1
|
Chunhui G, Yanqiu Y, Jibing C, Ning L, Fujun L. Exosomes and non-coding RNAs: bridging the gap in Alzheimer's pathogenesis and therapeutics. Metab Brain Dis 2025; 40:84. [PMID: 39754674 PMCID: PMC11700052 DOI: 10.1007/s11011-024-01520-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 12/27/2024] [Indexed: 01/06/2025]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that primarily affects the elderly population and is the leading cause of dementia. Meanwhile, the vascular hypothesis suggests that vascular damage occurs in the early stages of the disease, leading to neurodegeneration and hindered waste clearance, which in turn triggers a series of events including the accumulation of amyloid plaques and Tau protein tangles. Non-coding RNAs (ncRNAs), including long noncoding RNAs (lncRNAs), microRNAs (miRNAs), and circular RNAs (circRNAs), have been found to be involved in the regulation of AD. Furthermore, lncRNAs and circRNAs can act as competitive endogenous RNAs to inhibit miRNAs, and their interactions can form a complex regulatory network. Exosomes, which are extracellular vesicles (EVs), are believed to be able to transfer ncRNAs between cells, thus playing a regulatory role in the brain by crossing the blood-brain barrier (BBB). Exosomes are part of the intercellular carrier system; therefore, utilizing exosomes to deliver drugs to recipient cells might not activate the immune system, making it a potential strategy to treat central nervous system diseases. In this review, we review that AD is a multifactorial neurological disease and that ncRNAs can regulate its multiple pathogenic mechanisms to improve our understanding of the etiology of AD and to simultaneously regulate multiple pathogenic mechanisms of AD through the binding of ncRNAs to exosomes to improve the treatment of AD.
Collapse
Affiliation(s)
- Guo Chunhui
- Graduate School, Guangxi University of Chinese Medicine, Nanning, 530000, China
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, 530000, China
| | - You Yanqiu
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, 530000, China
| | - Chen Jibing
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, 530000, China.
| | - Luo Ning
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, 530000, China.
| | - Li Fujun
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, 530000, China.
| |
Collapse
|
2
|
Abuduwaili Z, Fan Y, Tao W, Chen Y, Xu Y, Zhu X. The Role of circRNAs in the Pathological Mechanisms of Alzheimer's Disease: Potential Biomarkers for Diagnosis. Curr Neuropharmacol 2025; 23:635-649. [PMID: 39449333 DOI: 10.2174/011570159x337659241014140824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 08/02/2024] [Accepted: 08/20/2024] [Indexed: 10/26/2024] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease leading to dementia in the elderly, and the mechanisms of AD have not been fully defined. Circular RNAs (circRNAs), covalently closed RNAs produced by reverse splicing, have critical effects in the pathogenesis of AD. CircRNAs participate in production and clearance of Aβ and tau, regulate neuroinflammation, synaptic plasticity and the process of apoptosis and autophagy, indicating that circRNAs may be alternative biomarkers and therapeutic targets. Our review summarizes the functions of circRNAs in the progression and development of AD, which provide insights into the prospect of circRNAs in the diagnosis and treatment of AD.
Collapse
Affiliation(s)
- Zulalai Abuduwaili
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Yingao Fan
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
| | - Wenyuan Tao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, China
| | - Yanting Chen
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, China
| | - Xiaolei Zhu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, Jiangsu, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, Jiangsu, China
| |
Collapse
|
3
|
Goodrich JM, Furlong MA, Urwin DJ, Gabriel J, Hughes J, Jung AM, Calkins MM, DuBose KN, Caban‐Martinez AJ, Solle NS, Beitel SC, Burgess JL. Epigenetic Modifications Associated With Wildland-Urban Interface (WUI) Firefighting. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2025; 66:22-33. [PMID: 39968828 PMCID: PMC11905879 DOI: 10.1002/em.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/05/2025] [Accepted: 02/07/2025] [Indexed: 02/20/2025]
Abstract
Wildland-urban interface (WUI) firefighting involves exposure to burning vegetation, structures, and other human-made hazards, often without respiratory protection. Response activities can last for long periods of time, spanning multiple days or weeks. Epigenetic modifications, including microRNA (miRNA) expression and DNA methylation, are responsive to toxicant exposures and are part of the development of cancers and other diseases. Epigenetic modifications have not been studied in relation to WUI fires. Firefighters (n = 99) from southern California, including 79 firefighters who responded to at least one WUI fire, provided blood samples at baseline and approximately 10 months later. We quantified the relative abundance of 800 miRNAs in blood samples using the nCounter Human v3 miRNA expression panel and blood leukocyte DNA methylation throughout the genome via the Infinium EPIC array. We used linear mixed models to compare the expression of each miRNA across time and DNA methylation at each locus, adjusting for potential confounders. In the miRNA analysis among all firefighters, 65 miRNAs were significantly different at follow-up compared to baseline at a false discovery rate of 5%. Results were similar when restricted to firefighters with a recorded WUI fire exposure during the interim period, although only 50 were significant. Expression of miRNA hsa-miR-518c-3p, a tumor suppressor, was significantly associated with WUI fire response (fold change 0.77, 95% CI = [0.69, 0.87]). In the DNA methylation analysis, no statistically significant changes over time were identified. In summary, WUI fire exposures over a wildfire season altered miRNA expression but did not substantially impact DNA methylation.
Collapse
Affiliation(s)
- Jaclyn M. Goodrich
- Department of Environmental Health SciencesUniversity of Michigan School of Public HealthAnn ArborMichiganUSA
| | - Melissa A. Furlong
- Department of Community, Environment and PolicyUniversity of Arizona Mel and Enid Zuckerman College of Public HealthTucsonArizonaUSA
| | - Derek J. Urwin
- Department of Chemistry & BiochemistryUniversity of California, Los AngelesLos AngelesCaliforniaUSA
- Los Angeles County Fire DepartmentLos AngelesCaliforniaUSA
| | - Jamie Gabriel
- Los Angeles County Fire DepartmentLos AngelesCaliforniaUSA
| | - Jeff Hughes
- Orange County Fire AuthorityIrvineCaliforniaUSA
| | - Alesia M. Jung
- Department of Community, Environment and PolicyUniversity of Arizona Mel and Enid Zuckerman College of Public HealthTucsonArizonaUSA
- Exponent, Inc.Menlo ParkCaliforniaUSA
| | - Miriam M. Calkins
- National Institute for Occupational Safety and Health (NIOSH), Centers for Disease Control and Prevention (CDC)CincinnatiOhioUSA
| | - Kathleen N. DuBose
- United States Department of InteriorOffice of Wildland FireBoiseIdahoUSA
| | | | - Natasha Schaefer Solle
- Department of Public Health SciencesUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Shawn C. Beitel
- Department of Community, Environment and PolicyUniversity of Arizona Mel and Enid Zuckerman College of Public HealthTucsonArizonaUSA
| | - Jefferey L. Burgess
- Department of Community, Environment and PolicyUniversity of Arizona Mel and Enid Zuckerman College of Public HealthTucsonArizonaUSA
| |
Collapse
|
4
|
Yang JY. miR-574-5p in epigenetic regulation and Toll-like receptor signaling. Cell Commun Signal 2024; 22:567. [PMID: 39593070 PMCID: PMC11600836 DOI: 10.1186/s12964-024-01934-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024] Open
Abstract
miR-574-5p is an unusual microRNA (miRNA) that is often upregulated or downregulated following exposure to irradiation or toxic chemicals; bacterial, parasitic or viral infection; and a variety of other disease conditions. Canonically, miR-574-5p epigenetically regulates the expression of many messenger RNAs (mRNAs) through miRNA-mediated posttranscriptional regulation, thereby affecting cellular physiology or pathophysiology and contributing to the pathogenesis or progression of a variety of diseases. However, recent studies have established that in addition to serving as a fine-tuning repressor of gene expression, miR-574-5p also stimulates gene expression as an endogenous ligand for Toll-like receptor-8/7 (TLR8/7). Indeed, the binding of miR-574-5p to TLR8/7 triggers the TLR signaling pathway, leading to the induction of interferons, inflammatory cytokines and autoimmune signaling. These findings suggest that miR-574-5p is not only an important epigenetic regulator of gene expression, but also an important regulator of immune and inflammatory responses. Abnormal miR-574-5p-TLR8/7 signaling has been shown to be tightly associated with inflammation-related cancers and a number of autoimmune disorders. miR-574-5p can serve as a potential biomarker for many diseases. Most importantly, miR-574-5p is a promising therapeutic target for the treatment or prevention of human disorders, especially infectious diseases, cancers and autoimmune diseases.
Collapse
Affiliation(s)
- James Y Yang
- Kidney Health Institute, Health Science Center, East China Normal University, Minhang, Shanghai, 200241, China.
- Wuhu Hospital of East China Normal University, Wuhu, 241000, Anhui, China.
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiang'an, Xiamen, 361102, China.
| |
Collapse
|
5
|
Deng X, Qiu Z, Chen X, Liu J, Wang X, Li J, Zhang J, Cui X, Fu Y, Jiang M. Exploring the potential mechanism of ginsenoside Rg1 to regulate ferroptosis in Alzheimer's disease based on network pharmacology. Eur J Pharmacol 2024; 979:176859. [PMID: 39067563 DOI: 10.1016/j.ejphar.2024.176859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
OBJECTIVES To explore the pathogenesis of Alzheimer's disease (AD), the potential targets and signaling pathways of ginsenoside Rg1 against AD were investigated by network pharmacology. METHODS Ginsenoside Rg1 targets were identified through PubChem, PharmMapper, and Uniprot databases, while the GeneCards database was used to examine the respective targets of amyloid precursor protein (APP) and AD. Then, the common targets between ginsenoside Rg1 and APP were explored by the Venny tool, the interaction network diagram between the active components and the targets was built via Cytoscape software, as well as GO enrichment and KEGG pathway annotation analysis were performed. Furthermore, genes associated with ferroptosis were found by the GeneCards and FerrDb databases. Besides, the connection among ginsenoside Rg1, APP, ferroptosis, and AD was predicted and analyzed. Finally, the effects of ginsenosides Rg1 and liproxstain-1 on the proliferation and differentiation of APP/PS1 mice were evaluated by immunohistochemistry. RESULTS Ginsenoside Rg1, APP, ferroptosis, and AD had 12 hub genes. GO enrichment and KEGG pathway annotation analysis showed that EGFR, SRC, protein hydrolysis, protein phosphorylation, the Relaxin pathway, and the FoxO signaling pathway play an important role in the potential mechanism of ginsenoside Rg1's under regulation of ferroptosis anti-AD through the modulation of APP-related signaling pathways. The APP/PS1 mice experiment verified that ginsenosides Rg1 and liproxstain-1 can promote the proliferation and differentiation. CONCLUSION Ginsenoside Rg1, APP and ferroptosis may act on EGFR, SRC, the Relaxin and FoxO signaling pathways to regulate protein metabolism, protein phosphorylation and other pathways to improve AD symptoms.
Collapse
Affiliation(s)
- Xu Deng
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Zixiong Qiu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaoshuai Chen
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jiangxiu Liu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaowei Wang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jie Li
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Jiankai Zhang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China
| | - Xiaojun Cui
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| | - Yuan Fu
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| | - Mei Jiang
- Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Department of Human Anatomy, Dongguan Campus, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
6
|
Xu YR, Zhao J, Huang HY, Lin YCD, Lee TY, Huang HD, Yang Y, Wang YF. Recent insights into breast milk microRNA: their role as functional regulators. Front Nutr 2024; 11:1366435. [PMID: 38689935 PMCID: PMC11058965 DOI: 10.3389/fnut.2024.1366435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/02/2024] [Indexed: 05/02/2024] Open
Abstract
Breast milk (BM) is a primary biofluid that plays a crucial role in infant development and the regulation of the immune system. As a class of rich biomolecules in BM, microRNAs (miRNAs) are regarded as active factors contributing to infant growth and development. Surprisingly, these molecules exhibit resilience in harsh conditions, providing an opportunity for infants to absorb them. In addition, many studies have shown that miRNAs in breast milk, when absorbed into the gastrointestinal system, can act as a class of functional regulators to effectively regulate gene expression. Understanding the absorption pattern of BM miRNA may facilitate the creation of formula with a more optimal miRNA balance and pave the way for novel drug delivery techniques. In this review, we initially present evidence of BM miRNA absorption. Subsequently, we compile studies that integrate both in vivo and in vitro findings to illustrate the bioavailability and biodistribution of BM miRNAs post-absorption. In addition, we evaluate the strengths and weaknesses of previous studies and discuss potential variables contributing to discrepancies in their outcomes. This literature review indicates that miRNAs can be absorbed and act as regulatory agents.
