101
|
Na HK, Wi JS, Son HY, Ok JG, Huh YM, Lee TG. Discrimination of single nucleotide mismatches using a scalable, flexible, and transparent three-dimensional nanostructure-based plasmonic miRNA sensor with high sensitivity. Biosens Bioelectron 2018; 113:39-45. [DOI: 10.1016/j.bios.2018.04.033] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 04/06/2018] [Accepted: 04/16/2018] [Indexed: 01/20/2023]
|
102
|
Hu J, Lin C, Liu M, Tong Q, Xu S, Wang D, Zhao Y. Analysis of the microRNA transcriptome of Daphnia pulex during aging. Gene 2018; 664:101-110. [PMID: 29684489 DOI: 10.1016/j.gene.2018.04.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 03/13/2018] [Accepted: 04/12/2018] [Indexed: 01/30/2023]
Abstract
Daphnia pulex is an important food organism that exhibits a particular mode of reproduction known as cyclical parthenogenesis (asexual) and sexual reproduction. Regulation of the aging process by microRNAs (miRNAs) is a research hotspot in miRNA studies. To investigate a possible role of miRNAs in regulating aging and senescence, we used Illumina HiSeq to sequence two miRNA libraries from 1-day-old (1d) and 25-day-old (25d) D. pulex specimens. In total, we obtained 11,218,097 clean reads and 28,569 unique miRNAs from 1d specimens and 11,819,106 clean reads and 44,709 unique miRNAs from 25d specimens. Bioinformatic analyses was used to identify 1335 differentially expressed miRNAs from known miRNAs, including 127 miRNAs that exhibited statistically significant differences (P < 0.01); 92 miRNAs were upregulated and 35 were downregulated. Quantitative real-time (qRT)-PCR experiments were performed for nine miRNAs from five samples (1d, 5d, 10d, 15d, 20d and 25d) during the aging process, and the sequencing and qRT-PCR data were found to be consistent. Ninety-four miRNAs were predicted to correspond to 2014 target genes in known miRNAs with 4032 target gene sites. Sixteen pathways changed significantly (P < 0.05) at different developmental stages, revealing many important principles of the miRNA regulatory aging network of D. pulex. Overall, the difference in miRNA expression profile during aging of D. pulex forms a basis for further studies aimed at understanding the role of miRNAs in regulating aging, reproductive transformation, senescence, and longevity.
Collapse
Affiliation(s)
- Jiabao Hu
- School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Chongyuan Lin
- School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Mengdi Liu
- School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Qiaoqiong Tong
- School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Shanliang Xu
- School of Marine Sciences, Ningbo University, Ningbo 315211, China
| | - Danli Wang
- School of Marine Sciences, Ningbo University, Ningbo 315211, China.
| | - Yunlong Zhao
- School of Life Science, East China Normal University, Shanghai 200062, China.
| |
Collapse
|
103
|
Zhou L, Lim MYT, Kaur P, Saj A, Bortolamiol-Becet D, Gopal V, Tolwinski N, Tucker-Kellogg G, Okamura K. Importance of miRNA stability and alternative primary miRNA isoforms in gene regulation during Drosophila development. eLife 2018; 7:e38389. [PMID: 30024380 PMCID: PMC6066331 DOI: 10.7554/elife.38389] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 07/04/2018] [Indexed: 12/19/2022] Open
Abstract
Mature microRNAs (miRNAs) are processed from primary transcripts (pri-miRNAs), and their expression is controlled at transcriptional and post-transcriptional levels. However, how regulation at multiple levels achieves precise control remains elusive. Using published and new datasets, we profile a time course of mature and pri-miRNAs in Drosophila embryos and reveal the dynamics of miRNA production and degradation as well as dynamic changes in pri-miRNA isoform selection. We found that 5' nucleotides influence stability of mature miRNAs. Furthermore, distinct half-lives of miRNAs from the mir-309 cluster shape their temporal expression patterns, and the importance of rapid degradation of the miRNAs in gene regulation is detected as distinct evolutionary signatures at the target sites in the transcriptome. Finally, we show that rapid degradation of miR-3/-309 may be important for regulation of the planar cell polarity pathway component Vang. Altogether, the results suggest that complex mechanisms regulate miRNA expression to support normal development.
Collapse
Affiliation(s)
- Li Zhou
- Temasek Life Sciences LaboratorySingaporeSingapore
- Department of Biological Sciences, Faculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Mandy Yu Theng Lim
- Temasek Life Sciences LaboratorySingaporeSingapore
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
| | - Prameet Kaur
- Division of ScienceYale-NUS CollegeSingaporeSingapore
| | - Abil Saj
- Cancer Therapeutics and Stratified OncologyGenome Institute of SingaporeSingaporeSingapore
| | | | - Vikneswaran Gopal
- Department of Statistics and Applied Probability, Faculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Nicholas Tolwinski
- Department of Biological Sciences, Faculty of ScienceNational University of SingaporeSingaporeSingapore
- Division of ScienceYale-NUS CollegeSingaporeSingapore
| | - Greg Tucker-Kellogg
- Department of Biological Sciences, Faculty of ScienceNational University of SingaporeSingaporeSingapore
| | - Katsutomo Okamura
- Temasek Life Sciences LaboratorySingaporeSingapore
- School of Biological SciencesNanyang Technological UniversitySingaporeSingapore
| |
Collapse
|
104
|
Maeda RK, Sitnik JL, Frei Y, Prince E, Gligorov D, Wolfner MF, Karch F. The lncRNA male-specific abdominal plays a critical role in Drosophila accessory gland development and male fertility. PLoS Genet 2018; 14:e1007519. [PMID: 30011265 PMCID: PMC6067764 DOI: 10.1371/journal.pgen.1007519] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 07/31/2018] [Accepted: 06/27/2018] [Indexed: 12/19/2022] Open
Abstract
Although thousands of long non-coding RNAs (lncRNA) have been identified in the genomes of higher eukaryotes, the precise function of most of them is still unclear. Here, we show that a >65 kb, male-specific, lncRNA, called male-specific abdominal (msa) is required for the development of the secondary cells of the Drosophila male accessory gland (AG). msa is transcribed from within the Drosophila bithorax complex and shares much of its sequence with another lncRNA, the iab-8 lncRNA, which is involved in the development of the central nervous system (CNS). Both lncRNAs perform much of their functions via a shared miRNA embedded within their sequences. Loss of msa, or of the miRNA it contains, causes defects in secondary cell morphology and reduces male fertility. Although both lncRNAs express the same miRNA, the phenotype in the secondary cells and the CNS seem to reflect misregulation of different targets in the two tissues. In many animals, the male seminal fluid induces physiology changes in the mated female that increase a male’s reproductive success. These changes are often referred to as the post-mating response (PMR). In Drosophila, the seminal fluid proteins responsible for generating the PMR are made in a specialized gland, analogous to the mammalian seminal vesicle and prostate, called the accessory gland (AG). In this work, we show that a male-specific, long, non-coding RNA (lncRNA), called msa, plays a critical role in the development and function of this gland, primarily through a microRNA (miRNA) encoded within its sequence. This same miRNA had previously been shown to be expressed in the central nervous system (CNS) via an alternative promoter, where its ability to repress homeotic genes is required for both male and female fertility. Here, we present evidence that the targets of this miRNA in the AG are likely different from those found in the CNS. Thus, the same miRNA seems to have been selected to affect Drosophila fertility through two different mechanisms. Although many non-coding RNAs have now been identified, very few can be shown to have function. Our work highlights a lncRNA that has multiple biological functions, affecting cellular morphology and fertility.
Collapse
Affiliation(s)
- Robert K. Maeda
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland
- * E-mail: (RKM); (FK)
| | - Jessica L. Sitnik
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Yohan Frei
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland
| | - Elodie Prince
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland
| | - Dragan Gligorov
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland
| | - Mariana F. Wolfner
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - François Karch
- Department of Genetics and Evolution, University of Geneva, Geneva, Switzerland
- * E-mail: (RKM); (FK)
| |
Collapse
|
105
|
Qu Z, Bendena WG, Nong W, Siggens KW, Noriega FG, Kai ZP, Zang YY, Koon AC, Chan HYE, Chan TF, Chu KH, Lam HM, Akam M, Tobe SS, Lam Hui JH. MicroRNAs regulate the sesquiterpenoid hormonal pathway in Drosophila and other arthropods. Proc Biol Sci 2018; 284:rspb.2017.1827. [PMID: 29237851 DOI: 10.1098/rspb.2017.1827] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 11/14/2017] [Indexed: 12/12/2022] Open
Abstract
Arthropods comprise the majority of all described animal species, and understanding their evolution is a central question in biology. Their developmental processes are under the precise control of distinct hormonal regulators, including the sesquiterpenoids juvenile hormone (JH) and methyl farnesoate. The control of the synthesis and mode of action of these hormones played important roles in the evolution of arthropods and their adaptation to diverse habitats. However, the precise roles of non-coding RNAs, such as microRNAs (miRNAs), controlling arthropod hormonal pathways are unknown. Here, we investigated the miRNA regulation of the expression of the juvenile hormone acid methyltransferase gene (JHAMT), which encodes a rate-determining sesquiterpenoid biosynthetic enzyme. Loss of function of the miRNA bantam in the fly Drosophila melanogaster increased JHAMT expression, while overexpression of the bantam repressed JHAMT expression and resulted in pupal lethality. The male genital organs of the pupae were malformed, and exogenous sesquiterpenoid application partially rescued the genital deformities. The role of the bantam in the regulation of sesquiterpenoid biosynthesis was validated by transcriptomic, qPCR and hormone titre (JHB3 and JH III) analyses. In addition, we found a conserved set of miRNAs that interacted with JHAMT, and the sesquiterpenoid receptor methoprene-tolerant (Met) in different arthropod lineages, including insects (fly, mosquito and beetle), crustaceans (water flea and shrimp), myriapod (centipede) and chelicerate (horseshoe crab). This suggests that these miRNAs might have conserved roles in the post-transcriptional regulation of genes in sesquiterpenoid pathways across the Panarthropoda. Some of the identified lineage-specific miRNAs are potential targets for the development of new strategies in aquaculture and agricultural pest control.
Collapse
Affiliation(s)
- Zhe Qu
- School of Life Sciences, Simon F.S. Li Marine Science Laboratory, Partner State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | | | - Wenyan Nong
- School of Life Sciences, Simon F.S. Li Marine Science Laboratory, Partner State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | | | - Fernando G Noriega
- Department of Biological Sciences and Biomolecular Sciences Institute, Florida International University, Miami, FL 33199, USA
| | - Zhen-Peng Kai
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, People's Republic of China
| | - Yang-Yang Zang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai 201418, People's Republic of China
| | - Alex C Koon
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Ho Yin Edwin Chan
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Ting Fung Chan
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Ka Hou Chu
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Hon Ming Lam
- School of Life Sciences, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Michael Akam
- Department of Zoology, University of Cambridge, Cambridge, UK
| | - Stephen S Tobe
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada M5S 3G5
| | - Jerome Ho Lam Hui
- School of Life Sciences, Simon F.S. Li Marine Science Laboratory, Partner State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Sha Tin, Hong Kong
| |
Collapse
|
106
|
Ma Z, Wang H, Cai Y, Wang H, Niu K, Wu X, Ma H, Yang Y, Tong W, Liu F, Liu Z, Zhang Y, Liu R, Zhu ZJ, Liu N. Epigenetic drift of H3K27me3 in aging links glycolysis to healthy longevity in Drosophila. eLife 2018; 7:e35368. [PMID: 29809154 PMCID: PMC5991832 DOI: 10.7554/elife.35368] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 04/23/2018] [Indexed: 12/17/2022] Open
Abstract
Epigenetic alteration has been implicated in aging. However, the mechanism by which epigenetic change impacts aging remains to be understood. H3K27me3, a highly conserved histone modification signifying transcriptional repression, is marked and maintained by Polycomb Repressive Complexes (PRCs). Here, we explore the mechanism by which age-modulated increase of H3K27me3 impacts adult lifespan. Using Drosophila, we reveal that aging leads to loss of fidelity in epigenetic marking and drift of H3K27me3 and consequential reduction in the expression of glycolytic genes with negative effects on energy production and redox state. We show that a reduction of H3K27me3 by PRCs-deficiency promotes glycolysis and healthy lifespan. While perturbing glycolysis diminishes the pro-lifespan benefits mediated by PRCs-deficiency, transgenic increase of glycolytic genes in wild-type animals extends longevity. Together, we propose that epigenetic drift of H3K27me3 is one of the molecular mechanisms that contribute to aging and that stimulation of glycolysis promotes metabolic health and longevity.
