Konya, Turkey
| | | | - Camilo Bulla
- Department of Pathobiology and Population Medicine, College of Veterinary Medicine, Mississippi State University, Mississippi State, MS, USA
| | - Kamilah E Grant
- Center for Biotechnology and Department of Agriculture School of Agriculture & Applied Sciences, Alcorn State University, Lorman, MS, USA
| | - Olga Bolden-Tiller
- Department of Agricultural and Environmental Sciences, Tuskegee University, Tuskegee, AL, USA
| | - Arlindo A Moura
- Department of Animal Sciences, Federal University of Ceara, Fortaleza, Brazil
| | - Erdoğan Memili
- Department of Animal and Dairy Sciences, Mississippi State University, Mississippi State, MS, USA
| |
Collapse
|
13
|
Velasco MX, Kosti A, Guardia GDA, Santos MC, Tegge A, Qiao M, Correa BRS, Hernández G, Kokovay E, Galante PAF, Penalva LOF. Antagonism between the RNA-binding protein Musashi1 and miR-137 and its potential impact on neurogenesis and glioblastoma development. RNA (NEW YORK, N.Y.) 2019; 25:768-782. [PMID: 31004009 PMCID: PMC6573790 DOI: 10.1261/rna.069211.118] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 04/15/2019] [Indexed: 06/09/2023]
Abstract
RNA-binding proteins (RBPs) and miRNAs are critical gene expression regulators that interact with one another in cooperative and antagonistic fashions. We identified Musashi1 (Msi1) and miR-137 as regulators of a molecular switch between self-renewal and differentiation. Msi1 and miR-137 have opposite expression patterns and functions, and Msi1 is repressed by miR-137. Msi1 is a stem-cell protein implicated in self-renewal while miR-137 functions as a proneuronal differentiation miRNA. In gliomas, miR-137 functions as a tumor suppressor while Msi1 is a prooncogenic factor. We suggest that the balance between Msi1 and miR-137 is a key determinant in cell fate decisions and disruption of this balance could contribute to neurodegenerative diseases and glioma development. Genomic analyses revealed that Msi1 and miR-137 share 141 target genes associated with differentiation, development, and morphogenesis. Initial results pointed out that these two regulators have an opposite impact on the expression of their target genes. Therefore, we propose an antagonistic model in which this network of shared targets could be either repressed by miR-137 or activated by Msi1, leading to different outcomes (self-renewal, proliferation, tumorigenesis).
Collapse
Affiliation(s)
- Mitzli X Velasco
- Greheey Children's Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
- Translation and Cancer Laboratory, Unit of Biomedical Research on Cancer, National Institute of Cancer (INCan), Mexico City 14080, Mexico
| | - Adam Kosti
- Greheey Children's Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
| | - Gabriela D A Guardia
- Centro de Oncologia Molecular-Hospital Sírio-Libanês, São Paulo 01308-050, Brazil
| | - Marcia C Santos
- Greheey Children's Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
| | - Allison Tegge
- Department of Statistics, Virginia Tech, Blacksburg, Virginia 14080, USA
| | - Mei Qiao
- Greheey Children's Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
| | - Bruna R S Correa
- Greheey Children's Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
- Centro de Oncologia Molecular-Hospital Sírio-Libanês, São Paulo 01308-050, Brazil
| | - Greco Hernández
- Translation and Cancer Laboratory, Unit of Biomedical Research on Cancer, National Institute of Cancer (INCan), Mexico City 14080, Mexico
| | - Erzsebet Kokovay
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
| | - Pedro A F Galante
- Centro de Oncologia Molecular-Hospital Sírio-Libanês, São Paulo 01308-050, Brazil
| | - Luiz O F Penalva
- Greheey Children's Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
| |
Collapse
|
14
|
Santerre M, Bagashev A, Gorecki L, Lysek KZ, Wang Y, Shrestha J, Del Carpio-Cano F, Mukerjee R, Sawaya BE. HIV-1 Tat protein promotes neuronal dysregulation by inhibiting E2F transcription factor 3 (E2F3). J Biol Chem 2018; 294:3618-3633. [PMID: 30591585 DOI: 10.1074/jbc.ra118.003744] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 12/17/2018] [Indexed: 12/29/2022] Open
Abstract
Individuals who are infected with HIV-1 accumulate damage to cells and tissues (e.g. neurons) that are not directly infected by the virus. These include changes known as HIV-associated neurodegenerative disorder (HAND), leading to the loss of neuronal functions, including synaptic long-term potentiation (LTP). Several mechanisms have been proposed for HAND, including direct effects of viral proteins such as the Tat protein. Searching for the mechanisms involved, we found here that HIV-1 Tat inhibits E2F transcription factor 3 (E2F3), CAMP-responsive element-binding protein (CREB), and brain-derived neurotropic factor (BDNF) by up-regulating the microRNA miR-34a. These changes rendered murine neurons dysfunctional by promoting neurite retraction, and we also demonstrate that E2F3 is a specific target of miR-34a. Interestingly, bioinformatics analysis revealed the presence of an E2F3-binding site within the CREB promoter, which we validated with ChIP and transient transfection assays. Of note, luciferase reporter assays revealed that E2F3 up-regulates CREB expression and that Tat interferes with this up-regulation. Further, we show that miR-34a inhibition or E2F3 overexpression neutralizes Tat's effects and restores normal distribution of the synaptic protein synaptophysin, confirming that Tat alters these factors, leading to neurite retraction inhibition. Our results suggest that E2F3 is a key player in neuronal functions and may represent a good target for preventing the development of HAND.
Collapse
Affiliation(s)
- Maryline Santerre
- From the Molecular Studies of Neurodegenerative Diseases Laboratory, FELS Institute for Cancer Research and Molecular Biology
| | - Asen Bagashev
- From the Molecular Studies of Neurodegenerative Diseases Laboratory, FELS Institute for Cancer Research and Molecular Biology.,the Department of Anatomy and Cell Biology, and
| | - Laura Gorecki
- From the Molecular Studies of Neurodegenerative Diseases Laboratory, FELS Institute for Cancer Research and Molecular Biology
| | - Kyle Z Lysek
- From the Molecular Studies of Neurodegenerative Diseases Laboratory, FELS Institute for Cancer Research and Molecular Biology
| | - Ying Wang
- From the Molecular Studies of Neurodegenerative Diseases Laboratory, FELS Institute for Cancer Research and Molecular Biology
| | - Jenny Shrestha
- From the Molecular Studies of Neurodegenerative Diseases Laboratory, FELS Institute for Cancer Research and Molecular Biology
| | - Fabiola Del Carpio-Cano
- From the Molecular Studies of Neurodegenerative Diseases Laboratory, FELS Institute for Cancer Research and Molecular Biology
| | - Ruma Mukerjee
- From the Molecular Studies of Neurodegenerative Diseases Laboratory, FELS Institute for Cancer Research and Molecular Biology
| | - Bassel E Sawaya
- From the Molecular Studies of Neurodegenerative Diseases Laboratory, FELS Institute for Cancer Research and Molecular Biology, .,the Department of Anatomy and Cell Biology, and.,the Department of Neurology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| |
Collapse
|
15
|
Song B, Du J, Song DF, Ren JC, Feng Y. Dysregulation of NCAPG, KNL1, miR-148a-3p, miR-193b-3p, and miR-1179 may contribute to the progression of gastric cancer. Biol Res 2018; 51:44. [PMID: 30390708 PMCID: PMC6215350 DOI: 10.1186/s40659-018-0192-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 10/16/2018] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Emerging evidence indicate that miRNAs play an important role on gastric cancer (GC) progression via regulating several downstream targets, but it is still partially uncovered. This study aimed to explore the molecular mechanisms of GC by comprehensive analysis of mRNAs and miRNA expression profiles. METHODS The mRNA and miRNA expression profiles of GSE79973 and GSE67354 downloaded from Gene Expression Omnibus were used to analyze the differentially expressed genes (DEGs) and DE-miRNAs among GC tissues and normal tissues. Then, targets genes of DE-miRNAs were predicted and the DE-miRNA-DEG regulatory network was constructed. Next, function enrichment analysis of the overlapped genes between the predicted DE-miRNAs targets and DEGs was performed and a protein-protein interactions network of overlapped genes was constructed. Finally, RT-PCR analysis was performed to detect the expression levels of several key DEGs and DE-miRNAs. RESULTS A set of 703 upregulated and 600 downregulated DEGs, as well as 8 upregulated DE-miRNAs and 27 downregulated DE-miRNAs were identified in GC tissue. hsa-miR-193b-3p and hsa-miR-148a-3p, which targeted most DEGs, were highlighted in the DE-miRNA-DEG regulatory network, as well as hsa-miR-1179, which targeted KNL1, was newly predicted to be associated with GC. In addition, NCAPG, which is targeted by miR-193b-3p, and KNL1, which is targeted by hsa-miR-1179, had higher degrees in the PPI network. RT-qPCR results showed that hsa-miR-148a-3p, hsa-miR-193b-3p, and hsa-miR-1179 were downregulated, and NCAPG and KNL1 were upregulated in GC tissues; this is consistent with our bioinformatics-predicted results. CONCLUSIONS The downregulation of miR-193b-3p might contribute to GC cell proliferation by mediating the upregulation of NCAPG; as additionally, the downregulation of miR-193b-3p might contribute to the mitotic nuclear division of GC cells by mediating the upregulation of KNL1.
