1
|
Ma D, Lin KY, Suresh D, Lin J, Gujar MR, Aung HY, Tan YS, Gao Y, Vincent AS, Chen T, Wang H. Arl2 GTPase associates with the centrosomal protein Cdk5rap2 to regulate cortical development via microtubule organization. PLoS Biol 2024; 22:e3002751. [PMID: 39137170 PMCID: PMC11321591 DOI: 10.1371/journal.pbio.3002751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 07/11/2024] [Indexed: 08/15/2024] Open
Abstract
ADP ribosylation factor-like GTPase 2 (Arl2) is crucial for controlling mitochondrial fusion and microtubule assembly in various organisms. Arl2 regulates the asymmetric division of neural stem cells in Drosophila via microtubule growth. However, the function of mammalian Arl2 during cortical development was unknown. Here, we demonstrate that mouse Arl2 plays a new role in corticogenesis via regulating microtubule growth, but not mitochondria functions. Arl2 knockdown (KD) leads to impaired proliferation of neural progenitor cells (NPCs) and neuronal migration. Arl2 KD in mouse NPCs significantly diminishes centrosomal microtubule growth and delocalization of centrosomal proteins Cdk5rap2 and γ-tubulin. Moreover, Arl2 physically associates with Cdk5rap2 by in silico prediction using AlphaFold multimer, which was validated by co-immunoprecipitation and proximity ligation assay. Remarkably, Cdk5rap2 overexpression significantly rescues the neurogenesis defects caused by Arl2 KD. Therefore, Arl2 plays an important role in mouse cortical development through microtubule growth via the centrosomal protein Cdk5rap2.
Collapse
Affiliation(s)
- Dongliang Ma
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore
| | - Kun-Yang Lin
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore
| | - Divya Suresh
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore
| | - Jiaen Lin
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore
| | - Mahekta R. Gujar
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore
| | - Htet Yamin Aung
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore
| | - Ye Sing Tan
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore
| | - Yang Gao
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore
| | - Anselm S. Vincent
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore
| | - Teng Chen
- College of Forensic Medicine, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, PR China
- The Key Laboratory of Health Ministry for Forensic Science, Xi’an Jiaotong University, Shaanxi, PR China
| | - Hongyan Wang
- Program in Neuroscience and Behavioural Disorders, Duke-NUS Medical School, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore
| |
Collapse
|
2
|
Yang L, Fang C, Song C, Zhang Y, Zhang R, Zhou S. Mesenchymal Stem Cell-Derived Exosomes are Effective for Radiation Enteritis and Essential for the Proliferation and Differentiation of Lgr5 + Intestinal Epithelial Stem Cells by Regulating Mir-195/Akt/β-Catenin Pathway. Tissue Eng Regen Med 2023; 20:739-751. [PMID: 37326937 PMCID: PMC10352229 DOI: 10.1007/s13770-023-00541-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/25/2023] [Accepted: 03/29/2023] [Indexed: 06/17/2023] Open
Abstract
BACKGROUND Radiation enteritis (RE) is a common complication of abdominal or pelvic radiotherapy, which when severe, could be life-threatening. Currently, there are no effective treatments. Studies have shown that mesenchymal stem cells (MSCs)-derived exosomes (MSC-exos) exhibit promising therapeutic effects in inflammatory diseases. However, the specific role of MSC-exos in RE and the regulatory mechanisms remain elusive. METHODS In vivo assay was carried out by injecting MSC-exos into the total abdominal irradiation (TAI)-induced RE mouse model. For in vitro assay, Lgr5-positive intestinal epithelial stem cells (Lgr5+ IESC) were extracted from mice, followed by irradiation along with MSC-exos treatment. HE staining was performed to measure histopathological changes. mRNA expression of inflammatory factors TNF-α and IL-6 and stem cell markers LGR5, and OCT4 were quantified by RT-qPCR. EdU and TUNEL staining was performed to estimate cell proliferation and apoptosis. MiR-195 expression in TAI mice and radiation-induced Lgr5+ IESC was tested. RESULTS We found that the injection of MSC-exos inhibited inflammatory reaction, increased stem cell marker expression, and maintained intestinal epithelial integrity in TAI mice. Furthermore, MSC-exos treatment increased the proliferation and simultaneously suppressed apoptosis in radiation-stimulated Lgr5+ IESC. MiR-195 expression increased by radiation exposure was decreased by MSC-exos therapy. MiR-195 overexpression facilitated the progress of RE by counteracting the effect of MSC-exos. Mechanistically, the Akt and Wnt/β-catenin pathways inhibited by MSC-exos were activated by miR-195 upregulation. CONCLUSION MSC-Exos are effective in treating RE and are essential for the proliferation and differentiation of Lgr5+ IESCs. Moreover, MSC-exos mediates its function by regulating miR-195 Akt β-catenin pathways.
Collapse
Affiliation(s)
- Leilei Yang
- Department of Gastrointestinal Surgery, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, No. 150, Ximen Street, Linhai, Taizhou, 317000, Zhejiang, China
| | - Chengfeng Fang
- Department of Gastrointestinal Surgery, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, No. 150, Ximen Street, Linhai, Taizhou, 317000, Zhejiang, China
| | - Caifang Song
- Department of Gastrointestinal Surgery, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, No. 150, Ximen Street, Linhai, Taizhou, 317000, Zhejiang, China
| | - Yaya Zhang
- Department of Gastrointestinal Surgery, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, No. 150, Ximen Street, Linhai, Taizhou, 317000, Zhejiang, China
| | - Ruili Zhang
- Department of Gastrointestinal Surgery, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, No. 150, Ximen Street, Linhai, Taizhou, 317000, Zhejiang, China.
| | - Shenkang Zhou
- Department of Gastrointestinal Surgery, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, No. 150, Ximen Street, Linhai, Taizhou, 317000, Zhejiang, China.
| |
Collapse
|
3
|
Gerstner CD, Reed M, Dahl TM, Ying G, Frederick JM, Baehr W. Arf-like Protein 2 (ARL2) Controls Microtubule Neogenesis during Early Postnatal Photoreceptor Development. Cells 2022; 12:147. [PMID: 36611941 PMCID: PMC9818799 DOI: 10.3390/cells12010147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/12/2022] [Accepted: 12/21/2022] [Indexed: 12/31/2022] Open
Abstract
Arf-like protein 2 (ARL2) is a ubiquitously expressed small GTPase with multiple functions. In a cell culture, ARL2 participates with tubulin cofactor D (TBCD) in the neogenesis of tubulin αβ-heterodimers, the building blocks of microtubules. To evaluate this function in the retina, we conditionally deleted ARL2 in mouse retina at two distinct stages, either during the embryonic development (retArl2-/-) or after ciliogenesis specifically in rods (rodArl2-/-). retArl2-/- retina sections displayed distorted nuclear layers and a disrupted microtubule cytoskeleton (MTC) as early as postnatal day 6 (P6). Rod and cone outer segments (OS) did not form. By contrast, the rod ARL2 knockouts were stable at postnatal day 35 and revealed normal ERG responses. Cytoplasmic dynein is reduced in retArl2-/- inner segments (IS), suggesting that dynein may be unstable in the absence of a normal MTC. We investigated the microtubular stability in the absence of either ARL2 (retARL2-/-) or DYNC1H1 (retDync1h1-/-), the dynein heavy chain, and found that both the retArl2-/- and retDync1h1-/- retinas exhibited reduced microtubules and nuclear layer distortion. The results suggest that ARL2 and dynein depend on each other to generate a functional MTC during the early photoreceptor development.
Collapse
Affiliation(s)
- Cecilia D. Gerstner
- Department of Ophthalmology, University of Utah Health Science Center, Salt Lake City, UT 84132, USA
| | - Michelle Reed
- Department of Ophthalmology, University of Utah Health Science Center, Salt Lake City, UT 84132, USA
| | - Tiffanie M. Dahl
- Department of Ophthalmology, University of Utah Health Science Center, Salt Lake City, UT 84132, USA
| | - Guoxin Ying
- Department of Ophthalmology, University of Utah Health Science Center, Salt Lake City, UT 84132, USA
| | - Jeanne M. Frederick
- Department of Ophthalmology, University of Utah Health Science Center, Salt Lake City, UT 84132, USA
| | - Wolfgang Baehr
- Department of Ophthalmology, University of Utah Health Science Center, Salt Lake City, UT 84132, USA
- Department of Neurobiology & Anatomy, University of Utah, Salt Lake City, UT 84112, USA
- Department of Biology, University of Utah, Salt Lake City, UT 84112, USA
| |
Collapse
|
4
|
Zhu F, Luo E, Yi F, Xiong J, Huang C, Li R. LncRNA ITSN1-2 knockdown inhibits OGD/R-induced inflammation and apoptosis in mouse hippocampal neurons via sponging miR-195-5p. Neuroreport 2021; 32:1325-1334. [PMID: 34554938 DOI: 10.1097/wnr.0000000000001732] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The upregulation of long noncoding RNA intersectin 1-2 (lnc ITSN1-2) is associated with poor prognosis in acute ischemic stroke (AIS) patients, but the role and mechanism of lnc ITSN1-2 in AIS are rarely reported, which, thus, are highlighted in this study. METHODS AIS cell model was constructed by oxygen glucose deprivation and reoxygenation (OGD/R). The quantitative real-time PCR was used to detect the expression of lnc ITSN1-2 in HT22 cells. The effects of lnc ITSN1-2 overexpression or knockdown on viability, LDH release, apoptosis, inflammatory and apoptotic factor expressions in OGD/R-induced HT22 cells were measured by cell counting kit-8 assay, LDH release kit, flow cytometry, ELISA and western blot, respectively. Starbase was used to screen the target genes of lnc ITSN1-2. The targeting relationship between lnc ITSN1-2 and miR-195-5p was predicted by starbase and verified by dual-luciferase report assay. The above assays were conducted again to study the function of miR-195-5p. Lastly, the levels of activated mitogen-activated protein kinases (MAPK) pathway-related proteins were determined by western blot. RESULTS OGD/R treatment reduced the HT22 cell viability and enhanced LDH release rate and lnc ITSN1-2 expression. Lnc ITSN1-2 overexpression promoted the cell injury, apoptosis and inflammation in OGD/R-induced HT22 cells, while lnc ITSN1-2 knockdown generated the opposite effect and deactivated the MAPK pathways. However, the effect of lnc ITSN1-2 knockdown in OGD/R-induced HT22 cells was reversed by miR-195-5p inhibitor. CONCLUSION Lnc ITSN1-2 knockdown suppressed the inflammation and apoptosis in OGD/R-induced HT22 cells by regulating the miR-195-5p-mediated MAPK pathways.
Collapse
Affiliation(s)
| | | | | | | | | | - Runying Li
- Department of Stomatology, PingXiang People's Hospital, Pingxiang, China
| |
Collapse
|
5
|
Carrella S, Massa F, Indrieri A. The Role of MicroRNAs in Mitochondria-Mediated Eye Diseases. Front Cell Dev Biol 2021; 9:653522. [PMID: 34222230 PMCID: PMC8249810 DOI: 10.3389/fcell.2021.653522] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022] Open
Abstract
The retina is among the most metabolically active tissues with high-energy demands. The peculiar distribution of mitochondria in cells of retinal layers is necessary to assure the appropriate energy supply for the transmission of the light signal. Photoreceptor cells (PRs), retinal pigment epithelium (RPE), and retinal ganglion cells (RGCs) present a great concentration of mitochondria, which makes them particularly sensitive to mitochondrial dysfunction. To date, visual loss has been extensively correlated to defective mitochondrial functions. Many mitochondrial diseases (MDs) show indeed neuro-ophthalmic manifestations, including retinal and optic nerve phenotypes. Moreover, abnormal mitochondrial functions are frequently found in the most common retinal pathologies, i.e., glaucoma, age-related macular degeneration (AMD), and diabetic retinopathy (DR), that share clinical similarities with the hereditary primary MDs. MicroRNAs (miRNAs) are established as key regulators of several developmental, physiological, and pathological processes. Dysregulated miRNA expression profiles in retinal degeneration models and in patients underline the potentiality of miRNA modulation as a possible gene/mutation-independent strategy in retinal diseases and highlight their promising role as disease predictive or prognostic biomarkers. In this review, we will summarize the current knowledge about the participation of miRNAs in both rare and common mitochondria-mediated eye diseases. Definitely, given the involvement of miRNAs in retina pathologies and therapy as well as their use as molecular biomarkers, they represent a determining target for clinical applications.