Collapse
Affiliation(s)
- Zaijun Ma
- Interdisciplinary Research Center on Biology and ChemistryShanghai Institute of Organic ChemistryPudongChina
- University of Chinese Academy of SciencesBeijingChina
| | - Hui Wang
- Interdisciplinary Research Center on Biology and ChemistryShanghai Institute of Organic ChemistryPudongChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yuping Cai
- Interdisciplinary Research Center on Biology and ChemistryShanghai Institute of Organic ChemistryPudongChina
- University of Chinese Academy of SciencesBeijingChina
| | - Han Wang
- Interdisciplinary Research Center on Biology and ChemistryShanghai Institute of Organic ChemistryPudongChina
- University of Chinese Academy of SciencesBeijingChina
| | - Kongyan Niu
- Interdisciplinary Research Center on Biology and ChemistryShanghai Institute of Organic ChemistryPudongChina
| | - Xiaofen Wu
- Interdisciplinary Research Center on Biology and ChemistryShanghai Institute of Organic ChemistryPudongChina
- University of Chinese Academy of SciencesBeijingChina
| | - Huanhuan Ma
- Interdisciplinary Research Center on Biology and ChemistryShanghai Institute of Organic ChemistryPudongChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yun Yang
- Interdisciplinary Research Center on Biology and ChemistryShanghai Institute of Organic ChemistryPudongChina
- University of Chinese Academy of SciencesBeijingChina
| | - Wenhua Tong
- Interdisciplinary Research Center on Biology and ChemistryShanghai Institute of Organic ChemistryPudongChina
| | - Feng Liu
- National Research Center for Translational MedicineState Key Laboratory of Medical Genomics, Rui-Jin Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Zhandong Liu
- Jan and Dan Duncan Neurological Research InstituteTexas Children’s HospitalTexasUnited States
- Department of PediatricsBaylor College of MedicineHoustonUnited States
- Computational and Integrative Biomedical Research CenterBaylor College of MedicineHoustonUnited States
| | - Yaoyang Zhang
- Interdisciplinary Research Center on Biology and ChemistryShanghai Institute of Organic ChemistryPudongChina
| | - Rui Liu
- Singlera GenomicsPudong, ShanghaiChina
| | - Zheng-Jiang Zhu
- Interdisciplinary Research Center on Biology and ChemistryShanghai Institute of Organic ChemistryPudongChina
| | - Nan Liu
- Interdisciplinary Research Center on Biology and ChemistryShanghai Institute of Organic ChemistryPudongChina
| |
Collapse
|
107
|
Lidzbarsky G, Gutman D, Shekhidem HA, Sharvit L, Atzmon G. Genomic Instabilities, Cellular Senescence, and Aging: In Vitro, In Vivo and Aging-Like Human Syndromes. Front Med (Lausanne) 2018; 5:104. [PMID: 29719834 PMCID: PMC5913290 DOI: 10.3389/fmed.2018.00104] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Accepted: 03/29/2018] [Indexed: 12/20/2022] Open
Abstract
As average life span and elderly people prevalence in the western world population is gradually increasing, the incidence of age-related diseases such as cancer, heart diseases, diabetes, and dementia is increasing, bearing social and economic consequences worldwide. Understanding the molecular basis of aging-related processes can help extend the organism’s health span, i.e., the life period in which the organism is free of chronic diseases or decrease in basic body functions. During the last few decades, immense progress was made in the understanding of major components of aging and healthy aging biology, including genomic instability, telomere attrition, epigenetic changes, proteostasis, nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and intracellular communications. This progress has been made by three spear-headed strategies: in vitro (cell and tissue culture from various sources), in vivo (includes diverse model and non-model organisms), both can be manipulated and translated to human biology, and the study of aging-like human syndromes and human populations. Herein, we will focus on current repository of genomic “senescence” stage of aging, which includes health decline, structural changes of the genome, faulty DNA damage response and DNA damage, telomere shortening, and epigenetic alterations. Although aging is a complex process, many of the “hallmarks” of aging are directly related to DNA structure and function. This review will illustrate the variety of these studies, done in in vitro, in vivo and human levels, and highlight the unique potential and contribution of each research level and eventually the link between them.
Collapse
Affiliation(s)
| | - Danielle Gutman
- Department of Human Biology, University of Haifa, Haifa, Israel
| | | | - Lital Sharvit
- Department of Human Biology, University of Haifa, Haifa, Israel
| | - Gil Atzmon
- Department of Human Biology, University of Haifa, Haifa, Israel
| |
Collapse
|
108
|
Mustafin RN, Khusnutdinova EK. The Role of Transposons in Epigenetic Regulation of Ontogenesis. Russ J Dev Biol 2018. [DOI: 10.1134/s1062360418020066] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
109
|
Miya Shaik M, Tamargo IA, Abubakar MB, Kamal MA, Greig NH, Gan SH. The Role of microRNAs in Alzheimer's Disease and Their Therapeutic Potentials. Genes (Basel) 2018; 9:genes9040174. [PMID: 29561798 PMCID: PMC5924516 DOI: 10.3390/genes9040174] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/05/2018] [Accepted: 03/05/2018] [Indexed: 12/19/2022] Open
Abstract
MicroRNAs (miRNAs) are short, endogenous, non-coding RNAs that post-transcriptionally regulate gene expression by base pairing with mRNA targets. Altered miRNA expression profiles have been observed in several diseases, including neurodegeneration. Multiple studies have reported altered expressions of miRNAs in the brains of individuals with Alzheimer’s disease (AD) as compared to those of healthy elderly adults. Some of the miRNAs found to be dysregulated in AD have been reported to correlate with neuropathological changes, including plaque and tangle accumulation, as well as altered expressions of species that are known to be involved in AD pathology. To examine the potentially pathogenic functions of several dysregulated miRNAs in AD, we review the current literature with a focus on the activities of ten miRNAs in biological pathways involved in AD pathogenesis. Comprehensive understandings of the expression profiles and activities of these miRNAs will illuminate their roles as potential therapeutic targets in AD brain and may lead to the discovery of breakthrough treatment strategies for AD.
Collapse
Affiliation(s)
- Munvar Miya Shaik
- School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia.
| | - Ian A Tamargo
- Drug Design and Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Murtala B Abubakar
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Usmanu Danfodiyo University, PMB 2254 Sokoto, Nigeria.
| | - Mohammad A Kamal
- Metabolomics and Enzymology Unit, Fundamental and Applied Biology Group, King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia.
| | - Nigel H Greig
- Drug Design and Development Section, Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Siew Hua Gan
- School of Pharmacy, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor, Malaysia.
| |
Collapse
|
110
|
Aw SS, Lim IKH, Tang MXM, Cohen SM. A Glio-Protective Role of mir-263a by Tuning Sensitivity to Glutamate. Cell Rep 2018; 19:1783-1793. [PMID: 28564598 DOI: 10.1016/j.celrep.2017.05.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 03/14/2017] [Accepted: 05/02/2017] [Indexed: 12/25/2022] Open
Abstract
Glutamate is a ubiquitous neurotransmitter, mediating information flow between neurons. Defects in the regulation of glutamatergic transmission can result in glutamate toxicity, which is associated with neurodegeneration. Interestingly, glutamate receptors are expressed in glia, but little is known about their function, and the effects of their misregulation, in these non-neuronal cells. Here, we report a glio-protective role for Drosophila mir-263a mediated by its regulation of glutamate receptor levels in glia. mir-263a mutants exhibit a pronounced movement defect due to aberrant overexpression of CG5621/Grik, Nmdar1, and Nmdar2. mir-263a mutants exhibit excitotoxic death of a subset of astrocyte-like and ensheathing glia in the CNS. Glial-specific normalization of glutamate receptor levels restores cell numbers and suppresses the movement defect. Therefore, microRNA-mediated regulation of glutamate receptor levels protects glia from excitotoxicity, ensuring CNS health. Chronic low-level glutamate receptor overexpression due to mutations affecting microRNA (miRNA) regulation might contribute to glial dysfunction and CNS impairment.
Collapse
Affiliation(s)
- Sherry Shiying Aw
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore 138673, Singapore.
| | - Isaac Kok Hwee Lim
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Melissa Xue Mei Tang
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Stephen Michael Cohen
- Institute of Molecular and Cell Biology, 61 Biopolis Drive, Singapore 138673, Singapore.
| |
Collapse
|
111
|
Xu Y, Chen P, Wang X, Yao J, Zhuang S. miR-34a deficiency in APP/PS1 mice promotes cognitive function by increasing synaptic plasticity via AMPA and NMDA receptors. Neurosci Lett 2018; 670:94-104. [PMID: 29378298 DOI: 10.1016/j.neulet.2018.01.045] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/19/2018] [Accepted: 01/23/2018] [Indexed: 11/19/2022]
Abstract
MicroRNA (miR)-34a was recently determined to contribute to the pathological development of Alzheimer's disease (AD). miR-34a deficiency significantly attenuates cognitive deficits in amyloid precursor protein (APP)/presenilin 1 (PS1) mice; however, its role in early AD pathology and the underlying mechanisms remain elusive. Here, we confirmed that the increase of miR-34a expression in APP/PS1 mice was earlier than the relevant AD pathological characteristics, such as amyloid-β production, amyloid plaque deposition, and cognitive deficits. Furthermore, because predicted miR-34a target genes were broadly linked to α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and N-methyl-d-aspartate (NMDA) receptors, we evaluated synaptic plasticity by investigating high-frequency conditioning tetanus-induced excitatory postsynaptic potential, which revealed that synaptic plasticity was promoted in miR-34a knockout/APP/PS1 mice. Therefore, we assessed the expression of the presynaptic components synaptophysin and postsynaptic density protein 95 (PSD95) and found that synaptophysin and PSD95 were not altered by miR-34a deficiency. Additionally, the synaptic strength (vesicular fusion, vesicular docking, and transporting) was either not significantly changed. We also evaluated the levels of AMPA and NMDA receptors, which showed that the expression of AMPA and NMDA receptors was markedly upregulated in APP/PS1 mice with miR-34a deficiency. We conclude that miR-34a is involved in synaptic deficits in AD pathological development, which was, at least in part, due to the inhibition of NMDA (by miR-34a-5p) and AMPA (by miR-34a-3p) receptor expression.