Collapse
Affiliation(s)
- Bin Song
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital, Jilin University, No.126, Xiantai Street, Changchun, 130033, China
| | - Juan Du
- Internal Medicine 2, The Tumor Hospital of Jilin Province, Changchun, 130012, China
| | - De-Feng Song
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital, Jilin University, No.126, Xiantai Street, Changchun, 130033, China
| | - Ji-Chen Ren
- Internal Medicine 2, The Tumor Hospital of Jilin Province, Changchun, 130012, China
| | - Ye Feng
- Department of Gastrointestinal and Colorectal Surgery, China-Japan Union Hospital, Jilin University, No.126, Xiantai Street, Changchun, 130033, China.
| |
Collapse
|
16
|
Sun J, Lv J, Zhang W, Li L, Lv J, Geng Y, Yin A. Combination with miR-124a improves the protective action of BMSCs in rescuing injured rat podocytes from abnormal apoptosis and autophagy. J Cell Biochem 2018; 119:7166-7176. [PMID: 29904949 DOI: 10.1002/jcb.26771] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 02/02/2018] [Indexed: 12/13/2022]
Abstract
This in vitro study was performed to identify the role of miR-124a in bone marrow stromal stem cells (BMSCs) therapy for H2 O2 -induced rat podocyte injury, and determine whether combination treatment with miR-124a could improve the protective effect of BMSCs. Cell viability of podocytes was detected by CCK-8 assay. Detection of ROS level, apoptotic rate, and autophagy rate was carried out using flow cytometry assays. Oxidative stress parameters were analyzed using the ELISA assays. MiR-124a and mRNA levels were determined using real-time PCR. Protein expression was detected using Western blotting. Our study revealed a pivotal role of miR-124a in the protective action of BMSCs on podocyte injury driven by oxidative stress. BMSCs could rescue injured podocytes from aberrant apoptosis and autophagy by regulating cleaved caspase-3, Bax, Bcl-2, LC3-II/I, and p62. Suppression of the PI3 K/Akt/mTOR signaling pathway is likely one of the main mechanisms underlying the protective action of BMSCs transfected with miR-124a. Our study revealed that miR-124a further improves the protective effect of BMSCs in injured podocytes. Thus, the combination of BMSCs and microRNAs could be a beneficial treatment for renal diseases in the near future.
Collapse
Affiliation(s)
- Jiping Sun
- Department of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jing Lv
- Department of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Wenjing Zhang
- Department of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lili Li
- Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Jia Lv
- Department of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yingzhou Geng
- Department of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Aiping Yin
- Department of Nephrology, The First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
17
|
Su L, Wang C, Zheng C, Wei H, Song X. A meta-analysis of public microarray data identifies biological regulatory networks in Parkinson's disease. BMC Med Genomics 2018; 11:40. [PMID: 29653596 PMCID: PMC5899355 DOI: 10.1186/s12920-018-0357-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 03/26/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a long-term degenerative disease that is caused by environmental and genetic factors. The networks of genes and their regulators that control the progression and development of PD require further elucidation. METHODS We examine common differentially expressed genes (DEGs) from several PD blood and substantia nigra (SN) microarray datasets by meta-analysis. Further we screen the PD-specific genes from common DEGs using GCBI. Next, we used a series of bioinformatics software to analyze the miRNAs, lncRNAs and SNPs associated with the common PD-specific genes, and then identify the mTF-miRNA-gene-gTF network. RESULT Our results identified 36 common DEGs in PD blood studies and 17 common DEGs in PD SN studies, and five of the genes were previously known to be associated with PD. Further study of the regulatory miRNAs associated with the common PD-specific genes revealed 14 PD-specific miRNAs in our study. Analysis of the mTF-miRNA-gene-gTF network about PD-specific genes revealed two feed-forward loops: one involving the SPRK2 gene, hsa-miR-19a-3p and SPI1, and the second involving the SPRK2 gene, hsa-miR-17-3p and SPI. The long non-coding RNA (lncRNA)-mediated regulatory network identified lncRNAs associated with PD-specific genes and PD-specific miRNAs. Moreover, single nucleotide polymorphism (SNP) analysis of the PD-specific genes identified two significant SNPs, and SNP analysis of the neurodegenerative disease-specific genes identified seven significant SNPs. Most of these SNPs are present in the 3'-untranslated region of genes and are controlled by several miRNAs. CONCLUSION Our study identified a total of 53 common DEGs in PD patients compared with healthy controls in blood and brain datasets and five of these genes were previously linked with PD. Regulatory network analysis identified PD-specific miRNAs, associated long non-coding RNA and feed-forward loops, which contribute to our understanding of the mechanisms underlying PD. The SNPs identified in our study can determine whether a genetic variant is associated with PD. Overall, these findings will help guide our study of the complex molecular mechanism of PD.
Collapse
Affiliation(s)
- Lining Su
- Department of Biology of Basic Medical Science College, Hebei North University, Zhangjiakou, 075000, Hebei, China
| | - Chunjie Wang
- Department of Basic Medicine, Zhangjiakou University, Zhangjiakou, 75000, Hebei, China
| | - Chenqing Zheng
- Shenzhen RealOmics (Biotech) Co., Ltd, Shenzhen, 518081, Guangdong, China
| | - Huiping Wei
- Department of Biology of Basic Medical Science College, Hebei North University, Zhangjiakou, 075000, Hebei, China.
| | - Xiaoqing Song
- Department of Biology of Basic Medical Science College, Hebei North University, Zhangjiakou, 075000, Hebei, China
| |
Collapse
|
18
|
Avansini SH, Torres FR, Vieira AS, Dogini DB, Rogerio F, Coan AC, Morita ME, Guerreiro MM, Yasuda CL, Secolin R, Carvalho BS, Borges MG, Almeida VS, Araújo PAOR, Queiroz L, Cendes F, Lopes-Cendes I. Dysregulation of NEUROG2 plays a key role in focal cortical dysplasia. Ann Neurol 2018; 83:623-635. [PMID: 29461643 PMCID: PMC5901021 DOI: 10.1002/ana.25187] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 02/16/2018] [Accepted: 02/16/2018] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Focal cortical dysplasias (FCDs) are an important cause of drug-resistant epilepsy. In this work, we aimed to investigate whether abnormal gene regulation, mediated by microRNA, could be involved in FCD type II. METHODS We used total RNA from the brain tissue of 16 patients with FCD type II and 28 controls. MicroRNA expression was initially assessed by microarray. Quantitative polymerase chain reaction, in situ hybridization, luciferase reporter assays, and deep sequencing for genes in the mTOR pathway were performed to validate and further explore our initial study. RESULTS hsa-let-7f (p = 0.039), hsa-miR-31 (p = 0.0078), and hsa-miR34a (p = 0.021) were downregulated in FCD type II, whereas a transcription factor involved in neuronal and glial fate specification, NEUROG2 (p < 0.05), was upregulated. We also found that the RND2 gene, a NEUROG2-target, is upregulated (p < 0.001). In vitro experiments showed that hsa-miR-34a downregulates NEUROG2 by binding to its 5'-untranslated region. Moreover, we observed strong nuclear expression of NEUROG2 in balloon cells and dysmorphic neurons and found that 28.5% of our patients presented brain somatic mutations in genes of the mTOR pathway. INTERPRETATION Our findings suggest a new molecular mechanism, in which NEUROG2 has a pivotal and central role in the pathogenesis of FCD type II. In this way, we found that the downregulation of hsa-miR-34a leads to upregulation of NEUROG2, and consequently to overexpression of the RND2 gene. These findings indicate that a faulty coupling in neuronal differentiation and migration mechanisms may explain the presence of aberrant cells and complete dyslamination in FCD type II. Ann Neurol 2018;83:623-635.
Collapse
Affiliation(s)
- Simoni H Avansini
- Department of Medical Genetics, University of Campinas and Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Fábio R Torres
- Department of Medical Genetics, University of Campinas and Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - André S Vieira
- Department of Medical Genetics, University of Campinas and Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Danyella B Dogini
- Department of Medical Genetics, University of Campinas and Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Fabio Rogerio
- Department of Anatomical Pathology, University of Campinas and Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Ana C Coan
- Department of Neurology, University of Campinas and Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Marcia E Morita
- Department of Neurology, University of Campinas and Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Marilisa M Guerreiro
- Department of Neurology, University of Campinas and Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Clarissa L Yasuda
- Department of Neurology, University of Campinas and Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Rodrigo Secolin
- Department of Medical Genetics, University of Campinas and Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Benilton S Carvalho
- Department of Medical Genetics, University of Campinas and Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Murilo G Borges
- Department of Medical Genetics, University of Campinas and Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Vanessa S Almeida
- Department of Medical Genetics, University of Campinas and Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Patrícia A O R Araújo
- Department of Medical Genetics, University of Campinas and Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Luciano Queiroz
- Department of Anatomical Pathology, University of Campinas and Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Fernando Cendes
- Department of Neurology, University of Campinas and Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| | - Iscia Lopes-Cendes
- Department of Medical Genetics, University of Campinas and Brazilian Institute of Neuroscience and Neurotechnology, Campinas, Brazil
| |
Collapse
|
19
|
Identification of microRNAs differentially expressed in glioblastoma stem-like cells and their association with patient survival. Sci Rep 2018; 8:2836. [PMID: 29434344 PMCID: PMC5809429 DOI: 10.1038/s41598-018-20929-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 01/25/2018] [Indexed: 01/29/2023] Open
Abstract
Glioblastoma stem-like cells (GSCs) are critical for the aggressiveness and progression of glioblastoma (GBM) and contribute to its resistance to adjuvant treatment. MicroRNAs (miRNAs) are small, non-coding RNAs controlling gene expression at the post-transcriptional level, which are known to be important regulators of the stem-like features. Moreover, miRNAs have been previously proved to be promising diagnostic biomarkers in several cancers including GBM. Using global expression analysis of miRNAs in 10 paired in-vitro as well as in-vivo characterized primary GSC and non-stem glioblastoma cultures, we identified a miRNA signature associated with the stem-like phenotype in GBM. 51 most deregulated miRNAs classified the cell cultures into GSC and non-stem cell clusters and identified a subgroup of GSC cultures with more pronounced stem-cell characteristics. The importance of the identified miRNA signature was further supported by demonstrating that a Risk Score based on the expression of seven miRNAs overexpressed in GSC predicted overall survival in GBM patients in the TCGA dataset independently of the IDH1 status. In summary, we identified miRNAs differentially expressed in GSCs and described their association with GBM patient survival. We propose that these miRNAs participate on GSC features and could represent helpful prognostic markers and potential therapeutic targets in GBM.