Collapse
Affiliation(s)
- Yi-Ran Xu
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Jinglu Zhao
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Hsi-Yuan Huang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Yang-Chi-Dung Lin
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Tzong-Yi Lee
- Institute of Bioinformatics and Systems Biology and Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Hsien-Da Huang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Yi Yang
- Department of Nephrology, Center for Regeneration and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
- Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu, China
| | - Yong-Fei Wang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| |
Collapse
|
7
|
Barreda-Manso MA, Soto A, Muñoz-Galdeano T, Reigada D, Nieto-Díaz M, Maza RM. MiR-138-5p Upregulation during Neuronal Maturation Parallels with an Increase in Neuronal Survival. Int J Mol Sci 2023; 24:16509. [PMID: 38003699 PMCID: PMC10671628 DOI: 10.3390/ijms242216509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Neuronal maturation is a process that plays a key role in the development and regeneration of the central nervous system. Although embryonic brain development and neurodegeneration have received considerable attention, the events that govern postnatal neuronal maturation are less understood. Among the mechanisms influencing such neuronal maturation processes, apoptosis plays a key role. Several regulators have been described to modulate apoptosis, including post-transcriptional regulation by microRNAs. This study aimed to analyze endogenous expression changes of miR-138-5p, as well as its main validated pro-apoptotic target caspase3, during the maturation of neuronal cultures and their response under apoptotic challenge. Our results point out that the observed opposite expression of miR-138-5p and its target caspase3 might modulate apoptosis favoring neuronal survival at distinct maturation stages. The unchanged expression of miR-138-5p in mature neurons contrasts with the significant downregulation in immature neurons upon apoptotic stimulation. Similarly, immunoblot and individual cellular assays confirmed that during maturation, not only the expression but processing of CASP-3 and caspase activity is reduced after apoptotic stimulation which results in a reduction of neuronal death. Further studies would be needed to determine a more detailed role of miR-138-5p in apoptosis during neuronal maturation and the synergistic action of several microRNAs acting cooperatively on caspase3 or other apoptotic targets.
Collapse
Affiliation(s)
- María Asunción Barreda-Manso
- Research Unit, Molecular Neuroprotection Group, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain; (A.S.); (T.M.-G.); (D.R.); (M.N.-D.)
- Research Unit, Functional Exploration and Neuromodulation of the Central Nervous System (FENNSI) Group, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain
| | - Altea Soto
- Research Unit, Molecular Neuroprotection Group, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain; (A.S.); (T.M.-G.); (D.R.); (M.N.-D.)
| | - Teresa Muñoz-Galdeano
- Research Unit, Molecular Neuroprotection Group, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain; (A.S.); (T.M.-G.); (D.R.); (M.N.-D.)
| | - David Reigada
- Research Unit, Molecular Neuroprotection Group, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain; (A.S.); (T.M.-G.); (D.R.); (M.N.-D.)
| | - Manuel Nieto-Díaz
- Research Unit, Molecular Neuroprotection Group, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain; (A.S.); (T.M.-G.); (D.R.); (M.N.-D.)
| | - Rodrigo M. Maza
- Research Unit, Molecular Neuroprotection Group, Hospital Nacional de Parapléjicos, SESCAM, 45071 Toledo, Spain; (A.S.); (T.M.-G.); (D.R.); (M.N.-D.)
| |
Collapse
|
8
|
Chen R, Yang H, Dai J, Zhang M, Lu G, Zhang M, Yu H, Zheng M, He Q. The biological functions of maternal-derived extracellular vesicles during pregnancy and lactation and its impact on offspring health. Clin Nutr 2023; 42:493-504. [PMID: 36857958 DOI: 10.1016/j.clnu.2023.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 01/25/2023] [Accepted: 02/09/2023] [Indexed: 02/18/2023]
Abstract
During pregnancy and lactation, mothers provide not only nutrients, but also many bioactive components for their offspring through placenta and breast milk, which are essential for offspring development. Extracellular vesicles (EVs) are nanovesicles containing a variety of biologically active molecules and participate in the intercellular communication. In the past decade, an increasing number of studies have reported that maternal-derived EVs play a crucial role in offspring growth, development, and immune system establishment. Hereby, we summarized the characteristics of EVs; biological functions of maternal-derived EVs during pregnancy, including implantation, decidualization, placentation, embryo development and birth of offspring; biological function of breast milk-derived EVs (BMEs) on infant oral and intestinal diseases, immune system, neurodevelopment, and metabolism. In summary, emerging studies have revealed that maternal-derived EVs play a pivotal role in offspring health. As such, maternal-derived EVs may be used as promising biomarkers in offspring disease diagnosis and treatment. However, existing research on maternal-derived EVs and offspring health is largely limited to animal and cellular studies. Evidence from human studies is needed.
Collapse
Affiliation(s)
- Rui Chen
- School of Public Health, Wuhan University, Wuhan, China
| | | | - Jie Dai
- School of Public Health, Wuhan University, Wuhan, China
| | - Minzhe Zhang
- School of Public Health, Wuhan University, Wuhan, China
| | - Gaolei Lu
- School of Public Health, Wuhan University, Wuhan, China
| | - Minjie Zhang
- School of Public Health, Wuhan University, Wuhan, China
| | - Hongjie Yu
- School of Public Health, Wuhan University, Wuhan, China
| | - Miaobing Zheng
- School of Nutrition and Exercise, Deakin University, Melbourne, Australia
| | - Qiqiang He
- School of Public Health, Wuhan University, Wuhan, China; Wuhan University Shenzhen Research Institute, Shenzhen, China; Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, Wuhan University, Wuhan, China.
| |
Collapse
|
9
|
Niu D, Zhang X, Zhang S, Fan T, Zhou X, Wang H, Zhang X, Nan F, Jiang S, Liu F, Wang Y, Wang B. Human Cytomegalovirus IE2 Disrupts Neural Progenitor Development and Induces Microcephaly in Transgenic Mouse. Mol Neurobiol 2023; 60:3883-3897. [PMID: 36991278 DOI: 10.1007/s12035-023-03310-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/25/2023] [Indexed: 03/31/2023]
Abstract
Human cytomegalovirus (HCMV) is a significant contributor to congenital birth defects. Limited by the lack of animal models, the pathogenesis of neurological damage in vivo caused by HCMV infection and the role of individual viral genes remain to be elucidated. Immediate early (IE2) protein may play a function in neurodevelopmental problems caused by HCMV infection. Here, this study intended to investigate IE2's long-term effects on development of the brain in IE2-expressing transgenic mice (Rosa26-LSL-IE2+/-, Camk2α-Cre) aimed to observe the phenotype of postnatal mice. The expression of IE2 in transgenic mice was confirmed by PCR and Western blot technology. We collected mouse brain tissue at 2, 4, 6, 8, and 10 days postpartum to analyze the developmental process of neural stem cells by immunofluorescence. We discovered that transgenic mice (Rosa26-LSL-IE2+/-, Camk2α-Cre) can reliably produce IE2 in the brain at various postpartum phases. Furthermore, we also observed the symptoms of microcephaly in postnatal transgenic mice, and IE2 can damage the amount of neural stem cells, prevent them from proliferating and differentiating, and activate microglia and astrocytes, creating an unbalanced environment in the brain's neurons. In conclusion, we demonstrate that long-term expression of HCMV-IE2 can cause microcephaly through molecular mechanisms affecting the differentiation and development of neural stem cells in vivo. This work establishes a theoretical and experimental foundation for elucidating the molecular mechanism of fetal microcephaly brought by HCMV infection in throughout the period of neural development of pregnancy.
Collapse
Affiliation(s)
- Delei Niu
- Department of Pathogenic Biology, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China
| | - Xianjuan Zhang
- Department of Pathogenic Biology, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China
| | - Shuyun Zhang
- Department of Pathogenic Biology, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China
| | - Tianyu Fan
- Department of Immunology, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China
| | - Xiaoqiong Zhou
- Department of Pathogenic Biology, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China
| | - Hui Wang
- Department of Special Medicine, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China
| | - Xueming Zhang
- Department of Pathogenic Biology, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China
| | - Fulong Nan
- Department of Special Medicine, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China
| | - Shasha Jiang
- Department of Pathogenic Biology, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China
| | - Fengjun Liu
- Department of Special Medicine, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China
| | - Yunyang Wang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China.
| | - Bin Wang
- Department of Pathogenic Biology, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China.
- Department of Special Medicine, College of Basic Medicine, Qingdao University, Qingdao, 266000, Shandong, China.
| |
Collapse
|
10
|
Daoutsali E, Pepers BA, Stamatakis S, van der Graaf LM, Terwindt GM, Parfitt DA, Buijsen RAM, van Roon-Mom WMC. Amyloid beta accumulations and enhanced neuronal differentiation in cerebral organoids of Dutch-type cerebral amyloid angiopathy patients. Front Aging Neurosci 2023; 14:1048584. [PMID: 36733499 PMCID: PMC9887998 DOI: 10.3389/fnagi.2022.1048584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 12/29/2022] [Indexed: 01/18/2023] Open
Abstract
Introduction ADutch-type cerebral amyloid angiopathy (D-CAA) is a hereditary brain disorder caused by a point mutation in the amyloid precursor protein (APP) gene. The mutation is located within the amyloid beta (Aβ) domain of APP and leads to Aβ peptide accumulation in and around the cerebral vasculature. There lack of disease models to study the cellular and molecular pathological mechanisms of D-CAA together with the absence of a disease phenotype in vitro in overexpression cell models, as well as the limited availability of D-CAA animal models indicates the need for a D-CAA patient-derived model. Methods We generated cerebral organoids from four D-CAA patients and four controls, cultured them up to 110 days and performed immunofluorescent and targeted gene expression analyses at two time points (D52 and D110). Results D-CAA cerebral organoids exhibited Aβ accumulations, showed enhanced neuronal and astrocytic gene expression and TGFβ pathway de-regulation. Conclusions These results illustrate the potential of cerebral organoids as in vitro disease model of D-CAA that can be used to understand disease mechanisms of D-CAA and can serve as therapeutic intervention platform for various Aβ-related disorders.
Collapse
Affiliation(s)
- Elena Daoutsali
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands,*Correspondence: Willeke M. C. van Roon-Mom, ; Elena Daoutsali,
| | - Barry A. Pepers
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Stavros Stamatakis
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | | | - Gisela M. Terwindt
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands
| | - David A. Parfitt
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Ronald A. M. Buijsen
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Willeke M. C. van Roon-Mom
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands,*Correspondence: Willeke M. C. van Roon-Mom, ; Elena Daoutsali,
| |
Collapse
|
11
|
Zhang ZW, Tu H, Jiang M, Vanan S, Chia SY, Jang SE, Saw WT, Ong ZW, Ma DR, Zhou ZD, Xu J, Guo KH, Yu WP, Ling SC, Margolin RA, Chain DG, Zeng L, Tan EK. The APP intracellular domain promotes LRRK2 expression to enable feed-forward neurodegenerative mechanisms in Parkinson's disease. Sci Signal 2022; 15:eabk3411. [PMID: 35998231 DOI: 10.1126/scisignal.abk3411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Gain-of-function mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are common in familial forms of Parkinson's disease (PD), which is characterized by progressive neurodegeneration that impairs motor and cognitive function. We previously demonstrated that LRRK2-mediated phosphorylation of β-amyloid precursor protein (APP) triggers the production and nuclear translocation of the APP intracellular domain (AICD). Here, we connected LRRK2 to AICD in a feed-forward cycle that enhanced LRRK2-mediated neurotoxicity. In cooperation with the transcription factor FOXO3a, AICD promoted LRRK2 expression, thus increasing the abundance of LRRK2 that promotes AICD activation. APP deficiency in LRRK2G2019S mice suppressed LRRK2 expression, LRRK2-mediated mitochondrial dysfunction, α-synuclein accumulation, and tyrosine hydroxylase (TH) loss in the brain, phenotypes associated with toxicity and loss of dopaminergic neurons in PD. Conversely, AICD overexpression increased LRRK2 expression and LRRK2-mediated neurotoxicity in LRRK2G2019S mice. In LRRK2G2019S mice or cultured dopaminergic neurons from LRRK2G2019S patients, treatment with itanapraced reduced LRRK2 expression and was neuroprotective. Itanapraced showed similar effects in a neurotoxin-induced PD mouse model, suggesting that inhibiting the AICD may also have therapeutic benefits in idiopathic PD. Our findings reveal a therapeutically targetable, feed-forward mechanism through which AICD promotes LRRK2-mediated neurotoxicity in PD.