Collapse
Affiliation(s)
- Yuelong Xu
- Department of Neurology, Linyi Central Hospital, No. 17 Jiankang Road, Linyi, Shandong, 276400, PR China
| | - Ping Chen
- Department of Pharmacy, Affiliated Hospital of Shandong Medical College, No. 80 Jintan Road, Linyi, Shandong, 276000, PR China
| | - Xianjun Wang
- Department of Neurology, Linyi People's Hospital, No. 49 Yizhou Road, Linyi, Shandong, 276000, PR China
| | - Jinguo Yao
- Department of Neurology, Linyi Central Hospital, No. 17 Jiankang Road, Linyi, Shandong, 276400, PR China
| | - Sujing Zhuang
- Department of Neurology, Linyi Central Hospital, No. 17 Jiankang Road, Linyi, Shandong, 276400, PR China.
| |
Collapse
|
112
|
Liang H, Ding B, Liang J, Shi X, Jiang X, Gao Y. MicroRNA-10a inhibits A30P α-synuclein aggregation and toxicity by targeting proapoptotic protein BCL2L11. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:624-633. [PMID: 31938148 PMCID: PMC6958036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Accepted: 12/22/2017] [Indexed: 06/10/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder around the world, and is characterized by progressive loss of nigrostriatal dopaminergic neurons. Certain microRNAs (miRNAs) are aberrantly expressed in the post-mortem brain tissues of patients with PD and in vivo PD model mice. However, the role of brain-enriched miRNA (miR)-10a in PD has not been studied. To investigate the regulatory role of miR-10a on α-synuclein (α-syn) in the pathology of PD, the present study aimed to examine whether upregulation of miR-10a attenuated A30P α-syn mutant aggregation and cellular toxicity. miRNA expression analysis by reverse transcription-quantitative polymerase chain reaction demonstrated that miR-10a expression was decreased in the midbrain of A30P α-syn transgenic mice and in SH-SY5Y human neuroblastoma cells transfected with A30P α-syn. In addition, miR-10a mimics were used to upregulate miR-10a expression. It was revealed that the upregulation of miR-10a suppressed α-syn intracellular accumulation and toxicity in α-syn-overexpressing SH-SY5Y cells. In addition, miR-10a overexpression resulted in a reversal of the A30P α-syn-induced upregulation of proapoptotic protein Bcl-2-associated X protein and cleaved caspase-3 expression and downregulation of antiapoptotic protein B-cell lymphoma-2 (BCL2) expression. A luciferase reporter assay demonstrated that BCL2-like 11 (BCL2L11), an apoptosis inducer, was a novel target gene of miR-10a. A30P α-syn aggregation and toxicity were alleviated by knocking down endogenous BCL2L11 in SH-SY5Y cells using a small interfering RNA specific for BCL2L11. In conclusion, these results demonstrate that miR-10a may serve a functional role in α-syn-induced neuronal pathology by inhibiting expression of BCL2L11 and that upregulation of miR-10a expression may be a useful therapeutic strategy for the treatment of PD.
Collapse
Affiliation(s)
- Huimin Liang
- Department of Neurology, Huaihe Hospital of Henan UniversityKaifeng, Henan, P. R. China
| | - Bingqian Ding
- Department of Neurology, Huaihe Hospital of Henan UniversityKaifeng, Henan, P. R. China
| | - Junhui Liang
- Department of Laboratory Medicine, Wugang People’s HospitalPingdingshan, Henan, P. R. China
| | - Xiaoyan Shi
- College of Pharmacy, Henan UniversityKaifeng, Henan, P. R. China
| | - Xin Jiang
- Department of Neurology, Huaihe Hospital of Henan UniversityKaifeng, Henan, P. R. China
| | - Yang Gao
- Department of Neurology, Huaihe Hospital of Henan UniversityKaifeng, Henan, P. R. China
| |
Collapse
|
113
|
Hu W, Wu J, Jiang W, Tang J. MicroRNAs and Presbycusis. Aging Dis 2018; 9:133-142. [PMID: 29392088 PMCID: PMC5772851 DOI: 10.14336/ad.2017.0119] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/19/2017] [Indexed: 12/19/2022] Open
Abstract
Presbycusis (age-related hearing loss) is the most universal sensory degenerative disease in elderly people caused by the degeneration of cochlear cells. Non-coding microRNAs (miRNAs) play a fundamental role in gene regulation in almost every multicellular organism, and control the aging processes. It has been identified that various miRNAs are up- or down-regulated during mammalian aging processes in tissue-specific manners. Most miRNAs bind to specific sites on their target messenger-RNAs (mRNAs) and decrease their expression. Germline mutation may lead to dysregulation of potential miRNAs expression, causing progressive hair cell degeneration and age-related hearing loss. Therapeutic innovations could emerge from a better understanding of diverse function of miRNAs in presbycusis. This review summarizes the relationship between miRNAs and presbycusis, and presents novel miRNAs-targeted strategies against presbycusis.
Collapse
Affiliation(s)
- Weiming Hu
- 1Department of Otolaryngology-Head and Neck Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Junwu Wu
- 2Department of Otolaryngology-Head and Neck Surgery, Yiwu traditional Chinese Medicine Hospital, Yiwu 322000, China.,3Department of Otolaryngology-Head and Neck Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou 310016, China
| | - Wenjing Jiang
- 1Department of Otolaryngology-Head and Neck Surgery, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou 310014, China
| | - Jianguo Tang
- 3Department of Otolaryngology-Head and Neck Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou 310016, China
| |
Collapse
|
114
|
Nakamura S, Yoshimori T. Autophagy and Longevity. Mol Cells 2018; 41:65-72. [PMID: 29370695 PMCID: PMC5792715 DOI: 10.14348/molcells.2018.2333] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 12/26/2017] [Accepted: 12/29/2017] [Indexed: 12/12/2022] Open
Abstract
Autophagy is an evolutionally conserved cytoplasmic degradation system in which varieties of materials are sequestered by a double membrane structure, autophagosome, and delivered to the lysosomes for the degradation. Due to the wide varieties of targets, autophagic activity is essential for cellular homeostasis. Recent genetic evidence indicates that autophagy has a crucial role in the regulation of animal lifespan. Basal level of autophagic activity is elevated in many longevity paradigms and the activity is required for lifespan extension. In most cases, genes involved in autophagy and lysosomal function are induced by several transcription factors including HLH-30/TFEB, PHA-4/FOXA and MML-1/Mondo in long-lived animals. Pharmacological treatments have been shown to extend lifespan through activation of autophagy, indicating autophagy could be a potential and promising target to modulate animal lifespan. Here we summarize recent progress regarding the role of autophagy in lifespan regulation.
Collapse
Affiliation(s)
- Shuhei Nakamura
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka,
Japan
- Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka,
Japan
| | - Tamotsu Yoshimori
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka,
Japan
- Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka,
Japan
| |
Collapse
|
115
|
Ueda M, Sato T, Ohkawa Y, Inoue YH. Identification of miR-305, a microRNA that promotes aging, and its target mRNAs in Drosophila. Genes Cells 2018; 23:80-93. [PMID: 29314553 DOI: 10.1111/gtc.12555] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 12/01/2017] [Indexed: 01/07/2023]
Abstract
MicroRNAs (miRNAs) are involved in the regulation of important biological processes. Here, we describe a novel Drosophila miRNAs involved in aging. We selected eight Drosophila miRNAs, displaying high homology with seed sequences of aging-related miRNAs characterized in other species, and investigated whether the over-expression of these miRNAs affected aging in Drosophila adult flies. The lifespan of adults over-expressing miR-305, a miRNA showing high homology with miR-239 in C. elegans, was significantly shorter. Conversely, a reduction in miR-305 expression led to a longer lifespan than that in control flies. miR-305 over-expression accelerated the impairment of locomotor activity and promoted the age-dependent accumulation of poly-ubiquitinated protein aggregates in the muscle, as flies aged. Thus, we show that the ectopic expression of miR-305 has a deleterious effect on aging in Drosophila. To identify the targets of miR-305, we performed RNA-Seq. We discovered several mRNAs encoding antimicrobial peptides and insulin-like peptides, whose expression changed in adults upon miR-305 over-expression. We further confirmed, by qRT-PCR, that miR-305 over-expression significantly decreases the mRNA levels of four antimicrobial peptides. As these mRNAs contain multiple sequences matching the seed sequence of miR-305, we speculate that a reduction in target mRNA levels, caused by ectopic miRNA expression, promotes aging.
Collapse
Affiliation(s)
- Makiko Ueda
- Department of Insect Biomedical Research, Center for Advanced Insect Research Promotion, Kyoto Institute of Technology, Kyoto, Japan
| | - Tetsuya Sato
- Medical Institute of Bioregulation, Kyushu University, Kyushu, Japan
| | - Yasuyuki Ohkawa
- Medical Institute of Bioregulation, Kyushu University, Kyushu, Japan
| | - Yoshihiro H Inoue
- Department of Insect Biomedical Research, Center for Advanced Insect Research Promotion, Kyoto Institute of Technology, Kyoto, Japan
| |
Collapse
|
116
|
Bai Y, Zhang Y, Han B, Yang L, Chen X, Huang R, Wu F, Chao J, Liu P, Hu G, Zhang JH, Yao H. Circular RNA DLGAP4 Ameliorates Ischemic Stroke Outcomes by Targeting miR-143 to Regulate Endothelial-Mesenchymal Transition Associated with Blood-Brain Barrier Integrity. J Neurosci 2018; 38:32-50. [PMID: 29114076 PMCID: PMC6705810 DOI: 10.1523/jneurosci.1348-17.2017] [Citation(s) in RCA: 296] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 10/24/2017] [Accepted: 10/29/2017] [Indexed: 01/01/2023] Open
Abstract
Circular RNAs (circRNAs) are highly expressed in the CNS and regulate physiological and pathophysiological processes. However, the potential role of circRNAs in stroke remains largely unknown. Here, we show that the circRNA DLGAP4 (circDLGAP4) functions as an endogenous microRNA-143 (miR-143) sponge to inhibit miR-143 activity, resulting in the inhibition of homologous to the E6-AP C-terminal domain E3 ubiquitin protein ligase 1 expression. circDLGAP4 levels were significantly decreased in the plasma of acute ischemic stroke patients (13 females and 13 males) and in a mouse stroke model. Upregulation of circDLGAP4 expression significantly attenuated neurological deficits and decreased infarct areas and blood-brain barrier damage in the transient middle cerebral artery occlusion mouse stroke model. Endothelial-mesenchymal transition contributes to blood-brain barrier disruption and circDLGAP4 overexpression significantly inhibited endothelial-mesenchymal transition by regulating tight junction protein and mesenchymal cell marker expression. Together, the results of our study are illustrative of the involvement of circDLGAP4 and its coupling mechanism in cerebral ischemia, providing translational evidence that circDLGAP4 serves as a novel therapeutic target for acute cerebrovascular protection.SIGNIFICANCE STATEMENT Circular RNAs (circRNAs) are involved in the regulation of physiological and pathophysiological processes. However, whether circRNAs are involved in ischemic injury, particularly cerebrovascular disorders, remains largely unknown. Here, we demonstrate a critical role for circular RNA DLGAP4 (circDLGAP4), a novel circular RNA originally identified as a sponge for microRNA-143 (miR-143), in ischemic stroke outcomes. Overexpression of circDLGAP4 significantly attenuated neurological deficits and decreased infarct areas and blood-brain barrier damage in the transient middle cerebral artery occlusion mouse stroke model. To our knowledge, this is the first report describing the efficacy of circRNA injection in an ischemic stroke model. Our investigation suggests that circDLGAP4 may serve as a novel therapeutic target for acute ischemic injury.
Collapse
Affiliation(s)
- Ying Bai
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Yuan Zhang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Bing Han
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Li Yang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Xufeng Chen
- Emergency Department, Jiangsu Province Hospital, Nanjing 210029, China
| | - Rongrong Huang
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Fangfang Wu
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Jie Chao
- Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| | - Pei Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Southeast University, Nanjing 210009, China
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing 210029, China
| | - John H Zhang
- Department of Anesthesiology, School of Medicine, Loma Linda University, Loma Linda, California 92354, and
| | - Honghong Yao
- Department of Pharmacology, School of Medicine, Southeast University, Nanjing 210009, China,
- Institute of Life Sciences, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing 210096, China
| |
Collapse
|
117
|
Abstract
The dominant polyglutamine (polyQ) disorders are a group of progressive and incurable neurodegenerative disorders, which are caused by unstable expanded CAG trinucleotide repeats in the coding regions of their respective causative genes. The most prevalent polyQ disorders worldwide are Huntington’s disease and spinocerebellar ataxia type 3. Epigenetic mechanisms, such as DNA methylation, histone modifications and chromatin remodeling and noncoding RNA regulation, regulate gene expression or genome function. Epigenetic dysregulation has been suggested to play a pivotal role in the pathogenesis of polyQ disorders. Here, we summarize the current knowledge of epigenetic changes present in several representative polyQ disorders and discuss the potentiality of miRNAs as therapeutic targets for the clinic therapy of these disorders.