Collapse
|
20
|
Guo W, Xie B, Xiong S, Liang X, Gui JF, Mei J. miR-34a Regulates Sperm Motility in Zebrafish. Int J Mol Sci 2017; 18:E2676. [PMID: 29232857 PMCID: PMC5751278 DOI: 10.3390/ijms18122676] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 11/29/2017] [Accepted: 12/07/2017] [Indexed: 01/03/2023] Open
Abstract
Increasing attention has been focused on the role of microRNAs in post-transcription regulation during spermatogenesis. Recently, the miR-34 family has been shown to be involved in the spermatogenesis, but the clear function of the miR-34 family in spermatogenesis is still obscure. Here we analyzed the function of miR-34a, a member of the miR-34 family, during spermatogenesis using miR-34a knockout zebrafish generated by the clustered regularly interspaced short palindromic repeats/associated protein 9 (CRISPR/Cas9) system. miR-34a knockout zebrafish showed no obvious defects on testis morphology and sperm quantity. However, we found a significant increase in progressive sperm motility that is one of the pivotal factors influencing in vitro fertilization rates, in the knockout zebrafish. Moreover, breeding experiments showed that, when miR-34a-knockout male zebrafish mated with the wide-type females, they had a higher fertilization rate than did the wide-type males. Glycogen synthase kinase-3a (gsk3a), a potential sperm motility regulatory gene was predicted to be targeted by miR-34a, which was further supported by luciferase reporter assays, since a significant decrease of luciferase activity was detected upon ectopic overexpression of miR-34a. Our findings suggest that miR-34a downregulates gsk3a by targeting its 3' untranslated region, and miR-34a/gsk3a interaction modulates sperm motility in zebrafish. This study will help in understanding in the role of the miR-34 family during spermatogenesis and will set paths for further studies.
Collapse
Affiliation(s)
- Wenjie Guo
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China.
| | - Binyue Xie
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China.
| | - Shuting Xiong
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China.
| | - Xufang Liang
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China.
| | - Jian-Fang Gui
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China.
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Wuhan 430072, China.
| | - Jie Mei
- College of Fisheries, Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
21
|
Zhao X, Ji Z, Xie Y, Liu G, Li H. MicroRNA-154 as a prognostic factor in bladder cancer inhibits cellular malignancy by targeting RSF1 and RUNX2. Oncol Rep 2017; 38:2727-2734. [PMID: 29048677 PMCID: PMC5780025 DOI: 10.3892/or.2017.5992] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 09/11/2017] [Indexed: 12/28/2022] Open
Abstract
Recent studies have demonstrated that microRNA-154 (miR-154) is involved in tumorigenesis, progression, invasion and metastasis in several types of human cancer. However, whether it plays a role in bladder cancer (BC) is unclear. The aim of the present study was to determine miR-154 levels in human BC tissues and investigate the correlation between miR-154 levels and clinicopathological characteristics as well as patient outcome. Using RT-qPCR, we found that the expression levels of miR-154 were significantly lower in BC tissues compared to adjacent normal tissues. We also demonstrated that downregulation of miR-154 was associated with advanced clinicopathological features and worse prognoses for patients with BC. Using a variety of integrated approaches, we demonstrated that both runt-related transcription factor 2 (RUNX2) and remodeling and spacing factor 1 (RSF1) were miR-154 targets. Notably, there was an inverse correlation between RSF1, RUNX2 and miR-154 expression in BC tissues. The biological functions of miR-154 were examined in vitro using Cell Counting Kit-8 (CCK-8), wound healing, and Transwell assays with T24 human bladder carcinoma cells transfected with miR-154 mimics or negative controls. These assays demonstrated that miR-154 significantly suppressed proliferation, migration and invasion of T24 cells (P<0.05). Furthermore, overexpression of RSF1 and RUNX2 rescued miR-154-induced inhibition of these aggressive behaviors. Our results indicated that miR-154, and its downstream targets RSF1 and RUNX2, are promising options for future BC therapies.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Zhigang Ji
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Yi Xie
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Guanghua Liu
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Hanzhong Li
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|
22
|
Venkat P, Shen Y, Chopp M, Chen J. Cell-based and pharmacological neurorestorative therapies for ischemic stroke. Neuropharmacology 2017; 134:310-322. [PMID: 28867364 DOI: 10.1016/j.neuropharm.2017.08.036] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 08/22/2017] [Accepted: 08/24/2017] [Indexed: 01/09/2023]
Abstract
Ischemic stroke remains one of most common causes of death and disability worldwide. Stroke triggers a cascade of events leading to rapid neuronal damage and death. Neuroprotective agents that showed promise in preclinical experiments have failed to translate to the clinic. Even after decades of research, tPA remains the only FDA approved drug for stroke treatment. However, tPA is effective when administered 3-4.5 h after stroke onset and the vast majority of stroke patients do not receive tPA therapy. Therefore, there is a pressing need for novel therapies for ischemic stroke. Since stroke induces rapid cell damage and death, neuroprotective strategies that aim to salvage or replace injured brain tissue are challenged by treatment time frames. To overcome the barriers of neuroprotective therapies, there is an increasing focus on neurorestorative therapies for stroke. In this review article, we provide an update on neurorestorative treatments for stroke using cell therapy such as bone marrow derived mesenchymal stromal cells (BMSCs), human umbilical cord blood cells (HUCBCs) and select pharmacological approaches including Minocycline and Candesartan that have been employed in clinical trials. This review article discusses the present understanding of mechanisms of neurorestorative therapies and summarizes ongoing clinical trials. This article is part of the Special Issue entitled 'Cerebral Ischemia'.
Collapse
Affiliation(s)
- Poornima Venkat
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA
| | - Yi Shen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA; Gerontology Institute, Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA; Department of Physics, Oakland University, Rochester, MI, 48309, USA
| | - Jieli Chen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, 48202, USA; Gerontology Institute, Department of Neurology, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neurotrauma Neurorepair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, China.
| |
Collapse
|
23
|
Insights into the post-transcriptional regulation of the mitochondrial electron transport chain. Biochem Soc Trans 2017; 44:1491-1498. [PMID: 27911731 PMCID: PMC5095899 DOI: 10.1042/bst20160100] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/24/2016] [Accepted: 06/28/2016] [Indexed: 12/11/2022]
Abstract
The regulation of the mitochondrial electron transport chain is central to the control of cellular homeostasis. There are significant gaps in our understanding of how the expression of the mitochondrial and nuclear genome-encoded components of the electron transport chain are co-ordinated, and how the assembly of the protein complexes that constitute the electron transport chain are regulated. Furthermore, the role post-transcriptional gene regulation may play in modulating these processes needs to be clarified. This review summarizes the current knowledge regarding the post-transcriptional gene regulation of the electron transport chain and highlights how noncoding RNAs may contribute significantly both to complex electron transport chain regulatory networks and to mitochondrial dysfunction.
Collapse
|
24
|
Hauberg ME, Roussos P, Grove J, Børglum AD, Mattheisen M. Analyzing the Role of MicroRNAs in Schizophrenia in the Context of Common Genetic Risk Variants. JAMA Psychiatry 2016; 73:369-77. [PMID: 26963595 PMCID: PMC7005318 DOI: 10.1001/jamapsychiatry.2015.3018] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
IMPORTANCE The recent implication of 108 genomic loci in schizophrenia marked a great advancement in our understanding of the disease. Against the background of its polygenic nature there is a necessity to identify how schizophrenia risk genes interplay. As regulators of gene expression, microRNAs (miRNAs) have repeatedly been implicated in schizophrenia etiology. It is therefore of interest to establish their role in the regulation of schizophrenia risk genes in disease-relevant biological processes. OBJECTIVE To examine the role of miRNAs in schizophrenia in the context of disease-associated genetic variation. DESIGN, SETTING, AND PARTICIPANTS The basis of this study was summary statistics from the largest schizophrenia genome-wide association study meta-analysis to date (83 550 individuals in a meta-analysis of 52 genome-wide association studies) completed in 2014 along with publicly available data for predicted miRNA targets. We examined whether schizophrenia risk genes were more likely to be regulated by miRNA. Further, we used gene set analyses to identify miRNAs that are regulators of schizophrenia risk genes. MAIN OUTCOMES AND MEASURES Results from association tests for miRNA targetomes and related analyses. RESULTS In line with previous studies, we found that similar to other complex traits, schizophrenia risk genes were more likely to be regulated by miRNAs (P < 2 × 10-16). Further, the gene set analyses revealed several miRNAs regulating schizophrenia risk genes, with the strongest enrichment for targets of miR-9-5p (P = .0056 for enrichment among the top 1% most-associated single-nucleotide polymorphisms, corrected for multiple testing). It is further of note that MIR9-2 is located in a genomic region showing strong evidence for association with schizophrenia (P = 7.1 × 10-8). The second and third strongest gene set signals were seen for the targets of miR-485-5p and miR-137, respectively. CONCLUSIONS AND RELEVANCE This study provides evidence for a role of miR-9-5p in the etiology of schizophrenia. Its implication is of particular interest as the functions of this neurodevelopmental miRNA tie in with established disease biology: it has a regulatory loop with the fragile X mental retardation homologue FXR1 and regulates dopamine D2 receptor density.
Collapse
Affiliation(s)
- Mads Engel Hauberg
- Department of Biomedicine, Aarhus University, Aarhus, Denmark2Lundbeck Foundation Initiative of Integrative Psychiatric Research, Lundbeck, Denmark3Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark
| | - Panos Roussos
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York5Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York6Institute for Genomics and Multiscale Biology, Icahn School of M
| | - Jakob Grove
- Department of Biomedicine, Aarhus University, Aarhus, Denmark2Lundbeck Foundation Initiative of Integrative Psychiatric Research, Lundbeck, Denmark3Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark9Bioinformatics Research Centre, Aarhu
| | - Anders Dupont Børglum
- Department of Biomedicine, Aarhus University, Aarhus, Denmark2Lundbeck Foundation Initiative of Integrative Psychiatric Research, Lundbeck, Denmark3Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark10Research Department P, Aarhus Univer
| | - Manuel Mattheisen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark2Lundbeck Foundation Initiative of Integrative Psychiatric Research, Lundbeck, Denmark3Centre for Integrative Sequencing, Aarhus University, Aarhus, Denmark
| |
Collapse
|
25
|
Tsai CH, Liao KH, Shih CC, Chan CH, Hsieh JY, Tsai CF, Wang HW, Chang SJ. Small RNA and RNA-IP Sequencing Identifies and Validates Novel MicroRNAs in Human Mesenchymal Stem Cells. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2016; 20:191-8. [PMID: 26910904 DOI: 10.1089/omi.2015.0136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Organ regeneration therapies using multipotent mesenchymal stem cells (MSCs) are currently being investigated for a variety of common complex diseases. Understanding the molecular regulation of MSC biology will benefit regenerative medicine. MicroRNAs (miRNAs) act as regulators in MSC stemness. There are approximately 2500 currently known human miRNAs that have been recorded in the miRBase v21 database. In the present study, we identified novel microRNAs involved in MSC stemness and differentiation by obtaining the global microRNA expression profiles (miRNomes) of MSCs from two anatomical locations bone marrow (BM-MSCs) and umbilical cord Wharton's jelly (WJ-MSCs) and from osteogenically and adipogenically differentiated progenies of BM-MSCs. Small RNA sequencing (smRNA-seq) and bioinformatics analyses predicted that 49 uncharacterized miRNA candidates had high cellular expression values in MSCs. Another independent batch of Ago1/2-based RNA immunoprecipitation (RNA-IP) sequencing datasets validated the existence of 40 unreported miRNAs in cells and their associations with the RNA-induced silencing complex (RISC). Nine of these 40 new miRNAs were universally overexpressed in both MSC types; nine others were overexpressed in differentiated cells. A novel miRNA (UNI-118-3p) was specifically expressed in BM-MSCs, as verified using RT-qPCR. Taken together, this report offers comprehensive miRNome profiles for two MSC types, as well as cells differentiated from BM-MSCs. MSC transplantation has the potential to ameliorate degenerative disorders and repair damaged tissues. Interventions involving the above 40 new microRNA members in transplanted MSCs may potentially guide future clinical applications.