Collapse
Affiliation(s)
| | - Filomena Massa
- Telethon Institute of Genetics and Medicine, Naples, Italy
| | - Alessia Indrieri
- Telethon Institute of Genetics and Medicine, Naples, Italy.,Institute for Genetic and Biomedical Research, National Research Council (CNR), Milan, Italy
| |
Collapse
|
6
|
Zhu X, Wu T, Chi Y, Ge Y, Jiao Y, Zhu F, Cui L. MicroRNA-195 suppresses enterovirus A71-induced pyroptosis in human neuroblastoma cells through targeting NLRX1. Virus Res 2020; 292:198245. [PMID: 33253716 DOI: 10.1016/j.virusres.2020.198245] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 12/09/2022]
Abstract
Enterovirus A71 (EV-A71) emerged as a leading cause of virus derived infant encephalitis in most Asian countries. Some recent studies point out the critical role of microRNA (miRNA) in the regulation of pyroptosis. However, the role of miRNAs in the regulation of EV-A71 infection-induced pyroptosis was not previously explored. In this study, we utilized microRNA array and real-time PCR to verify that miR-195 significantly down-regulate in EV-A71-infected SH-SY5Y human neuroblastoma cells. An inverse correlation of NLRX1 with miR-195 expression in EV-A71-infected SH-SY5Y cells was found. Target prediction of miR-195 showed that NLRX1 could directly interact with miR-195. Results from luciferase reporter assays, qRT-PCR and western blotting demonstrated the negative regulation between miR-195 and NLRX1. Silencing NLRX1 expression with small interfering RNAs (siRNAs-NLRX1) and over-expression of miR-195 also attenuate the EV-A71 associated pyroptosis. Our findings provided evidence showed that miR-195 can regulate EV-A71 infection-induced pyroptosis, by directly targeting NLRX1.
Collapse
Affiliation(s)
- Xiaojuan Zhu
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| | - Tao Wu
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| | - Ying Chi
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| | - Yiyue Ge
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| | - Yongjun Jiao
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| | - Fengcai Zhu
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China
| | - Lunbiao Cui
- NHC Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, China.
| |
Collapse
|
7
|
Singh T, Yadav S. Role of microRNAs in neurodegeneration induced by environmental neurotoxicants and aging. Ageing Res Rev 2020; 60:101068. [PMID: 32283224 DOI: 10.1016/j.arr.2020.101068] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 03/02/2020] [Accepted: 04/04/2020] [Indexed: 02/06/2023]
Abstract
The progressive loss of neuronal structure and functions resulting in the death of neurons is considered as neurodegeneration. Environmental toxicants induced degeneration of neurons is accelerated with aging. In adult brains, most of the neurons are post-mitotic, and their loss results in the development of diseases like amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), Alzheimer's disease (AD), and Huntington's disease (HD). Neurodegenerative diseases have several similarities at the sub-cellular and molecular levels, such as synaptic degeneration, oxidative stress, inflammation, and cognitive decline, which are also known in brain aging. Identification of these similarities at the molecular level offers hope for the development of new therapeutics to ameliorate all neurodegenerative diseases simultaneously. Aging is known as the most strongly associated additive factor in the pathogenesis of neurodegenerative diseases. Studies carried out so far identified several genes, which are responsible for selective degeneration of neurons in different neurodegenerative diseases. Countless efforts have been made in identifying therapeutics for neurodegenerative diseases; however, the discovery of effective therapy remains elusive. Findings made in the last two decades identified microRNAs (miRNAs) as the most potent post-transcription regulatory RNA molecule, which can condition protein levels in the cell and tissue-specific manner. Identification of miRNAs, which regulate both neurotoxicant and aging-associated degeneration of brain cells, raises the possibility that roads leading to aging and neurotoxicant induced neurodegeneration cross at some point. Identification of miRNAs, which are common to aging and neurotoxicant induced neurodegeneration, will help in understanding the complex mechanism of neurodegenerative disease development. In the future, the use of natural miRNAs in vivo in therapy will be able to tackle several issues of aging and neurodegeneration. In the present review, we have provided a summary of findings made on the role of miRNAs in neurodegeneration and explored the common link made by miRNAs between aging and neurotoxicants induced neurodegeneration.
Collapse
Affiliation(s)
- Tanisha Singh
- Developmental Toxicology Division, Systems Toxicology and Health Risk Assessment Group, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan,31 Mahatma Gandhi Marg, Lucknow-226001, Uttar Pradesh, India; Department of Neurological Surgery, School of Medicine, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, Pennsylvania-15213, USA.
| | - Sanjay Yadav
- Developmental Toxicology Division, Systems Toxicology and Health Risk Assessment Group, CSIR- Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan,31 Mahatma Gandhi Marg, Lucknow-226001, Uttar Pradesh, India; Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Raebareli, Munsiganj, Raebareli 229405, UP, India.
| |
Collapse
|
8
|
Rittig AH, Johansen C, Celis P, Odum N, Litman T, Woetmann A, Lindahl LM, Iversen L. Suppressed microRNA-195-5p expression in mycosis fungoides promotes tumor cell proliferation. Exp Dermatol 2020; 30:1141-1149. [PMID: 32492224 DOI: 10.1111/exd.14124] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/19/2020] [Accepted: 05/24/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Several cancers, including mycosis fungoides (MF), have reported dysregulation of miR-195-5p. miR-195-5p plays a role in cell cycle regulation in several malignant diseases. OBJECTIVES This study aimed to investigate: (a) the expression level of miR-195-5p in lesional MF skin biopsies and (b) the potential regulatory roles of miR-195-5p in MF. METHODS Quantitative real-time polymerase chain reaction (RT-qPCR) was used to determine miR-195-5p expression in MF skin biopsies and cell lines. The effect of miR-195-5p and ADP-ribosylation factor-like protein 2 (ARL2) on cell cycle and apoptosis was measured by flow cytometry assays. Changes in ARL2 expression were determined by RT-qPCR and Western blotting (WB). RESULTS We found lower expression levels of miR-195-5p in lesional skin from MF patients compared with non-lesional MF skin and skin from healthy volunteers. Additionally, miR-195-5p showed lower expression levels in the skin from patients with disease progression compared with patients with stable disease. In vitro studies showed that overexpression of miR-195-5p induced a cell cycle arrest in G0G1. Using microarray analysis, we identified several genes that were regulated after miR-195-5p overexpression. The most downregulated gene after miR-195-5p mimic transfection was ARL2. RT-qPCR and WB analyses confirmed downregulation of ARL2 following transfection with miR-195-5p mimic. Lastly, transfection with siRNA against ARL2 also induced a G0G1 arrest. CONCLUSION Upregulation of miR-195-5p in MF inhibits cycle arrest by downregulation of ARL2. miR-195-5p may thus function as a tumor suppressor in MF and low miR-195-5p expression in lesional MF skin may promote disease progression.
Collapse
Affiliation(s)
- Anne H Rittig
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Claus Johansen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Pamela Celis
- Department of Molecular Medicine, Aarhus University, Aarhus, Denmark
| | - Niels Odum
- Department of Immunology and Microbiology, Leo Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Litman
- Department of Immunology and Microbiology, Leo Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
| | - Anders Woetmann
- Department of Immunology and Microbiology, Leo Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
| | - Lise M Lindahl
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| | - Lars Iversen
- Department of Dermatology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
9
|
Chang L, Zhang W, Shi S, Peng Y, Wang D, Zhang L, Zhang J. microRNA-195 attenuates neuronal apoptosis in rats with ischemic stroke through inhibiting KLF5-mediated activation of the JNK signaling pathway. Mol Med 2020; 26:31. [PMID: 32272873 PMCID: PMC7146986 DOI: 10.1186/s10020-020-00150-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 02/18/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Accumulating evidence has implicated the regulation of microRNAs (miRs) in ischemia stroke. The current study aimed to elucidate the role of microRNA-195 (miR-195) in neuronal apoptosis and brain plasticity in rats with ischemic stroke via the JNK signaling pathway/KLF5 axis. METHODS Ischemic stroke rat models were established by middle cerebral artery occlusion (MCAO), and oxygen deprivation (OGD) models were constructed in rat neuronal cells, followed by gain- or loss-of-function of miR-195 and/or KLF5 in rats and cells. Infarct volume, neuronal loss and ultrastructure, the expression of GAP-43, SYP and KLF5 protein as well as cell apoptosis were determined in the rats. Caspase-3 activity as well as the expression of miR-195, KLF5, GAP-43, SYP, JNK, phosphorylated JNK, Bax and Bcl-2 was measured in the cells. RESULTS The infarct size, expression of GAP-43 and SYP protein and apoptotic cells were increased in the miR-195-/- MCAO rats, while reductions were detected in the miR-195 mimic MCAO and KLF5-/- MCAO rats. Bcl-2 expression was increased, Bax and Caspase-3 expression as well as the ratio of phosphorylated JNK/JNK was decreased in response to miR-195 overexpression or KLF5 knockdown. Interestingly, the silencing of KLF5 reversed the effects exerted by the miR-195 inhibitor on the expression of Bcl-2, phosphorylated JNK/JNK, Bax and Caspase-3. CONCLUSIONS Collectively, our study unraveled that miR-195 could down-regulate KLF5 and block the JNK signaling pathway, ultimately inhibiting neuronal apoptosis in rats with ischemic stroke.
Collapse
Affiliation(s)
- Lisha Chang
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, No. 73, Jianshe South Road, Tangshan, 063000, Hebei Province, People's Republic of China
| | - Wan Zhang
- Quality Control Office, North China University of Science and Technology Affiliated Hospital, Tangshan, 063000, People's Republic of China
| | - Songxin Shi
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, No. 73, Jianshe South Road, Tangshan, 063000, Hebei Province, People's Republic of China
| | - Yanbo Peng
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, No. 73, Jianshe South Road, Tangshan, 063000, Hebei Province, People's Republic of China
| | - Dali Wang
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, No. 73, Jianshe South Road, Tangshan, 063000, Hebei Province, People's Republic of China
| | - Li Zhang
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, No. 73, Jianshe South Road, Tangshan, 063000, Hebei Province, People's Republic of China
| | - Jiang Zhang
- Department of Neurology, North China University of Science and Technology Affiliated Hospital, No. 73, Jianshe South Road, Tangshan, 063000, Hebei Province, People's Republic of China.
| |
Collapse
|
10
|
Li K, Zhao B, Wei D, Cui Y, Qian L, Wang W, Liu G. Long non-coding RNA ANRIL enhances mitochondrial function of hepatocellular carcinoma by regulating the MiR-199a-5p/ARL2 axis. ENVIRONMENTAL TOXICOLOGY 2020; 35:313-321. [PMID: 31670868 DOI: 10.1002/tox.22867] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 10/11/2019] [Accepted: 10/16/2019] [Indexed: 06/10/2023]
Abstract
Although the roles of long non-coding RNA (lncRNA) ANRIL (Antisense non-coding RNA in the INK4A locus) have been established in various tumors, its roles in mitochondrial metabolic reprogramming of hepatocellular carcinoma (HCC) cells are still unclear. This work aims to explore lncRNA ANRIL roles in regulating the mitochondrial metabolic reprogramming of liver cancer cells. First, we found that lncRAN ANRIL expression was significantly increased in HCC tissues or cells compared with the normal adjacent tissues and normal tissues or cells. Functional experiment showed that overexpression of lncRNA ANRIL promoted mitochondrial function in HCC cells, evident by the increased mitochondrial DNA copy numbers, ATP (Adenosine triphosphate) level, mitochondrial membrane potential, and the expression levels of mitochondrial markers, while ANRIL knockdown exerted the opposite effects. Mechanistically, lncRNA ANRIL acted as a competing endogenous RNA to increase ARL2 (ADP-ribosylationfactor-like 2) expression via sponging miR-199a-5p. Notably, the miR-199a-5p/ARL2 axis is necessary for ANRIL-mediated promoting effects on HCC cell mitochondrial function. This work reveals a novel ANRIL-miR-199a-5p-ARL2 axis in HCC cell progression, which might provide potential targets for HCC treatment.