Collapse
Affiliation(s)
- Zhi-Wei Zhang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore
| | - Haitao Tu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore
| | - Mei Jiang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore.,Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Sarivin Vanan
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore
| | - Sook Yoong Chia
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore
| | - Se-Eun Jang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore
| | - Wuan-Ting Saw
- Research Department, National Neuroscience Institute, Singapore General Hospital (SGH) Campus, Singapore 169856, Singapore
| | - Zhi-Wei Ong
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore
| | - Dong-Rui Ma
- Department of Neurology, Singapore General Hospital, Singapore 169609, Singapore
| | - Zhi-Dong Zhou
- Research Department, National Neuroscience Institute, Singapore General Hospital (SGH) Campus, Singapore 169856, Singapore.,Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore 169857, Singapore
| | - Jie Xu
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Kai-Hua Guo
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, Guangdong 510080, China
| | - Wei-Ping Yu
- Animal Gene Editing Laboratory, Biological Resource Center, A*STAR, Singapore 138673, Singapore.,Institute of Molecular and Cell Biology, A*STAR, Singapore 138673, Singapore
| | - Shuo-Chien Ling
- Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore 169857, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | | | | | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore.,Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore 169857, Singapore.,Centre for Molecular Neuropathology, Lee Kong Chian School of Medicine, Nanyang Technology University, Novena Campus, Singapore 308232, Singapore
| | - Eng-King Tan
- Research Department, National Neuroscience Institute, Singapore General Hospital (SGH) Campus, Singapore 169856, Singapore.,Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore 169857, Singapore.,Department of Neurology, National Neuroscience Institute, Singapore 308433, Singapore
| |
Collapse
|
12
|
Chia SY, Vipin A, Ng KP, Tu H, Bommakanti A, Wang BZ, Tan YJ, Zailan FZ, Ng ASL, Ling SC, Okamura K, Tan EK, Kandiah N, Zeng L. Upregulated Blood miR-150-5p in Alzheimer’s Disease Dementia Is Associated with Cognition, Cerebrospinal Fluid Amyloid-β, and Cerebral Atrophy. J Alzheimers Dis 2022; 88:1567-1584. [DOI: 10.3233/jad-220116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: There is an urgent need for noninvasive, cost-effective biomarkers for Alzheimer’s disease (AD), such as blood-based biomarkers. They will not only support the clinical diagnosis of dementia but also allow for timely pharmacological and nonpharmacological interventions and evaluations. Objective: To identify and validate a novel blood-based microRNA biomarker for dementia of the Alzheimer’s type (DAT). Methods: We conducted microRNA sequencing using peripheral blood mononuclear cells isolated from a discovery cohort and validated the identified miRNAs in an independent cohort and AD postmortem tissues. miRNA correlations with AD pathology and AD clinical-radiological imaging were conducted. We also performed bioinformatics and cell-based assay to identify miRNA target genes. Results: We found that miR-150-5p expression was significantly upregulated in DAT compared to mild cognitive impairment and healthy subjects. Upregulation of miR-150-5p was observed in AD hippocampus. We further found that higher miR-150-5p levels were correlated with the clinical measures of DAT, including lower global cognitive scores, lower CSF Aβ 42, and higher CSF total tau. Interestingly, we observed that higher miR-150-5p levels were associated with MRI brain volumes within the default mode and executive control networks, two key networks implicated in AD. Furthermore, pathway analysis identified the targets of miR-150-5p to be enriched in the Wnt signaling pathway, including programmed cell death 4 (PDCD4). We found that PDCD4 was downregulated in DAT blood and was downregulated by miR-150-5p at both the transcriptional and protein levels Conclusion: Our findings demonstrated that miR-150-5p is a promising clinical blood-based biomarker for DAT
Collapse
Affiliation(s)
- Sook-Yoong Chia
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Ashwati Vipin
- Department of Neurology, National Neuroscience Institute, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technology University, Novena Campus, Singapore
| | - Kok Pin Ng
- Department of Neurology, National Neuroscience Institute, Singapore
- Duke-NUS Medical School, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technology University, Novena Campus, Singapore
| | - Haitao Tu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Ananth Bommakanti
- Temasek Life Sciences Laboratory, 1 Research Link National University of Singapore, Singapore
| | | | - Yi Jayne Tan
- Department of Neurology, National Neuroscience Institute, Singapore
| | - Fatin Zahra Zailan
- Department of Neurology, National Neuroscience Institute, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technology University, Novena Campus, Singapore
| | - Adeline Su-Lyn Ng
- Department of Neurology, National Neuroscience Institute, Singapore
- Duke-NUS Medical School, Singapore
| | - Shuo-Chian Ling
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Neuroscience & Behavioral Disorders Program, Duke-NUS Medical School, Singapore
| | - Katsutomo Okamura
- Temasek Life Sciences Laboratory, 1 Research Link National University of Singapore, Singapore
- Nara Institute of Science and Technology, Takayama, Ikoma, Nara, Japan
| | - Eng-King Tan
- Neuroscience & Behavioral Disorders Program, Duke-NUS Medical School, Singapore
- Research Department, National Neuroscience Institute, Singapore General Hospital Campus, Singapore
| | - Nagaendran Kandiah
- Department of Neurology, National Neuroscience Institute, Singapore
- Duke-NUS Medical School, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technology University, Novena Campus, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
- Neuroscience & Behavioral Disorders Program, Duke-NUS Medical School, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technology University, Novena Campus, Singapore
| |
Collapse
|
13
|
Subbaiah KCV, Wu J, Tang WHW, Yao P. FAM114A1 influences cardiac pathological remodeling by regulating angiotensin II signaling. JCI Insight 2022; 7:152783. [PMID: 35671117 PMCID: PMC9310534 DOI: 10.1172/jci.insight.152783] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 06/01/2022] [Indexed: 11/30/2022] Open
Abstract
Cardiac pathological remodeling, a primary contributor to heart failure (HF) and death, is an important target for HF therapy. However, the signaling pathways that govern cardiac remodeling are not fully elucidated. Here, we found that a functionally unannotated human myocardial infarction–associated (MI-associated) gene, family with sequence similarity 114 member A1 (FAM114A1), is induced in failing human and mouse hearts compared with nonfailing hearts. Homozygous KO of Fam114a1 (Fam114a1–/–) in the mouse genome reduces cardiomyocyte hypertrophy, inflammation, and cardiac fibrosis while restoring cardiac function in angiotensin II–induced (Ang II–induced) and MI-induced HF mouse models. Cardiac fibroblasts (CFs) exhibit the highest FAM114A1 expression among different cardiac cell types. FAM114A1 is a critical autonomous factor for CF proliferation, activation, and migration. Mechanistically, FAM114A1 interacts with angiotensin receptor–associated protein (AGTRAP) and regulates the expression of angiotensin type 1 receptor (AT1R) and downstream Ang II signaling transduction, and it subsequently influences profibrotic response. Our results indicate that FAM114A1 regulates Ang II signaling, thereby activating CFs and other cardiac cells and augmenting pathological cardiac remodeling. These findings provide potentially novel insights into the regulation of cardiac remodeling and identify FAM114A1 as a therapeutic target for the treatment of heart disease.
Collapse
Affiliation(s)
- Kadiam C Venkata Subbaiah
- Department of Medicine, University of Rochester School of Medicine, Rochester, United States of America
| | - Jiangbin Wu
- Department of Medicine, University of Rochester School of Medicine, Rochester, United States of America
| | - Wai Hong Wilson Tang
- Department of Cardiovascular Medicine, Cleveland Clinic Foundation, Cleveland, United States of America
| | - Peng Yao
- Department of Medicine, University of Rochester School of Medicine, Rochester, United States of America
| |
Collapse
|
14
|
Lim J, Kim S, Oh SJ, Han SM, Moon SY, Kang B, Seo SB, Jang S, Son SU, Jung J, Kang T, Park SA, Moon M, Lim EK. miRNA sensing hydrogels capable of self-signal amplification for early diagnosis of Alzheimer's disease. Biosens Bioelectron 2022; 209:114279. [PMID: 35447599 DOI: 10.1016/j.bios.2022.114279] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/21/2022] [Accepted: 04/09/2022] [Indexed: 11/02/2022]
Abstract
Alzheimer's disease (AD), one of the leading senile disorders in the world, causes severe memory loss and cognitive impairment. To date, there is no clear cure for AD. However, early diagnosis and monitoring can help mitigate the effects of this disease. In this study, we reported a platform for diagnosing early-stage AD using microRNAs (miRNAs) in the blood as biomarkers. First, we selected an appropriate target miRNA (miR-574-5p) using AD model mice (4-month-old 5XFAD mice) and developed a hydrogel-based sensor that enabled high-sensitivity detection of the target miRNA. This hydrogel contained catalytic hairpin assembly (CHA) reaction-based probes, leading to fluorescence signal amplification without enzymes and temperature changes, at room temperature. This sensor exhibited high sensitivity and selectivity, as evidenced by its picomolar-level detection limit (limit of detection: 1.29 pM). Additionally, this sensor was evaluated using the plasma of AD patients and non-AD control to validate its clinical applicability. Finally, to use this sensor as a point-of-care-testing (POCT) diagnostic system, a portable fluorometer was developed and verified for feasibility of application.
Collapse
Affiliation(s)
- Jaewoo Lim
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea; Department of Nanobiotechnology, KRIBB School of Biotechnology, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34113, South Korea
| | - Sujin Kim
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, South Korea
| | - Seung Jae Oh
- YUHS-KRIBB Medical Convergence Research Institute, College of Medicine, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul, 03722, South Korea
| | - Song Mi Han
- Lab for Neurodegenerative Dementia, Department of Anatomy, Ajou University School of Medicine, 164 World Cup-ro, Yeongtong-gu, Suwon, 16499, South Korea; Department of Neurology, Ajou University School of Medicine, 164 World Cup-ro, Yeongtong-gu, Suwon, 16499, South Korea; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, 164 World Cup-ro, Yeongtong-gu, Suwon, 16499, South Korea
| | - So Young Moon
- Department of Neurology, Ajou University School of Medicine, 164 World Cup-ro, Yeongtong-gu, Suwon, 16499, South Korea
| | - Byunghoon Kang
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Seung Beom Seo
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea; Department of Cogno-Mechatronics Engineering, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan, 46241, South Korea
| | - Soojin Jang
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea; Department of Nanobiotechnology, KRIBB School of Biotechnology, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34113, South Korea
| | - Seong Uk Son
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea; Department of Nanobiotechnology, KRIBB School of Biotechnology, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34113, South Korea
| | - Juyeon Jung
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea; Department of Nanobiotechnology, KRIBB School of Biotechnology, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34113, South Korea
| | - Taejoon Kang
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea
| | - Sun Ah Park
- Lab for Neurodegenerative Dementia, Department of Anatomy, Ajou University School of Medicine, 164 World Cup-ro, Yeongtong-gu, Suwon, 16499, South Korea; Department of Neurology, Ajou University School of Medicine, 164 World Cup-ro, Yeongtong-gu, Suwon, 16499, South Korea; Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of Medicine, 164 World Cup-ro, Yeongtong-gu, Suwon, 16499, South Korea
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, 158, Gwanjeodong-ro, Seo-gu, Daejeon, 35365, South Korea.
| | - Eun-Kyung Lim
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, South Korea; Department of Nanobiotechnology, KRIBB School of Biotechnology, University of Science and Technology, 125 Gwahak-ro, Yuseong-gu, Daejeon, 34113, South Korea.
| |
Collapse
|
15
|
Genes Associated with Disturbed Cerebral Neurogenesis in the Embryonic Brain of Mouse Models of Down Syndrome. Genes (Basel) 2021; 12:genes12101598. [PMID: 34680993 PMCID: PMC8535956 DOI: 10.3390/genes12101598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/05/2021] [Accepted: 10/07/2021] [Indexed: 02/06/2023] Open
Abstract
Down syndrome (DS), also known as trisomy 21, is the most frequent genetic cause of intellectual disability. Although the mechanism remains unknown, delayed brain development is assumed to be involved in DS intellectual disability. Analyses with human with DS and mouse models have shown that defects in embryonic cortical neurogenesis may lead to delayed brain development. Cre-loxP-mediated chromosomal engineering has allowed the generation of a variety of mouse models carrying various partial Mmu16 segments. These mouse models are useful for determining genotype–phenotype correlations and identifying dosage-sensitive genes involved in the impaired neurogenesis. In this review, we summarize several candidate genes and pathways that have been linked to defective cortical neurogenesis in DS.