Collapse
Affiliation(s)
- Chin-Han Tsai
- 1 Department of Obstetrics and Gynecology, Hsinchu Mackay Memorial Hospital , Hsinchu, Taiwan
| | | | - Chuan-Chi Shih
- 1 Department of Obstetrics and Gynecology, Hsinchu Mackay Memorial Hospital , Hsinchu, Taiwan
| | - Chia-Hao Chan
- 1 Department of Obstetrics and Gynecology, Hsinchu Mackay Memorial Hospital , Hsinchu, Taiwan
| | | | | | - Hsei-Wei Wang
- 2 Institute of Microbiology and Immunology.,3 VGH-YM Genome Research Center, National Yang-Ming University , Taipei, Taiwan .,4 Departments of Education and Research, Taipei City Hospital , Taipei, Taiwan
| | - Shing-Jyh Chang
- 1 Department of Obstetrics and Gynecology, Hsinchu Mackay Memorial Hospital , Hsinchu, Taiwan
| |
Collapse
|
26
|
Davis GM, Haas MA, Pocock R. MicroRNAs: Not "Fine-Tuners" but Key Regulators of Neuronal Development and Function. Front Neurol 2015; 6:245. [PMID: 26635721 PMCID: PMC4656843 DOI: 10.3389/fneur.2015.00245] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 11/09/2015] [Indexed: 12/21/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of short non-coding RNAs that operate as prominent post-transcriptional regulators of eukaryotic gene expression. miRNAs are abundantly expressed in the brain of most animals and exert diverse roles. The anatomical and functional complexity of the brain requires the precise coordination of multilayered gene regulatory networks. The flexibility, speed, and reversibility of miRNA function provide precise temporal and spatial gene regulatory capabilities that are crucial for the correct functioning of the brain. Studies have shown that the underlying molecular mechanisms controlled by miRNAs in the nervous systems of invertebrate and vertebrate models are remarkably conserved in humans. We endeavor to provide insight into the roles of miRNAs in the nervous systems of these model organisms and discuss how such information may be used to inform regarding diseases of the human brain.
Collapse
Affiliation(s)
- Gregory M. Davis
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| | - Matilda A. Haas
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| | - Roger Pocock
- Development and Stem Cells Program, Monash Biomedicine Discovery Institute, Department of Anatomy and Developmental Biology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
27
|
Saghazadeh A, Rezaei N. MicroRNA machinery in Parkinson's disease: a platform for neurodegenerative diseases. Expert Rev Neurother 2015; 22:427-453. [PMID: 26574782 DOI: 10.1586/14737175.2015.1114886] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
MicroRNAs (miRNAs) are noncoding RNAs that recognize their protein-coding target genes and whereby subjugate them after transcription. Despite the infancy of this field of science, the role of miRNAs in neurodegeneration is well-acknowledged. This review was conducted to indicate that Parkinson's disease (PD) is not excluded from this rule. To this end, we evaluated the existing literature and arranged PD-associated miRNAs according to their mechanism of action, particularly apoptosis, autophagy, inflammation, mitochondrial dysfunction and oxidative stress. According to this arrangement, a majority of PD-associated miRNAs were indicated to influence autophagic/apoptotic pathways. We also categorized PD-associated miRNAs according to that they could exert detrimental or beneficial or both into three sets, activator, inhibitor, and double-edged, correspondingly. Considering this criterion, a majority of PD-associated miRNAs were included in the activator category. In addition, evidences from genetic association studies investigating genetic variants of or related to miRNAs in PD patients are presented. Finally, possible applications of the miRNA machinery in PD, including mechanistic networks, diagnostic, prognostic and therapeutic potentials, are discussed. But there may be additional miRNAs involved in the pathogenesis of PD which have hitherto remained unknown and thus further studies are needed to explore the issue and to extend this platform.
Collapse
Affiliation(s)
- Amene Saghazadeh
- a Molecular Immunology Research Center and Department of Immunology, School of Medicine , Tehran University of Medical Sciences , Tehran , Iran
| | - Nima Rezaei
- a Molecular Immunology Research Center and Department of Immunology, School of Medicine , Tehran University of Medical Sciences , Tehran , Iran.,b Research Center for Immunodeficiencies, Children's Medical Center , Tehran University of Medical Sciences , Tehran , Iran.,c Universal Scientific Education and Research Network (USERN) , Tehran , Iran
| |
Collapse
|
28
|
Zheng J, Ji C, Lu X, Tong W, Fan X, Gao Y. Integrated expression profiles of mRNA and microRNA in the liver of Fructus Meliae Toosendan water extract injured mice. Front Pharmacol 2015; 6:236. [PMID: 26539117 PMCID: PMC4609846 DOI: 10.3389/fphar.2015.00236] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 10/02/2015] [Indexed: 02/06/2023] Open
Abstract
Liver toxicity is a severe problem associated with Traditional Chinese Medicine (TCM). Fructus Meliae Toosendan (FMT) is a known hepatotoxic TCM, however, the toxicological mechanisms of liver injury caused by FMT treatment still remain largely unknown. In this study, we aimed to reveal possible mechanisms of FMT water extract-induced liver injury using a systemic approach. After three consecutive daily dosing of FMT water extract, significant increases of alanine transaminase, aspartate transaminase, and alkaline phosphatase activities, along with elevated total bilirubin and total cholesterol levels and a decrease of triglyceride level, were detected in mice serum. Moreover, hydropic degeneration was observed in hepatocytes, suggesting the presence of FMT-induced liver injury. mRNA and microRNA expression profiles of liver samples from injured mice were analyzed and revealed 8 miRNAs and 1,723 mRNAs were significantly changed after FMT water extract treatment. For the eight differentially expressed miRNAs, their predicted target genes were collected and a final set of 125 genes and 4 miRNAs (miR-139-5p, miR-199a-5p, miR-2861, and miR-3960) was selected to investigate important processes involved in FMT hepatotoxicity. Our results demonstrated several cellular functions were disordered after FMT treatment, such as cellular growth and proliferation, gene expression and cellular development. We hypothesized that liver cell necrosis was the main liver toxicity of FMT water extract, which was possibly caused by oxidative stress responses.
Collapse
Affiliation(s)
- Jie Zheng
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University Hangzhou, China
| | - Cai Ji
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University Hangzhou, China
| | - Xiaoyan Lu
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University Hangzhou, China
| | - Wei Tong
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University Hangzhou, China
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University Hangzhou, China
| | - Yue Gao
- Department of Pharmacology and Toxicology, Beijing Institute of Radiation Medicine Beijing, China
| |
Collapse
|
29
|
Taheriazam A, Talaei AJ, Jamshidi M, Shakeri M, Khoshbakht S, Yahaghi E, Shokrani M. Up-regulation of miR-130b expression level and down-regulation of miR-218 serve as potential biomarker in the early detection of human osteosarcoma. Diagn Pathol 2015; 10:184. [PMID: 26446495 PMCID: PMC4596511 DOI: 10.1186/s13000-015-0422-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 10/01/2015] [Indexed: 01/02/2023] Open
Abstract
Background Osteosarcoma (OS) is a primary malignant bone tumor with high morbidity that principally emerges in children and adolescents. MiRNAs regulate a variety of normal physiologic processes and are involved in tumorigenesis and development of multiple malignancies, including OS. This study was aimed to evaluate the clinical significance of miR-130b and miR-218 in osteosarcoma patient. Methods We utilized quantitative real-time PCR to evaluate the level of miR-130b and miR-218 expressions in OS patients and normal tissues and their relationship with clinicopathological features and survival in OS patients. Results QRT-PCR indicated that miR-130b expression in tumor tissues was strongly elevated than adjacent non-tumor tissues (P < 0.001), while the level of miR-218 expression in osteosarcoma tissues was down-regulated than adjacent non-tumor tissues (P < 0.001). We evaluated the clinical significance of miR-130b and miR-218 in osteosarcoma. Clinical correlation analysis showed that increased expression of miR-130b and decreased expression of miR-218 were significantly associated with advanced tumor stage (x2 = 6.285, P < 0.009; x2 = 7.172, P < 0.007), distant metastasis (x2
= 5.528; P < 0.001; x2 = 4.617, P < 0.001) and size of tumor (x2 = 5.01, P = 0.013; x2 = 4.271, P = 0.019). Conclusions Taken together, our data indicated that high miR-130b level and low level of miR-218 are associated with poor clinicopathological characteristics. Furthermore, miR-130b may play a key role in the progression of osteosarcoma.