Collapse
Affiliation(s)
- Kun Li
- Institute of Biomedical and Health Science, School of Life and Health Science, Anhui Science and Technology University, Fengyang, China
| | - Bao Zhao
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Diandian Wei
- Institute of Biomedical and Health Science, School of Life and Health Science, Anhui Science and Technology University, Fengyang, China
| | - Yixuan Cui
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Lisheng Qian
- Institute of Biomedical and Health Science, School of Life and Health Science, Anhui Science and Technology University, Fengyang, China
| | - Wenrui Wang
- School of Life Science and Technology, Bengbu Medical College, Bengbu, China
| | - Guodong Liu
- Institute of Biomedical and Health Science, School of Life and Health Science, Anhui Science and Technology University, Fengyang, China
| |
Collapse
|
11
|
Exosomal microRNA-122 mediates obesity-related cardiomyopathy through suppressing mitochondrial ADP-ribosylation factor-like 2. Clin Sci (Lond) 2019; 133:1871-1881. [PMID: 31434696 DOI: 10.1042/cs20190558] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/14/2019] [Accepted: 08/21/2019] [Indexed: 12/23/2022]
Abstract
Emerging studies have demonstrated that microRNAs (miRs) participate in the development of multiple metabolic complications including cardiovascular diseases. Among them, circulating level of liver-secreted miR-122 was closely correlated with several consequence of heart diseases in clinical studies, and overexpression of miR-122 impaired cardiomyocyte function. However, it was unknown whether miR-122 could regulate cardiac biology in obesity. Therefore, present study was to disclose the role of miR-122 in cardiac metabolic disorders and potential molecular mechanisms. Through utilizing clinical samples and high fat diet-fed mice, we investigated the physiological roles of miR-122 in obesity-related cardiomyopathy. Besides, present study explored the mitochondrial function under exosomal miR-122 stimulation in mouse primary cardiomyocytes. In clinical samples and obese mice, the circulating level of exosomal miR-122 was positively correlated with cardiac dysfunctional parameters, including reduction in ejection fraction (EF) and increased levels of NT-proBNP. Human plasma exosomes transported miR-122 into mouse primary cardiomyocytes, and impaired mitochondrial ATP production and oxygen consumption, whereas miR-122 sponge improved these inhibitory effects. In dietary-induced mice, increased hepatic and circulating exosomal miR-122 deteriorated cardiac structure and functional index, and inhibited mitochondrial function. Liver-specific blockage of miR-122 attenuated abnormal cardiac remodeling. Mechanistically, miR-122 directly bound and suppressed mitochondrial protein ADP-ribosylation factor-like 2 (Arl-2) in vitro and in vivo Knockdown of Arl-2 abolished the mitochondrial benefits of miR-122 sponge in exosome-treated mouse primary cardiomyocytes.In conclusions, our present study firstly showed that liver-secreted exosomal miR-122 played a critical role in the development of metabolic cardiomyopathy, and miR-122/mitochondrial Arl-2 signaling affected cardiac energy homeostasis.
Collapse
|
12
|
Xuan W, Yu H, Zhang X, Song D. Crosstalk between the lncRNA UCA1 and microRNAs in cancer. FEBS Lett 2019; 593:1901-1914. [PMID: 31166011 DOI: 10.1002/1873-3468.13470] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 05/28/2019] [Accepted: 05/29/2019] [Indexed: 12/11/2022]
Abstract
Long non-coding RNAs (lncRNAs) are a major subset of highly conserved non-coding RNAs (ncRNAs) that consist of at least 200 nucleotides and have limited protein-coding potential. Cumulative data have shown that lncRNAs are deregulated in many types of cancer and may control pathophysiological processes of cancer at various levels, including transcription, post-transcription and translation. Recently, lncRNAs have been demonstrated to interact with microRNAs (miRNAs), another major subset of ncRNAs, which regulate physiological and pathological processes by inhibiting target mRNA translation or promoting mRNA degradation. The lncRNA urothelial carcinoma-associated 1 (UCA1) has recently gained much attention as it is overexpressed in many types of cancer and is involved in carcinogenesis. Here, we review the crosstalk between UCA1 and miRNAs during the pathogenesis of cancer, with a focus on cancer-cell proliferation, invasion, drug resistance, and metabolism.
Collapse
Affiliation(s)
- Wei Xuan
- Department of Hepatopancreaticobiliary Surgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Hongyu Yu
- Department of Nephrology, Second Hospital of Jilin University, Changchun, China
| | - Xiaoling Zhang
- The First Hospital and Institute of Immunology, Jilin University, Changchun, China
| | - Dandan Song
- Department of Clinical Laboratory, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
13
|
Clotaire DZJ, Du X, Wei Y, Yang D, Hua J. miR-19b-3p integrates Jak-Stat signaling pathway through Plzf to regulate self-renewal in dairy goat male germline stem cells. Int J Biochem Cell Biol 2018; 105:104-114. [PMID: 30393202 DOI: 10.1016/j.biocel.2018.10.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 10/19/2018] [Accepted: 10/25/2018] [Indexed: 12/27/2022]
|
14
|
Wang Y, Guan G, Cheng W, Jiang Y, Shan F, Wu A, Cheng P, Guo Z. ARL2 overexpression inhibits glioma proliferation and tumorigenicity via down-regulating AXL. BMC Cancer 2018; 18:599. [PMID: 29843637 PMCID: PMC5975491 DOI: 10.1186/s12885-018-4517-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 05/18/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Glioma is the most common primary brain tumor in adults with a poor prognosis. As a member of ARF subfamily GTPase, ARL2 plays a key role in regulating the dynamics of microtubules and mitochondrial functions. Recently, ARL2 has been identified as a prognostic and therapeutic target in a variety range of malignant tumors. However, the biological functional role of ARL2 in glioma still remains unknown. The aim of this study was to explore the expression and functional role of ARL2 in glioma. METHODS In this study, we investigated the expression of ARL2 in glioma samples by using RT-PCR, immunohistochemistry and western blot. The correlation between ARL2 expression and the outcomes of glioma patients was evaluated with survival data from TCGA, CGGA and Rembrandt dataset. Lentiviral technique was used for ARL2 overexpression in U87 and U251 cells. CCK8 assay, colony formation assay, wound healing test, transwell invasion assay and in vivo subcutaneous xenograft model were performed to investigated the biological functions of ARL2. RESULTS ARL2 expression was down-regulated in glioma, and was inversely associated with poor prognosis in glioma patients. Furthermore, exogenous ARL2 overexpression attenuated the growth and colony-formation abilities of glioma cells, as well as their migration and invasive capabilities. Moreover, elevated expression of ARL2 inhibited in vivo tumorigenicity of glioma cells. Mechanistically, ARL2 regulated AXL expression, which was known as an important functional regulator of proliferation and tumorigenicity in glioma cells. CONCLUSION Our study suggests that ARL2 inhibits the proliferation, migration and tumorigenicity of glioma cells by regulating the expression of AXL and may conduct as a new prognostic and therapeutic target for glioma.
Collapse
Affiliation(s)
- Yulin Wang
- Department of Neurosurgery, The First Hospital of China Medical University, 155 Nanjingbei Street, Heping, Shenyang, Liaoning, 110001, People's Republic of China
| | - Gefei Guan
- Department of Neurosurgery, The First Hospital of China Medical University, 155 Nanjingbei Street, Heping, Shenyang, Liaoning, 110001, People's Republic of China
| | - Wen Cheng
- Department of Neurosurgery, The First Hospital of China Medical University, 155 Nanjingbei Street, Heping, Shenyang, Liaoning, 110001, People's Republic of China
| | - Yang Jiang
- Department of Neurosurgery, The First Hospital of China Medical University, 155 Nanjingbei Street, Heping, Shenyang, Liaoning, 110001, People's Republic of China
| | - Fengping Shan
- Department of Immunology, School of Basic Medical Science, China Medical University, Shenyang, 110122, Liaoning, China
| | - Anhua Wu
- Department of Neurosurgery, The First Hospital of China Medical University, 155 Nanjingbei Street, Heping, Shenyang, Liaoning, 110001, People's Republic of China
| | - Peng Cheng
- Department of Neurosurgery, The First Hospital of China Medical University, 155 Nanjingbei Street, Heping, Shenyang, Liaoning, 110001, People's Republic of China.
| | - Zongze Guo
- Department of Neurosurgery, The First Hospital of China Medical University, 155 Nanjingbei Street, Heping, Shenyang, Liaoning, 110001, People's Republic of China.
| |
Collapse
|
15
|
Li C, Ma Y, Zhang K, Gu J, Tang F, Chen S, Cao L, Li S, Jin Y. Aberrant transcriptional networks in step-wise neurogenesis of paroxysmal kinesigenic dyskinesia-induced pluripotent stem cells. Oncotarget 2018; 7:53611-53627. [PMID: 27449084 PMCID: PMC5288209 DOI: 10.18632/oncotarget.10680] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 06/30/2016] [Indexed: 12/31/2022] Open
Abstract
Paroxysmal kinesigenic dyskinesia (PKD) is an episodic movement disorder with autosomal-dominant inheritance and marked variability in clinical manifestations. Proline-rich transmembrane protein 2 (PRRT2) has been identified as a causative gene of PKD, but the molecular mechanism underlying the pathogenesis of PKD still remains a mystery. The phenotypes and transcriptional patterns of the PKD disease need further clarification. Here, we report the generation and neural differentiation of iPSC lines from two familial PKD patients with c.487C>T (p. Gln163X) and c.573dupT (p. Gly192Trpfs*8) PRRT2 mutations, respectively. Notably, an extremely lower efficiency in neural conversion from PKD-iPSCs than control-iPSCs is observed by a step-wise neural differentiation method of dual inhibition of SMAD signaling. Moreover, we show the high expression level of PRRT2 throughout the human brain and the expression pattern of PRRT2 in other human tissues for the first time. To gain molecular insight into the development of the disease, we conduct global gene expression profiling of PKD cells at four different stages of neural induction and identify altered gene expression patterns, which peculiarly reflect dysregulated neural transcriptome signatures and a differentiation tendency to mesodermal development, in comparison to control-iPSCs. Additionally, functional and signaling pathway analyses indicate significantly different cell fate determination between PKD-iPSCs and control-iPSCs. Together, the establishment of PKD-specific in vitro models and the illustration of transcriptome features in PKD cells would certainly help us with better understanding of the defects in neural conversion as well as further investigations in the pathogenesis of the PKD disease.