Collapse
|
16
|
Donzelli J, Proestler E, Riedel A, Nevermann S, Hertel B, Guenther A, Gattenlöhner S, Savai R, Larsson K, Saul MJ. Small extracellular vesicle-derived miR-574-5p regulates PGE2-biosynthesis via TLR7/8 in lung cancer. J Extracell Vesicles 2021; 10:e12143. [PMID: 34596365 PMCID: PMC8485338 DOI: 10.1002/jev2.12143] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 07/20/2021] [Accepted: 08/19/2021] [Indexed: 12/30/2022] Open
Abstract
Intercellular communication plays an essential role in lung cancer (LC). One of the major players in cell-cell-communication is small extracellular vesicles (sEV). SEV trigger various biological responses by transporting cellular cargo to target cells. One essential sEV component are microRNAs (miRs), whose transport has recently attracted increasing research interest. We report that prostaglandin E2 (PGE2 ), a key inflammatory lipid mediator, specifically induces the sorting of miR-574-5p in sEV of A549 and 2106T cells. We found that sEV-derived miR-574-5p activates Toll-like receptors (TLR) 7/8, thereby decreasing PGE2 -levels. In contrast, intracellular miR-574-5p induces PGE2 -biosynthesis. Consequently, the combination of intracellular and sEV-derived miR-574-5p controls PGE2 -levels via a feedback loop. This was only observed in adeno- but not in squamous cell carcinoma, indicating a cell-specific response to sEV-derived miRs, which might be due to unique tetraspanin compositions. Hence, we describe a novel function of miR-574-5p unique to adenocarcinoma. Intracellular miR-574-5p induces PGE2 and thus the secretion of sEV-derived miR-574-5p, which in turn decreases PGE2 -biosynthesis in recipient cells.
Collapse
Affiliation(s)
- Julia Donzelli
- Department of BiologyTechnische Universität DarmstadtDarmstadtGermany
| | - Eva Proestler
- Department of BiologyTechnische Universität DarmstadtDarmstadtGermany
| | - Anna Riedel
- Department of BiologyTechnische Universität DarmstadtDarmstadtGermany
| | - Sheila Nevermann
- Department of BiologyTechnische Universität DarmstadtDarmstadtGermany
| | - Brigitte Hertel
- Department of BiologyTechnische Universität DarmstadtDarmstadtGermany
| | - Andreas Guenther
- Department of Internal MedicineMember of the German Centre for Lung Research (DZL)Member of Cardio‐Pulmonary Institute (CPI)Justus Liebig UniversityGiessenGermany
| | | | - Rajkumar Savai
- Department of Internal MedicineMember of the German Centre for Lung Research (DZL)Member of Cardio‐Pulmonary Institute (CPI)Justus Liebig UniversityGiessenGermany
- Department of Lung Development and RemodellingMember of the DZLMember of CPIMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
- Lung Microenvironmental Niche in CancerogenesisInstitute for Lung Health (ILH)Justus Liebig UniversityGiessenGermany
| | - Karin Larsson
- Rheumatology UnitDepartment of MedicineKarolinska University HospitalStockholmSweden
| | - Meike J. Saul
- Department of BiologyTechnische Universität DarmstadtDarmstadtGermany
| |
Collapse
|
17
|
Hu Y, Zhang Y, Ren R, Dammer EB, Xie X, Chen S, Huang Q, Huang W, Zhang R, Chen H, Wang H, Wang G. microRNA-425 loss mediates amyloid plaque microenvironment heterogeneity and promotes neurodegenerative pathologies. Aging Cell 2021; 20:e13454. [PMID: 34510683 PMCID: PMC8520725 DOI: 10.1111/acel.13454] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 04/19/2021] [Accepted: 08/05/2021] [Indexed: 01/09/2023] Open
Abstract
Different cellular and molecular changes underlie the pathogenesis of Alzheimer's disease (AD). Among these, neuron‐specific dysregulation is a necessary event for accumulation of classic pathologies including amyloid plaques. Here, we show that AD‐associated pathophysiology including neuronal cell death, inflammatory signaling, and endolysosomal dysfunction is spatially colocalized to amyloid plaques in regions with abnormal microRNA‐425 (miR‐425) levels and this change leads to focal brain microenvironment heterogeneity, that is, an amyloid plaque‐associated microenvironment (APAM). APAM consists of multiple specific neurodegenerative signature pathologies associated with senile plaques that contribute to the heterogeneity and complexity of AD. Remarkably, miR‐425, a neuronal‐specific regulator decreased in AD brain, maintains a normal spatial transcriptome within brain neurons. We tested the hypothesis that miR‐425 loss correlates with enhanced levels of mRNA targets downstream, supporting APAM and AD progression. A miR‐425‐deficient mouse model has enhanced APP amyloidogenic processing, neuroinflammation, neuron loss, and cognitive impairment. In the APP/PS1 mouse model, intervening with miR‐425 supplementation ameliorated APAM changes and memory deficits. This study reveals a novel mechanism of dysregulation of spatial transcriptomic changes in AD brain, identifying a probable neuronal‐specific microRNA regulator capable of staving off amyloid pathogenesis. Moreover, our findings provide new insights for developing AD treatment strategies with miRNA oligonucleotide(s).
Collapse
Affiliation(s)
- Yong‐Bo Hu
- Department of Neurology and Neuroscience Institute Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
- Department of Pharmacology and Chemical Biology Shanghai Jiao Tong University School of Medicine Shanghai China
- Department of Neurology,Shanghai East Hospital School of Medicine,Tongji University Shanghai China
| | - Yong‐Fang Zhang
- Department of Pharmacology and Chemical Biology Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Ru‐Jing Ren
- Department of Neurology and Neuroscience Institute Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Eric B. Dammer
- Department of Biochemistry and Center for Neurodegenerative Disease Emory University School of Medicine Atlanta Georgia USA
| | - Xin‐Yi Xie
- Department of Neurology and Neuroscience Institute Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Shi‐Wu Chen
- Department of Neurology and Neuroscience Institute Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Qiang Huang
- Department of Neurology and Neuroscience Institute Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Wan‐Ying Huang
- Department of Pharmacology and Chemical Biology Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Rui Zhang
- Department of Pharmacology and Chemical Biology Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Hong‐Zhuan Chen
- Institute of Interdisciplinary Science Shuguang Hospital Shanghai University of Traditional Chinese Medicine Shanghai China
| | - Hao Wang
- Department of Pharmacology and Chemical Biology Shanghai Jiao Tong University School of Medicine Shanghai China
| | - Gang Wang
- Department of Neurology and Neuroscience Institute Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai China
| |
Collapse
|
18
|
Leroux C, Chervet ML, German JB. Perspective: Milk microRNAs as Important Players in Infant Physiology and Development. Adv Nutr 2021; 12:1625-1635. [PMID: 34022770 PMCID: PMC8483967 DOI: 10.1093/advances/nmab059] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/08/2021] [Accepted: 04/14/2021] [Indexed: 12/16/2022] Open
Abstract
Evolutionary selective pressure on lactation has resulted in milk that provides far more than simply essential nutrients, delivering a complex repertoire of agents from hormones to intact cells. Human infants are born with low barrier integrity of their gut, which means that many of the complex biopolymer components of milk enter and circulate in lymph and blood, reaching organs throughout the body. Due to this state of gut maturation, all components of milk are potentially part of the crosstalk between mother and infants. This article highlights the functions of milk's complex biopolymers, more specifically the potential role of microRNAs (miRNAs) contained in extracellular vesicles in human milk. miRNAs are key effectors in the regulation of many biological processes during early-age development, and consequently milk-sourced miRNAs must be considered to provide unique biological assets to the infant during breastfeeding. This article interprets the evidence of the potential action of human milk miRNAs on infant development, taking into account their abundance in milk based on the literature and current knowledge. Human milk miRNAs appear to influence lipid and glucose metabolism, gut maturation, neurogenesis, and immunity. We also show growing evidence that human milk miRNAs are epigenetic modulators that play a pivotal role in the regulation of tissue-specific gene expression throughout life. Furthermore, this article addresses the ongoing debate regarding the potential influence of human milk miRNAs on viral infection as a new research area. This article highlights that these bioactive molecules are now being incorporated into our overall understanding of nutrient needs for healthy infant development, preparing each individual infant to succeed as a healthy and protected adult throughout its life. In essence, miRNAs are a new language in the Rosetta stone of health that is mammalian lactation.
Collapse
Affiliation(s)
| | - Mathilde Lea Chervet
- Foods for Health Institute, Department of Food Science and Technology, University of California, Davis, Davis, CA, USA
| | - J Bruce German
- Foods for Health Institute, Department of Food Science and Technology, University of California, Davis, Davis, CA, USA
| |
Collapse
|
19
|
Kinoshita C, Kubota N, Aoyama K. Interplay of RNA-Binding Proteins and microRNAs in Neurodegenerative Diseases. Int J Mol Sci 2021; 22:ijms22105292. [PMID: 34069857 PMCID: PMC8157344 DOI: 10.3390/ijms22105292] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/14/2021] [Accepted: 05/15/2021] [Indexed: 02/07/2023] Open
Abstract
The number of patients with neurodegenerative diseases (NDs) is increasing, along with the growing number of older adults. This escalation threatens to create a medical and social crisis. NDs include a large spectrum of heterogeneous and multifactorial pathologies, such as amyotrophic lateral sclerosis, frontotemporal dementia, Alzheimer’s disease, Parkinson’s disease, Huntington’s disease and multiple system atrophy, and the formation of inclusion bodies resulting from protein misfolding and aggregation is a hallmark of these disorders. The proteinaceous components of the pathological inclusions include several RNA-binding proteins (RBPs), which play important roles in splicing, stability, transcription and translation. In addition, RBPs were shown to play a critical role in regulating miRNA biogenesis and metabolism. The dysfunction of both RBPs and miRNAs is often observed in several NDs. Thus, the data about the interplay among RBPs and miRNAs and their cooperation in brain functions would be important to know for better understanding NDs and the development of effective therapeutics. In this review, we focused on the connection between miRNAs, RBPs and neurodegenerative diseases.
Collapse
Affiliation(s)
- Chisato Kinoshita
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan;
- Correspondence: (C.K.); (K.A.); Tel.: +81-3-3964-3794 (C.K.); +81-3-3964-3793 (K.A.)
| | - Noriko Kubota
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan;
- Teikyo University Support Center for Women Physicians and Researchers, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan
| | - Koji Aoyama
- Department of Pharmacology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi, Tokyo 173-8605, Japan;
- Correspondence: (C.K.); (K.A.); Tel.: +81-3-3964-3794 (C.K.); +81-3-3964-3793 (K.A.)
| |
Collapse
|
20
|
Liu J, Sala MA, Kim J. Dampening the Fire: A Negative Feedback Loop in Acute Respiratory Distress Syndrome. Am J Respir Cell Mol Biol 2021; 64:158-160. [PMID: 33522886 PMCID: PMC7874398 DOI: 10.1165/rcmb.2020-0487ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Jing Liu
- Department of Surgery, College of Medicine, Cancer Center, University of Illinois at Chicago, Chicago, Illinois
| | - Marc A Sala
- Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, and
| | - Jiyeon Kim
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois
| |
Collapse
|
21
|
Wu J, Venkata Subbaiah KC, Jiang F, Hedaya O, Mohan A, Yang T, Welle K, Ghaemmaghami S, Tang WHW, Small E, Yan C, Yao P. MicroRNA-574 regulates FAM210A expression and influences pathological cardiac remodeling. EMBO Mol Med 2021; 13:e12710. [PMID: 33369227 PMCID: PMC7863409 DOI: 10.15252/emmm.202012710] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/23/2022] Open
Abstract
Aberrant expression of mitochondrial proteins impairs cardiac function and causes heart disease. The mechanism of regulation of mitochondria encoded protein expression during cardiac disease, however, remains underexplored. Here, we show that multiple pathogenic cardiac stressors induce the expression of miR-574 guide and passenger strands (miR-574-5p/3p) in both humans and mice. miR-574 knockout mice exhibit severe cardiac disorder under different pathogenic cardiac stresses while miR-574-5p/3p mimics that are delivered systematically using nanoparticles reduce cardiac pathogenesis under disease insults. Transcriptomic analysis of miR-574-null hearts uncovers family with sequence similarity 210 member A (FAM210A) as a common target mRNA of miR-574-5p and miR-574-3p. The interactome capture analysis suggests that FAM210A interacts with mitochondrial translation elongation factor EF-Tu. Manipulating miR-574-5p/3p or FAM210A expression changes the protein expression of mitochondrial-encoded electron transport chain (ETC) genes but not nuclear-encoded mitochondrial ETC genes in both human AC16 cardiomyocyte cells and miR-574-null murine hearts. Together, we discovered that miR-574 regulates FAM210A expression and modulates mitochondrial-encoded protein expression, which may influence cardiac remodeling in heart failure.