Collapse
Affiliation(s)
- Hongmei Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Tie-Shan Tang
- State Key Laboratory of Membrane Biology, Institute of Zoology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| | - Caixia Guo
- CAS Key Laboratory of Genomics & Precision Medicine, Beijing Institute of Genomics, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
118
|
Gendron CM, Pletcher SD. MicroRNAs mir-184 and let-7 alter Drosophila metabolism and longevity. Aging Cell 2017; 16:1434-1438. [PMID: 28963741 PMCID: PMC5676060 DOI: 10.1111/acel.12673] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2017] [Indexed: 11/29/2022] Open
Abstract
MicroRNAs (miRNAs) are small RNA molecules that regulate gene expression associated with many complex biological processes. By comparing miRNA expression between long‐lived cohorts of Drosophila melanogaster that were fed a low‐nutrient diet with normal‐lived control animals fed a high‐nutrient diet, we identified miR‐184, let‐7, miR‐125, and miR‐100 as candidate miRNAs involved in modulating aging. We found that ubiquitous, adult‐specific overexpression of these individual miRNAs led to significant changes in fat metabolism and/or lifespan. Most impressively, adult‐specific overexpression of let‐7 in female nervous tissue increased median fly lifespan by ~22%. We provide evidence that this lifespan extension is not due to alterations in nutrient intake or to decreased insulin signaling.
Collapse
Affiliation(s)
- Christi M. Gendron
- Department of Molecular and Integrative Physiology and the Geriatrics Center University of Michigan Ann Arbor Michigan 48109 USA
| | - Scott D. Pletcher
- Department of Molecular and Integrative Physiology and the Geriatrics Center University of Michigan Ann Arbor Michigan 48109 USA
| |
Collapse
|
119
|
Green CD, Huang Y, Dou X, Yang L, Liu Y, Han JDJ. Impact of Dietary Interventions on Noncoding RNA Networks and mRNAs Encoding Chromatin-Related Factors. Cell Rep 2017; 18:2957-2968. [PMID: 28329687 DOI: 10.1016/j.celrep.2017.03.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 01/17/2017] [Accepted: 02/28/2017] [Indexed: 01/15/2023] Open
Abstract
Dietary interventions dramatically affect metabolic disease and lifespan in various aging models. Here, we profiled liver microRNA (miRNA), coding, and long non-coding RNA (lncRNA) expression by high-throughput deep sequencing in mice across multiple energy intake and expenditure interventions. Strikingly, three dietary intervention network design patterns were uncovered: (1) lifespan-extending interventions largely repressed the expression of miRNAs, lncRNAs, and transposable elements; (2) protein-coding mRNAs with expression positively correlated with long lifespan are highly targeted by miRNAs; and (3) miRNA-targeting interactions mainly target chromatin-related functions. We experimentally validated miR-34a, miR-107, and miR-212-3p targeting of the chromatin remodeler Chd1 and further demonstrate that Chd1 knockdown mimics high-fat diet and aging-induced gene expression changes and activation of transposons. Our findings demonstrate lifespan-extending diets repress miRNA-chromatin remodeler interactions and safeguard against deregulated transcription induced by aging and lifespan shortening diets, events linked by microRNA, chromatin, and ncRNA crosstalk.
Collapse
Affiliation(s)
- Christopher D Green
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Yi Huang
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Xiaoyang Dou
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liu Yang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yong Liu
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jing-Dong J Han
- Key Laboratory of Computational Biology, CAS Center for Excellence in Molecular Cell Science, Collaborative Innovation Center for Genetics and Developmental Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China.
| |
Collapse
|
120
|
Kato M, Kashem MA, Cheng C. An intestinal microRNA modulates the homeostatic adaptation to chronic oxidative stress in C. elegans. Aging (Albany NY) 2017; 8:1979-2005. [PMID: 27623524 PMCID: PMC5076448 DOI: 10.18632/aging.101029] [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: 07/06/2016] [Accepted: 08/19/2016] [Indexed: 12/22/2022]
Abstract
Adaptation to an environmental or metabolic perturbation is a feature of the evolutionary process. Recent insights into microRNA function suggest that microRNAs serve as key players in a robust adaptive response against stress in animals through their capacity to fine-tune gene expression. However, it remains largely unclear how a microRNA-modulated downstream mechanism contributes to the process of homeostatic adaptation. Here we show that loss of an intestinally expressed microRNA gene, mir-60, in the nematode C. elegans promotes an adaptive response to chronic - a mild and long-term - oxidative stress exposure. The pathway involved appears to be unique since the canonical stress-responsive factors, such as DAF-16/FOXO, are dispensable for mir-60 loss to enhance oxidative stress resistance. Gene expression profiles revealed that genes encoding lysosomal proteases and those involved in xenobiotic metabolism and pathogen defense responses are up-regulated by the loss of mir-60. Detailed genetic studies and computational microRNA target prediction suggest that endocytosis components and a bZip transcription factor gene zip-10, which functions in innate immune response, are directly modulated by miR-60 in the intestine. Our findings suggest that the mir-60 loss facilitates adaptive response against chronic oxidative stress by ensuring the maintenance of cellular homeostasis.
Collapse
Affiliation(s)
- Masaomi Kato
- The Laboratory of Ageing, Centenary Institute, Camperdown, NSW 2050, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Mohammed Abul Kashem
- The Laboratory of Ageing, Centenary Institute, Camperdown, NSW 2050, Australia.,Sydney Medical School, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Chao Cheng
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Lebanon, NH 03756, USA.,Department of Molecular and Systems Biology, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
| |
Collapse
|
121
|
Zhao W, Siegel D, Biton A, Tonqueze OL, Zaitlen N, Ahituv N, Erle DJ. CRISPR-Cas9-mediated functional dissection of 3'-UTRs. Nucleic Acids Res 2017; 45:10800-10810. [PMID: 28985357 PMCID: PMC5737544 DOI: 10.1093/nar/gkx675] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/25/2017] [Indexed: 11/21/2022] Open
Abstract
Many studies using reporter assays have demonstrated that 3′ untranslated regions (3′-UTRs) regulate gene expression by controlling mRNA stability and translation. Due to intrinsic limitations of heterologous reporter assays, we sought to develop a gene editing approach to investigate the regulatory activity of 3′-UTRs in their native context. We initially used dual-CRISPR (clustered, regularly interspaced, short palindromic repeats)-Cas9 targeting to delete DNA regions corresponding to nine chemokine 3′-UTRs that destabilized mRNA in a reporter assay. Targeting six chemokine 3′-UTRs increased chemokine mRNA levels as expected. However, targeting CXCL1, CXCL6 and CXCL8 3′-UTRs unexpectedly led to substantial mRNA decreases. Metabolic labeling assays showed that targeting these three 3′-UTRs increased mRNA stability, as predicted by the reporter assay, while also markedly decreasing transcription, demonstrating an unexpected role for 3′-UTR sequences in transcriptional regulation. We further show that CRISPR–Cas9 targeting of specific 3′-UTR elements can be used for modulating gene expression and for highly parallel localization of active 3′-UTR elements in the native context. Our work demonstrates the duality and complexity of 3′-UTR sequences in regulation of gene expression and provides a useful approach for modulating gene expression and for functional annotation of 3′-UTRs in the native context.
Collapse
Affiliation(s)
- Wenxue Zhao
- Lung Biology Center, Department of Medicine, University of California San Francisco, 4th St, San Francisco, CA 94158, USA
| | - David Siegel
- Lung Biology Center, Department of Medicine, University of California San Francisco, 4th St, San Francisco, CA 94158, USA
| | - Anne Biton
- Lung Biology Center, Department of Medicine, University of California San Francisco, 4th St, San Francisco, CA 94158, USA.,Centre de Bioinformatique, Biostatistique et Biologie Intégrative, C3BI, USR 3756 Institut Pasteur et CNRS, 25-28 Rue du Dr Roux, Paris 75015, France
| | - Olivier Le Tonqueze
- Lung Biology Center, Department of Medicine, University of California San Francisco, 4th St, San Francisco, CA 94158, USA
| | - Noah Zaitlen
- Lung Biology Center, Department of Medicine, University of California San Francisco, 4th St, San Francisco, CA 94158, USA
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, Institute for Human Genetics, University of California San Francisco, 4th St, San Francisco, CA 94158, USA
| | - David J Erle
- Lung Biology Center, Department of Medicine, University of California San Francisco, 4th St, San Francisco, CA 94158, USA
| |
Collapse
|
122
|
Victoria B, Nunez Lopez YO, Masternak MM. MicroRNAs and the metabolic hallmarks of aging. Mol Cell Endocrinol 2017; 455:131-147. [PMID: 28062199 PMCID: PMC5724961 DOI: 10.1016/j.mce.2016.12.021] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 10/10/2016] [Accepted: 12/16/2016] [Indexed: 12/12/2022]
Abstract
Aging, the natural process of growing older, is characterized by a progressive deterioration of physiological homeostasis at the cellular, tissue, and organismal level. Metabolically, the aging process is characterized by extensive changes in body composition, multi-tissue/multi-organ insulin resistance, and physiological declines in multiple signaling pathways including growth hormone, insulin/insulin-like growth factor 1, and sex steroids regulation. With this review, we intend to consolidate published information about microRNAs that regulate critical metabolic processes relevant to aging. In certain occasions we uncover relationships likely relevant to aging, which has not been directly described before, such as the miR-451/AMPK axis. We have also included a provocative section highlighting the potential role in aging of a new designation of miRNAs, namely fecal miRNAs, recently discovered to regulate intestinal microbiota in mammals.
Collapse
Affiliation(s)
- Berta Victoria
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, USA.
| | - Yury O Nunez Lopez
- Translational Research Institute for Metabolism & Diabetes. Florida Hospital, 301 East Princeton St, Orlando, FL 32804, USA.
| | - Michal M Masternak
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, USA; Department of Head and Neck Surgery, The Greater Poland Cancer Centre, 15 Garbary St., 61-866, Poznan, Poland.
| |
Collapse
|
123
|
Sirtuins, epigenetics and longevity. Ageing Res Rev 2017; 40:11-19. [PMID: 28789901 DOI: 10.1016/j.arr.2017.08.001] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 07/26/2017] [Accepted: 08/02/2017] [Indexed: 12/31/2022]
Abstract
Aging of organisms begins from a single cell at the molecular level. It includes changes related to telomere shortening, cell senescence and epigenetic modifications. These processes accumulate over the lifespan. Research studies show that epigenetic signaling contributes to human disease, tumorigenesis and aging. Epigenetic DNA modifications involve changes in the gene activity but not in the DNA sequence. An epigenome consists of chemical modifications to the DNA and histone proteins without the changes in the DNA sequence. These modifications strongly depend on the environment, could be reversible and are potentially transmittable to daughter cells. Epigenetics includes DNA methylation, noncoding RNA interference, and modifications of histone proteins. Sirtuins, a family of nicotine adenine dinucleotide (NAD+)-dependent enzymes, are involved in the cell metabolism and can regulate many cellular functions including DNA repair, inflammatory response, cell cycle or apoptosis. Literature shows the strong interconnection between sirtuin expression and aging processes. However, the direct relationship is still unknown. Here, we would like to summarize the existing knowledge about epigenetic processes in aging, especially those related to sirtuin expression. Another objective is to explain why some negative correlations between sirtuin activity and the rate of aging can be assumed.