Collapse
Affiliation(s)
- Chun Li
- Laboratory of Molecular Developmental Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yu Ma
- Laboratory of Molecular Developmental Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Kunshan Zhang
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China
| | - Junjie Gu
- Laboratory of Molecular Developmental Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Fan Tang
- Laboratory of Molecular Developmental Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shengdi Chen
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Key Laboratory of Stem Cell Biology, Center for The Excellence in Molecular and Cell Sciences, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Li Cao
- Department of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Siguang Li
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China.,Collaborative Innovation Center for Brain Science, Tongji University, Shanghai 200092, China
| | - Ying Jin
- Laboratory of Molecular Developmental Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,Key Laboratory of Stem Cell Biology, Center for The Excellence in Molecular and Cell Sciences, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences and Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| |
Collapse
|
16
|
Zammit V, Baron B, Ayers D. MiRNA Influences in Neuroblast Modulation: An Introspective Analysis. Genes (Basel) 2018; 9:genes9010026. [PMID: 29315268 PMCID: PMC5793179 DOI: 10.3390/genes9010026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 12/22/2017] [Accepted: 12/29/2017] [Indexed: 02/07/2023] Open
Abstract
Neuroblastoma (NB) is the most common occurring solid paediatric cancer in children under the age of five years. Whether of familial or sporadic origin, chromosome abnormalities contribute to the development of NB and cause dysregulation of microRNAs (miRNAs). MiRNAs are small non-coding, single stranded RNAs that target messenger RNAs at the post-transcriptional levels by repressing translation within all facets of human physiology. Such gene 'silencing' activities by miRNAs allows the development of regulatory feedback loops affecting multiple functions within the cell, including the possible differentiation of neural stem cell (NSC) lineage selection. Neurogenesis includes stages of self-renewal and fate specification of NSCs, migration and maturation of young neurones, and functional integration of new neurones into the neural circuitry, all of which are regulated by miRNAs. The role of miRNAs and their interaction in cellular processes are recognised aspects of cancer genetics, and miRNAs are currently employed as biomarkers for prognosis and tumour characterisation in multiple cancer models. Consequently, thorough understanding of the mechanisms of how these miRNAs interplay at the transcriptomic level will definitely lead to the development of novel, bespoke and efficient therapeutic measures, with this review focusing on the influences of miRNAs on neuroblast modulations leading to neuroblastoma.
Collapse
Affiliation(s)
- Vanessa Zammit
- National Blood Transfusion Service, St. Luke's Hospital, PTA1010 G'Mangia, Malta.
- School of Biomedical Science and Physiology, University of Wolverhampton, Wolverhampton WV1 1LY, UK.
| | - Byron Baron
- Centre for Molecular Medicine and Biobanking, Faculty of Medicine and Surgery, University of Malta, MSD2080 Msida, Malta.
| | - Duncan Ayers
- Centre for Molecular Medicine and Biobanking, Faculty of Medicine and Surgery, University of Malta, MSD2080 Msida, Malta.
- School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PL, UK.
| |
Collapse
|
17
|
Shan J, Zhao B, Shan Z, Nie J, Deng R, Xiong R, Tsun A, Pan W, Zhao H, Chen L, Jin Y, Qian Z, Lui K, Liang R, Li D, Sun B, Lavillette D, Xu K, Li B. Histone demethylase LSD1 restricts influenza A virus infection by erasing IFITM3-K88 monomethylation. PLoS Pathog 2017; 13:e1006773. [PMID: 29281729 PMCID: PMC5760097 DOI: 10.1371/journal.ppat.1006773] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 01/09/2018] [Accepted: 11/27/2017] [Indexed: 11/30/2022] Open
Abstract
The histone demethylase LSD1 has been known as a key transcriptional coactivator for DNA viruses such as herpes virus. Inhibition of LSD1 was found to block viral genome transcription and lytic replication of DNA viruses. However, RNA virus genomes do not rely on chromatin structure and histone association, and the role of demethylase activity of LSD1 in RNA virus infections is not anticipated. Here, we identify that, contrary to its role in enhancing DNA virus replication, LSD1 limits RNA virus replication by demethylating and activating IFITM3 which is a host restriction factor for many RNA viruses. We have found that LSD1 is recruited to demethylate IFITM3 at position K88 under IFNα treatment. However, infection by either Vesicular Stomatitis Virus (VSV) or Influenza A Virus (IAV) triggers methylation of IFITM3 by promoting its disassociation from LSD1. Accordingly, inhibition of the enzymatic activity of LSD1 by Trans-2-phenylcyclopropylamine hydrochloride (TCP) increases IFITM3 monomethylation which leads to more severe disease outcomes in IAV-infected mice. In summary, our findings highlight the opposite role of LSD1 in fighting RNA viruses comparing to DNA viruses infection. Our data suggest that the demethylation of IFITM3 by LSD1 is beneficial for the host to fight against RNA virus infection. The viral genomes of DNA viruses but not RNA viruses form chromatin structure during infection. Thus, epigenetic modulators are not expected to have crucial roles in RNA viral infection. However, here, we identify for the first time, that, opposite to its role in enhancing DNA virus replication, LSD1, a histone demethylase, limits RNA virus replication. We show that, under IFNα treatment, LSD1 is involved in the demethylation of IFITM3, a well-known host restriction factor for many RNA viruses. To counteract IFITM3 activation by demethylation, several RNA viruses, such as VSV and IAV, but not Zika virus, have developed strategy to inactive IFITM3 by promoting its dissociation from LSD1. In agreement with our findings, the inhibition of the enzymatic activity of LSD1 by small molecule leads to more severe disease outcomes in IAV-infected mice. Our data suggest that although LSD1 inhibitor is beneficial for treating DNA virus infection, it could be harmful to the host suffering from RNA virus infection. On the contrary, developing strategies to stimulate LSD1 activity to demethylate of IFITM3 is essential to fight RNA viruses.
Collapse
Affiliation(s)
- Jiaoyu Shan
- CAS Center for Excellence in Molecular Cell Science, CAS Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Human Parasitology Department of Basic Medicine College, Xinjiang Medical University, Urumqi, China
| | - Binbin Zhao
- CAS Center for Excellence in Molecular Cell Science, CAS Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Zhao Shan
- CAS Center for Excellence in Molecular Cell Science, CAS Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jia Nie
- CAS Center for Excellence in Molecular Cell Science, CAS Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rong Deng
- CAS Center for Excellence in Molecular Cell Science, CAS Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rui Xiong
- CAS Key Laboratory of Molecular Virology and Immunology, Unit of interspecies transmission of arboviruses and therapeutics research, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Andy Tsun
- CAS Center for Excellence in Molecular Cell Science, CAS Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Weiqi Pan
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Hanzhi Zhao
- CAS Center for Excellence in Molecular Cell Science, Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Ling Chen
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ying Jin
- CAS Center for Excellence in Molecular Cell Science, Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Zhikang Qian
- CAS Key Laboratory of Molecular Virology and Immunology, Unit of Herpesvirus and Molecular Virology Research, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Kawing Lui
- CAS Center for Excellence in Molecular Cell Science, CAS Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rui Liang
- CAS Center for Excellence in Molecular Cell Science, CAS Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Dan Li
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai JiaoTong University, Shanghai, China
| | - Bing Sun
- CAS Center for Excellence in Molecular Cell Science, CAS Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Virology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Dimitri Lavillette
- CAS Key Laboratory of Molecular Virology and Immunology, Unit of interspecies transmission of arboviruses and therapeutics research, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ke Xu
- CAS Center for Excellence in Molecular Cell Science, CAS Key Laboratory of Molecular Virology and Immunology, Unit of Molecular Immunology, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- CAS Key Laboratory of Molecular Virology and Immunology, Unit of interspecies transmission of arboviruses and therapeutics research, Institut Pasteur of Shanghai, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- * E-mail: (BL); (KX)
| | - Bin Li
- Shanghai Institute of Immunology, Shanghai JiaoTong University School of Medicine, Shanghai, China
- Department of Immunology and Microbiology, Shanghai JiaoTong University School of Medicine, Shanghai JiaoTong University, Shanghai, China
- * E-mail: (BL); (KX)
| |
Collapse
|
18
|
Miao X, Wu X, Shi W. MicroRNA-346 regulates neural stem cell proliferation and differentiation by targeting KLF4. Am J Transl Res 2017; 9:5400-5410. [PMID: 29312492 PMCID: PMC5752890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Accepted: 10/28/2017] [Indexed: 06/07/2023]
Abstract
MicroRNAs have been shown to play an important role in stem cell fate determination and self-renewal. However, the role of miRNAs in neural stem cells (NSCs) remains poorly understood. In this study, we showed that miR-346, a less characterized microRNA, promoted NSCs proliferation, differentiation and apoptosis by targeting KLF4, a core transcriptional factor in stem cell fate determination. Our data suggested that miR-346 could directly target the 3'-untranslated region of KLF4. Overexpression of miR-346 decreased KLF4 expression at both mRNA and protein levels in NSCs. More importantly, Overexpression of miR-346 repressed NSC proliferation and induced the expression of lineage markers including GFAP and Tuj1. Additionally, overexpression of miR-346 promoted apoptosis of NSCs. In concert, suppressing its expression by an antisense RNA, anti-miR-346, promoted NSC proliferation, and meanwhile inhibited its differentiation and apoptosis. We also showed that the effects of miR-346 overexpression could be reversed by re-expression of KLF4. Taken together, Those data suggest that miR-346 is a novel miRNA that regulates NSC proliferation and differentiation by targeting KLF4.
Collapse
Affiliation(s)
- Xingyu Miao
- Department of Neurosurgery, The Third Affiliated Hospital, School of Medicine, Xi’an Jiaotong UniversityXi’an, China
- Department of Neurosurgery, Shaanxi Provincial People’s HospitalXi’an, China
| | - Xiaoying Wu
- Department of Orthopedics, Shaanxi Provincial People’s HospitalXi’an, China
| | - Wei Shi
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Xi’an Jiaotong UniversityXi’an, China
| |
Collapse
|
19
|
Liu X, Wu D, Wen S, Zhao S, Xia A, Li F, Ji X. Mild therapeutic hypothermia protects against cerebral ischemia/reperfusion injury by inhibiting miR-15b expression in rats. Brain Circ 2017; 3:219-226. [PMID: 30276328 PMCID: PMC6057705 DOI: 10.4103/bc.bc_15_17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 07/10/2017] [Accepted: 07/24/2017] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Mild hypothermia has a protective effect on ischemic stroke, but the mechanisms remain elusive. Here, we investigated microRNA (miRNA) profiles and the specific role of miRNAs in ischemic stroke treated with mild hypothermia. MATERIALS AND METHODS Male adult Sprague Dawley rats were subjected to focal transient cerebral ischemia. Mild hypothermia was induced by applying ice packs around the neck and head of the animals. miRNAs expression profiles were detected in ischemic stroke treated with mild therapeutic hypothermia through miRNA chips. Reverse transcription-polymerase chain reaction (RT-PCR) was used to verify the change of miRNA array. Western blot and adenosine triphosphate (ATP) assay kits were used to detect the changes of protein expression and ATP levels, respectively. miR-15b mimic and its control were injected into the right lateral ventricle 60 min before the induction of ischemia. RESULTS The results showed that mild hypothermia affected miRNAs profiles expression. We verified the expression of miR-15b and miR-598-3p by miRNA RT-PCR. miR-15b mimic inhibited the expression of its target, ADP ribosylation factor-like 2 (Arl2) protein, and decreased ATP levels in PC12 cells. Compared with the control, miR-15b mimic increased the infarct volume and aggravated the neurological function under normothermia or hypothermia treatment. Furthermore, the expression of Arl2 was decreased in the miR-15b mimic group under normothermia or hypothermia treatment. CONCLUSIONS Mild therapeutic hypothermia affected miRNA profiles and protected against cerebral ischemia/reperfusion by inhibiting miR-15b expression in rats. miR-15b may be a potential target for therapeutic intervention in stroke.