Collapse
Affiliation(s)
- Jiangbin Wu
- Department of MedicineAab Cardiovascular Research InstituteUniversity of Rochester School of Medicine & DentistryRochester, New YorkNYUSA
| | - Kadiam C Venkata Subbaiah
- Department of MedicineAab Cardiovascular Research InstituteUniversity of Rochester School of Medicine & DentistryRochester, New YorkNYUSA
| | - Feng Jiang
- Department of MedicineAab Cardiovascular Research InstituteUniversity of Rochester School of Medicine & DentistryRochester, New YorkNYUSA
- Department of Biochemistry & BiophysicsUniversity of Rochester School of Medicine & DentistryRochester, New YorkNYUSA
| | - Omar Hedaya
- Department of MedicineAab Cardiovascular Research InstituteUniversity of Rochester School of Medicine & DentistryRochester, New YorkNYUSA
- Department of Biochemistry & BiophysicsUniversity of Rochester School of Medicine & DentistryRochester, New YorkNYUSA
| | - Amy Mohan
- Department of MedicineAab Cardiovascular Research InstituteUniversity of Rochester School of Medicine & DentistryRochester, New YorkNYUSA
| | - Tingting Yang
- Department of OphthalmologyColumbia UniversityNew YorkNYUSA
| | - Kevin Welle
- Mass Spectrometry Resource LabUniversity of Rochester School of Medicine & DentistryRochester, New YorkNYUSA
| | - Sina Ghaemmaghami
- Mass Spectrometry Resource LabUniversity of Rochester School of Medicine & DentistryRochester, New YorkNYUSA
| | | | - Eric Small
- Department of MedicineAab Cardiovascular Research InstituteUniversity of Rochester School of Medicine & DentistryRochester, New YorkNYUSA
| | - Chen Yan
- Department of MedicineAab Cardiovascular Research InstituteUniversity of Rochester School of Medicine & DentistryRochester, New YorkNYUSA
| | - Peng Yao
- Department of MedicineAab Cardiovascular Research InstituteUniversity of Rochester School of Medicine & DentistryRochester, New YorkNYUSA
- Department of Biochemistry & BiophysicsUniversity of Rochester School of Medicine & DentistryRochester, New YorkNYUSA
- The Center for RNA BiologyUniversity of Rochester School of Medicine & DentistryRochester, New YorkNYUSA
- The Center for Biomedical InformaticsUniversity of Rochester School of Medicine & DentistryRochester, New YorkNYUSA
| |
Collapse
|
22
|
Liu L, Chen X, Chen YH, Zhang K. Identification of Circular RNA hsa_Circ_0003391 in Peripheral Blood Is Potentially Associated With Alzheimer's Disease. Front Aging Neurosci 2020; 12:601965. [PMID: 33424579 PMCID: PMC7793744 DOI: 10.3389/fnagi.2020.601965] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/01/2020] [Indexed: 12/28/2022] Open
Abstract
Circular RNAs (circRNAs) have recently been discovered as a novel type of endogenous non-coding RNA that may regulate gene expression in mammals. In the central nervous system (CNS), circRNAs are relevant to many neurological disorders such as Alzheimer's disease (AD). In this study, we attempted to identify an aberrant circRNA, hsa_circ_0003391, which is significantly downregulated in the peripheral blood of patients with AD, and to explore the relationship between hsa_circ_0003391 and the clinical manifestation of AD. The expression of hsa_circ_0003391 had a specific decrease in the peripheral blood of patients with AD compared to those with other types of dementia. To evaluate the potential diagnostic value of the circRNA, we performed a receiver operating characteristic (ROC) curve analysis. The area under the curve (AUC) value was 0.7283 for hsa_circ_0003391, which was statistically significant. The natural form of hsa_circ_0003391 in the peripheral blood was a loop structure with good stability. We found a potential correlation between the expression of hsa_circ_0003391 and the clinical manifestations of AD. Bioinformatic analysis was carried out to predict the latent target microRNAs (miRNA) of hsa_circ_0003391. Furthermore, microRNAs targeted by hsa_circ_0003391 were successfully detected, and miR-574-5p had an expected elevation in the AD groups, suggesting that miR-574-5p might be a potential microRNA target for hsa_circ_0003391. Our results suggest that the downregulation of hsa_circ_0003391 in the peripheral blood has a potential relationship with AD. Our findings not only provide an important latent biomarker but also highlight an important perspective for the following study into AD pathogenesis. This may promote the process of novel therapeutics targeting non-coding RNA.
Collapse
Affiliation(s)
- Li Liu
- Department of Neurology, Shenyang Fifth People Hospital, Shenyang, China.,Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Xi Chen
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Yu-Hua Chen
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| | - Ke Zhang
- Department of Developmental Cell Biology, Key Laboratory of Cell Biology, Ministry of Public Health, Key Laboratory of Medical Cell Biology, Ministry of Education, China Medical University, Shenyang, China
| |
Collapse
|
23
|
Disouky A, Lazarov O. Adult hippocampal neurogenesis in Alzheimer's disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 177:137-156. [PMID: 33453939 DOI: 10.1016/bs.pmbts.2020.09.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
New neurons are generated in the dentate gyrus of the adult brain throughout life. They incorporate in the granular cell layer of the dentate gyrus and integrate in the hippocampal circuitry. Increasing evidence suggests that new neurons play a role in learning and memory. In turn, a large body of evidence suggests that neurogenesis is impaired in Alzheimer's disease, contributing to memory deficits characterizing the disease. We outline here current knowledge about the biology of adult hippocampal neurogenesis and its function in learning and memory. In addition, we discuss evidence that neurogenesis is dysfunctional in Alzheimer's disease, address the controversy in the literature concerning the persistence of hippocampal neurogenesis in the adult and aging human brain, and evaluate the therapeutic potential of neurogenesis-based drug development for the treatment of cognitive deficits in Alzheimer's disease.
Collapse
Affiliation(s)
- Ahmed Disouky
- Departments of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Orly Lazarov
- Departments of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States.
| |
Collapse
|
24
|
Jiang M, Vanan S, Tu HT, Zhang W, Zhang ZW, Chia SY, Jang SE, Zeng XX, Yu WP, Xu J, Guo KH, Zeng L. Amyloid precursor protein intracellular domain-dependent regulation of FOXO3a inhibits adult hippocampal neurogenesis. Neurobiol Aging 2020; 95:250-263. [PMID: 32866886 DOI: 10.1016/j.neurobiolaging.2020.07.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 07/14/2020] [Accepted: 07/29/2020] [Indexed: 10/23/2022]
Abstract
The amyloid precursor protein (APP) intracellular domain (AICD) is a metabolic by-product of APP produced through sequential proteolytic cleavage by α-, β-, and γ-secretases. The interaction between AICD and Fe65 has been reported to impair adult neurogenesis in vivo. However, the exact role of AICD in mediating neural stem cell fate remains unclear. To identify the role of AICD in neuronal proliferation and differentiation, as well as to clarify the molecular mechanisms underlying the role of AICD in neurogenesis, we first generated a mouse model expressing the Rosa26-based AICD transgene. AICD overexpression did not alter the spatiotemporal expression pattern of full-length APP or accumulation of its metabolites. In addition, AICD decreased the newly generated neural progenitor cell (NPC) pool, inhibited the proliferation and differentiation efficiency of NPCs, and increased cell death both in vitro and in vivo. Given that abnormal neurogenesis is often associated with depression-like behavior in adult mice, we conducted a forced swim test and tail suspension test with AICD mice and found a depression-like behavioral phenotype in AICD transgenic mice. Moreover, AICD stimulated FOXO3a transcriptional activation, which in turn negatively regulated AICD. In addition, functional loss of FOXO3a in NPCs derived from the hippocampal dentate gyrus of adult AICD transgenic mice rescued neurogenesis defects. AICD also increased the mRNA expression of FOXO3a target genes related to neurogenesis and cell death. These results suggest that FOXO3a is the functional target of AICD in neurogenesis regulation. Our study reveals the role of AICD in mediating neural stem cell fate to maintain homeostasis during brain development via interaction with FOXO3a.
Collapse
Affiliation(s)
- Mei Jiang
- Department of Neurobiology and Anatomy, Sun Yat-Sen University Zhongshan School of Medicine, Guangzhou, PR China; Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Sarivin Vanan
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Hai-Tao Tu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Wei Zhang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Zhi-Wei Zhang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Sook-Yoong Chia
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Se Eun Jang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Xiao-Xia Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Wei-Ping Yu
- Animal Gene Editing Laboratory, Biological resource Centre, A∗STAR, Singapore; Institute of Molecular and Cell Biology, A∗STAR, Proteos, Singapore
| | - Jie Xu
- Department of Neurobiology and Anatomy, Sun Yat-Sen University Zhongshan School of Medicine, Guangzhou, PR China.
| | - Kai-Hua Guo
- Department of Neurobiology and Anatomy, Sun Yat-Sen University Zhongshan School of Medicine, Guangzhou, PR China.
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore; Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore; Lee Kong Chian School of Medicine, Nanyang Technology University, Novena Campus, Singapore.
| |
Collapse
|
25
|
Ono C, Fukuhara T, Li S, Wang J, Sato A, Izumi T, Fauzyah Y, Yamamoto T, Morioka Y, Dokholyan NV, Standley DM, Matsuura Y. Various miRNAs compensate the role of miR-122 on HCV replication. PLoS Pathog 2020; 16:e1008308. [PMID: 32574204 PMCID: PMC7337399 DOI: 10.1371/journal.ppat.1008308] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 07/06/2020] [Accepted: 05/18/2020] [Indexed: 02/07/2023] Open
Abstract
One of the determinants for tissue tropism of hepatitis C virus (HCV) is miR-122, a liver-specific microRNA. Recently, it has been reported that interaction of miR-122 to HCV RNA induces a conformational change of the 5'UTR internal ribosome entry site (IRES) structure to form stem-loop II structure (SLII) and hijack of translating 80S ribosome through the binding of SLIII to 40S subunit, which leads to efficient translation. On the other hand, low levels of HCV-RNA replication have also been detected in some non-hepatic cells; however, the details of extrahepatic replication remain unknown. These observations suggest the possibility that miRNAs other than miR-122 can support efficient replication of HCV-RNA in non-hepatic cells. Here, we identified a number of such miRNAs and show that they could be divided into two groups: those that bind HCV-RNA at two locations (miR-122 binding sites I and II), in a manner similar to miR-122 (miR-122-like), and those that target a single site that bridges sites I and II and masking both G28 and C29 in the 5'UTR (non-miR-122-like). Although the enhancing activity of these non-hepatic miRNAs were lower than those of miR-122, substantial expression was detected in various normal tissues. Furthermore, structural modeling indicated that both miR-122-like and non-miR-122-like miRNAs not only can facilitate the formation of an HCV IRES SLII but also can stabilize IRES 3D structure in order to facilitate binding of SLIII to the ribosome. Together, these results suggest that HCV facilitates miR-122-independent replication in non-hepatic cells through recruitment of miRNAs other than miR-122. And our findings can provide a more detailed mechanism of miR-122-dependent enhancement of HCV-RNA translation by focusing on IRES tertiary structure.
Collapse
Affiliation(s)
- Chikako Ono
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Takasuke Fukuhara
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Songling Li
- Department of Genome Informatics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Jian Wang
- Department of Pharmacology, Penn State University College of Medicine, Hershey, Pennsylvania, United States of America
| | - Asuka Sato
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Takuma Izumi
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yuzy Fauzyah
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Takuya Yamamoto
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yuhei Morioka
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Nikolay V. Dokholyan
- Department of Pharmacology, Penn State University College of Medicine, Hershey, Pennsylvania, United States of America
- Department of Biochemistry & Molecular Biology, Penn State University College of Medicine, Hershey, Pennsylvania, United States of America
| | - Daron M. Standley
- Department of Genome Informatics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yoshiharu Matsuura
- Department of Molecular Virology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| |
Collapse
|
26
|
Jeon SG, Yoo A, Chun DW, Hong SB, Chung H, Kim JI, Moon M. The Critical Role of Nurr1 as a Mediator and Therapeutic Target in Alzheimer's Disease-related Pathogenesis. Aging Dis 2020; 11:705-724. [PMID: 32489714 PMCID: PMC7220289 DOI: 10.14336/ad.2019.0718] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/18/2019] [Indexed: 01/16/2023] Open
Abstract
Several studies have revealed that the transcription factor nuclear receptor related 1 (Nurr1) plays several roles not only in the regulation of gene expression related to dopamine synthesis, but also in alternative splicing, and miRNA targeting. Moreover, it regulates cognitive functions and protects against inflammation-induced neuronal death. In particular, the role of Nurr1 in the pathogenesis of Parkinson's disease (PD) has been well investigated; for example, it has been shown that it restores behavioral and histological impairments in PD models. Although many studies have evaluated the connection between Nurr1 and PD pathogenesis, the role of Nurr1 in Alzheimer's disease (AD) remain to be studied. There have been several studies describing Nurr1 protein expression in the AD brain. However, only a few studies have examined the role of Nurr1 in the context of AD. Therefore, in this review, we highlight the overall effects of Nurr1 under the neuropathologic conditions related to AD. Furthermore, we suggest the possibility of using Nurr1 as a therapeutic target for AD or other neurodegenerative disorders.