Collapse
Affiliation(s)
- Afshin Taheriazam
- Department of Orthopedics Surgery, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Amir Jouya Talaei
- Department of Genetics, Faculty of Life Sciences, Azad University of Tehran Medical Sciences Branch, Tehran, Iran
| | - Mohammad Jamshidi
- Cellular and Molecular Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.,Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mohammadreza Shakeri
- Department of Orthopaedic and Trauma Surgery, Birjand University of Medical Sciences, Birjand, Iran
| | - Samaneh Khoshbakht
- Department of Statistics, Faculty of Sciences, Islamic Azad University, Mashhad Branch, Mashhad, Iran
| | - Emad Yahaghi
- Department of Molecular Biology, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Marjan Shokrani
- Graduate, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
30
|
Gallagher MF, Salley Y, Spillane CD, Ffrench B, El Baruni S, Blacksheilds G, Smyth P, Martin C, Sheils O, Watson W, O'Leary JJ. Enhanced regulation of cell cycle and suppression of osteoblast differentiation molecular signatures by prostate cancer stem-like holoclones. J Clin Pathol 2015; 68:692-702. [PMID: 26038242 DOI: 10.1136/jclinpath-2015-203001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/12/2015] [Indexed: 12/25/2022]
Abstract
AIMS Targeting the stem cell properties of tumor-initiating cells is an avenue through which cancer treatment may be improved. Before this can be achieved, so-called 'cancer stem cell' (CSC) models must be developed and characterized in specific malignancies. METHODS In this study, holoclone formation assays were used to characterise stem-like molecular signatures in prostate cancer (PCa) cells. RESULTS LNCaP and PC3 parent cells were capable of responding to stem cell differentiation morphogen retinoic acid (RA), suggesting the presence of inherent stem-like properties. LNCaP cells, which represent early, androgen-responsive disease, formed holoclones after twenty six days. PC3 cells, which represent advanced, metastatic, castration-resistant disease, formed holoclones after only six days. Holoclones displayed decreased expression of RA-genes, suggesting a more immature, less differentiated phenotype. Gene and microRNA arrays demonstrated that holoclones downregulated a number of stem cell differentiation regulators while displaying enhanced regulation of G2 to M transition and the mitotic spindle checkpoint components of the cell cycle. PC3 holoclones displayed pronounced downregulation of known regulators of osteoblast differentiation from mesenchymal stem cells and Epithelial Mesenchymal Transition. CONCLUSIONS Our results suggest that some PCa cells retain the ability to transition to a more immature state in which differentiation and metastatic mechanisms are suppressed. The highlighting of osteoblast differentiation regulators in this mechanism is particularly notable, considering the propensity of PCa to metastasise to bone.
Collapse
Affiliation(s)
- Michael F Gallagher
- Department of Histopathology, Central Pathology Laboratory, University of Dublin, Trinity College, St James Hospital, Dublin 8, Ireland Pathology Department, Coombe Women's and Infants University Hospital, Dublin 8, Ireland
| | - Yvonne Salley
- Department of Histopathology, Central Pathology Laboratory, University of Dublin, Trinity College, St James Hospital, Dublin 8, Ireland Pathology Department, Coombe Women's and Infants University Hospital, Dublin 8, Ireland
| | - Cathy D Spillane
- Department of Histopathology, Central Pathology Laboratory, University of Dublin, Trinity College, St James Hospital, Dublin 8, Ireland Pathology Department, Coombe Women's and Infants University Hospital, Dublin 8, Ireland
| | - Brendan Ffrench
- Department of Histopathology, Central Pathology Laboratory, University of Dublin, Trinity College, St James Hospital, Dublin 8, Ireland Pathology Department, Coombe Women's and Infants University Hospital, Dublin 8, Ireland
| | - Salah El Baruni
- Department of Histopathology, Central Pathology Laboratory, University of Dublin, Trinity College, St James Hospital, Dublin 8, Ireland Pathology Department, Coombe Women's and Infants University Hospital, Dublin 8, Ireland
| | - Gordon Blacksheilds
- Department of Histopathology, Central Pathology Laboratory, University of Dublin, Trinity College, St James Hospital, Dublin 8, Ireland
| | - Paul Smyth
- Department of Histopathology, Central Pathology Laboratory, University of Dublin, Trinity College, St James Hospital, Dublin 8, Ireland
| | - Cara Martin
- Department of Histopathology, Central Pathology Laboratory, University of Dublin, Trinity College, St James Hospital, Dublin 8, Ireland Pathology Department, Coombe Women's and Infants University Hospital, Dublin 8, Ireland
| | - Orla Sheils
- Department of Histopathology, Central Pathology Laboratory, University of Dublin, Trinity College, St James Hospital, Dublin 8, Ireland
| | - William Watson
- School of Medicine and Medical Science, Conway Institute, University College Dublin, Dublin 4, Ireland
| | - John J O'Leary
- Department of Histopathology, Central Pathology Laboratory, University of Dublin, Trinity College, St James Hospital, Dublin 8, Ireland Pathology Department, Coombe Women's and Infants University Hospital, Dublin 8, Ireland
| |
Collapse
|
31
|
Hu J, Lv G, Zhou S, Zhou Y, Nie B, Duan H, Zhang Y, Yuan X. The Downregulation of MiR-182 Is Associated with the Growth and Invasion of Osteosarcoma Cells through the Regulation of TIAM1 Expression. PLoS One 2015; 10:e0121175. [PMID: 25973950 PMCID: PMC4431740 DOI: 10.1371/journal.pone.0121175] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 01/28/2015] [Indexed: 01/28/2023] Open
Abstract
Background Osteosarcoma is the most common primary bone malignancy in children and young adults. Increasing results suggest that discovery of microRNAs (miRNAs) might provide a novel therapeutical target for osteosarcoma. Methods MiR-182 expression level in osteosarcoma cell lines and tissues were assayed by qRT-PCR. MiRNA mimics or inhibitor were transfected for up-regulation or down-regulation of miR-182 expression. Cell function was assayed by CCK8, migration assay and invasion assay. The target genes of miR-182 were predicated by bioinformatics algorithm (TargetScan Human). Results MiR-182 was down-regulated in osteosarcoma tissues and cell lines. Overexpression of miR-182 inhibited tumor growth, migration and invasion. Subsequent investigation revealed that TIAM1 was a direct and functional target of miR-182 in osteosarcoma cells. Overexpression of miR-182 impaired TIAM1-induced inhibition of proliferation and invasion in osteosarcoma cells. Conclusions Down-expression of miR-182 in osteosarcoma promoted tumor growth, migration and invasion by targeting TIAM1. MiR-182 might act as a tumor suppressor gene whose down-regulation contributes to the progression and metastasis of osteosarcoma, providing a potential therapy target for osteosarcoma patients.
Collapse
Affiliation(s)
- Jun Hu
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Orthopedics, The First Hospital of Kunming, Kunming, Yunnan, China
| | - Guohua Lv
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- * E-mail:
| | - Shuguang Zhou
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Orthopedics, The Affiliated Hospital of Hunan University of Medicine and The Third People’s Hospital of Huaihua, Huaihua, Hunan, China
| | - Yucheng Zhou
- Department of Orthopedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Orthopedics, The People’s Hospital of Qingyuan, Qingyuan, Guangdong, China
| | - Bangxu Nie
- Department of Orthopedics, The First Hospital of Kunming, Kunming, Yunnan, China
| | - Hong Duan
- Department of Orthopedics, The First Hospital of Kunming, Kunming, Yunnan, China
| | - Yunfeng Zhang
- Department of Orthopedics, The First Hospital of Kunming, Kunming, Yunnan, China
| | - Xiaofeng Yuan
- Department of Orthopedics, The First Hospital of Kunming, Kunming, Yunnan, China
| |
Collapse
|
32
|
Ren J, Wang H, Tran K, Civini S, Jin P, Castiello L, Feng J, Kuznetsov SA, Robey PG, Sabatino M, Stroncek DF. Human bone marrow stromal cell confluence: effects on cell characteristics and methods of assessment. Cytotherapy 2015; 17:897-911. [PMID: 25882666 DOI: 10.1016/j.jcyt.2015.03.607] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 03/09/2015] [Indexed: 01/05/2023]
Abstract
BACKGROUND AIMS Ex vivo expansion and serial passage of human bone marrow stromal cells (BMSCs, also known as bone marrow-derived mesenchymal stem cells) is required to obtain sufficient quantities for clinical therapy. The BMSC confluence criteria used to determine passage and harvest timing vary widely, and the impact of confluence on BMSC properties remains controversial. The effects of confluence on BMSC properties were studied and confluence-associated markers were identified. METHODS BMSC characteristics were analyzed as they grew from 50% to 100% confluence, including viability, population doubling time, apoptosis, colony formation, immunosuppression, surface marker expression, global gene expression and microRNA expression. In addition, culture supernatant protein, glucose, lactate and pH levels were analyzed. RESULTS Confluence-dependent changes were detected in the expression of several cell surface markers: 39 culture supernatant proteins, 26 microRNAs and 2078 genes. Many of these surface markers, proteins, microRNAs and genes have been reported to be important in BMSC function. The pigment epithelium-derived factor/vascular endothelial growth factor ratio increased with confluence, but 80% and 100% confluent BMSCs demonstrated a similar level of immunosuppression of mixed lymphocyte reactions. In addition, changes in lactate and glucose levels correlated with BMSC density. CONCLUSIONS BMSC characteristics change as confluence increases. 100% confluent BMSCs may have compromised pro-angiogenesis properties but may retain their immunomodulatory properties. Supernatant lactate and glucose levels can be used to estimate confluence and ensure consistency in passage and harvest timing. Flow cytometry or microRNA expression can be used to confirm that the BMSCs have been harvested at the appropriate confluence.
Collapse
Affiliation(s)
- Jiaqiang Ren
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Huan Wang
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Katherine Tran
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Sara Civini
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Ping Jin
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Luciano Castiello
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Ji Feng
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - Sergei A Kuznetsov
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Pamela G Robey
- Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Marianna Sabatino
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | - David F Stroncek
- Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
33
|
Dulamea AO. The potential use of mesenchymal stem cells in stroke therapy--From bench to bedside. J Neurol Sci 2015; 352:1-11. [PMID: 25818674 DOI: 10.1016/j.jns.2015.03.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 03/09/2015] [Accepted: 03/10/2015] [Indexed: 12/11/2022]
Abstract
Stroke is the second main cause of morbidity and mortality worldwide. The rationale for the use of mesenchymal stem cells (MSCs) in stroke is based on the capacity of MSCs to secrete a large variety of bioactive molecules such as growth factors, cytokines and chemokines leading to reduction of inflammation, increased neurogenesis from the germinative niches of central nervous system, increased angiogenesis, effects on astrocytes, oligodendrocytes and axons. This review presents the data derived from experimental studies and the evidence available from clinical trials about the use of MSCs in stroke therapy.