Collapse
|
124
|
Valera E, Spencer B, Mott J, Trejo M, Adame A, Mante M, Rockenstein E, Troncoso JC, Beach TG, Masliah E, Desplats P. MicroRNA-101 Modulates Autophagy and Oligodendroglial Alpha-Synuclein Accumulation in Multiple System Atrophy. Front Mol Neurosci 2017; 10:329. [PMID: 29089869 PMCID: PMC5650998 DOI: 10.3389/fnmol.2017.00329] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 09/29/2017] [Indexed: 01/09/2023] Open
Abstract
Synucleinopathies, neurodegenerative disorders with alpha-synuclein (α-syn) accumulation, are the second leading cause of neurodegeneration in the elderly, however no effective disease-modifying alternatives exist for these diseases. Multiple system atrophy (MSA) is a fatal synucleinopathy characterized by the accumulation of toxic aggregates of α-syn within oligodendroglial cells, leading to demyelination and neurodegeneration, and the reduction of this accumulation might halt the fast progression of MSA. In this sense, the involvement of microRNAs (miRNAs) in synucleinopathies is yet poorly understood, and the potential of manipulating miRNA levels as a therapeutic tool is underexplored. In this study, we analyzed the levels of miRNAs that regulate the expression of autophagy genes in MSA cases, and investigated the mechanistic correlates of miRNA dysregulation in in vitro models of synucleinopathy. We found that microRNA-101 (miR-101) was significantly increased in the striatum of MSA patients, together with a reduction in the expression of its predicted target gene RAB5A. Overexpression of miR-101 in oligodendroglial cell cultures resulted in a significant increase in α-syn accumulation, along with autophagy deficits. Opposite results were observed upon expression of an antisense construct targeting miR-101. Stereotaxic delivery of a lentiviral construct expressing anti-miR-101 into the striatum of the MBP-α-syn transgenic (tg) mouse model of MSA resulted in reduced oligodendroglial α-syn accumulation and improved autophagy. These results suggest that miRNA dysregulation contributes to MSA pathology, with miR-101 alterations potentially mediating autophagy impairments. Therefore, therapies targeting miR-101 may represent promising approaches for MSA and related neuropathologies with autophagy dysfunction.
Collapse
Affiliation(s)
- Elvira Valera
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| | - Brian Spencer
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| | - Jennifer Mott
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| | - Margarita Trejo
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| | - Anthony Adame
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| | - Michael Mante
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| | - Edward Rockenstein
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| | - Juan C Troncoso
- Department of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Thomas G Beach
- Banner Sun Health Research Institute, Sun City, AZ, United States
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States.,Department of Pathology, University of California, San Diego, La Jolla, CA, United States
| | - Paula Desplats
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States.,Department of Pathology, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
125
|
Consales C, Cirotti C, Filomeni G, Panatta M, Butera A, Merla C, Lopresto V, Pinto R, Marino C, Benassi B. Fifty-Hertz Magnetic Field Affects the Epigenetic Modulation of the miR-34b/c in Neuronal Cells. Mol Neurobiol 2017; 55:5698-5714. [PMID: 29039021 DOI: 10.1007/s12035-017-0791-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 09/26/2017] [Indexed: 12/24/2022]
Abstract
The exposure to extremely low-frequency magnetic fields (ELF-MFs) has been associated to increased risk of neurodegenerative diseases, although the underlying molecular mechanisms are still undefined. Since epigenetic modulation has been recently encountered among the key events leading to neuronal degeneration, we here aimed at assessing if the control of gene expression mediated by miRNAs, namely miRs-34, has any roles in driving neuronal cell response to 50-Hz (1 mT) magnetic field in vitro. We demonstrate that ELF-MFs drive an early reduction of the expression level of miR-34b and miR-34c in SH-SY5Y human neuroblastoma cells, as well as in mouse primary cortical neurons, by affecting the transcription of the common pri-miR-34. This modulation is not p53 dependent, but attributable to the hyper-methylation of the CpG island mapping within the miR-34b/c promoter. Incubation with N-acetyl-l-cysteine or glutathione ethyl-ester fails to restore miR-34b/c expression, suggesting that miRs-34 are not responsive to ELF-MF-induced oxidative stress. By contrast, we show that miRs-34 control reactive oxygen species production and affect mitochondrial oxidative stress triggered by ELF-MFs, likely by modulating mitochondria-related miR-34 targets identified by in silico analysis. We finally demonstrate that ELF-MFs alter the expression of the α-synuclein, which is specifically stimulated upon ELF-MFs exposure via both direct miR-34 targeting and oxidative stress. Altogether, our data highlight the potential of the ELF-MFs to tune redox homeostasis and epigenetic control of gene expression in vitro and shed light on the possible mechanism(s) producing detrimental effects and predisposing neurons to degeneration.
Collapse
Affiliation(s)
- Claudia Consales
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy.
| | - Claudia Cirotti
- Department of Biology, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Giuseppe Filomeni
- Department of Biology, University of Rome Tor Vergata, 00133, Rome, Italy.,Cell Stress and Survival Unit, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Society Research Center, 2100, Copenhagen, Denmark
| | - Martina Panatta
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy
| | - Alessio Butera
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy
| | - Caterina Merla
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy.,Vectorology and Anticancer Therapies, UMR 8203, CNRS, Gustave Roussy, Univ. Paris-Sud, Université Paris-Saclay, 94805, Villejuif, France
| | - Vanni Lopresto
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy
| | - Rosanna Pinto
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy
| | - Carmela Marino
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy
| | - Barbara Benassi
- Division of Health Protection Technologies, ENEA-Italian National Agency for New Technologies, Energy and Sustainable Economic Development, ENEA-Casaccia, Via Anguillarese 301, 00123, Rome, Italy.
| |
Collapse
|
126
|
Pang J, Xiong H, Lin P, Lai L, Yang H, Liu Y, Huang Q, Chen S, Ye Y, Sun Y, Zheng Y. Activation of miR-34a impairs autophagic flux and promotes cochlear cell death via repressing ATG9A: implications for age-related hearing loss. Cell Death Dis 2017; 8:e3079. [PMID: 28981097 PMCID: PMC5680584 DOI: 10.1038/cddis.2017.462] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 07/27/2017] [Accepted: 08/02/2017] [Indexed: 12/12/2022]
Abstract
Age-related hearing loss is a major unresolved public health problem. We have previously elucidated that the activation of cochlear miR-34a is correlated with age-related hearing loss in C57BL/6 mice. A growing body of evidence points that aberrant autophagy promotes cell death during the development of multiple age-related diseases. The aim of this study was to investigate the role of miR-34a-involved disorder of autophagy in the pathogenesis of age-related hearing loss. Our results showed that miR-34a expression was markedly upregulated in the aging cochlea accompanied with impairment of autophagic flux. In the inner ear HEI-OC1 cell line, miR-34a overexpression resulted in an accumulation of phagophores and impaired autophagosome-lysosome fusion, and led to cell death subsequently. Notably, autophagy-related protein 9A (ATG9A), an autophagy protein, was significantly decreased after miR-34a overexpression. Knockdown of ATG9A inhibited autophagy flux, which is similar to the effects of miR-34a overexpression. Moreover, ursodeoxycholic acid significantly rescued miR-34a-induced HEI-OC1 cell death by restoring autophagy activity. Collectively, these findings increase our understanding of the biological effects of miR-34a in the development of age-related hearing loss and highlight miR-34a as a promising therapeutic target for its treatment.
Collapse
Affiliation(s)
- Jiaqi Pang
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hao Xiong
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Peiliang Lin
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, China
| | - Lan Lai
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haidi Yang
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, China
| | - Yimin Liu
- Guangzhou Occupational Disease Prevention and Treatment Center, Guangzhou, China
| | - Qiuhong Huang
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, China
| | - Suijun Chen
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, China
| | - Yongyi Ye
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yingfeng Sun
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, China
| | - Yiqing Zheng
- Department of Otolaryngology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.,Institute of Hearing and Speech-Language Science, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
127
|
MicroRNA-dependent regulation of metamorphosis and identification of microRNAs in the red flour beetle, Tribolium castaneum. Genomics 2017. [DOI: 10.1016/j.ygeno.2017.06.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
128
|
|
129
|
Brunquell J, Snyder A, Cheng F, Westerheide SD. HSF-1 is a regulator of miRNA expression in Caenorhabditis elegans. PLoS One 2017; 12:e0183445. [PMID: 28837599 PMCID: PMC5570370 DOI: 10.1371/journal.pone.0183445] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 08/06/2017] [Indexed: 12/16/2022] Open
Abstract
The ability of an organism to sense and adapt to environmental stressors is essential for proteome maintenance and survival. The highly conserved heat shock response is a survival mechanism employed by all organisms, including the nematode Caenorhabditis elegans, upon exposure to environmental extremes. Transcriptional control of the metazoan heat shock response is mediated by the heat shock transcription factor HSF-1. In addition to regulating global stress-responsive genes to promote stress-resistance and survival, HSF-1 has recently been shown to regulate stress-independent functions in controlling development, metabolism, and longevity. However, the indirect role of HSF-1 in coordinating stress-dependent and -independent processes through post-transcriptional regulation is largely unknown. MicroRNAs (miRNAs) have emerged as a class of post-transcriptional regulators that control gene expression through translational repression or mRNA degradation. To determine the role of HSF-1 in regulating miRNA expression, we have performed high-throughput small RNA-sequencing in C. elegans grown in the presence and absence of hsf-1 RNAi followed by treatment with or without heat shock. This has allowed us to uncover the miRNAs regulated by HSF-1 via heat-dependent and -independent mechanisms. Integrated miRNA/mRNA target-prediction analyses suggest HSF-1 as a post-transcriptional regulator of development, metabolism, and longevity through regulating miRNA expression. This provides new insight into the possible mechanism by which HSF-1 controls these processes. We have also uncovered oxidative stress response factors and insulin-like signaling factors as a common link between processes affected by HSF-1-regulated miRNAs in stress-dependent and -independent mechanisms, respectively. This may provide a role for miRNAs in regulating cross-talk between various stress responses. Our work therefore uncovers an interesting potential role for HSF-1 in post-transcriptionally controlling gene expression in C. elegans, and suggests a mechanism for cross-talk between stress responses.
Collapse
Affiliation(s)
- Jessica Brunquell
- Department of Cell Biology, Microbiology, and Molecular Biology, College of Arts and Sciences, University of South Florida, Tampa, Florida, United States of America
| | - Alana Snyder
- Department of Cell Biology, Microbiology, and Molecular Biology, College of Arts and Sciences, University of South Florida, Tampa, Florida, United States of America
| | - Feng Cheng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, Florida, United States of America
- Department of Biostatistics, College of Public Health, University of South Florida, Tampa, Florida, United States of America
- * E-mail: (SDW); (FC)
| | - Sandy D. Westerheide
- Department of Cell Biology, Microbiology, and Molecular Biology, College of Arts and Sciences, University of South Florida, Tampa, Florida, United States of America
- * E-mail: (SDW); (FC)
| |
Collapse
|
130
|
Ashapkin VV, Kutueva LI, Vanyushin BF. Aging as an Epigenetic Phenomenon. Curr Genomics 2017; 18:385-407. [PMID: 29081695 PMCID: PMC5635645 DOI: 10.2174/1389202918666170412112130] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 01/17/2016] [Accepted: 02/09/2016] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Hypermethylation of genes associated with promoter CpG islands, and hypomethylation of CpG poor genes, repeat sequences, transposable elements and intergenic genome sections occur during aging in mammals. Methylation levels of certain CpG sites display strict correlation to age and could be used as "epigenetic clock" to predict biological age. Multi-substrate deacetylases SIRT1 and SIRT6 affect aging via locus-specific modulations of chromatin structure and activity of multiple regulatory proteins involved in aging. Random errors in DNA methylation and other epigenetic marks during aging increase the transcriptional noise, and thus lead to enhanced phenotypic variation between cells of the same tissue. Such variation could cause progressive organ dysfunction observed in aged individuals. Multiple experimental data show that induction of NF-κB regulated gene sets occurs in various tissues of aged mammals. Upregulation of multiple miRNAs occurs at mid age leading to downregulation of enzymes and regulatory proteins involved in basic cellular functions, such as DNA repair, oxidative phosphorylation, intermediate metabolism, and others. CONCLUSION Strong evidence shows that all epigenetic systems contribute to the lifespan control in various organisms. Similar to other cell systems, epigenome is prone to gradual degradation due to the genome damage, stressful agents, and other aging factors. But unlike mutations and other kinds of the genome damage, age-related epigenetic changes could be fully or partially reversed to a "young" state.