Collapse
Affiliation(s)
- Seong Gak Jeon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Anji Yoo
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Dong Wook Chun
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Sang Bum Hong
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Hyunju Chung
- Department of Core Research Laboratory, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea
| | - Jin-il Kim
- Department of Nursing, College of Nursing, Jeju National University, Jeju-si 63243, Republic of Korea
| | - Minho Moon
- Department of Biochemistry, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| |
Collapse
|
27
|
MicroRNA-574-5p in gastric cancer cells promotes angiogenesis by targeting protein tyrosine phosphatase non-receptor type 3 (PTPN3). Gene 2020; 733:144383. [PMID: 31972307 DOI: 10.1016/j.gene.2020.144383] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/28/2022]
Abstract
We elucidate in this study that up-regulation of miR-574-5p in gastric cancer cells under hypoxic conditions contributed to angiogenesis. We found that miR-574-5p and HIF-1α were up-regulated in gastric cancer cells cultured under 2% O2 or in medium containing CoCl2, and in muscle tissues of mice injected with NaNO2, indicating up-regulation of miR-574-5p in vitro or in vivo in response to hypoxic conditions. We hypothesized that up-regulation of miR-574-5p could promote angiogenesis. Transfection of gastric cancer cells with miR-574-5p mimics or inhibitor resulted in increase or decrease in the expression of VEGFA. Viability, migration, invasion and tube formation of HUVECs cultured with conditioned medium from SGC/574 cells transfected with miR-574-5p inhibitor were reduced. Tube formation of HUVECs cultured with conditioned medium from SGC-7901 cells transfected with miR-574-5p mimics was increased. An in vivo study demonstrated that inhibition of miR-574-5p in the tumor xenografts of mice reduced the expression of CD31 one of the endothelial cell markers. We identified PTPN3 a tyrosine phosphatase as a target of miR-574-5p that bound to the 3'UTR of PTPN3 mRNA to inhibit the expression of PTPN3. Furthermore, the data in this study demonstrated that inhibition of PTPN3 in gastric cancer cells enhanced phosphorylation of p44/42 MAPKs and promoted angiogenesis. We conclude that miR-574-5p in gastric cancer cells promoted angiogenesis via enhancing phosphorylation of p44/42 MAPKs by miR-574-5p inhibition of PTPN3 expression.
Collapse
|
28
|
Gao Y, Zhang R, Wei G, Dai S, Zhang X, Yang W, Li X, Bai C. Long Non-coding RNA Maternally Expressed 3 Increases the Expression of Neuron-Specific Genes by Targeting miR-128-3p in All-Trans Retinoic Acid-Induced Neurogenic Differentiation From Amniotic Epithelial Cells. Front Cell Dev Biol 2019; 7:342. [PMID: 31921854 PMCID: PMC6936004 DOI: 10.3389/fcell.2019.00342] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 12/03/2019] [Indexed: 12/16/2022] Open
Abstract
MicroRNA (miR)-128-3p is a brain-enriched miRNA that participates in the regulation of neural cell differentiation and the protection of neurons, but the mechanisms by which miR-128-3p regulates its target and downstream genes to influence cell fate from adult stem cells are poorly understood. In this study, we show down-regulation of miR-128-3p during all-trans retinoic acid (ATRA)-induced neurogenic differentiation from amniotic epithelial cells (AECs). We investigated miR-128-3p in both the Notch pathway and in the expression of neuron-specific genes predicted to be involved in miR-128-3p signaling to elucidate its role in the genetic regulation of downstream neurogenic differentiation. Our results demonstrate that miR-128-3p is a negative regulator for the transcription of the neuron-specific genes β III-tubulin, neuron-specific enolase (NSE), and polysialic acid-neural cell adhesion molecule (PSA-NCAM) via targeting Jagged 1 to inhibit activation of the Notch signaling pathway. We also used bioinformatics algorithms to screen for miR-128-3p interactions with long non-coding (lnc) RNA and circular RNA as competing endogenous RNAs to further elucidate underlying down-regulated molecular mechanisms. The lncRNA maternally expressed 3 is up-regulated by the ATRA/cAMP/CREB pathway, and it, in turn, is directly down-regulated by miR-128-3p to increase the amount of neuron differentiation. Endogenous miRNAs are, therefore, involved in neurogenic differentiation from AECs and should be considered during the development of effective cell transplant therapies for the treatment of neurodegenerative disease.
Collapse
Affiliation(s)
- Yuhua Gao
- Institute of Precision Medicine, School of Clinical Medicine, Jining Medical University, Jining, China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ranxi Zhang
- Department of Spine Surgery, Qingdao Municipal Hospital, Qingdao, China
| | - Guanghe Wei
- Institute of Precision Medicine, School of Clinical Medicine, Jining Medical University, Jining, China
| | - Shanshan Dai
- Institute of Precision Medicine, School of Clinical Medicine, Jining Medical University, Jining, China
| | - Xue Zhang
- Institute of Precision Medicine, School of Clinical Medicine, Jining Medical University, Jining, China
| | - Wancai Yang
- Institute of Precision Medicine, School of Clinical Medicine, Jining Medical University, Jining, China.,Department of Pathology, University of Illinois at Chicago, Chicago, IL, United States
| | - Xiangchen Li
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China.,College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang A&F University, Lin'an, China
| | - Chunyu Bai
- Institute of Precision Medicine, School of Clinical Medicine, Jining Medical University, Jining, China.,Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
29
|
Circulating Levels of miR-574-5p Are Associated with Neurological Outcome after Cardiac Arrest in Women: A Target Temperature Management (TTM) Trial Substudy. DISEASE MARKERS 2019; 2019:1802879. [PMID: 31275442 PMCID: PMC6589199 DOI: 10.1155/2019/1802879] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 03/18/2019] [Accepted: 03/26/2019] [Indexed: 01/10/2023]
Abstract
Purpose Postresuscitation neuroprognostication is guided by neurophysiological tests, biomarker measurement, and clinical examination. Recent investigations suggest that circulating microRNAs (miRNA) may help in outcome prediction after cardiac arrest. We assessed the ability of miR-574-5p to predict neurological outcome after cardiac arrest, in a sex-specific manner. Methods In this substudy of the Target Temperature Management (TTM) Trial, we enrolled 590 cardiac arrest patients for which blood samples were available. Expression levels of miR-574-5p were measured by quantitative PCR in plasma samples collected 48 h after cardiac arrest. The endpoint of the study was poor neurological outcome at 6 months (cerebral performance category scores 3 to 5). Results Eighty-one percent of patients were men, and 49% had a poor neurological outcome. Circulating levels of miR-574-5p at 48 h were higher in patients with a poor neurological outcome at 6 months (p < 0.001), both in women and in men. Circulating levels of miR-574-5p were univariate predictors of neurological outcome (odds ratio (OR) [95% confidence interval (CI)]: 1.5 [1.26-1.78]). After adjustment with clinical variables and NSE, circulating levels of miR-574-5p predicted neurological outcome in women (OR [95% CI]: 1.9 [1.09-3.45]), but not in men (OR [95% CI]: 1.0 [0.74-1.28]). Conclusion miR-574-5p is associated with neurological outcome after cardiac arrest in women.
Collapse
|
30
|
Zhang W, Tan YW, Yam WK, Tu H, Qiu L, Tan EK, Chu JJH, Zeng L. In utero infection of Zika virus leads to abnormal central nervous system development in mice. Sci Rep 2019; 9:7298. [PMID: 31086212 PMCID: PMC6513999 DOI: 10.1038/s41598-019-43303-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 04/16/2019] [Indexed: 12/14/2022] Open
Abstract
The World Health Organization has declared ZIKA virus (ZIKV) a global public health emergency, prompted by the association of ZIKV infections with severe brain abnormalities in the human fetus. ZIKV preferentially targets human neuronal precursor cells (NPCs) in both monolayer and cortical brain organoid culture systems and stunts their growth. Although ZIKV is well recognized to cause microcephaly, there is no systematic analysis to demonstrate the effect of ZIKV on central nervous system (CNS) development, including brain malformations and spinal cord dysfunction. Here, we conducted a longitudinal analysis to show that a novel mouse model (infected in utero and monitored after birth until adulthood) recapitulates the effects of ZIKV infection affecting neural stem cells fate and leads to a thinner cortex and a smaller brain. Furthermore, we demonstrate the effect of ZIKV on spinal cord function. Specifically, we found significant reductions in neuron numbers in the anterior horn of grey matter of the spinal cord and muscle dystrophy with a significant decrease in forepaw grip strength in the ZIKV group. Thus, the established mouse model of ZIKV infection leading to abnormal CNS development will help to further advance our understanding of the disease pathogenesis.
Collapse
Affiliation(s)
- Wei Zhang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Yong Wah Tan
- Collaborative Translation Unit for HFMD, Institute of Molecular and Cell Biology, Agency of Science, Technology & Research (A STAR), Singapore, 138673, Singapore
| | - Wan Keat Yam
- Collaborative Translation Unit for HFMD, Institute of Molecular and Cell Biology, Agency of Science, Technology & Research (A STAR), Singapore, 138673, Singapore
| | - Haitao Tu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Lifeng Qiu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Eng King Tan
- Research Department, National Neuroscience Institute, SGH Campus, Singapore, 169856, Singapore.,Department of Neurology, National Neuroscience Institute, SGH Campus, Singapore, 169856, Singapore.,Neuroscience & Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore, 169857, Singapore
| | - Justin Jang Hann Chu
- Collaborative Translation Unit for HFMD, Institute of Molecular and Cell Biology, Agency of Science, Technology & Research (A STAR), Singapore, 138673, Singapore.,Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore. .,Neuroscience & Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore, 169857, Singapore. .,Lee Kong Chian School of Medicine, Novena Campus, 11 Mandalay Road, Singapore, 308232, Singapore.
| |
Collapse
|
31
|
Patel AA, Ganepola GA, Rutledge JR, Chang DH. The Potential Role of Dysregulated miRNAs in Alzheimer’s Disease Pathogenesis and Progression. J Alzheimers Dis 2019; 67:1123-1145. [DOI: 10.3233/jad-181078] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Ankur A. Patel
- Department of Research, Center for Cancer Research and Genomic Medicine, The Valley Hospital, Paramus, NJ, USA
| | - Ganepola A.P. Ganepola
- Department of Research, Center for Cancer Research and Genomic Medicine, The Valley Hospital, Paramus, NJ, USA
| | - John R. Rutledge
- Department of Oncology Special Program, The Daniel and Gloria Blumenthal Cancer Center, The Valley Hospital, Paramus, NJ, USA
| | - David H. Chang
- Department of Research, Center for Cancer Research and Genomic Medicine, The Valley Hospital, Paramus, NJ, USA
| |
Collapse
|
32
|
Belarbi Y, Mejhert N, Gao H, Arner P, Rydén M, Kulyté A. MicroRNAs-361-5p and miR-574-5p associate with human adipose morphology and regulate EBF1 expression in white adipose tissue. Mol Cell Endocrinol 2018; 472:50-56. [PMID: 29191698 DOI: 10.1016/j.mce.2017.11.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/24/2017] [Accepted: 11/23/2017] [Indexed: 12/21/2022]
Abstract
Reduced adipose expression of the transcription factor Early B cell factor 1 (EBF1) is linked to white adipose tissue (WAT) hypertrophy. We aimed to identify microRNAs (miRNAs) associated with WAT hypertrophy and EBF1 regulation. We mapped WAT miRNA expression from 26 non-obese women discordant in WAT morphology and determined EBF1 activity in the non-obese and 30 obese women. Expression of 15 miRNAs was higher in hypertrophy and 10 were predicted to target EBF1. Binding of miR-365-5p/miR-574-5p were validated with 3'-UTR assay. Overexpression of miR-365-5p or miR-574-5p reduced EBF1 while inhibition of miR-574 increased EBF1 expression in human adipocytes in vitro. Additive effects on EBF1 were observed when concomitantly overexpressing both miRNAs. EBF1 targets were affected by over expression/inhibition of either miRNAs. Finally, miR-365-5p/miR-574-5p expression in 56 individuals correlated significantly with EBF1 activity. Our results suggest that miR-365-5p and miR-574-5p may be linked to WAT hypertrophy via effects on EBF1 expression.