Collapse
Affiliation(s)
- Xiangrong Liu
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, PR China
- China-America Joint Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, PR China
| | - Di Wu
- China-America Joint Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, PR China
| | - Shaohong Wen
- China-America Joint Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, PR China
| | - Shunying Zhao
- China-America Joint Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, PR China
| | - Ao Xia
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, PR China
- China-America Joint Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China
| | - Fang Li
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, PR China
- China-America Joint Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China
| | - Xunming Ji
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, Beijing 100053, PR China
- China-America Joint Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing 100053, PR China
- Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, PR China
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing 100053, PR China
| |
Collapse
|
20
|
Zhao H, Han Z, Liu X, Gu J, Tang F, Wei G, Jin Y. The chromatin remodeler Chd4 maintains embryonic stem cell identity by controlling pluripotency- and differentiation-associated genes. J Biol Chem 2017; 292:8507-8519. [PMID: 28298436 DOI: 10.1074/jbc.m116.770248] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 03/13/2017] [Indexed: 12/23/2022] Open
Abstract
The unique properties of embryonic stem cells (ESCs), including unlimited self-renewal and pluripotent differentiation potential, are sustained by integrated genetic and epigenetic networks composed of transcriptional factors and epigenetic modulators. However, the molecular mechanisms underlying the function of these regulators are not fully elucidated. Chromodomain helicase DNA-binding protein 4 (Chd4), an ATPase subunit of the nucleosome remodeling and deacetylase (NuRD) complex, is highly expressed in ESCs. However, its function in ESC regulation remains elusive. Here we report that Chd4 is required for the maintenance of ESC self-renewal. RNAi-mediated silencing of Chd4 disrupted self-renewal and up-regulated lineage commitment-associated genes under self-renewal culture conditions. During ESC differentiation in embryoid body formation, we observed significantly stronger induction of differentiation-associated genes in Chd4-deficient cells. The phenotype was different from that caused by the deletion of Mbd3, another subunit of the NuRD complex. Transcriptomic analyses revealed that Chd4 secured ESC identity by controlling the expression of subsets of pluripotency- and differentiation-associated genes. Importantly, Chd4 repressed the transcription of T box protein 3 (Tbx3), a transcription factor with important functions in ESC fate determination. Tbx3 knockdown partially rescued aberrant activation of differentiation-associated genes, especially of endoderm-associated genes, induced by Chd4 depletion. Moreover, we identified an interaction of Chd4 with the histone variant H2A.Z. This variant stabilized Chd4 by inhibiting Chd4 protein degradation through the ubiquitin-proteasome pathway. Collectively, this study identifies the Chd4-Tbx3 axis in controlling ESC fate and a role of H2A.Z in maintaining the stability of Chd4 proteins.
Collapse
Affiliation(s)
- Haixin Zhao
- Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, 320 Yueyang Road, Shanghai 200031, China; University of the Chinese Academy of Sciences
| | - Zhijun Han
- Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xinyuan Liu
- Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, 320 Yueyang Road, Shanghai 200031, China
| | - Junjie Gu
- Laboratory of Molecular Developmental Biology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Fan Tang
- Laboratory of Molecular Developmental Biology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Gang Wei
- Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ying Jin
- Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, 320 Yueyang Road, Shanghai 200031, China; Laboratory of Molecular Developmental Biology, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
21
|
Yang J, Zhao H, Ma Y, Shi G, Song J, Tang Y, Li S, Li T, Liu N, Tang F, Gu J, Zhang L, Zhang Z, Zhang X, Jin Y, Le W. Early pathogenic event of Alzheimer's disease documented in iPSCs from patients with PSEN1 mutations. Oncotarget 2017; 8:7900-7913. [PMID: 27926491 PMCID: PMC5352369 DOI: 10.18632/oncotarget.13776] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 11/21/2016] [Indexed: 11/25/2022] Open
Abstract
Alzheimer's disease (AD) is the most common age-related dementia characterized by progressive neuronal loss. However, the molecular mechanisms for the neuronal loss is still debated. Here, we used induced pluripotent stem cells (iPSCs) derived from somatic cells of familial AD patients carrying PSEN1 mutations to study the early pathogenic event of AD. We found that premature neuronal differentiation with decreased proliferation and increased apoptosis occured in AD-iPSC-derived neural progenitor cells (AD-NPCs) once neuronal differentiation was initiated, together with higher levels of Aβ42 and phosphorylated tau. Premature neuronal differentiation in AD-NPCs was caused by PSEN1 mutations and might be correlated to multiple dysregulated processes including but not limited to Wnt-Notch pathway. Our study documented previously unappreciated early NPC dysfunction in AD-NPCs, providing valuable new insights into the early mechanisms underlying AD pathogenesis.
Collapse
Affiliation(s)
- Juan Yang
- Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai 200031, China
| | - Hanzhi Zhao
- Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai 200031, China
| | - Yu Ma
- Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai 200031, China
- Shanghai Stem Cell Institute, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Guilai Shi
- Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai 200031, China
| | - Jian Song
- Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai 200031, China
| | - Yu Tang
- Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai 200031, China
| | - Song Li
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian 116021, China
| | - Ting Li
- Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai 200031, China
| | - Nan Liu
- Institute of Neuroscience, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Fan Tang
- Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai 200031, China
- Shanghai Stem Cell Institute, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Junjie Gu
- Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai 200031, China
- Shanghai Stem Cell Institute, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Lingling Zhang
- Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai 200031, China
| | - Zhuohua Zhang
- Institute of Precision Medicine, The Xiangya Hospital, State Key Laboratory of Medical Genetics, Xiangya Medical School, Central South University, Changsha 410078, China
| | - Xiaohui Zhang
- Institute of Neuroscience, Shanghai Institutes of Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ying Jin
- Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai 200031, China
- Shanghai Stem Cell Institute, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
| | - Weidong Le
- Key Laboratory of Stem Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai JiaoTong University School of Medicine, Shanghai 200031, China
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian 116021, China
- Institute of Neurology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200025, China
- Collaborative Innovation Center for Brain Science, the First Affiliated Hospital, Dalian Medical University, Dalian 116011, China
| |
Collapse
|
22
|
Agrawal R, Dale TP, Al-Zubaidi MA, Benny Malgulwar P, Forsyth NR, Kulshreshtha R. Pluripotent and Multipotent Stem Cells Display Distinct Hypoxic miRNA Expression Profiles. PLoS One 2016; 11:e0164976. [PMID: 27783707 PMCID: PMC5081191 DOI: 10.1371/journal.pone.0164976] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 10/04/2016] [Indexed: 12/20/2022] Open
Abstract
MicroRNAs are reported to have a crucial role in the regulation of self-renewal and differentiation of stem cells. Hypoxia has been identified as a key biophysical element of the stem cell culture milieu however, the link between hypoxia and miRNA expression in stem cells remains poorly understood. We therefore explored miRNA expression in hypoxic human embryonic and mesenchymal stem cells (hESCs and hMSCs). A total of 50 and 76 miRNAs were differentially regulated by hypoxia (2% O2) in hESCs and hMSCs, respectively, with a negligible overlap of only three miRNAs. We found coordinate regulation of precursor and mature miRNAs under hypoxia suggesting their regulation mainly at transcriptional level. Hypoxia response elements were located upstream of 97% of upregulated hypoxia regulated miRNAs (HRMs) suggesting hypoxia-inducible-factor (HIF) driven transcription. HIF binding to the candidate cis-elements of specific miRNAs under hypoxia was confirmed by Chromatin immunoprecipitation coupled with qPCR. Role analysis of a subset of upregulated HRMs identified linkage to reported inhibition of differentiation while a downregulated subset of HRMs had a putative role in the promotion of differentiation. MiRNA-target prediction correlation with published hypoxic hESC and hMSC gene expression profiles revealed HRM target genes enriched in the cytokine:cytokine receptor, HIF signalling and pathways in cancer. Overall, our study reveals, novel and distinct hypoxia-driven miRNA signatures in hESCs and hMSCs with the potential for application in optimised culture and differentiation models for both therapeutic application and improved understanding of stem cell biology.
Collapse
Affiliation(s)
- Rahul Agrawal
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi, India-110016
| | - Tina P. Dale
- Guy Hilton Research Centre, Institute of Science and Technology in Medicine, University of Keele, Thornburrow Drive, Hartshill, Stoke-on-Trent, Staffordshire, ST4 7QB, United Kingdom
| | - Mohammed A. Al-Zubaidi
- Guy Hilton Research Centre, Institute of Science and Technology in Medicine, University of Keele, Thornburrow Drive, Hartshill, Stoke-on-Trent, Staffordshire, ST4 7QB, United Kingdom
- College of Pharmacy, Al-Mustansiriyah University, Baghdad, Iraq
| | - Prit Benny Malgulwar
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India-110029
| | - Nicholas R. Forsyth
- Guy Hilton Research Centre, Institute of Science and Technology in Medicine, University of Keele, Thornburrow Drive, Hartshill, Stoke-on-Trent, Staffordshire, ST4 7QB, United Kingdom
| | - Ritu Kulshreshtha
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi, India-110016
| |
Collapse
|
23
|
Kobeissy FH, Hansen K, Neumann M, Fu S, Jin K, Liu J. Deciphering the Role of Emx1 in Neurogenesis: A Neuroproteomics Approach. Front Mol Neurosci 2016; 9:98. [PMID: 27799894 PMCID: PMC5065984 DOI: 10.3389/fnmol.2016.00098] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 09/26/2016] [Indexed: 12/18/2022] Open
Abstract
Emx1 has long been implicated in embryonic brain development. Previously we found that mice null of Emx1 gene had smaller dentate gyri and reduced neurogenesis, although the molecular mechanisms underlying this defect was not well understood. To decipher the role of Emx1 gene in neural regeneration and the timing of its involvement, we determine the frequency of neural stem cells (NSCs) in embryonic and adult forebrains of Emx1 wild type (WT) and knock out (KO) mice in the neurosphere assay. Emx1 gene deletion reduced the frequency and self-renewal capacity of NSCs of the embryonic brain but did not affect neuronal or glial differentiation. Emx1 KO NSCs also exhibited a reduced migratory capacity in response to serum or vascular endothelial growth factor (VEGF) in the Boyden chamber migration assay compared to their WT counterparts. A thorough comparison between NSC lysates from Emx1 WT and KO mice utilizing 2D-PAGE coupled with tandem mass spectrometry revealed 38 proteins differentially expressed between genotypes, including the F-actin depolymerization factor Cofilin. A global systems biology and cluster analysis identified several potential mechanisms and cellular pathways implicated in altered neurogenesis, all involving Cofilin1. Protein interaction network maps with functional enrichment analysis further indicated that the differentially expressed proteins participated in neural-specific functions including brain development, axonal guidance, synaptic transmission, neurogenesis, and hippocampal morphology, with VEGF as the upstream regulator intertwined with Cofilin1 and Emx1. Functional validation analysis indicated that apart from the overall reduced level of phosphorylated Cofilin1 (p-Cofilin1) in the Emx1 KO NSCs compared to WT NSCs as demonstrated in the western blot analysis, VEGF was able to induce more Cofilin1 phosphorylation and FLK expression only in the latter. Our results suggest that a defect in Cofilin1 phosphorylation induced by VEGF or other growth factors might contribute to the reduced neurogenesis in the Emx1 null mice during brain development.