Collapse
Affiliation(s)
- Vasily V Ashapkin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Lyudmila I Kutueva
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Boris F Vanyushin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
131
|
Salminen A, Kaarniranta K, Kauppinen A. Regulation of longevity by FGF21: Interaction between energy metabolism and stress responses. Ageing Res Rev 2017; 37:79-93. [PMID: 28552719 DOI: 10.1016/j.arr.2017.05.004] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/28/2017] [Accepted: 05/18/2017] [Indexed: 12/11/2022]
Abstract
Fibroblast growth factor 21 (FGF21) is a hormone-like member of FGF family which controls metabolic multiorgan crosstalk enhancing energy expenditure through glucose and lipid metabolism. In addition, FGF21 acts as a stress hormone induced by endoplasmic reticulum stress and dysfunctions of mitochondria and autophagy in several tissues. FGF21 also controls stress responses and metabolism by modulating the functions of somatotropic axis and hypothalamic-pituitary-adrenal (HPA) pathway. FGF21 is a potent longevity factor coordinating interactions between energy metabolism and stress responses. Recent studies have revealed that FGF21 treatment can alleviate many age-related metabolic disorders, e.g. atherosclerosis, obesity, type 2 diabetes, and some cardiovascular diseases. In addition, transgenic mice overexpressing FGF21 have an extended lifespan. However, chronic metabolic and stress-related disorders involving inflammatory responses can provoke FGF21 resistance and thus disturb healthy aging process. First, we will describe the role of FGF21 in interorgan energy metabolism and explain how its functions as a stress hormone can improve healthspan. Next, we will examine both the induction of FGF21 expression via the integrated stress response and the molecular mechanism through which FGF21 enhances healthy aging. Finally, we postulate that FGF21 resistance, similarly to insulin resistance, jeopardizes human healthspan and accelerates the aging process.
Collapse
|
132
|
Dato S, Rose G, Crocco P, Monti D, Garagnani P, Franceschi C, Passarino G. The genetics of human longevity: an intricacy of genes, environment, culture and microbiome. Mech Ageing Dev 2017; 165:147-155. [DOI: 10.1016/j.mad.2017.03.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 03/04/2017] [Accepted: 03/30/2017] [Indexed: 12/13/2022]
|
133
|
Jian C, Lu M, Zhang Z, Liu L, Li X, Huang F, Xu N, Qin L, Zhang Q, Zou D. miR-34a knockout attenuates cognitive deficits in APP/PS1 mice through inhibition of the amyloidogenic processing of APP. Life Sci 2017; 182:104-111. [PMID: 28533191 DOI: 10.1016/j.lfs.2017.05.023] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 05/09/2017] [Accepted: 05/18/2017] [Indexed: 11/25/2022]
Abstract
The noncoding miRNA-34a (miR-34a) is involved in Alzheimer's disease (AD) pathologenesis and shows potential for application as a biomarker for early diagnosis and intervention. Here, we established miR-34a knockout mice in an APP/PS1 background (APP/PS1-miR-34a KO mice) by crossbreeding miR-34a-/- mice with APP/PS1 mice. We then investigated cognitive impairment and related pathologies. The results showed that the level of miR-34a was increased at about 6months in APP/PS1 mice, consistent with the increase in amyloid β (Aβ), and cognitive function was significantly improved in mice when miR-34a was knocked out in 9-month-old and 12-month-old mice, indicating that miR-34a is a potential candidate for determining the progression of AD. Furthermore, we assessed the processing of amyloid precursor protein (APP) and the results suggest that cognitive improvement by miR-34a knock out was mainly triggered by depression of γ-secretase activity, without affecting β- and α-secretase activities, indicating that miR-34a plays an important role in AD pathology, mainly by inhibiting the amyloidogenic processing of APP, without altering the non-amyloidogenic processing of APP.
Collapse
Affiliation(s)
- Chongdong Jian
- Youjiang Medical University for Nationalities, Baise, Guangxi 533000, People's Republic of China
| | - Mengru Lu
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, China; Department of Neurology, The First People's Hospital of Nanning, Nanning, Guangxi 530022, China
| | - Zhao Zhang
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, China; Department of Neurology, The First People's Hospital of Nanning, Nanning, Guangxi 530022, China
| | - Long Liu
- Department of Chemistry and Biology, College of Science, National University of Defense Technology, Changsha, Hunan 410073, China
| | - Xianfeng Li
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, China; Department of Neurology, The First People's Hospital of Nanning, Nanning, Guangxi 530022, China
| | - Fang Huang
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, China; Department of Neurology, The First People's Hospital of Nanning, Nanning, Guangxi 530022, China
| | - Ning Xu
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, China; Department of Neurology, The First People's Hospital of Nanning, Nanning, Guangxi 530022, China
| | - Lina Qin
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, China; Department of Neurology, The First People's Hospital of Nanning, Nanning, Guangxi 530022, China
| | - Qian Zhang
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, China; Department of Neurology, The First People's Hospital of Nanning, Nanning, Guangxi 530022, China.
| | - Donghua Zou
- Department of Neurology, The Fifth Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530022, China; Department of Neurology, The First People's Hospital of Nanning, Nanning, Guangxi 530022, China.
| |
Collapse
|
134
|
Kogure A, Uno M, Ikeda T, Nishida E. The microRNA machinery regulates fasting-induced changes in gene expression and longevity in Caenorhabditis elegans. J Biol Chem 2017; 292:11300-11309. [PMID: 28507100 DOI: 10.1074/jbc.m116.765065] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 04/29/2017] [Indexed: 12/23/2022] Open
Abstract
Intermittent fasting (IF) is a dietary restriction regimen that extends the lifespans of Caenorhabditis elegans and mammals by inducing changes in gene expression. However, how IF induces these changes and promotes longevity remains unclear. One proposed mechanism involves gene regulation by microRNAs (miRNAs), small non-coding RNAs (∼22 nucleotides) that repress gene expression and whose expression can be altered by fasting. To test this proposition, we examined the role of the miRNA machinery in fasting-induced transcriptional changes and longevity in C. elegans We revealed that fasting up-regulated the expression of the miRNA-induced silencing complex (miRISC) components, including Argonaute and GW182, and the miRNA-processing enzyme DRSH-1 (the ortholog of the Drosophila Drosha enzyme). Our lifespan measurements demonstrated that IF-induced longevity was suppressed by knock-out or knockdown of miRISC components and was completely inhibited by drsh-1 ablation. Remarkably, drsh-1 ablation inhibited the fasting-induced changes in the expression of the target genes of DAF-16, the insulin/IGF-1 signaling effector in C. elegans Fasting-induced transcriptome alterations were substantially and modestly suppressed in the drsh-1 null mutant and the null mutant of ain-1, a gene encoding GW182, respectively. Moreover, miRNA array analyses revealed that the expression levels of numerous miRNAs changed after 2 days of fasting. These results indicate that components of the miRNA machinery, especially the miRNA-processing enzyme DRSH-1, play an important role in mediating IF-induced longevity via the regulation of fasting-induced changes in gene expression.
Collapse
Affiliation(s)
- Akiko Kogure
- From the Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan
| | - Masaharu Uno
- From the Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan
| | - Takako Ikeda
- From the Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan
| | - Eisuke Nishida
- From the Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Sakyo-ku, Kyoto 606-8502, Japan
| |
Collapse
|
135
|
Pervasive Behavioral Effects of MicroRNA Regulation in Drosophila. Genetics 2017; 206:1535-1548. [PMID: 28468905 PMCID: PMC5500149 DOI: 10.1534/genetics.116.195776] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 04/23/2017] [Indexed: 11/18/2022] Open
Abstract
Picao-Osorio et al. reveal pervasive effects of microRNA regulation on complex locomotor behaviors in Drosophila larvae: over 40% of microRNAs display... The effects of microRNA (miRNA) regulation on the genetic programs underlying behavior remain largely unexplored. Despite this, recent work in Drosophila shows that mutation of a single miRNA locus (miR-iab4/iab8) affects the capacity of the larva to correct its orientation if turned upside down (self-righting, SR), suggesting that other miRNAs might also be involved in behavioral control. Here we explore this possibility, studying early larval SR behavior in a collection of 81 Drosophila miRNA mutants covering almost the entire miRNA complement of the late embryo. Unexpectedly, we observe that >40% of all miRNAs tested significantly affect SR time, revealing pervasive behavioral effects of miRNA regulation in the early larva. Detailed analyses of those miRNAs affecting SR behavior (SR-miRNAs) show that individual miRNAs can affect movement in different ways, suggesting that specific molecular and cellular elements are affected by individual miRNA mutations. Furthermore, gene expression analysis shows that the Hox gene Abdominal-B (Abd-B) represents one of the targets deregulated by several SR-miRNAs. Our work thus reveals pervasive effects of miRNA regulation on a complex innate behavior in Drosophila and suggests that miRNAs may be core components of the genetic programs underlying behavioral control in other animals too.
Collapse
|
136
|
Wang X, Suofu Y, Akpinar B, Baranov SV, Kim J, Carlisle DL, Zhang Y, Friedlander RM. Systemic antimiR-337-3p delivery inhibits cerebral ischemia-mediated injury. Neurobiol Dis 2017; 105:156-163. [PMID: 28461247 DOI: 10.1016/j.nbd.2017.04.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 04/20/2017] [Accepted: 04/27/2017] [Indexed: 11/15/2022] Open
Abstract
Modulation of miRNA expression has been shown to be beneficial in the context of multiple diseases. The purpose of this study was to determine if an inhibitor of miR-337-3p is neuroprotective for hypoxic injury after tail vein injection. We evaluated miR-337-3p expression levels and in brain tissue in vivo before and after permanent middle cerebral artery occlusion (pMCAO) in mice. Subsequently, a custom locked nucleic acid (LNA) antimir-337-3p oligonucleotide was developed and tested in vitro after induction of oxygen glucose-deprivation (OGD) and in vivo by injection into the mouse tail vein for 3 consecutive days before pMCAO. Ischemic lesion volume was measured by TTC staining. We show that systemically administered LNA antimir-337-3p crosses the blood brain-brain-barrier (BBB), penetrates into neurosn, downregulates endogenous miR-337-3p expression and reduces ischemic brain injury. The findings support the use of similar antimir-LNA constructs as novel therapies in neurological disease.
Collapse
Affiliation(s)
- Xiaomin Wang
- Department of Neurological Surgery, Neuroapoptosis Laboratory, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, United States
| | - Yalikun Suofu
- Department of Neurological Surgery, Neuroapoptosis Laboratory, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, United States
| | - Berkcan Akpinar
- Department of Neurological Surgery, Neuroapoptosis Laboratory, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, United States
| | - Sergei V Baranov
- Department of Neurological Surgery, Neuroapoptosis Laboratory, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, United States
| | - Jinho Kim
- Department of Neurological Surgery, Neuroapoptosis Laboratory, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, United States
| | - Diane L Carlisle
- Department of Neurological Surgery, Neuroapoptosis Laboratory, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, United States
| | - Yu Zhang
- Department of Neurological Surgery, Neuroapoptosis Laboratory, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, United States.
| | - Robert M Friedlander
- Department of Neurological Surgery, Neuroapoptosis Laboratory, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, United States.
| |
Collapse
|
137
|
Wu N, Zhu Q, Chen B, Gao J, Xu Z, Li D. High-throughput sequencing of pituitary and hypothalamic microRNA transcriptome associated with high rate of egg production. BMC Genomics 2017; 18:255. [PMID: 28335741 PMCID: PMC5364632 DOI: 10.1186/s12864-017-3644-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Accepted: 03/18/2017] [Indexed: 01/21/2023] Open
Abstract
Background MicroRNAs exist widely in viruses, plants and animals. As endogenous small non-coding RNAs, miRNAs regulate a variety of biological processes. Tissue miRNA expression studies have discovered numerous functions for miRNAs in various tissues of chicken, but the regulation of miRNAs in chicken pituitary and hypothalamic development related to high and low egg-laying performance has remained unclear. Results In this study, using high-throughput sequencing technology, we sequenced two tissues (pituitary and hypothalamus) in 3 high- and 3 low-rate egg production Luhua chickens at the age of 300 days. By comparing low- and high-rate egg production chickens, 46 known miRNAs and 27 novel miRNAs were identified as differentially expressed (P < 0.05). Six differentially expressed known miRNAs, which are expressed in both tissues, were used in RT-qPCR validation and SNP detection. Among them, seven SNPs in two miRNA precursors (gga-miR-1684a and gga-miR-1434) were found that might enhance or reduce the production of the mature miRNAs. In addition, 124 and 30 reciprocally expressed miRNA-target pairs were identified by RNA-seq in pituitary and hypothalamic tissues, respectively and randomly selected candidate miRNA and miRNA-target pairs were validated by RT-qPCR in Jiuyuan black fowl. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway annotation illustrated that a large number of egg laying-related pathways were enriched in the high-rate egg production chickens, including ovarian steroidogenesis and steroid hormone biosynthesis. Conclusions These differentially expressed miRNAs and their predicted target genes, especially identified reciprocally expressed miRNA-target pairs, advance the study of miRNA function and egg production associated miRNA identification. The analysis of the miRNA-related SNPs and their effects provided insights into the effects of SNPs on miRNA biogenesis and function. The data generated in this study will further our understanding of miRNA regulation mechanisms in the chicken egg-laying process. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3644-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Nan Wu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, People's Republic of China, 610000
| | - Qing Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, People's Republic of China, 610000
| | - Binlong Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, People's Republic of China, 610000
| | - Jian Gao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, People's Republic of China, 610000
| | - Zhongxian Xu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, People's Republic of China, 610000
| | - Diyan Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, People's Republic of China, 610000.