Collapse
Affiliation(s)
- Yasmina Belarbi
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, SE-14186 Stockholm, Sweden
| | - Niklas Mejhert
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, SE-14186 Stockholm, Sweden
| | - Hui Gao
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, SE-14186 Stockholm, Sweden; Department of Biosciences & Nutrition, Karolinska Institutet, SE-141 Stockholm, Sweden
| | - Peter Arner
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, SE-14186 Stockholm, Sweden
| | - Mikael Rydén
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, SE-14186 Stockholm, Sweden
| | - Agné Kulyté
- Lipid Laboratory, Department of Medicine Huddinge, Karolinska Institutet, SE-14186 Stockholm, Sweden.
| |
Collapse
|
33
|
Huang SK, Luo Q, Peng H, Li J, Zhao M, Wang J, Gu YY, Li Y, Yuan P, Zhao GH, Huang CZ. A Panel of Serum Noncoding RNAs for the Diagnosis and Monitoring of Response to Therapy in Patients with Breast Cancer. Med Sci Monit 2018; 24:2476-2488. [PMID: 29683112 PMCID: PMC5935014 DOI: 10.12659/msm.909453] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The aim of this study was to identify a panel of serum noncoding RNAs (ncRNAs) as potential diagnostic and prognostic biomarkers for breast cancer. MATERIAL AND METHODS Patients with breast cancer (n=30), and normal controls (n=30) were included in the 'training set.' A 'validation set' included cases of breast cancer (n=128) and controls (n=77). All cases provided blood samples for serum analysis. All cases of breast cancer were confirmed histologically and were staged. Quantitative reverse transcription polymerase chain reaction (RT-qPCR) was used to detect the expression of 11 candidate ncRNAs, including long noncoding RNAs (lncRNAs) and microRNAs (miRNAs), in the serum. The expression of the panel of ncRNAs was further analyzed following surgery or chemotherapy. RESULTS The four ncRNAs identified in the serum of patients with breast cancer included let-7a, miR-155, miR-574-5p, and metastasis-associated lung adenocarcinoma transcript 1 (MALAT1). Analysis based on the risk score showed that the panel of these four ncRNAs could effectively distinguish between patients with breast cancer and the control group. For the training set and the validation set, analysis of the receiver-operating characteristic (ROC) curve showed that the areas under the curve (AUCs) were 0.960 and 0.968, respectively. Also, the serum expression levels of the four ncRNAs differed in the pre-treatment and the post-treatment patients with breast cancer, with levels of miR-155 showing a significant decrease following chemotherapy. CONCLUSIONS A panel of serum ncRNAs, including let-7a, miR-155, miR-574-5p, and MALAT1, was shown to be present in patients with breast cancer.
Collapse
Affiliation(s)
- Sheng-Kai Huang
- Department of Clinical Laboratory, National Cancer Center and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (mainland).,State Key Laboratory of Molecular Oncology,Department of Etiology and Carcinogenesis, National Cancer Center and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (mainland).,Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, Beijing, China (mainland)
| | - Qing Luo
- Department of Oncology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China (mainland)
| | - Hua Peng
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (mainland).,Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, Beijing, China (mainland)
| | - Jia Li
- Department of Clinical Laboratory, National Cancer Center and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (mainland)
| | - Mei Zhao
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (mainland).,Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, Beijing, China (mainland)
| | - Jia Wang
- Department of Clinical Laboratory, Meitan General Hospital, Beijing, China (mainland)
| | - Yu-Yu Gu
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (mainland).,Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, Beijing, China (mainland)
| | - Yan Li
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (mainland).,Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, Beijing, China (mainland)
| | - Peng Yuan
- Department of Medical Oncology, National Cancer Center and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (mainland)
| | - Guo-Hua Zhao
- Department of Transfusion Medicine, National Cancer Center and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (mainland)
| | - Chang-Zhi Huang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, National Cancer Center and Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (mainland).,Beijing Key Laboratory for Carcinogenesis and Cancer Prevention, Beijing, China (mainland)
| |
Collapse
|
34
|
Zhang W, Zeng X, Zeng L. Functional Analysis of MicroRNAs in Neurogenesis During Mouse Cortical Development. Methods Mol Biol 2018; 1680:191-203. [PMID: 29030850 DOI: 10.1007/978-1-4939-7339-2_13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The advantage of using in utero electroporation is that it can study the gene function during neurodevelopment in vivo. Using functional analysis of a microRNA (miRNA) gene as an example, this protocol describes a set of techniques that are crucial for the success of neurogenesis studies, including mice time mating, plasmid preparation, utero electroporation following miRNA injection into mice embryonic brain ventricle, labeling of proliferating cells with EDU (ethynyldeoxyuridine), cryosectioning, immunofluorescence staining, and confocal microscopic analysis. This chapter also provides detailed technical tips regarding experimental planning, mouse surgery, multi-embryo injection with different plasmids, electroporation, and maintenance of pregnant mother with post-electroporated embryo.
Collapse
Affiliation(s)
- Wei Zhang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Xiaoxia Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, 308433, Singapore. .,Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore, 169857, Singapore.
| |
Collapse
|
35
|
Ku T, Li B, Gao R, Zhang Y, Yan W, Ji X, Li G, Sang N. NF-κB-regulated microRNA-574-5p underlies synaptic and cognitive impairment in response to atmospheric PM 2.5 aspiration. Part Fibre Toxicol 2017; 14:34. [PMID: 28851397 PMCID: PMC5575838 DOI: 10.1186/s12989-017-0215-3] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 08/20/2017] [Indexed: 12/12/2022] Open
Abstract
Background PM2.5 (particulate matter ≤ 2.5 μm) is one of the leading environmental risk factors for the global burden of disease. Whereas increasing evidence has linked the adverse roles of PM2.5 with cardiovascular and respiratory diseases, limited but growing emerging evidence suggests that PM2.5 exposure can affect the nervous system, causing neuroinflammation, synaptic dysfunction and cognitive deterioration. However, the molecular mechanisms underlying the synaptic and cognitive deficits elicited by PM2.5 exposure are largely unknown. Methods C57BL/6 mice received oropharyngeal aspiration of PM2.5 (1 and 5 mg/kg bw) every other day for 4 weeks. The mice were also stereotaxically injected with β-site amyloid precursor protein cleaving enzyme 1 (β-secretase, BACE1) shRNA or LV-miR-574-5p lentiviral constructs in the absence or presence of PM2.5 aspiration at 5 mg/kg bw every other day for 4 weeks. Spatial learning and memory were assessed with the Morris water maze test, and synaptic function integrity was evaluated with electrophysiological recordings of long-term potentiation (LTP) and immunoblot analyses of glutamate receptor subunit expression. The expression of α-secretase (ADAM10), BACE1, and γ-secretase (nicastrin) and the synthesis and accumulation of amyloid β (Aβ) were measured by immunoblot and enzyme-linked immunosorbent assay (ELISA). MicroRNA (miRNA) expression was screened with a microRNA microarray analysis and confirmed by real-time quantitative reverse transcription PCR (qRT-PCR) analysis. Dual-luciferase reporter gene and chromatin immunoprecipitation (ChIP) analyses were used to detect the binding of miR-574-5p in the 3’UTR of BACE1 and NF-κB p65 in the promoter of miR-574-5p, respectively. Results PM2.5 aspiration caused neuroinflammation and deteriorated synaptic function integrity and spatial learning and memory, and the effects were associated with the induction of BACE1. The action was mediated by NF-κB p65-regulated downregulation of miR-574-5p, which targets BACE1. Overexpression of miR-574-5p in the hippocampal region decreased BACE1 expression, restored synaptic function, and improved spatial memory and learning following PM2.5 exposure. Conclusions Taken together, our findings reveal a novel molecular mechanism underlying impaired synaptic and cognitive function following exposure to PM2.5, suggesting that miR-574-5p is a potential intervention target for the prevention and treatment of PM2.5-induced neurological disorders. Electronic supplementary material The online version of this article (10.1186/s12989-017-0215-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tingting Ku
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Ben Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Rui Gao
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Yingying Zhang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Wei Yan
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Xiaotong Ji
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Guangke Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi, 030006, China.
| |
Collapse
|
36
|
Rajman M, Schratt G. MicroRNAs in neural development: from master regulators to fine-tuners. Development 2017; 144:2310-2322. [DOI: 10.1242/dev.144337] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The proper formation and function of neuronal networks is required for cognition and behavior. Indeed, pathophysiological states that disrupt neuronal networks can lead to neurodevelopmental disorders such as autism, schizophrenia or intellectual disability. It is well-established that transcriptional programs play major roles in neural circuit development. However, in recent years, post-transcriptional control of gene expression has emerged as an additional, and probably equally important, regulatory layer. In particular, it has been shown that microRNAs (miRNAs), an abundant class of small regulatory RNAs, can regulate neuronal circuit development, maturation and function by controlling, for example, local mRNA translation. It is also becoming clear that miRNAs are frequently dysregulated in neurodevelopmental disorders, suggesting a role for miRNAs in the etiology and/or maintenance of neurological disease states. Here, we provide an overview of the most prominent regulatory miRNAs that control neural development, highlighting how they act as ‘master regulators’ or ‘fine-tuners’ of gene expression, depending on context, to influence processes such as cell fate determination, cell migration, neuronal polarization and synapse formation.
Collapse
Affiliation(s)
- Marek Rajman
- Biochemisch-Pharmakologisches Centrum, Institut für Physiologische Chemie, Philipps-Universität Marburg, Marburg 35043, Germany
| | - Gerhard Schratt
- Biochemisch-Pharmakologisches Centrum, Institut für Physiologische Chemie, Philipps-Universität Marburg, Marburg 35043, Germany
| |
Collapse
|
37
|
Qiu L, Liao MC, Chen AK, Wei S, Xie S, Reuveny S, Zhou ZD, Hunziker W, Tan EK, Oh SKW, Zeng L. Immature Midbrain Dopaminergic Neurons Derived from Floor-Plate Method Improve Cell Transplantation Therapy Efficacy for Parkinson's Disease. Stem Cells Transl Med 2017. [PMID: 28650520 PMCID: PMC5689771 DOI: 10.1002/sctm.16-0470] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Recent reports have indicated human embryonic stem cells-derived midbrain dopamine (mDA) neurons as proper cell resources for use in Parkinson's disease (PD) therapy. Nevertheless, no detailed and systematic study has been conducted to identify which differentiation stages of mDA cells are most suitable for transplantation in PD therapy. Here, we transplanted three types of mDA cells, DA progenitors (differentiated in vitro for 16 days [D16]), immature DA neurons (D25), and DA neurons (D35), into PD mice and found that all three types of cells showed high viability and strong neuronal differentiation in vivo. Both D25 and D35 cells showed neuronal maturation and differentiation toward TH+ cells and, accordingly, satisfactory behavioral functional recovery. However, transplanted D16 cells were less capable of producing functional recovery. These findings provide a valuable guideline for standardizing the differentiation stage of the transplantable cells used in clinical cell therapy for PD. Stem Cells Translational Medicine 2017;6:1803-1814.
Collapse
Affiliation(s)
- Lifeng Qiu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Mei-Chih Liao
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore
| | - Allen K Chen
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore
| | - Shunhui Wei
- Epithelial Cell Biology Laboratory, Institute of Molecular and Cell Biology, Singapore
| | - Shaoping Xie
- Research Department, National Neuroscience Institute, Singapore
| | - Shaul Reuveny
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore
| | - Zhi Dong Zhou
- Research Department, National Neuroscience Institute, Singapore.,Neuroscience & Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore
| | - Walter Hunziker
- Epithelial Cell Biology Laboratory, Institute of Molecular and Cell Biology, Singapore.,Department of Physiology, National University of Singapore, Singapore
| | - Eng King Tan
- Research Department, National Neuroscience Institute, Singapore.,Neuroscience & Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore.,Department of Neurology, National Neuroscience Institute, Singapore
| | - Steve K W Oh
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore.,Neuroscience & Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore
| |
Collapse
|
38
|
Qiu L, Lim YM, Chen AK, Reuveny S, Oh SKW, Tan EK, Zeng L. Microcarrier-Expanded Neural Progenitor Cells Can Survive, Differentiate, and Innervate Host Neurons Better When Transplanted as Aggregates. Cell Transplant 2016; 25:1343-57. [DOI: 10.3727/096368915x690378] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Neuronal progenitor cells (NPCs) derived from human embryonic stem cells (hESCs) are an excellent cell source for transplantation therapy due to their availability and ethical acceptability. However, the traditional method of expansion and differentiation of hESCs into NPCs in monolayer cultures requires a long time, and the cell yield is low. A microcarrier (MC) platform can improve the expansion of hESCs and increase the yield of NPCs. In this study, for the first time, we transplanted microcarrier-expanded hESC-derived NPCs into the striatum of adult NOD-SCID IL2Rgc null mice, either as single cells or as cell aggregates. The recipient mice were perfused, and the in vivo survival, differentiation, and targeted innervation of the transplanted cells were assessed by immunostaining. We found that both the transplanted single NPCs and aggregate NPCs were able to survive 1 month posttransplantation, as revealed by human-specific neural cell adhesion molecule (NCAM) and human nuclear antigen staining. Compared to the single cells, the transplanted cell aggregates showed better survival over a 3-month period. In addition, both the transplanted single NPCs and the aggregate NPCs were able to differentiate into DCX-positive immature neurons and Tuj1-positive neurons in vivo by 1 month posttransplantation. However, only the transplantation of aggregate NPCs was shown to result in mature neurons at 3 months posttransplantation. Furthermore, we found that the cell aggregates were able to send long axons to innervate their targets. Our study provides preclinical evidence that the use of MCs to expand and differentiate hESC-derived NPCs and transplantation of these cells as aggregates produce longer survival in vivo.