Collapse
Affiliation(s)
- Firas H Kobeissy
- Department of Psychiatry, Center for Neuroproteomics and Biomarkers Research, University of Florida Gainesville, FL, USA
| | - Katharina Hansen
- Department of Neurological Surgery, University of California, San FranciscoSan Francisco, CA, USA; San Francisco VA Medical CenterSan Francisco, CA, USA
| | - Melanie Neumann
- Department of Neurological Surgery, University of California, San FranciscoSan Francisco, CA, USA; San Francisco VA Medical CenterSan Francisco, CA, USA
| | - Shuping Fu
- Department of Neurological Surgery, University of California, San FranciscoSan Francisco, CA, USA; San Francisco VA Medical CenterSan Francisco, CA, USA; Key Laboratory of Acupuncture and Medicine Research of Minister of Education, Nanjing University of Chinese MedicineNanjing, China
| | - Kulin Jin
- Pharmacology & Neuroscience, University of North Texas Health Science Center Fort Worth, TX, USA
| | - Jialing Liu
- Department of Neurological Surgery, University of California, San FranciscoSan Francisco, CA, USA; San Francisco VA Medical CenterSan Francisco, CA, USA
| |
Collapse
|
24
|
Francis JW, Turn RE, Newman LE, Schiavon C, Kahn RA. Higher order signaling: ARL2 as regulator of both mitochondrial fusion and microtubule dynamics allows integration of 2 essential cell functions. Small GTPases 2016; 7:188-196. [PMID: 27400436 PMCID: PMC5129891 DOI: 10.1080/21541248.2016.1211069] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 07/05/2016] [Accepted: 07/06/2016] [Indexed: 10/21/2022] Open
Abstract
ARL2 is among the most highly conserved proteins, predicted to be present in the last eukaryotic common ancestor, and ubiquitously expressed. Genetic screens in multiple model organisms identified ARL2, and its cytosolic binding partner cofactor D (TBCD), as important in tubulin folding and microtubule dynamics. Both ARL2 and TBCD also localize to centrosomes, making it difficult to dissect these effects. A growing body of evidence also has found roles for ARL2 inside mitochondria, as a regulator of mitochondrial fusion. Other studies have revealed roles for ARL2, in concert with its closest paralog ARL3, in the traffic of farnesylated cargos between membranes and specifically to cilia and photoreceptor cells. Details of each of these signaling processes continue to emerge. We summarize those data here and speculate about the potential for cross-talk or coordination of cell regulation, termed higher order signaling, based upon the use of a common GTPase in disparate cell functions.
Collapse
Affiliation(s)
- Joshua W. Francis
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Rachel E. Turn
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Laura E. Newman
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Cara Schiavon
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| | - Richard A. Kahn
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
25
|
Hang P, Sun C, Guo J, Zhao J, Du Z. BDNF-mediates Down-regulation of MicroRNA-195 Inhibits Ischemic Cardiac Apoptosis in Rats. Int J Biol Sci 2016; 12:979-89. [PMID: 27489501 PMCID: PMC4971736 DOI: 10.7150/ijbs.15071] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 06/07/2016] [Indexed: 11/23/2022] Open
Abstract
Background: Our previous studies suggested that brain-derived neurotrophic factor (BDNF)/tropomyosin-related kinase B (TrkB) axis inhibited cardiomyocyte apoptosis in myocardial infarction (MI). However, the relationship between BDNF and microRNA (miRNA) in cardiomyocytes are unclear. The present study was performed to investigate the role of miR-195 and the interplay between BDNF and miR-195 in ischemic cardiomyocyte apoptosis. Methods: Male Wistar rats were subjected to coronary artery ligation, and primary neonatal rat ventricular myocytes were treated with hypoxia or hydrogen peroxide (H2O2). BDNF level in rat ventricles was measured by enzyme linked immunosorbent assay (ELISA). miR-195 mimic, inhibitor or negative control was transfected into the cardiomyocytes. Cell viability and apoptosis were detected by MTT assay and TdT-mediated dUTP nick end labeling (TUNEL) staining, respectively. Cardiac function and apoptosis were detected in MI rats intravenously injected with antagomiR-195. Luciferase assay, Western blot and Real-time RT-PCR were employed to clarify the interplay between miR-195 and BDNF. Results: miR-195 level was dynamically regulated in response to MI and significantly increased in ischemic regions 24 h post-MI as well as in hypoxic or H2O2-treated cardiomyocytes. Meanwhile, BDNF protein level was rapidly increased in MI rats and H2O2-treated cardiomyocytes. Apoptosis in both hypoxic and H2O2-treated cardiomyocytes were markedly reduced and cell viability was increased by miR-195 inhibitor. Moreover, inhibition of miR-195 significantly improved cardiac function of MI rats. Bcl-2 but not BDNF was validated as the direct target of miR-195. Furthermore, BDNF abolished the pro-apoptotic role of miR-195, which was reversed by its scavenger TrkB-Fc. Conclusion: Up-regulation of miR-195 in ischemic cardiomyocytes promotes ischemic apoptosis by targeting Bcl-2. BDNF mitigated the pro-apoptotic effect of miR-195 in rat cardiomyocytes. These findings may provide better understanding of the pro-apoptotic role of miR-195 in MI and suggest that BDNF/miR-195/Bcl-2 axis may be beneficial for limiting myocardial ischemic injury.
Collapse
Affiliation(s)
- Pengzhou Hang
- 1. Institute of Clinical Pharmacology of the Second Affiliated Hospital (Key Laboratory of Drug Research, Heilongjiang Higher Education Institutions), Harbin Medical University, Harbin 150086, Heilongjiang Province, P. R. China
| | - Chuan Sun
- 1. Institute of Clinical Pharmacology of the Second Affiliated Hospital (Key Laboratory of Drug Research, Heilongjiang Higher Education Institutions), Harbin Medical University, Harbin 150086, Heilongjiang Province, P. R. China
| | - Jing Guo
- 1. Institute of Clinical Pharmacology of the Second Affiliated Hospital (Key Laboratory of Drug Research, Heilongjiang Higher Education Institutions), Harbin Medical University, Harbin 150086, Heilongjiang Province, P. R. China
| | - Jing Zhao
- 2. Department of Cardiology, the First Affiliated Hospital (Key Laboratory of Cardiac Diseases and Heart Failure), Harbin Medical University, Harbin 150001, Heilongjiang Province, P. R. China
| | - Zhimin Du
- 1. Institute of Clinical Pharmacology of the Second Affiliated Hospital (Key Laboratory of Drug Research, Heilongjiang Higher Education Institutions), Harbin Medical University, Harbin 150086, Heilongjiang Province, P. R. China
| |
Collapse
|
26
|
Identification of key regulators of pancreatic cancer progression through multidimensional systems-level analysis. Genome Med 2016; 8:38. [PMID: 27137215 PMCID: PMC4853852 DOI: 10.1186/s13073-016-0282-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 02/19/2016] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Pancreatic cancer is an aggressive cancer with dismal prognosis, urgently necessitating better biomarkers to improve therapeutic options and early diagnosis. Traditional approaches of biomarker detection that consider only one aspect of the biological continuum like gene expression alone are limited in their scope and lack robustness in identifying the key regulators of the disease. We have adopted a multidimensional approach involving the cross-talk between the omics spaces to identify key regulators of disease progression. METHODS Multidimensional domain-specific disease signatures were obtained using rank-based meta-analysis of individual omics profiles (mRNA, miRNA, DNA methylation) related to pancreatic ductal adenocarcinoma (PDAC). These domain-specific PDAC signatures were integrated to identify genes that were affected across multiple dimensions of omics space in PDAC (genes under multiple regulatory controls, GMCs). To further pin down the regulators of PDAC pathophysiology, a systems-level network was generated from knowledge-based interaction information applied to the above identified GMCs. Key regulators were identified from the GMC network based on network statistics and their functional importance was validated using gene set enrichment analysis and survival analysis. RESULTS Rank-based meta-analysis identified 5391 genes, 109 miRNAs and 2081 methylation-sites significantly differentially expressed in PDAC (false discovery rate ≤ 0.05). Bimodal integration of meta-analysis signatures revealed 1150 and 715 genes regulated by miRNAs and methylation, respectively. Further analysis identified 189 altered genes that are commonly regulated by miRNA and methylation, hence considered GMCs. Systems-level analysis of the scale-free GMCs network identified eight potential key regulator hubs, namely E2F3, HMGA2, RASA1, IRS1, NUAK1, ACTN1, SKI and DLL1, associated with important pathways driving cancer progression. Survival analysis on individual key regulators revealed that higher expression of IRS1 and DLL1 and lower expression of HMGA2, ACTN1 and SKI were associated with better survival probabilities. CONCLUSIONS It is evident from the results that our hierarchical systems-level multidimensional analysis approach has been successful in isolating the converging regulatory modules and associated key regulatory molecules that are potential biomarkers for pancreatic cancer progression.
Collapse
|
27
|
Chen K, Koe CT, Xing ZB, Tian X, Rossi F, Wang C, Tang Q, Zong W, Hong WJ, Taneja R, Yu F, Gonzalez C, Wu C, Endow S, Wang H. Arl2- and Msps-dependent microtubule growth governs asymmetric division. J Cell Biol 2016; 212:661-76. [PMID: 26953351 PMCID: PMC4792071 DOI: 10.1083/jcb.201503047] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 02/10/2016] [Indexed: 12/18/2022] Open
Abstract
Drosophila Arl2 governs neuroblast asymmetric cell division through regulation of microtubule growth and localization of Msps to centrosomes. Asymmetric division of neural stem cells is a fundamental strategy to balance their self-renewal and differentiation. It is long thought that microtubules are not essential for cell polarity in asymmetrically dividing Drosophila melanogaster neuroblasts (NBs; neural stem cells). Here, we show that Drosophila ADP ribosylation factor like-2 (Arl2) and Msps, a known microtubule-binding protein, control cell polarity and spindle orientation of NBs. Upon arl2 RNA intereference, Arl2-GDP expression, or arl2 deletions, microtubule abnormalities and asymmetric division defects were observed. Conversely, overactivation of Arl2 leads to microtubule overgrowth and depletion of NBs. Arl2 regulates microtubule growth and asymmetric division through localizing Msps to the centrosomes in NBs. Moreover, Arl2 regulates dynein function and in turn centrosomal localization of D-TACC and Msps. Arl2 physically associates with tubulin cofactors C, D, and E. Arl2 functions together with tubulin-binding cofactor D to control microtubule growth, Msps localization, and NB self-renewal. Therefore, Arl2- and Msps-dependent microtubule growth is a new paradigm regulating asymmetric division of neural stem cells.