| |
Collapse
|
138
|
Ripa R, Dolfi L, Terrigno M, Pandolfini L, Savino A, Arcucci V, Groth M, Terzibasi Tozzini E, Baumgart M, Cellerino A. MicroRNA miR-29 controls a compensatory response to limit neuronal iron accumulation during adult life and aging. BMC Biol 2017; 15:9. [PMID: 28193224 PMCID: PMC5304403 DOI: 10.1186/s12915-017-0354-x] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/25/2017] [Indexed: 02/07/2023] Open
Abstract
Background A widespread modulation of gene expression occurs in the aging brain, but little is known as to the upstream drivers of these changes. MicroRNAs emerged as fine regulators of gene expression in many biological contexts and they are modulated by age. MicroRNAs may therefore be part of the upstream drivers of the global gene expression modulation correlated with aging and aging-related phenotypes. Results Here, we show that microRNA-29 (miR-29) is induced during aging in short-lived turquoise killifish brain and genetic antagonism of its function induces a gene-expression signature typical of aging. Mechanicistically, we identified Ireb2 (a master gene for intracellular iron delivery that encodes for IRP2 protein), as a novel miR-29 target. MiR-29 is induced by iron loading and, in turn, it reduces IRP2 expression in vivo, therefore limiting intracellular iron delivery in neurons. Genetically modified fish with neuro-specific miR-29 deficiency exhibit increased levels of IRP2 and transferrin receptor, increased iron content, and oxidative stress. Conclusions Our results demonstrate that age-dependent miR-29 upregulation is an adaptive mechanism that counteracts the expression of some aging-related phenotypes and its anti-aging activity is primarily exerted by regulating intracellular iron homeostasis limiting excessive iron-exposure in neurons. Electronic supplementary material The online version of this article (doi:10.1186/s12915-017-0354-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Roberto Ripa
- Scuola Normale Superiore, Laboratory of Biology (Bio@SNS), c/o Istituto di Biofisica del CNR, via 17 Moruzzi 1, 56124, Pisa, Italy
| | - Luca Dolfi
- Scuola Normale Superiore, Laboratory of Biology (Bio@SNS), c/o Istituto di Biofisica del CNR, via 17 Moruzzi 1, 56124, Pisa, Italy
| | - Marco Terrigno
- Scuola Normale Superiore, Laboratory of Biology (Bio@SNS), c/o Istituto di Biofisica del CNR, via 17 Moruzzi 1, 56124, Pisa, Italy
| | - Luca Pandolfini
- Wellcome Trust/Cancer Research UK Gurdon Institute, Tennis Court Road, Cambridge, CB2 1QN, UK
| | | | - Valeria Arcucci
- Scuola Normale Superiore, Laboratory of Biology (Bio@SNS), c/o Istituto di Biofisica del CNR, via 17 Moruzzi 1, 56124, Pisa, Italy
| | - Marco Groth
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstr. 11, 07745, Jena, Germany
| | - Eva Terzibasi Tozzini
- Scuola Normale Superiore, Laboratory of Biology (Bio@SNS), c/o Istituto di Biofisica del CNR, via 17 Moruzzi 1, 56124, Pisa, Italy
| | - Mario Baumgart
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstr. 11, 07745, Jena, Germany
| | - Alessandro Cellerino
- Scuola Normale Superiore, Laboratory of Biology (Bio@SNS), c/o Istituto di Biofisica del CNR, via 17 Moruzzi 1, 56124, Pisa, Italy. .,Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), Beutenbergstr. 11, 07745, Jena, Germany.
| |
Collapse
|
139
|
Venø MT, Venø ST, Rehberg K, van Asperen JV, Clausen BH, Holm IE, Pasterkamp RJ, Finsen B, Kjems J. Cortical Morphogenesis during Embryonic Development Is Regulated by miR-34c and miR-204. Front Mol Neurosci 2017; 10:31. [PMID: 28232790 PMCID: PMC5299138 DOI: 10.3389/fnmol.2017.00031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 01/26/2017] [Indexed: 01/26/2023] Open
Abstract
The porcine brain closely resembles the human brain in aspects such as development and morphology. Temporal miRNA profiling in the developing embryonic porcine cortex revealed a distinct set of miRNAs, including miR-34c and miR-204, which exhibited a highly specific expression profile across the time of cortical folding. These miRNAs were found to target Doublecortin (DCX), known to be involved in neuron migration during cortical folding of gyrencephalic brains. In vivo modulation of miRNA expression in mouse embryos confirmed that miR-34c and miR-204 can control neuronal migration and cortical morphogenesis, presumably by posttranscriptional regulation of DCX.
Collapse
Affiliation(s)
- Morten T Venø
- Department of Molecular Biology and Genetics, Interdisciplinary Nanoscience Center, Aarhus University Aarhus, Denmark
| | - Susanne T Venø
- Department of Molecular Biology and Genetics, Interdisciplinary Nanoscience Center, Aarhus University Aarhus, Denmark
| | - Kati Rehberg
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht Utrecht, Netherlands
| | - Jessy V van Asperen
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht Utrecht, Netherlands
| | - Bettina H Clausen
- Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark Odense, Denmark
| | - Ida E Holm
- Laboratory for Experimental Neuropathology, Department of Pathology, Randers Hospital Randers, Denmark
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht Utrecht, Netherlands
| | - Bente Finsen
- Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark Odense, Denmark
| | - Jørgen Kjems
- Department of Molecular Biology and Genetics, Interdisciplinary Nanoscience Center, Aarhus University Aarhus, Denmark
| |
Collapse
|
140
|
MicroRNAs underlying memory deficits in neurodegenerative disorders. Prog Neuropsychopharmacol Biol Psychiatry 2017; 73:79-86. [PMID: 27117821 DOI: 10.1016/j.pnpbp.2016.04.011] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/01/2016] [Accepted: 04/22/2016] [Indexed: 11/23/2022]
Abstract
Neurodegenerative disorders are defined by neuronal loss and often associated with dementia. Understanding the multifactorial nature of cognitive decline is of particular interest. Cell loss is certainly a possibility but also an early imbalance in the complex gene networks involved in learning and memory. The small (~22nt) non-coding microRNAs play a major role in gene expression regulation and have been linked to neuronal survival and cognition. Interestingly, changes in microRNA signatures are associated with neurodegenerative disorders. In this review, we explore the role of three microRNAs, namely miR-132, miR-124 and miR-34, which are dysregulated in major neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease and Huntington's disease. Interestingly, these microRNAs have been associated with both memory impairment and neuronal survival, providing a potential common molecular mechanism contributing to dementia.
Collapse
|
141
|
Basavaraju M, de Lencastre A. Alzheimer's disease: presence and role of microRNAs. Biomol Concepts 2017; 7:241-52. [PMID: 27505094 DOI: 10.1515/bmc-2016-0014] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 07/01/2016] [Indexed: 11/15/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder that accounts for the most cases of dementia. AD affects more than 25 million people globally and is predicted to affect nearly one in 85 people worldwide by 2050. AD is characterized by the accumulation of dense plaques of β-amyloid peptide (Aβ) and neurofibrillary tangles of hyperphosphorylated tau that cause impairment in memory, cognition, and daily activities. Although early-onset AD has been linked to several mutations, reliable genetic markers for late-onset AD are lacking. Further, the diagnosis of AD biomarkers has its limitations and cannot detect early-stage AD. The identification of accurate, early, and non-invasive biomarkers for AD is, therefore, an unmet challenge. Recently, microRNAs (miRNAs) have emerged as a novel class of gene regulatory elements with conserved roles in development and disease. Recent discoveries have uncovered roles of miRNAs in several model organisms during aging and have identified potential miRNAs biomarkers of AD. Here we will discuss this emerging field of miRNAs associated with AD and prospects for the future.
Collapse
|
142
|
MicroRNAs in brain aging. Mech Ageing Dev 2017; 168:3-9. [PMID: 28119001 DOI: 10.1016/j.mad.2017.01.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Revised: 01/06/2017] [Accepted: 01/18/2017] [Indexed: 12/19/2022]
Abstract
Brain aging is one of the most crucial biological processes that affect the physiological balance between health and disease. Age-associated dysfunction of brain leads to severe health problems in current aging society. MicroRNAs (miRNAs) have emerged as important regulators in most physiological processes including fine-tuning of the short-term, cellular regulatory functions as well as modulation of long-term organismal lifespan. In this review, we discuss critical roles of miRNAs in the progression of normal and pathological brain aging. 50% of all known miRNAs are found in brain including cortex and hippocampus. A significant number of expressed miRNAs were differentially regulated during aging, implicating miRNAs as regulators of brain aging. The ability of miRNAs to regulate multiple targets within a pathway or even multiple pathways allows for coordinated regulation of brain functions. miRNA-mediated, brain functional changes are evident in cognition, inflammation, neuroprotection, lipid metabolism, mitochondrial function and lifespan. Dysregulation of brain miRNAs contributes to accelerated cognitive decline and increased neurological disorders. Elucidating mechanisms by which miRNAs and their multiple targets are temporally and spatially regulated in normal and pathological brain aging will provide a deeper understanding on the process of interrelated pathways of brain aging, and a new insight into therapeutic interventions.
Collapse
|
143
|
Regulation of miR-34 Family in Neuronal Development. Mol Neurobiol 2017; 55:936-945. [PMID: 28084588 DOI: 10.1007/s12035-016-0359-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 12/28/2016] [Indexed: 12/31/2022]
Abstract
Differentiation of neural stem cells (NSC's) to mature and functional neurons requires coordinated expression of mRNA, microRNAs (miRNAs) and regulatory proteins. Our earlier unbiased miRNA profiling studies have identified miR-200, miR-34 and miR-221/222 as maximally up-regulated miRNA families in differentiating PC12 cells and demonstrated the capability of miR-200 family in inducing neuronal differentiation (J. Neurochem, 2015, 133, 640-652). In present study, we have investigated role of miR-34 family in neuronal differentiation and identified P53 as mediator of nerve growth factor (NGF) induced miR-34a expression in differentiating PC12 cells. Our studies have shown that NGF induced miR-34a, arrests proliferating PC12 cells to G1 phase, which is pre-requisite for neuronal differentiation. Our studies have also shown that increased expression of miR-34a controls the P53 level in differentiated PC12 cells in feedback inhibition manner, which probably prevents differentiated cells from P53 induced apoptosis. Expression profiling of miR-34 family in different neuronal, non-neuronal and developing cells have identified differentiated and aged brain cells as richest source of miR-34, which also indicates that higher expression of miR-34 family helps in maintaining the mature neurons in non-proliferative stage. In conclusion, our studies have shown that miR-34 is brain enriched miRNA family, which up-regulates with neuronal maturation and brain ageing and co-operative regulation of P53 and miR-34a helps in neuronal differentiation by arresting cells in G1 phase.