Collapse
Affiliation(s)
- Adriana Octaviana Dulamea
- U.M.F. "Carol Davila", Fundeni Clinical Institute, Department of Neurology, 258 Sos. Fundeni, Sector 2, Bucharest, Romania.
| |
Collapse
|
34
|
miR-34a regulates cell proliferation, morphology and function of newborn neurons resulting in improved behavioural outcomes. Cell Death Dis 2015; 6:e1622. [PMID: 25633291 PMCID: PMC4669781 DOI: 10.1038/cddis.2014.589] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 12/05/2014] [Accepted: 12/10/2014] [Indexed: 12/16/2022]
Abstract
miR-34a is involved in the regulation of the fate of different cell types. However, the mechanism by which it controls the differentiation programme of neural cells remains largely unknown. Here, we investigated the role of miR-34a in neurogenesis and maturation of developing neurons and identified Doublecortin as a new miR-34a target. We found that the overexpression of miR-34a in vitro significantly increases precursor proliferation and influences morphology and function of developing neurons. Indeed, miR-34a overexpressing neurons showed a decreased expression of several synaptic proteins and receptor subunits, a decrement of NMDA-evoked current density and, interestingly, a more efficient response to synaptic stimulus. In vivo, miR-34a overexpression showed stage-specific effects. In neural progenitors, miR-34a overexpression promoted cell proliferation, in migratory neuroblasts reduced the migration and in differentiating newborn neurons modulated process outgrowth and complexity. Importantly, we found that rats overexpressing miR-34a in the brain have better learning abilities and reduced emotionality.
Collapse
|
35
|
Taran R, Mamidi MK, Singh G, Dutta S, Parhar IS, John JP, Bhonde R, Pal R, Das AK. In vitro and in vivo neurogenic potential of mesenchymal stem cells isolated from different sources. J Biosci 2014; 39:157-69. [PMID: 24499800 DOI: 10.1007/s12038-013-9409-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Regenerative medicine is an evolving interdisciplinary topic of research involving numerous technological methods that utilize stem cells to repair damaged tissues. Particularly, mesenchymal stem cells (MSCs) are a great tool in regenerative medicine because of their lack of tumorogenicity, immunogenicity and ability to perform immunomodulatory as well as anti-inflammatory functions. Numerous studies have investigated the role of MSCs in tissue repair and modulation of allogeneic immune responses. MSCs derived from different sources hold unique regenerative potential as they are self-renewing and can differentiate into chondrocytes, osteoblasts, adipocytes, cardiomyocytes, hepatocytes, endothelial and neuronal cells, among which neuronal-like cells have gained special interest. MSCs also have the ability to secrete multiple bioactive molecules capable of stimulating recovery of injured cells and inhibiting inflammation. In this review we focus on neural differentiation potential of MSCs isolated from different sources and how certain growth factors/small molecules can be used to derive neuronal phenotypes from MSCs. We also discuss the efficacy of MSCs when transplanted in vivo and how they can generate certain neurons and lead to relief or recovery of the diseased condition. Furthermore, we have tried to evaluate the appropriatemerits of different sources ofMSCs with respect to their propensity towards neurological differentiation as well as their effectiveness in preclinical studies.
Collapse
Affiliation(s)
- Ramyani Taran
- Manipal Institute of Regenerative Medicine, Manipal University Branch Campus, Bangalore, India
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Cowled C, Stewart CR, Likic VA, Friedländer MR, Tachedjian M, Jenkins KA, Tizard ML, Cottee P, Marsh GA, Zhou P, Baker ML, Bean AG, Wang LF. Characterisation of novel microRNAs in the Black flying fox (Pteropus alecto) by deep sequencing. BMC Genomics 2014; 15:682. [PMID: 25128405 PMCID: PMC4156645 DOI: 10.1186/1471-2164-15-682] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 08/07/2014] [Indexed: 12/11/2022] Open
Abstract
Background Bats are a major source of new and emerging viral diseases. Despite the fact that bats carry and shed highly pathogenic viruses including Ebola, Nipah and SARS, they rarely display clinical symptoms of infection. Host factors influencing viral replication are poorly understood in bats and are likely to include both pre- and post-transcriptional regulatory mechanisms. MicroRNAs are a major mechanism of post-transcriptional gene regulation, however very little is known about them in bats. Results This study describes 399 microRNAs identified by deep sequencing of small RNA isolated from tissues of the Black flying fox, Pteropus alecto, a confirmed natural reservoir of the human pathogens Hendra virus and Australian bat lyssavirus. Of the microRNAs identified, more than 100 are unique amongst vertebrates, including a subset containing mutations in critical seed regions. Clusters of rapidly-evolving microRNAs were identified, as well as microRNAs predicted to target genes involved in antiviral immunity, the DNA damage response, apoptosis and autophagy. Closer inspection of the predicted targets for several highly supported novel miRNA candidates suggests putative roles in host-virus interaction. Conclusions MicroRNAs are likely to play major roles in regulating virus-host interaction in bats, via dampening of inflammatory responses (limiting the effects of immunopathology), and directly limiting the extent of viral replication, either through restricting the availability of essential factors or by controlling apoptosis. Characterisation of the bat microRNA repertoire is an essential step towards understanding transcriptional regulation during viral infection, and will assist in the identification of mechanisms that enable bats to act as natural virus reservoirs. This in turn will facilitate the development of antiviral strategies for use in humans and other species. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-682) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christopher Cowled
- CSIRO Australian Animal Health Laboratory, 5 Portarlington Rd, Geelong East, Victoria 3220, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Huat TJ, Khan AA, Pati S, Mustafa Z, Abdullah JM, Jaafar H. IGF-1 enhances cell proliferation and survival during early differentiation of mesenchymal stem cells to neural progenitor-like cells. BMC Neurosci 2014; 15:91. [PMID: 25047045 PMCID: PMC4117972 DOI: 10.1186/1471-2202-15-91] [Citation(s) in RCA: 98] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 07/07/2014] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND There has been increasing interest recently in the plasticity of mesenchymal stem cells (MSCs) and their potential to differentiate into neural lineages. To unravel the roles and effects of different growth factors in the differentiation of MSCs into neural lineages, we have differentiated MSCs into neural lineages using different combinations of growth factors. Based on previous studies of the roles of insulin-like growth factor 1 (IGF-1) in neural stem cell isolation in the laboratory, we hypothesized that IGF-1 can enhance proliferation and reduce apoptosis in neural progenitor-like cells (NPCs) during differentiation of MSCs into NCPs.We induced MSCs differentiation under four different combinations of growth factors: (A) EGF + bFGF, (B) EGF + bFGF + IGF-1, (C) EGF + bFGF + LIF, (D) EGF + bFGF + BDNF, and (E) without growth factors, as a negative control. The neurospheres formed were characterized by immunofluorescence staining against nestin, and the expression was measured by flow cytometry. Cell proliferation and apoptosis were also studied by MTS and Annexin V assay, respectively, at three different time intervals (24 hr, 3 days, and 5 days). The neurospheres formed in the four groups were then terminally differentiated into neuron and glial cells. RESULTS The four derived NPCs showed a significantly higher expression of nestin than was shown by the negative control. Among the groups treated with growth factors, NPCs treated with IGF-1 showed the highest expression of nestin. Furthermore, NPCs derived using IGF-1 exhibited the highest cell proliferation and cell survival among the treated groups. The NPCs derived from IGF-1 treatment also resulted in a better yield after the terminal differentiation into neurons and glial cells than that of the other treated groups. CONCLUSIONS Our results suggested that IGF-1 has a crucial role in the differentiation of MSCs into neuronal lineage by enhancing the proliferation and reducing the apoptosis in the NPCs. This information will be beneficial in the long run for improving both cell-based and cell-free therapy for neurodegenerative diseases.
Collapse
Affiliation(s)
- Tee Jong Huat
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, 16150 Kubang Kerian, Kota Bharu, Kelantan, Malaysia.
| | | | | | | | | | | |
Collapse
|
38
|
Inhibition of Notch signaling facilitates the differentiation of human-induced pluripotent stem cells into neural stem cells. Mol Cell Biochem 2014; 395:291-8. [PMID: 24972705 DOI: 10.1007/s11010-014-2130-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 06/17/2014] [Indexed: 12/19/2022]
Abstract
Neural stem cells (NSCs) derived from induced pluripotent stem cells (iPSCs) are becoming an appealing source of cell-based therapies of brain diseases. As such, it is important to understand the molecular mechanisms that regulate the differentiation of iPSCs toward NSCs. It is well known that Notch signaling governs the retention of stem cell features and drives stem cells fate. However, further studies are required to investigate the role of Notch signaling in the NSCs differentiation of iPSCs. In this study, we successfully generated NSCs from human iPSCs using serum-free medium supplemented with retinoic acid (RA) in vitro. We then assessed changes in the expression of Notch signaling-related molecules and some miRNAs (9, 34a, 200b), which exert their regulation by targeting Notch signaling. Moreover, we used a γ-secretase inhibitor (DAPT) to disturb Notch signaling. Data revealed that the levels of the Notch signaling-related molecules decreased, whereas those miRNAs increased, during this differentiation process. Inhibition of Notch signaling accelerated the formation of the neural rosette structures and the expression of NSC and mature neurocyte marker genes. This suggests that Notch signaling negatively regulated the neuralization of human iPSCs, and that this process may be regulated by some miRNAs.
Collapse
|
39
|
Chen J, Venkat P, Zacharek A, Chopp M. Neurorestorative therapy for stroke. Front Hum Neurosci 2014; 8:382. [PMID: 25018718 PMCID: PMC4072966 DOI: 10.3389/fnhum.2014.00382] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 05/14/2014] [Indexed: 12/29/2022] Open
Abstract
Ischemic stroke is responsible for many deaths and long-term disability world wide. Development of effective therapy has been the target of intense research. Accumulating preclinical literature has shown that substantial functional improvement after stroke can be achieved using subacutely administered cell-based and pharmacological therapies. This review will discuss some of the latest findings on bone marrow-derived mesenchymal stem cells (BMSCs), human umbilical cord blood cells, and off-label use of some pharmacological agents, to promote recovery processes in the sub-acute and chronic phases following stroke. This review paper also focuses on molecular mechanisms underlying the cell-based and pharmacological restorative processes, which enhance angiogenesis, arteriogenesis, neurogenesis, and white matter remodeling following cerebral ischemia as well as an analysis of the interaction/coupling among these restorative events. In addition, the role of microRNAs mediating the intercellular communication between exogenously administered cells and parenchymal cells, and their effects on the regulation of angiogenesis and neuronal progenitor cell proliferation and differentiation, and brain plasticity after stroke are described.