Collapse
Affiliation(s)
- Keng Chen
- Neuroscience and Behavioral Disorders Program, Duke-National University of Singapore Graduate Medical School, Singapore 169857 National University of Singapore Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117456
| | - Chwee Tat Koe
- Neuroscience and Behavioral Disorders Program, Duke-National University of Singapore Graduate Medical School, Singapore 169857 National University of Singapore Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117456
| | - Zhanyuan Benny Xing
- Department of Cell Biology, Duke University, Duke University Medical Center, Durham, NC 27710
| | - Xiaolin Tian
- Neuroscience Center of Excellence, Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| | - Fabrizio Rossi
- Institute for Research in Biomedicine Barcelona, 08028 Barcelona, Spain
| | - Cheng Wang
- Neuroscience and Behavioral Disorders Program, Duke-National University of Singapore Graduate Medical School, Singapore 169857
| | - Quan Tang
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore 117604 Department of Biological Sciences, National University of Singapore, Singapore 117604
| | - Wenhui Zong
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore 117604 Department of Biological Sciences, National University of Singapore, Singapore 117604
| | - Wan Jin Hong
- Institute of Molecular and Cell Biology, Singapore 138673
| | - Reshma Taneja
- National University of Singapore Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117456 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| | - Fengwei Yu
- Neuroscience and Behavioral Disorders Program, Duke-National University of Singapore Graduate Medical School, Singapore 169857 National University of Singapore Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117456 Temasek Life Sciences Laboratory, National University of Singapore, Singapore 117604 Department of Biological Sciences, National University of Singapore, Singapore 117604
| | - Cayetano Gonzalez
- Institute for Research in Biomedicine Barcelona, 08028 Barcelona, Spain Institució Catalana de Recerca i Estudis Avançats, 08010 Barcelona, Spain
| | - Chunlai Wu
- Neuroscience Center of Excellence, Department of Cell Biology and Anatomy, Louisiana State University Health Sciences Center, New Orleans, LA 70112
| | - Sharyn Endow
- Neuroscience and Behavioral Disorders Program, Duke-National University of Singapore Graduate Medical School, Singapore 169857 Department of Cell Biology, Duke University, Duke University Medical Center, Durham, NC 27710
| | - Hongyan Wang
- Neuroscience and Behavioral Disorders Program, Duke-National University of Singapore Graduate Medical School, Singapore 169857 National University of Singapore Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore 117456 Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597
| |
Collapse
|
28
|
Comprehensive profiling reveals mechanisms of SOX2-mediated cell fate specification in human ESCs and NPCs. Cell Res 2016; 26:171-89. [PMID: 26809499 DOI: 10.1038/cr.2016.15] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 11/20/2015] [Accepted: 11/24/2015] [Indexed: 12/15/2022] Open
Abstract
SOX2 is a key regulator of multiple types of stem cells, especially embryonic stem cells (ESCs) and neural progenitor cells (NPCs). Understanding the mechanism underlying the function of SOX2 is of great importance for realizing the full potential of ESCs and NPCs. Here, through genome-wide comparative studies, we show that SOX2 executes its distinct functions in human ESCs (hESCs) and hESC-derived NPCs (hNPCs) through cell type- and stage-dependent transcription programs. Importantly, SOX2 suppresses non-neural lineages in hESCs and regulates neurogenesis from hNPCs by inhibiting canonical Wnt signaling. In hESCs, SOX2 achieves such inhibition by direct transcriptional regulation of important Wnt signaling modulators, WLS and SFRP2. Moreover, SOX2 ensures pluripotent epigenetic landscapes via interacting with histone variant H2A.Z and recruiting polycomb repressor complex 2 to poise developmental genes in hESCs. Together, our results advance our understanding of the mechanism by which cell type-specific transcription factors control lineage-specific gene expression programs and specify cell fate.
Collapse
|
29
|
Zheng D, Yu Y, Li M, Wang G, Chen R, Fan GC, Martin C, Xiong S, Peng T. Inhibition of MicroRNA 195 Prevents Apoptosis and Multiple-Organ Injury in Mouse Models of Sepsis. J Infect Dis 2015; 213:1661-70. [PMID: 26704614 DOI: 10.1093/infdis/jiv760] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 12/16/2015] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND MicroRNAs (miRs) are a class of short RNA molecules, which negatively regulate gene expression. The levels of circulating miR-15 family members are elevated in septic patients and may be associated with septic death. This study investigated whether inhibition of miR-195, a member of the miR-15 family, provided beneficial effects in sepsis. METHODS AND RESULTS Sepsis was induced by injection of feces into the peritoneum in mice. miR-195 was upregulated in the lung and liver of septic mice. Silencing of miR-195 increased the protein levels of BCL-2, Sirt1, and Pim-1; prevented apoptosis; reduced liver and lung injury; and improved the survival in septic mice. Silencing of miR-195 provided similar protection in lipopolysaccharide-induced endotoxemic mice. In endothelial cells, upregulation of miR-195 induced apoptosis, and inhibition of miR-195 prevented lipopolysaccharide-induced apoptosis. miR-195 repressed expression of its protein targets, BCL-2, Sirt1, and Pim-1. Furthermore, overexpression of Pim-1 prevented apoptosis induced by lipopolysaccharide and miR-195 mimic. Inhibition of Pim-1 attenuated the protective effects of miR-195 silencing in septic mice. CONCLUSIONS Silencing of miR-195 reduced multiple-organ injury and improved the survival in sepsis, and the protective effects of miR-195 inhibition were associated with upregulation of Bcl-2, Sirt1, and Pim-1. Thus, inhibition of miR-195 may represent a new therapeutic approach for sepsis.
Collapse
Affiliation(s)
- Dong Zheng
- Jiangsu Key Laboratory of Infection and Immunity, Institutes for Biology and Medical Sciences, Soochow University, Suzhou Lawson Health Research Institute Department of Medicine Department of Pathology, University of Western Ontario, London, Canada
| | - Yong Yu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China
| | - Minghui Li
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China
| | - Grace Wang
- Department of Pathology, University of Western Ontario, London, Canada
| | - Ruizhen Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China
| | - Guo-Chang Fan
- Department of Pharmacology and Cell Biophysics, University of Cincinnati College of Medicine, Ohio
| | | | - Sidong Xiong
- Jiangsu Key Laboratory of Infection and Immunity, Institutes for Biology and Medical Sciences, Soochow University, Suzhou
| | - Tianqing Peng
- Jiangsu Key Laboratory of Infection and Immunity, Institutes for Biology and Medical Sciences, Soochow University, Suzhou Lawson Health Research Institute Department of Medicine Department of Pathology, University of Western Ontario, London, Canada
| |
Collapse
|
30
|
Identification of a panel of five serum miRNAs as a biomarker for Parkinson's disease. Parkinsonism Relat Disord 2015; 22:68-73. [PMID: 26631952 DOI: 10.1016/j.parkreldis.2015.11.014] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 10/30/2015] [Accepted: 11/16/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND OBJECTIVE Parkinson's disease (PD) is the second most common age-related neurodegenerative disorder after Alzheimer's disease. The aim of this work was to determine whether the differences of serum miRNAs profiling could distinguish PD patients from healthy individuals. METHODS We collected serum samples from 106 sporadic PD patients and 91 age/gender-matched healthy controls. Serum miRNAs were analysed by Solexa sequencing followed by a qRT-PCR examination. The qRT-PCR assay, which was divided into two phases, was used to validate the expression of miRNAs screened by Solexa sequencing. Receiver operating characteristic (ROC) curve analysis and clustering analysis were performed to determine the diagnostic usefulness of the selected miRNAs for PD. RESULTS In this study, we generated a profile of 5 serum miRNAs: miR-195 was up-regulated, and miR-185, miR-15b, miR-221 and miR-181a were down-regulated. CONCLUSION This group of five miRNAs can precisely distinguish PD patients from health individuals and may be used as a potential serum-based biomarker for the diagnosis of PD.
Collapse
|
31
|
Shi X, Yan C, Liu B, Yang C, Nie X, Wang X, Zheng J, Wang Y, Zhu Y. miR-381 Regulates Neural Stem Cell Proliferation and Differentiation via Regulating Hes1 Expression. PLoS One 2015; 10:e0138973. [PMID: 26431046 PMCID: PMC4591969 DOI: 10.1371/journal.pone.0138973] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 09/08/2015] [Indexed: 11/18/2022] Open
Abstract
Neural stem cells are self-renewing, multipotent and undifferentiated precursors that retain the capacity for differentiation into both glial (astrocytes and oligodendrocytes) and neuronal lineages. Neural stem cells offer cell-based therapies for neurological disorders such as Alzheimer's disease, Parkinson's disease, Huntington's disease and spinal cord injuries. However, their cellular behavior is poorly understood. MicroRNAs (miRNAs) are a class of small noncoding RNAs involved in cell development, proliferation and differentiation through regulating gene expression at post-transcriptional level. The role of miR-381 in the development of neural stem cells remains unknown. In this study, we showed that overexpression of miR-381 promoted neural stem cells proliferation. It induced the neural stem cells differentiation to neurons and inhibited their differentiation to astrocytes. Furthermore, we identified HES1 as a direct target of miR-381 in neural stem cells. Moreover, re-expression of HES1 impaired miR-381-induced promotion of neural stem cells proliferation and induce neural stem cells differentiation to neurons. In conclusion, miR-381 played important role in neural stem cells proliferation and differentiation.
Collapse
Affiliation(s)
- Xiaodong Shi
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, PR China
| | - Chunhua Yan
- Department of Respiratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, PR China
| | - Baoquan Liu
- Department of anatomy, Harbin Medical University, Harbin, 150081, PR China
| | - Chunxiao Yang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, PR China
| | - Xuedan Nie
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, PR China
| | - Xiaokun Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, PR China
| | - Jiaolin Zheng
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, PR China
| | - Yue Wang
- Department of Occupational Health, College of Public Health, Harbin Medical University, Harbin, 150081, PR China
| | - Yulan Zhu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150081, PR China
| |
Collapse
|
32
|
Guay C, Menoud V, Rome S, Regazzi R. Horizontal transfer of exosomal microRNAs transduce apoptotic signals between pancreatic beta-cells. Cell Commun Signal 2015; 13:17. [PMID: 25880779 PMCID: PMC4371845 DOI: 10.1186/s12964-015-0097-7] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 03/06/2015] [Indexed: 12/22/2022] Open
Abstract
Background Diabetes mellitus is a common metabolic disorder characterized by dysfunction of insulin-secreting pancreatic beta-cells. MicroRNAs are important regulators of beta-cell activities. These non-coding RNAs have recently been discovered to exert their effects not only inside the cell producing them but, upon exosome-mediated transfer, also in other recipient cells. This novel communication mode remains unexplored in pancreatic beta-cells. In the present study, the microRNA content of exosomes released by beta-cells in physiological and physiopathological conditions was analyzed and the biological impact of their transfer to recipient cells investigated. Results Exosomes were isolated from the culture media of MIN6B1 and INS-1 derived 832/13 beta-cell lines and from mice, rat or human islets. Global profiling revealed that the microRNAs released in MIN6B1 exosomes do not simply reflect the content of the cells of origin. Indeed, while a subset of microRNAs was preferentially released in exosomes others were selectively retained in the cells. Moreover, exposure of MIN6B1 cells to inflammatory cytokines changed the release of several microRNAs. The dynamics of microRNA secretion and their potential transfer to recipient cells were next investigated. As a proof-of-concept, we demonstrate that if cel-miR-238, a C. Elegans microRNA not present in mammalian cells, is expressed in MIN6B1 cells a fraction of it is released in exosomes and is transferred to recipient beta-cells. Furthermore, incubation of untreated MIN6B1 or mice islet cells in the presence of microRNA-containing exosomes isolated from the culture media of cytokine-treated MIN6B1 cells triggers apoptosis of recipient cells. In contrast, exosomes originating from cells not exposed to cytokines have no impact on cell survival. Apoptosis induced by exosomes produced by cytokine-treated cells was prevented by down-regulation of the microRNA-mediating silencing protein Ago2 in recipient cells, suggesting that the effect is mediated by the non-coding RNAs. Conclusions Taken together, our results suggest that beta-cells secrete microRNAs that can be transferred to neighboring beta-cells. Exposure of donor cells to pathophysiological conditions commonly associated with diabetes modifies the release of microRNAs and affects survival of recipient beta-cells. Our results support the concept that exosomal microRNAs transfer constitutes a novel cell-to-cell communication mechanism regulating the activity of pancreatic beta-cells. Electronic supplementary material The online version of this article (doi:10.1186/s12964-015-0097-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Claudiane Guay
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, Lausanne, Switzerland.
| | - Véronique Menoud
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, Lausanne, Switzerland.
| | - Sophie Rome
- CarMeN Laboratory (INSERM U.1060/INRA 1397, INSA), University of Lyon, Faculty of Medicine Lyon-Sud, Ouillons, France.
| | - Romano Regazzi
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, Lausanne, Switzerland.