Collapse
|
144
|
Martinez-Sanchez A, Rutter GA, Latreille M. MiRNAs in β-Cell Development, Identity, and Disease. Front Genet 2017; 7:226. [PMID: 28123396 PMCID: PMC5225124 DOI: 10.3389/fgene.2016.00226] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 12/21/2016] [Indexed: 12/22/2022] Open
Abstract
Pancreatic β-cells regulate glucose metabolism by secreting insulin, which in turn stimulates the utilization or storage of the sugar by peripheral tissues. Insulin insufficiency and a prolonged period of insulin resistance are usually the core components of type 2 diabetes (T2D). Although, decreased insulin levels in T2D have long been attributed to a decrease in β-cell function and/or mass, this model has recently been refined with the recognition that a loss of β-cell “identity” and dedifferentiation also contribute to the decline in insulin production. MicroRNAs (miRNAs) are key regulatory molecules that display tissue-specific expression patterns and maintain the differentiated state of somatic cells. During the past few years, great strides have been made in understanding how miRNA circuits impact β-cell identity. Here, we review current knowledge on the role of miRNAs in regulating the acquisition of the β-cell fate during development and in maintaining mature β-cell identity and function during stress situations such as obesity, pregnancy, aging, or diabetes. We also discuss how miRNA function could be harnessed to improve our ability to generate β-cells for replacement therapy for T2D.
Collapse
Affiliation(s)
- Aida Martinez-Sanchez
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London London, UK
| | - Guy A Rutter
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Medicine, Imperial College London London, UK
| | - Mathieu Latreille
- Cellular Identity and Metabolism Group, MRC London Institute of Medical SciencesLondon, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College LondonLondon, UK
| |
Collapse
|
145
|
Neault M, Couteau F, Bonneau É, De Guire V, Mallette FA. Molecular Regulation of Cellular Senescence by MicroRNAs: Implications in Cancer and Age-Related Diseases. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 334:27-98. [DOI: 10.1016/bs.ircmb.2017.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
|
146
|
Lai YW, Chu SY, Wei JY, Cheng CY, Li JC, Chen PL, Chen CH, Yu HH. Drosophila microRNA-34 Impairs Axon Pruning of Mushroom Body γ Neurons by Downregulating the Expression of Ecdysone Receptor. Sci Rep 2016; 6:39141. [PMID: 28008974 PMCID: PMC5180235 DOI: 10.1038/srep39141] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/17/2016] [Indexed: 11/25/2022] Open
Abstract
MicroRNA-34 (miR-34) is crucial for preventing chronic large-scale neurite degeneration in the aged brain of Drosophila melanogaster. Here we investigated the role of miR-34 in two other types of large-scale axon degeneration in Drosophila: axotomy-induced axon degeneration in olfactory sensory neurons (OSNs) and developmentally related axon pruning in mushroom body (MB) neurons. Ectopically overexpressed miR-34 did not inhibit axon degeneration in OSNs following axotomy, whereas ectopically overexpressed miR-34 in differentiated MB neurons impaired γ axon pruning. Intriguingly, the miR-34-induced γ axon pruning defect resulted from downregulating the expression of ecdysone receptor B1 (EcR-B1) in differentiated MB γ neurons. Notably, the separate overexpression of EcR-B1 or a transforming growth factor- β receptor Baboon, whose activation can upregulate the EcR-B1 expression, in MB neurons rescued the miR-34-induced γ axon pruning phenotype. Future investigations of miR-34 targets that regulate the expression of EcR-B1 in MB γ neurons are warranted to elucidate pathways that regulate axon pruning, and to provide insight into mechanisms that control large-scale axon degeneration in the nervous system.
Collapse
Affiliation(s)
- Yen-Wei Lai
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.,Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan.,Institute of Molecular and Cellular Biology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Sao-Yu Chu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Jia-Yi Wei
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| | - Chu-Ya Cheng
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan
| | - Jian-Chiuan Li
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan
| | - Po-Lin Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan.,Institute of Molecular and Cellular Biology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Chun-Hong Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan.,Institute of Molecular and Cellular Biology, College of Life Science, National Taiwan University, Taipei, Taiwan
| | - Hung-Hsiang Yu
- Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
147
|
Isik M, Blackwell TK, Berezikov E. MicroRNA mir-34 provides robustness to environmental stress response via the DAF-16 network in C. elegans. Sci Rep 2016; 6:36766. [PMID: 27905558 PMCID: PMC5131338 DOI: 10.1038/srep36766] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/20/2016] [Indexed: 12/16/2022] Open
Abstract
Diverse stresses and aging alter expression levels of microRNAs, suggesting a role for these posttranscriptional regulators of gene expression in stress modulation and longevity. Earlier studies demonstrated a central role for the miR-34 family in promoting cell cycle arrest and cell death following stress in human cells. However, the biological significance of this response was unclear. Here we show that in C. elegans mir-34 upregulation is necessary for developmental arrest, correct morphogenesis, and adaptation to a lower metabolic state to protect animals against stress-related damage. Either deletion or overexpression of mir-34 lead to an impaired stress response, which can largely be explained by perturbations in DAF-16/FOXO target gene expression. We demonstrate that mir-34 expression is regulated by the insulin signaling pathway via a negative feedback loop between miR-34 and DAF-16/FOXO. We propose that mir-34 provides robustness to stress response programs by controlling noise in the DAF-16/FOXO-regulated gene network.
Collapse
Affiliation(s)
- Meltem Isik
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands.,Joslin Diabetes Center, Harvard Stem Cell Institute and Harvard Medical School Department of Genetics, Boston, Massachusetts, United States of America
| | - T Keith Blackwell
- Joslin Diabetes Center, Harvard Stem Cell Institute and Harvard Medical School Department of Genetics, Boston, Massachusetts, United States of America
| | - Eugene Berezikov
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands.,European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
148
|
Xiong XP, Kurthkoti K, Chang KY, Li JL, Ren X, Ni JQ, Rana TM, Zhou R. miR-34 Modulates Innate Immunity and Ecdysone Signaling in Drosophila. PLoS Pathog 2016; 12:e1006034. [PMID: 27893816 PMCID: PMC5125713 DOI: 10.1371/journal.ppat.1006034] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 10/31/2016] [Indexed: 12/31/2022] Open
Abstract
microRNAs are endogenous small regulatory RNAs that modulate myriad biological processes by repressing target gene expression in a sequence-specific manner. Here we show that the conserved miRNA miR-34 regulates innate immunity and ecdysone signaling in Drosophila. miR-34 over-expression activates antibacterial innate immunity signaling both in cultured cells and in vivo, and flies over-expressing miR-34 display improved survival and pathogen clearance upon Gram-negative bacterial infection; whereas miR-34 knockout animals are defective in antibacterial defense. In particular, miR-34 achieves its immune-stimulatory function, at least in part, by repressing the two novel target genes Dlg1 and Eip75B. In addition, our study reveals a mutual repression between miR-34 expression and ecdysone signaling, and identifies miR-34 as a node in the intricate interplay between ecdysone signaling and innate immunity. Lastly, we identify cis-regulatory genomic elements and trans-acting transcription factors required for optimal ecdysone-mediated repression of miR-34. Taken together, our study enriches the repertoire of immune-modulating miRNAs in animals, and provides new insights into the interplay between steroid hormone signaling and innate immunity.
Collapse
Affiliation(s)
- Xiao-Peng Xiong
- Tumor Initiation and Maintenance Program; Sanford Burnham Prebys Medical Discovery Institute, California, United States of America
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, California, United States of America
| | - Krishna Kurthkoti
- Tumor Initiation and Maintenance Program; Sanford Burnham Prebys Medical Discovery Institute, California, United States of America
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, California, United States of America
| | - Kung-Yen Chang
- Tumor Initiation and Maintenance Program; Sanford Burnham Prebys Medical Discovery Institute, California, United States of America
- Department of Pediatrics, University of California San Diego School of Medicine, California, United States of America
| | - Jian-Liang Li
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, United States of America
| | - Xingjie Ren
- Gene Regulatory Laboratory, School of Medicine, Tsinghua University, Beijing, China
| | - Jian-Quan Ni
- Gene Regulatory Laboratory, School of Medicine, Tsinghua University, Beijing, China
| | - Tariq M. Rana
- Tumor Initiation and Maintenance Program; Sanford Burnham Prebys Medical Discovery Institute, California, United States of America
- Department of Pediatrics, University of California San Diego School of Medicine, California, United States of America
| | - Rui Zhou
- Tumor Initiation and Maintenance Program; Sanford Burnham Prebys Medical Discovery Institute, California, United States of America
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, California, United States of America
- * E-mail:
| |
Collapse
|
149
|
Pereira P, Queiroz JA, Figueiras A, Sousa F. Current progress on microRNAs-based therapeutics in neurodegenerative diseases. WILEY INTERDISCIPLINARY REVIEWS-RNA 2016; 8. [PMID: 27882692 DOI: 10.1002/wrna.1409] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 10/07/2016] [Accepted: 10/17/2016] [Indexed: 12/17/2022]
Abstract
MicroRNAs (miRNAs)-based therapy has recently emerged as a promising strategy in the treatments of neurodegenerative diseases. Thus, in this review, the most recent and important challenges and advances on the development of miRNA therapeutics for brain targeting are discussed. In particular, this review highlights current knowledge and progress in the field of manufacturing, recovery, isolation, purification, and analysis of these therapeutic oligonucleotides. Finally, the available miRNA delivery systems are reviewed and an analysis is presented in what concerns to the current challenges that have to be addressed to ensure their specificity and efficacy. Overall, it is intended to provide a perspective on the future of miRNA-based therapeutics, focusing the biotechnological approach to obtain miRNAs. WIREs RNA 2017, 8:e1409. doi: 10.1002/wrna.1409 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Patrícia Pereira
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal
| | - João A Queiroz
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal
| | - Ana Figueiras
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal.,REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, Universidade de Coimbra, Coimbra, Portugal
| | - Fani Sousa
- CICS-UBI - Health Sciences Research Centre, Universidade da Beira Interior, Covilhã, Portugal
| |
Collapse
|
150
|
Wu N, Gaur U, Zhu Q, Chen B, Xu Z, Zhao X, Yang M, Li D. Expressed microRNA associated with high rate of egg production in chicken ovarian follicles. Anim Genet 2016; 48:205-216. [PMID: 27781291 DOI: 10.1111/age.12516] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2016] [Indexed: 01/28/2023]
Abstract
MicroRNA (miRNA) is a highly conserved class of small noncoding RNA about 19-24 nucleotides in length that function in a specific manner to post-transcriptionally regulate gene expression in organisms. Tissue miRNA expression studies have discovered a myriad of functions for miRNAs in various aspects, but a role for miRNAs in chicken ovarian tissue at 300 days of age has not hitherto been reported. In this study, we performed the first miRNA analysis of ovarian tissues in chickens with low and high rates of egg production using high-throughput sequencing. By comparing low rate of egg production chickens with high rate of egg production chickens, 17 significantly differentially expressed miRNAs were found (P < 0.05), including 11 known and six novel miRNAs. We found that all 11 known miRNAs were involved mainly in pathways of reproduction regulation, such as steroid hormone biosynthesis and dopaminergic synapse. Additionally, expression profiling of six randomly selected differentially regulated miRNAs were validated by quantitative real-time polymerase chain reaction (RT-qPCR). Some miRNAs, such as gga-miR-34b, gga-miR-34c and gga-miR-216b, were reported to regulate processes such as proliferation, cell cycle, apoptosis and metastasis and were expressed differentially in ovaries of chickens with high rates of egg production, suggesting that these miRNAs have an important role in ovary development and reproductive management of chicken. Furthermore, we uncovered that a significantly up-regulated miRNA-gga-miR-200a-3p-is ubiquitous in reproduction-regulation-related pathways. This miRNA may play a special central role in the reproductive management of chicken, and needs to be further studied for confirmation.
Collapse
Affiliation(s)
- N Wu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 610000, China
| | - U Gaur
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 610000, China
| | - Q Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 610000, China
| | - B Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 610000, China
| | - Z Xu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 610000, China
| | - X Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 610000, China
| | - M Yang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 610000, China
| | - D Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 610000, China
| |
Collapse
|