Collapse
Affiliation(s)
- Jieli Chen
- Department of Neurology, Henry Ford Hospital , Detroit, MI , USA
| | - Poornima Venkat
- Department of Neurology, Henry Ford Hospital , Detroit, MI , USA ; Department of Physics, Oakland University , Rochester, MI , USA
| | - Alex Zacharek
- Department of Neurology, Henry Ford Hospital , Detroit, MI , USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital , Detroit, MI , USA ; Department of Physics, Oakland University , Rochester, MI , USA
| |
Collapse
|
40
|
Huo H, Luo Z, Wang M, Yu X, Liao Z, Zhou X, Yue S. MicroRNA expression profile in intrauterine hypoxia-induced pulmonary hypoplasia in rats. Exp Ther Med 2014; 8:747-753. [PMID: 25120593 PMCID: PMC4113549 DOI: 10.3892/etm.2014.1796] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 06/11/2014] [Indexed: 01/15/2023] Open
Abstract
Hypoxia is necessary for fetal development; however, excess hypoxia is detrimental. The mechanisms underlying the effects of hypoxia on lung development remain unclear, although important roles of microRNAs (miRNAs) during lung development have recently been established. However, the effect on lung development at an miRNA expression level, following changes in oxygen tension, have not yet been studied. In the present study, pregnant rats were exposed to a fraction of inspired oxygen of 10.5 or 21% for two days on gestation day 19, following which the body weight, lung wet weight, radial alveolar count (RAC) and mean linear intercept (Lm) of the newborn pups were analyzed on postnatal day 1. To define the role of miRNAs during lung development following intrauterine hypoxia exposure, the miRNA expression pattern was profiled using a miRNA microarray. The newborn rats in the hypoxic group exhibited statistically significant decreases in body weight, lung weight and the RAC, as well as a marked increase in the Lm. A total of 69 miRNAs were identified to have significant changes in expression, including 55 upregulated and 14 downregulated miRNAs. Quantitative polymerase chain reaction was used to validate the microarray results of six selected miRNAs. Therefore, the results indicated that late gestation intrauterine hypoxia exposure may cause lung injury and miRNAs may play important roles in this process.
Collapse
Affiliation(s)
- Huiyi Huo
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Ziqiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan 410008, P.R. China
| | - Mingjie Wang
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xiaohe Yu
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Zhengchang Liao
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xiaocheng Zhou
- Department of Neonatology, The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, P.R. China
| | - Shaojie Yue
- Department of Neonatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
41
|
Luo J, Zhao Q, Zhang W, Zhang Z, Gao J, Zhang C, Li Y, Tian Y. A novel panel of microRNAs provides a sensitive and specific tool for the diagnosis of breast cancer. Mol Med Rep 2014; 10:785-91. [PMID: 24866395 DOI: 10.3892/mmr.2014.2274] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 03/24/2014] [Indexed: 11/05/2022] Open
Abstract
Breast cancer has a high fatality rate. Early diagnosis reduces the rate of mortality; therefore, novel diagnostic methods are urgently required. The present study investigated the correlation of serum microRNA (miRNA/miR) expression with breast cancer, and tested a panel of miRNAs as promising potential biomarkers for breast cancer. Six miRNAs (miR‑374, miR‑666‑5p, miR‑451, miR‑148a, miR‑27a and miR‑30b) were selected for analysis and their differential expression levels were quantified using qPCR. The results demonstrated that four out of the six candidate miRNAs were significantly downregulated in breast cancer patients (miR‑451, P=0.000; miR‑148a, P=0.021; miR‑27a, P=0.013 and miR‑30b, P=0.001). A panel of miRNAs consisting of the four downregulated miRNAs was able to distinguish breast cancer from healthy controls, with an area under the receiver operating characteristic curve of 95.3%, a sensitivity of 94.7% and a specificity of 82.8%. Thus, this panel of miRNAs may be used as a sensitive and specific tool for the diagnosis of breast cancer.
Collapse
Affiliation(s)
- Jinhua Luo
- Department of Clinical Biochemistry, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Qing Zhao
- Medical College of Nankai University, Tianjin 300071, P.R. China
| | - Wei Zhang
- Department of Gynecology and Obstetrics, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Zhuhong Zhang
- Medical College of Nankai University, Tianjin 300071, P.R. China
| | - Jing Gao
- Department of Clinical Biochemistry, Chinese PLA General Hospital, Beijing 100853, P.R. China
| | - Chunyan Zhang
- Medical College of Nankai University, Tianjin 300071, P.R. China
| | - Yali Li
- Department of Clinical Laboratory, Haidian Hospital, Beijing 100080, P.R. China
| | - Yaping Tian
- Department of Clinical Biochemistry, Chinese PLA General Hospital, Beijing 100853, P.R. China
| |
Collapse
|
42
|
Duan P, Sun S, Li B, Huang C, Xu Y, Han X, Xing Y, Yan W. miR-29a modulates neuronal differentiation through targeting REST in mesenchymal stem cells. PLoS One 2014; 9:e97684. [PMID: 24841827 PMCID: PMC4026383 DOI: 10.1371/journal.pone.0097684] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 04/23/2014] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE To investigate the modulation of microRNAs (miRNAs) upon the neuronal differentiation of mesenchymal stem cells (MSCs) through targeting RE-1 Silencing Factor (REST), a mature neuronal gene suppressor in neuronal and un-neuronal cells. METHODS Rat bone marrow derived-MSCs were induced into neuron-like cells (MSC-NCs) by DMSO and BHA in vitro. The expression of neuron specific enolase (NSE), microtubule-associated protein tau (Tau), REST and its target genes, including synaptosomal-associated protein 25 (SNAP25) and L1 cell adhesion molecular (L1CAM), were detected in MSCs and MSC-NCs. miRNA array analysis was conducted to screen for the upregulated miRNAs after neuronal differentiation. TargetScan was used to predict the relationship between these miRNAs and REST gene, and dual luciferase reporter assay was applied to validate it. Gain and loss of function experiments were used to study the role of miR-29a upon neuronal differentiation of MSCs. The knockdown of REST was conducted to show that miR-29a affected this process through targeting REST. RESULTS MSCs were induced into neuron-like cells which presented neuronal cell shape and expressed NSE and Tau. The expression of REST declined and the expression of SNAP25 and L1CAM increased upon the neuronal differentiation of MSCs. Among 14 upregulated miRNAs, miR-29a was validated to target REST gene. During the neuronal differentiation of MSCs, miR-29a inhibition blocked the downregulation of REST, as well as the upregulation of SNAP25, L1CAM, NSE and Tau. REST knockdown rescued the effect of miR-29a inhibition on the expression of NSE and Tau. Meanwhile, miR-29a knockin significantly decreased the expression of REST and increased the expression of SNAP25 and L1CMA in MSCs, but did not significantly affect the expression of NSE and Tau. CONCLUSION miR-29a regulates neurogenic markers through targeting REST in mesenchymal stem cells, which provides advances in neuronal differentiation research and stem cell therapy for neurodegenerative diseases.
Collapse
Affiliation(s)
- Ping Duan
- Institute of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Shiling Sun
- Hematology Department in the First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Bo Li
- Institute of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Chuntian Huang
- Institute of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Yan Xu
- Institute of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Xuefei Han
- Institute of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Ying Xing
- Department of Physiology, Xinxiang Medical University, Xinxiang, Henan, China
- * E-mail: (Y. Xing); (WY)
| | - Wenhai Yan
- Institute of Basic Medicine, Zhengzhou University, Zhengzhou, Henan, China
- * E-mail: (Y. Xing); (WY)
| |
Collapse
|
43
|
miRNA Expression in Mesenchymal Stem Cells. CURRENT PATHOBIOLOGY REPORTS 2014. [DOI: 10.1007/s40139-014-0045-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
44
|
Rokavec M, Li H, Jiang L, Hermeking H. The p53/miR-34 axis in development and disease. J Mol Cell Biol 2014; 6:214-30. [DOI: 10.1093/jmcb/mju003] [Citation(s) in RCA: 239] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
45
|
Cheng BB, Qu MJ, Wu LL, Shen Y, Yan ZQ, Zhang P, Qi YX, Jiang ZL. MicroRNA-34a targets Forkhead box j2 to modulate differentiation of endothelial progenitor cells in response to shear stress. J Mol Cell Cardiol 2014; 74:4-12. [PMID: 24792364 DOI: 10.1016/j.yjmcc.2014.04.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Revised: 04/14/2014] [Accepted: 04/21/2014] [Indexed: 10/25/2022]
Abstract
Flow shear stress plays important roles in modulating differentiation of endothelial progenitor cells (EPCs). MicroRNAs are crucial for diverse cellular processes, but the expressions and functions of microRNAs in EPCs responding to mechanical stimuli remain unclear. We sought to determine the effects of microRNA-34a (miR-34a) and a novel target Forkhead box j2 (Foxj2) on shear stress-induced EPC differentiation. Human umbilical cord blood-derived EPCs were exposed to laminar shear stress of 15dyn/cm(2) with parallel plate flow chamber system. Real time RT-PCR showed that shear stress significantly increased miR-34a expression, which was accompanied by the endothelial differentiation of EPCs. Whereas Foxj2, a putative target of miR-34a predicted by multiple algorithms, was suppressed in this process. Dual luciferase reporter assays, as well as miR-34a mimics and inhibitor treatment were used to confirm the interplay between miR-34a and Foxj2. Our results revealed an inverse correlation of miR-34a and Foxj2 expressions implicated in the endothelial differentiation of EPCs. MiR-34a contributed to this process by up-regulating the expressions of endothelial cell markers, and down-regulating smooth muscular cell markers. In addition, Foxj2 overexpression attenuated endothelial differentiation of EPCs, while Foxj2 siRNA had the opposite effect. These data suggested a unique mechanism that shear stress induces the expression of miR-34a, which targets to Foxj2 and promotes endothelial differentiation of EPCs. The results provide new insights into miR-34a/Foxj2 on shear stress-induced EPC differentiation.