| |
Collapse
|
33
|
Katoh M. Cardio-miRNAs and onco-miRNAs: circulating miRNA-based diagnostics for non-cancerous and cancerous diseases. Front Cell Dev Biol 2014; 2:61. [PMID: 25364765 PMCID: PMC4207049 DOI: 10.3389/fcell.2014.00061] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 09/29/2014] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases and cancers are the leading causes of morbidity and mortality in the world. MicroRNAs (miRNAs) are short non-coding RNAs that primarily repress target mRNAs. Here, miR-24, miR-125b, miR-195, and miR-214 were selected as representative cardio-miRs that are upregulated in human heart failure. To bridge the gap between miRNA studies in cardiology and oncology, the targets and functions of these miRNAs in cardiovascular diseases and cancers will be reviewed. ACVR1B, BCL2, BIM, eNOS, FGFR3, JPH2, MEN1, MYC, p16, and ST7L are miR-24 targets that have been experimentally validated in human cells. ARID3B, BAK1, BCL2, BMPR1B, ERBB2, FGFR2, IL6R, MUC1, SITR7, Smoothened, STAT3, TET2, and TP53 are representative miR-125b targets. ACVR2A, BCL2, CCND1, E2F3, GLUT3, MYB, RAF1, VEGF, WEE1, and WNT7A are representative miR-195 targets. BCL2L2, ß-catenin, BIM, CADM1, EZH2, FGFR1, NRAS, PTEN, TP53, and TWIST1 are representative miR-214 targets. miR-125b is a good cardio-miR that protects cardiomyocytes; miR-195 is a bad cardio-miR that elicits cardiomyopathy and heart failure; miR-24 and miR-214 are bi-functional cardio-miRs. By contrast, miR-24, miR-125b, miR-195, and miR-214 function as oncogenic or tumor suppressor miRNAs in a cancer (sub)type-dependent manner. Circulating miR-24 is elevated in diabetes, breast cancer and lung cancer. Circulating miR-195 is elevated in acute myocardial infarction, breast cancer, prostate cancer and colorectal adenoma. Circulating miR-125b and miR-214 are elevated in some cancers. Cardio-miRs and onco-miRs bear some similarities in functions and circulation profiles. miRNAs regulate WNT, FGF, Hedgehog and other signaling cascades that are involved in orchestration of embryogenesis and homeostasis as well as pathogenesis of human diseases. Because circulating miRNA profiles are modulated by genetic and environmental factors and are dysregulated by genetic and epigenetic alterations in somatic cells, circulating miRNA association studies (CMASs) within several thousands of cases each for common non-cancerous diseases and major cancers are necessary for miRNA-based diagnostics.
Collapse
Affiliation(s)
- Masaru Katoh
- Department of Omics Network, National Cancer Center Tokyo, Japan
| |
Collapse
|
34
|
Sun N, Ye L, Chang T, Li X, Li X. microRNA-195-Cdc42 axis acts as a prognostic factor of esophageal squamous cell carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:6871-6879. [PMID: 25400770 PMCID: PMC4230103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 09/01/2014] [Indexed: 06/04/2023]
Abstract
BACKGROUND AND PURPOSE Previous studies observed the downregulation of microRNA (miR)-195 in esophageal squamous cell carcinoma (ESCC) tissues, confirmed cell division cycle 42 (Cdc42) as one target gene of miR-195, and demonstrated that miR-195 may act as a tumor suppressor in ESCC by regulating Cdc42 expression. This study aimed to explore the association of miR-195 and Cdc42 combined expression with clinicopathologic factors and prognosis. METHODS Expression of miR-195 and Cdc42 mRNA in 98 pairs of ESCC and paracancerous tissues were detected using real-time quantitative RT-PCR. RESULTS miR-195 downregulation and Cdc42 upregulation were both prevalent in ESCC tissues, and negatively correlated with each other. In addition, miR-195 expression negatively correlated with TNM stage (P=0.008) and lymphatic metastasis (P=0.022), while Cdc42 expression positively correlated with TNM stage (P=0.011) and tumor differentiation (P=0.024). Moreover, combined expression of miR-195 and Cdc42 (miR-195/Cdc42) was found to be prognostic indicators for progression-free survival and overall survival of ESCC patients both in univariate and multivariate analyses. CONCLUSION The main findings of this study indicate the involvement of miR-195-Cdc42 axis in the progression of ESCC and suggest that the combined aberrant expression of miR-195 and Cdc42 mRNA can serve as a promising unfavorable prognostic biomarker in ESCC.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Biomarkers, Tumor/genetics
- Carcinoma, Squamous Cell/enzymology
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/mortality
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/therapy
- Cell Differentiation
- Chi-Square Distribution
- Disease Progression
- Disease-Free Survival
- Esophageal Neoplasms/enzymology
- Esophageal Neoplasms/genetics
- Esophageal Neoplasms/mortality
- Esophageal Neoplasms/pathology
- Esophageal Neoplasms/therapy
- Esophageal Squamous Cell Carcinoma
- Female
- Gene Expression Regulation, Enzymologic
- Gene Expression Regulation, Neoplastic
- Humans
- Kaplan-Meier Estimate
- Male
- MicroRNAs/genetics
- Middle Aged
- Multivariate Analysis
- Neoplasm Staging
- Proportional Hazards Models
- RNA, Messenger/analysis
- Real-Time Polymerase Chain Reaction
- Risk Factors
- Time Factors
- cdc42 GTP-Binding Protein/genetics
Collapse
Affiliation(s)
- Niuniu Sun
- School of Nursing, Xinxiang Medical UniversityXinxiang 453003, China
| | - Lin Ye
- School of Nursing, Xinxiang Medical UniversityXinxiang 453003, China
| | - Tingmin Chang
- The First Affiliated Hospital of Xinxiang Medical UniversityXinxiang 453003, China
| | - Xiaojing Li
- School of Nursing, Xinxiang Medical UniversityXinxiang 453003, China
- Henan Nursing Vocational CollegeAnyang 455000, China
| | - Xiumin Li
- Center for Cancer Research, School of Nursing, Xinxiang Medical UniversityXinxiang 453003, China
- The Third Affiliated Hospital of Xinxiang Medical UniversityXinxiang 453003, China
| |
Collapse
|
35
|
Quantitative proteomics analysis highlights the role of redox hemostasis and energy metabolism in human embryonic stem cell differentiation to neural cells. J Proteomics 2014; 101:1-16. [PMID: 24530625 DOI: 10.1016/j.jprot.2014.02.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2013] [Revised: 01/16/2014] [Accepted: 02/01/2014] [Indexed: 01/15/2023]
Abstract
UNLABELLED Neural differentiation of human embryonic stem cells (hESCs) is a unique opportunity for in vitro analyses of neurogenesis in humans. Extrinsic cues through neural plate formation are well described in the hESCs although intracellular mechanisms underlying neural development are largely unknown. Proteome analysis of hESC differentiation to neural cells will help to further define molecular mechanisms involved in neurogenesis in humans. Using a two-dimensional differential gel electrophoresis (2D-DIGE) system, we analyzed the proteome of hESC differentiation to neurons at three stages, early neural differentiation, neural ectoderm and mature neurons. Out of 137 differentially accumulated protein spots, 118 spots were identified using MALDI-TOF/TOF and LC MS/MS. We observed that proteins involved in redox hemostasis, vitamin and energy metabolism and ubiquitin dependent proteolysis were more abundant in differentiated cells, whereas the abundance of proteins associated with RNA processing and protein folding was higher in hESCs. Higher abundance of proteins involved in maintaining cellular redox state suggests the importance of redox hemostasis in neural differentiation. Furthermore, our results support the concept of a coupling mechanism between neuronal activity and glucose utilization. The protein network analysis showed that the majority of the interacting proteins were associated with the cell cycle and cellular proliferation. These results enhanced our understanding of the molecular dynamics that underlie neural commitment and differentiation. BIOLOGICAL SIGNIFICANCE In highlighting the role of redox and unique metabolic properties of neuronal cells, the present findings add insight to our understanding of hESC differentiation to neurons. The abundance of fourteen proteins involved in maintaining cellular redox state, including 10 members of peroxiredoxin (Prdx) family, mainly increased during differentiation, thus highlighting a link of neural differentiation to redox. Our results revealed markedly higher expression of genes encoding enzymes involved in the glycolysis and amino acid synthesis during differentiation. Protein network analysis predicted a number of critical mediators in hESC differentiation. These proteins included TP53, CTNNB1, SMARCA4, TNF, TERT, E2F1, MYC, RB1, and AR.
Collapse
|
36
|
Sharma S, Eghbali M. Influence of sex differences on microRNA gene regulation in disease. Biol Sex Differ 2014; 5:3. [PMID: 24484532 PMCID: PMC3912347 DOI: 10.1186/2042-6410-5-3] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 12/30/2013] [Indexed: 12/21/2022] Open
Abstract
Sexual dimorphism is observed in most human diseases. The difference in the physiology and genetics between sexes can contribute tremendously to the disease prevalence, severity, and outcome. Both hormonal and genetic differences between males and females can lead to differences in gene expression patterns that can influence disease risk and course. MicroRNAs have emerged as potential regulatory molecules in all organisms. They can have a broad effect on every aspect of physiology, including embryogenesis, metabolism, and growth and development. Numerous microRNAs have been identified and elucidated to play a key role in cardiovascular diseases, as well as in neurological and autoimmune disorders. This is especially important as microRNA-based tools can be exploited as beneficial therapies for disease treatment and prevention. Sex steroid hormones as well as X-linked genes can have a considerable influence on the regulation of microRNAs. However, there are very few studies highlighting the role of microRNAs in sex biased diseases. This review attempts to summarize differentially regulated microRNAs in males versus females in different diseases and calls for more attention in this underexplored area that should set the basis for more effective therapeutic strategies for sexually dimorphic diseases.
Collapse
Affiliation(s)
| | - Mansoureh Eghbali
- Department of Anesthesiology, Division of Molecular Medicine, and Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California Los Angeles, BH-160CHS, Los Angeles, CA 90095-7115, USA.
| |
Collapse
|
37
|
Saab BJ, Mansuy IM. Neuroepigenetics of memory formation and impairment: the role of microRNAs. Neuropharmacology 2014; 80:61-9. [PMID: 24486712 DOI: 10.1016/j.neuropharm.2014.01.026] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2013] [Revised: 01/19/2014] [Accepted: 01/20/2014] [Indexed: 12/13/2022]
Abstract
MicroRNAs (miRNAs) are a class of short non-coding RNAs that primarily regulate protein synthesis through reversible translational repression or mRNA degradation. MiRNAs can act by translational control of transcription factors or via direct action on the chromatin, and thereby contribute to the non-genetic control of gene-environment interactions. MiRNAs that regulate components of pathways required for learning and memory further modulate the influence of epigenetics on cognition in the normal and diseased brain. This review summarizes recent data exemplifying the known roles of miRNAs in memory formation in different model organisms, and describes how neuronal plasticity regulates miRNA biogenesis, activity and degradation. It also examines the relevance of miRNAs for memory impairment in human, using recent clinical observations related to neurodevelopmental and neurodegenerative diseases, and discusses the potential mechanisms by which these miRNAs may contribute to memory disorders.
Collapse
Affiliation(s)
- Bechara J Saab
- Brain Research Institute, Neuroscience Center Zürich, Faculty of Medicine of the University of Zürich and Department of Health Sciences and Technology of the Swiss Federal Institute of Technology, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland
| | - Isabelle M Mansuy
- Brain Research Institute, Neuroscience Center Zürich, Faculty of Medicine of the University of Zürich and Department of Health Sciences and Technology of the Swiss Federal Institute of Technology, Winterthurerstrasse 190, CH-8057 Zürich, Switzerland.
| |
Collapse
|