Collapse
Affiliation(s)
- Bin-Bin Cheng
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ming-Juan Qu
- Department of Cell Biology & Genetics, School of Life Sciences, Ludong University, Yantai, China
| | - Lei-Lei Wu
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Shen
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi-Qiang Yan
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Zhang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ying-Xin Qi
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| | - Zong-Lai Jiang
- Institute of Mechanobiology & Medical Engineering, School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
46
|
Contribution of murine bone marrow mesenchymal stem cells to pancreas regeneration after partial pancreatectomy in mice. Cell Biol Int 2014; 36:823-31. [PMID: 22574754 DOI: 10.1042/cbi20110680] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The implantation of BMSCs (bone marrow mesenchymal stem cells) has emerged as a potential method of treating tissue damage, but the in vivo differentiation of BMSCs in an injured pancreas and its therapeutic effects have not been determined. Our aim has been to investigate the potential of BMSCs to contribute to the parenchyma and mesenchymal components of the pancreas during rapid regeneration, with preliminary exploration of the molecular mechanisms of this process. GFP(+) (green fluorescent protein(+) ) BMSCs were intravenously infused into the tail veins of mice that had received a 65-70% partial pancreatectomy, while mice that had only received a partial pancreatectomy and mice that had only been injected with BMSCs served as controls. Four weeks later, the injected GFP(+) BMSCs were diffusely engrafted in the pancreatic parenchyma and mesenchyma of the recipient mice with pancreatic injuries and had differentiated into pancreatic ductal epithelial cells (accounting for 1.7±0.3%), vascular endothelial cells (3.2±0.6%) and PSCs (pancreatic stellate cells) (5.2±1.6%), but no β or neural cells. Significantly, more engrafted and differentiated GFP(+) BMSCs were observed in the regenerating pancreas than in the normal pancreas. For the mice that received a partial pancreatectomy, the pancreatic weight/body weight of the mice with BMSC treatment was greater than mice without BMSC treatment (P<0.05). In addition, real-time RT-PCR (reverse transcription-PCR) showed that the expression levels of miR-9 (microRNA 9) and miR-204 in the engrafted BMSCs (5.2- and 2.6-fold, P<0.05, respectively) were increased compared with wild-type BMSCs. We also observed a significant reduction in the expression of miR-375 (0.71-fold, P<0.05) in engrafted GFP(+) BMSCs compared with wild-type BMSCs. BMSCs can therefore be a potential cell bank for treating pancreatic injuries by contributing to a variety of cell types. This process might be related to the expression of miR-9, miR-204 and miR-375.
Collapse
|
47
|
Bao B, Azmi AS, Li Y, Ahmad A, Ali S, Banerjee S, Kong D, Sarkar FH. Targeting CSCs in tumor microenvironment: the potential role of ROS-associated miRNAs in tumor aggressiveness. Curr Stem Cell Res Ther 2014; 9:22-35. [PMID: 23957937 PMCID: PMC4493722 DOI: 10.2174/1574888x113089990053] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 08/01/2013] [Accepted: 08/06/2013] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) have been widely considered as critical cellular signaling molecules involving in various biological processes such as cell growth, differentiation, proliferation, apoptosis, and angiogenesis. The homeostasis of ROS is critical to maintain normal biological processes. Increased production of ROS, namely oxidative stress, due to either endogenous or exogenous sources causes irreversible damage of bio-molecules such as DNA, proteins, lipids, and sugars, leading to genomic instability, genetic mutation, and altered gene expression, eventually contributing to tumorigenesis. A great amount of experimental studies in vitro and in vivo have produced solid evidence supporting that oxidative stress is strongly associated with increased tumor cell growth, treatment resistance, and metastasis, and all of which contribute to tumor aggressiveness. More recently, the data have indicated that altered production of ROS is also associated with cancer stem cells (CSCs), epithelial-to-mesenchymal transition (EMT), and hypoxia, the most common features or phenomena in tumorigenesis and tumor progression. However, the exact mechanism by which ROS is involved in the regulation of CSC and EMT characteristics as well as hypoxia- and, especially, HIF-mediated pathways is not well known. Emerging evidence suggests the role of miRNAs in tumorigenesis and progression of human tumors. Recently, the data have indicated that altered productions of ROS are associated with deregulated expression of miRNAs, suggesting their potential roles in the regulation of ROS production. Therefore, targeting ROS mediated through the deregulation of miRNAs by novel approaches or by naturally occurring anti-oxidant agents such as genistein could provide a new therapeutic approach for the prevention and/or treatment of human malignancies. In this article, we will discuss the potential role of miRNAs in the regulation of ROS production during tumorigenesis. Finally, we will discuss the role of genistein, as a potent anti-tumor agent in the regulation of ROS production during tumorigenesis and tumor development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Fazlul H Sarkar
- Departments of Pathology and Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, 740 HWCRC, 4100 John R Street, Detroit, MI 48201, USA.
| |
Collapse
|
48
|
Li Y, Liu Z, Xin H, Chopp M. The role of astrocytes in mediating exogenous cell-based restorative therapy for stroke. Glia 2013; 62:1-16. [PMID: 24272702 DOI: 10.1002/glia.22585] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2013] [Revised: 08/08/2013] [Accepted: 09/18/2013] [Indexed: 12/19/2022]
Abstract
Astrocytes have not been a major therapeutic target for the treatment of stroke, with most research emphasis on the neuron. Given the essential role that astrocytes play in maintaining physiological function of the central nervous system and the very rapid and sensitive reaction astrocytes have in response to cerebral injury or ischemic insult, we propose to replace the neurocentric view for treatment with a more nuanced astrocytic centered approach. In addition, after decades of effort in attempting to develop neuroprotective therapies, which target reduction of the ischemic lesion, there are no effective clinical treatments for stroke, aside from thrombolysis with tissue plasminogen activator, which is used in a small minority of patients. A more promising therapeutic approach, which may affect nearly all stroke patients, may be in promoting endogenous restorative mechanisms, which enhance neurological recovery. A focus of efforts in stimulating recovery post stroke is the use of exogenously administered cells. The present review focuses on the role of the astrocyte in mediating the brain network, brain plasticity, and neurological recovery post stroke. As a model to describe the interaction of a restorative cell-based therapy with astrocytes, which drives recovery from stroke, we specifically highlight the subacute treatment of stroke with multipotent mesenchymal stromal cell therapy.
Collapse
Affiliation(s)
- Yi Li
- Department of Neurology, Henry Ford Hospital, Detroit, Michigan
| | | | | | | |
Collapse
|
49
|
Bao B, Li Y, Ahmad A, Azmi AS, Bao G, Ali S, Banerjee S, Kong D, Sarkar FH. Targeting CSC-related miRNAs for cancer therapy by natural agents. Curr Drug Targets 2013; 13:1858-68. [PMID: 23140295 DOI: 10.2174/138945012804545515] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 10/09/2012] [Accepted: 11/03/2012] [Indexed: 12/22/2022]
Abstract
The theory of cancer stem cells (CSCs) has provided evidence on fundamental clinical implications because of the involvement of CSCs in cell migration, invasion, metastasis, and treatment resistance, which leads to the poor clinical outcome of cancer patients. Therefore, targeting CSCs will provide a novel therapeutic strategy for the treatment and/or prevention of tumors. However, the regulation of CSCs and its signaling pathways during tumorigenesis are not well understood. MicroRNAs (miRNAs) have been proved to act as key regulators of the post-transcriptional regulation of genes, which involve in a wide array of biological processes including tumorigenesis. The altered expressions of miRNAs are associated with poor clinical outcome of patients diagnosed with a variety of tumors. Therefore, emerging evidence strongly suggest that miRMAs play critical roles in tumor development and progression. Emerging evidence also suggest that miRNAs participate in the regulation of tumor cell growth, migration, invasion, angiogenesis, drug resistance, and metastasis. Moreover, miRNAs such as let-7, miR-21, miR-22, miR-34, miR-101, miR-146a, and miR-200 have been found to be associated with CSC phenotype and function mediated through targeting oncogenic signaling pathways. In this article, we will discuss the role of miRNAs in the regulation of CSC phenotype and function during tumor development and progression. We will also discuss the potential role of naturally occurring agents (nutraceuticals) as potent anti-tumor agents that are believed to function by targeting CSC-related miRNAs.
Collapse
Affiliation(s)
- Bin Bao
- Departments of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Hsieh JY, Huang TS, Cheng SM, Lin WS, Tsai TN, Lee OK, Wang HW. miR-146a-5p circuitry uncouples cell proliferation and migration, but not differentiation, in human mesenchymal stem cells. Nucleic Acids Res 2013; 41:9753-63. [PMID: 23963696 PMCID: PMC3834804 DOI: 10.1093/nar/gkt666] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Administration of mesenchymal stem cells (MSCs) has the potential to ameliorate degenerative disorders and to repair damaged tissues. The homing of transplanted MSCs to injured sites is a critical property of engraftment. Our aim was to identify microRNAs involved in controlling MSC proliferation and migration. MSCs can be isolated from bone marrow and umbilical cord Wharton’s jelly (BM-MSCs and WJ-MSCs, respectively), and WJ-MSCs show poorer motility yet have a better amplification rate compared with BM-MSCs. Small RNA sequencing revealed that miR-146a-5p is significantly overexpressed and has high abundance in WJ-MSCs. Knockdown of miR-146a-5p in WJ-MSCs inhibited their proliferation yet enhanced their migration, whereas overexpression of miR-146a-5p in BM-MSCs did not influence their osteogenic and adipogenic potentials. Chemokine (C-X-C motif) ligand 12 (CXCL12), together with SIKE1, which is an I-kappa-B kinase epsilon (IKKε) suppressor, is a direct target of miR-146a-5p in MSCs. Knockdown of miR-146a-5p resulted in the down-regulation of nuclear factor kappa-B (NF-κB) activity, which is highly activated in WJ-MSCs and is known to activate miR-146a-5p promoter. miR-146a-5p is also downstream of CXCL12, and a negative feedback loop is therefore formed in MSCs. These findings suggest that miR-146a-5p is critical to the uncoupling of motility and proliferation of MSCs. Our miRNome data also provide a roadmap for further understanding MSC biology.
Collapse
Affiliation(s)
- Jui-Yu Hsieh
- Department of Life Sciences, Institute of Microbiology and Immunology, National Yang-Ming University, Linong St. Taipei, Taiwan 11221, Division of Cardiology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Chenggong Rd. Taipei, Taiwan 11490, Department of Medical Research and Education, Taipei Veteran General Hospital, Shipai Rd., Taipei, Taiwan 11217, Stem Cell Research Center, National Yang-Ming University, Linong St. Taipei, Taiwan 11221, Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan, Veteran General Hospital-Yang Ming Genome Research Center, National Yang-Ming University, Linong St. Taipei, Taiwan 11221, Department of Education and Research, Taipei City Hospital, Zhengzhou Rd. Taipei, Taiwan 10341 and Cancer Research Center and Genome Research Center, National Yang-Ming University, Linong St. Taipei, Taiwan 11221
| | | | | | | | | | | | | |
Collapse
|