Collapse
Affiliation(s)
- Lifeng Qiu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
| | - Yu Ming Lim
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A STAR), Singapore
| | - Allen K. Chen
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A STAR), Singapore
| | - Shaul Reuveny
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A STAR), Singapore
| | - Steve K. W. Oh
- Stem Cell Group, Bioprocessing Technology Institute, Agency for Science, Technology and Research (A STAR), Singapore
| | - Eng King Tan
- Department of Neurology, National Neuroscience Institute, SGH Campus, Singapore
- Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore
- Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore
| |
Collapse
|
39
|
Zhang W, Kim PJ, Chen Z, Lokman H, Qiu L, Zhang K, Rozen SG, Tan EK, Je HS, Zeng L. MiRNA-128 regulates the proliferation and neurogenesis of neural precursors by targeting PCM1 in the developing cortex. eLife 2016; 5. [PMID: 26883496 PMCID: PMC4769165 DOI: 10.7554/elife.11324] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 01/22/2016] [Indexed: 12/18/2022] Open
Abstract
During the development, tight regulation of the expansion of neural progenitor cells (NPCs) and their differentiation into neurons is crucial for normal cortical formation and function. In this study, we demonstrate that microRNA (miR)-128 regulates the proliferation and differentiation of NPCs by repressing pericentriolar material 1 (PCM1). Specifically, overexpression of miR-128 reduced NPC proliferation but promoted NPC differentiation into neurons both in vivo and in vitro. In contrast, the reduction of endogenous miR-128 elicited the opposite effects. Overexpression of miR-128 suppressed the translation of PCM1, and knockdown of endogenous PCM1 phenocopied the observed effects of miR-128 overexpression. Furthermore, concomitant overexpression of PCM1 and miR-128 in NPCs rescued the phenotype associated with miR-128 overexpression, enhancing neurogenesis but inhibiting proliferation, both in vitro and in utero. Taken together, these results demonstrate a novel mechanism by which miR-128 regulates the proliferation and differentiation of NPCs in the developing neocortex. DOI:http://dx.doi.org/10.7554/eLife.11324.001 The neurons that transmit information around the brain develop from cells called neural progenitor cells. These cells can either divide to form more progenitor cells or to become specific types of neurons. If these carefully regulated processes go wrong – for example, if progenitors fail to stop dividing in order to mature – a range of neurodevelopmental conditions may develop, including autism spectrum disorders. Small RNA molecules called microRNAs control gene activity and protein formation by targeting certain other RNA molecules for destruction. One such microRNA, called miR-128, helps newly formed neurons to move to the correct region of the cortex – the outer layer of the brain, which is essential for many cognitive processes including thought and language. However, it was not clear whether miR-128 plays any other roles in the development of neurons. Zhang, Kim et al. have now analysed the role of miR-128 in the developing cortex of mice. The findings suggest that miR-128 prevents cortical neural progenitor cells from dividing and supports their development into more specialized cells. Causing miR-128 to be over-produced in the progenitor cells caused the cells to divide less often and encouraged them to mature into neurons. Conversely, removing miR-128 from the progenitor cells caused them to divide more and resulted in fewer neurons forming. Further investigation revealed that miR-128 works by causing less of a protein called PCM1 to be produced. Without this protein, cells cannot divide properly. Future studies could now investigate in more detail how miR-128 and PCM1 affect how the neurons in the cortex develop and work. DOI:http://dx.doi.org/10.7554/eLife.11324.002
Collapse
Affiliation(s)
- Wei Zhang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, Singapore
| | - Paul Jong Kim
- Molecular Neurophysiology Laboratory, Signature Program in Neuroscience and Behavioral Disorders, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Zhongcan Chen
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, Singapore
| | - Hidayat Lokman
- Molecular Neurophysiology Laboratory, Signature Program in Neuroscience and Behavioral Disorders, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Lifeng Qiu
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, Singapore
| | - Ke Zhang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, Singapore
| | - Steven George Rozen
- Center for Computational Biology, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Eng King Tan
- Department of Neurology, National Neuroscience Institute, Singapore, Singapore.,Neuroscience and Behavioral Disorders program, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Hyunsoo Shawn Je
- Molecular Neurophysiology Laboratory, Signature Program in Neuroscience and Behavioral Disorders, Duke-NUS Graduate Medical School, Singapore, Singapore.,Neuroscience and Behavioral Disorders program, Duke-NUS Graduate Medical School, Singapore, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore, Singapore.,Neuroscience and Behavioral Disorders program, Duke-NUS Graduate Medical School, Singapore, Singapore
| |
Collapse
|
40
|
Seifan A, Assuras S, Huey ED, Mez J, Tsapanou A, Caccappolo E. Childhood Learning Disabilities and Atypical Dementia: A Retrospective Chart Review. PLoS One 2015; 10:e0129919. [PMID: 26106899 PMCID: PMC4481274 DOI: 10.1371/journal.pone.0129919] [Citation(s) in RCA: 14] [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: 10/21/2014] [Accepted: 05/14/2015] [Indexed: 11/19/2022] Open
Abstract
Objective To further our understanding of the association between self-reported childhood learning disabilities (LDs) and atypical dementia phenotypes (Atypical Dementia), including logopenic primary progressive aphasia (L-PPA), Posterior Cortical Atrophy (PCA), and Dysexecutive-type Alzheimer’s Disease (AD). Methods This retrospective case series analysis of 678 comprehensive neuropsychological assessments compared rates of self-reported LD between dementia patients diagnosed with Typical AD and those diagnosed with Atypical Dementia. 105 cases with neuroimaging or CSF data available and at least one neurology follow-up were identified as having been diagnosed by the neuropsychologist with any form of neurodegenerative dementia. These cases were subject to a consensus diagnostic process among three dementia experts using validated clinical criteria for AD and PPA. LD was considered Probable if two or more statements consistent with prior LD were documented within the Social & Developmental History of the initial neuropsychological evaluation. Results 85 subjects (Typical AD n=68, Atypical AD n=17) were included in the final analysis. In logistic regression models adjusted for age, gender, handedness, education and symptom duration, patients with Probable LD, compared to patients without Probable LD, were significantly more likely to be diagnosed with Atypical Dementia vs. Typical AD (OR 13.1, 95% CI 1.3-128.4). All three of the L-PPA cases reporting a childhood LD endorsed childhood difficulty with language. By contrast, both PCA cases reporting Probable childhood LD endorsed difficulty with attention and/or math. Conclusions In people who develop dementia, childhood LD may predispose to atypical phenotypes. Future studies are required to confirm whether atypical neurodevelopment predisposes to regional-specific neuropathology in AD and other dementias.
Collapse
Affiliation(s)
- Alon Seifan
- Department of Neurology Weill Cornell Medical College, New York, New York, United States of America
- * E-mail:
| | - Stephanie Assuras
- Department of Neuropsychology, Columbia University, New York, New York, United States of America
| | - Edward D. Huey
- Department of Neurology Columbia University, New York, New York, United States of America
- Gertrude H. Sergievsky Center, Columbia University, New York, New York, United States of America
- Cognitive neuroscience division, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Jesse Mez
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, United States of America
| | - Angeliki Tsapanou
- Cognitive neuroscience division, Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Elise Caccappolo
- Department of Neuropsychology, Columbia University, New York, New York, United States of America
| |
Collapse
|
41
|
Terranova C, Narla ST, Lee YW, Bard J, Parikh A, Stachowiak EK, Tzanakakis ES, Buck MJ, Birkaya B, Stachowiak MK. Global Developmental Gene Programing Involves a Nuclear Form of Fibroblast Growth Factor Receptor-1 (FGFR1). PLoS One 2015; 10:e0123380. [PMID: 25923916 PMCID: PMC4414453 DOI: 10.1371/journal.pone.0123380] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Accepted: 02/17/2015] [Indexed: 12/11/2022] Open
Abstract
Genetic studies have placed the Fgfr1 gene at the top of major ontogenic pathways that enable gastrulation, tissue development and organogenesis. Using genome-wide sequencing and loss and gain of function experiments the present investigation reveals a mechanism that underlies global and direct gene regulation by the nuclear form of FGFR1, ensuring that pluripotent Embryonic Stem Cells differentiate into Neuronal Cells in response to Retinoic Acid. Nuclear FGFR1, both alone and with its partner nuclear receptors RXR and Nur77, targets thousands of active genes and controls the expression of pluripotency, homeobox, neuronal and mesodermal genes. Nuclear FGFR1 targets genes in developmental pathways represented by Wnt/β-catenin, CREB, BMP, the cell cycle and cancer-related TP53 pathway, neuroectodermal and mesodermal programing networks, axonal growth and synaptic plasticity pathways. Nuclear FGFR1 targets the consensus sequences of transcription factors known to engage CREB-binding protein, a common coregulator of transcription and established binding partner of nuclear FGFR1. This investigation reveals the role of nuclear FGFR1 as a global genomic programmer of cell, neural and muscle development.
Collapse
Affiliation(s)
- Christopher Terranova
- Department of Pathology and Anatomical Sciences, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Sridhar T. Narla
- Department of Pathology and Anatomical Sciences, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Yu-Wei Lee
- Department of Pathology and Anatomical Sciences, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Jonathan Bard
- Next-Generation Sequencing and Expression Analysis Core, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Abhirath Parikh
- Department of Chemical and Biological Engineering, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Ewa K. Stachowiak
- Department of Pathology and Anatomical Sciences, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Emmanuel S. Tzanakakis
- Department of Chemical and Biological Engineering, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Michael J. Buck
- Department of Biochemistry, Genomics and Bioinformatics Core, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Barbara Birkaya
- Department of Pathology and Anatomical Sciences, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Michal K. Stachowiak
- Department of Pathology and Anatomical Sciences, Western New York Stem Cell Culture and Analysis Center, State University of New York at Buffalo, Buffalo, New York, United States of America
- * E-mail:
| |
Collapse
|
42
|
APP intracellular domain acts as a transcriptional regulator of miR-663 suppressing neuronal differentiation. Cell Death Dis 2015; 6:e1651. [PMID: 25695604 PMCID: PMC4669786 DOI: 10.1038/cddis.2015.10] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 12/29/2014] [Indexed: 12/21/2022]
Abstract
Amyloid precursor protein (APP) is best known for its involvement in the pathogenesis of Alzheimer's disease. We have previously demonstrated that APP intracellular domain (AICD) regulates neurogenesis; however, the mechanisms underlying AICD-mediated regulation of neuronal differentiation are not yet fully characterized. Using genome-wide chromatin immunoprecipitation approaches, we found that AICD is specifically recruited to the regulatory regions of several microRNA genes, and acts as a transcriptional regulator for miR-663, miR-3648 and miR-3687 in human neural stem cells. Functional assays show that AICD negatively modulates neuronal differentiation through miR-663, a primate-specific microRNA. Microarray data further demonstrate that miR-663 suppresses the expression of multiple genes implicated in neurogenesis, including FBXL18 and CDK6. Our results indicate that AICD has a novel role in suppression of neuronal differentiation via transcriptional regulation of miR-663 in human neural stem cells.
Collapse
|
43
|
Qiu L, Zhang W, Tan EK, Zeng L. Deciphering the function and regulation of microRNAs in Alzheimer's disease and Parkinson's disease. ACS Chem Neurosci 2014; 5:884-94. [PMID: 25210999 DOI: 10.1021/cn500149w] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are single stranded, noncoding RNA molecules that are encoded by eukaryotic nuclear DNA. miRNAs function through imperfect base-pairing with complementary sequences of target mRNA molecules, which is typically via the cleavage of target mRNA with transcriptional repression or translational degradation. An increasing number of studies identified dysregulation of miRNAs in neurodegenerative disease and suggest that alterations in the miRNA regulatory pathway could contribute to the disease pathogenesis. However, molecular mechanisms underlying the pathological implications of dysregulated miRNA expression and regulation of the key genes that are involved in neurodegenerative diseases remain largely unknown. Here, we review the evidence for the functional role of dysregulated miRNAs involved in disease pathogenesis, as well as how miRNAs govern neuronal functions either upstream or downstream of target genes that are disease pathogenic factors. Furthermore, we review the cellular feedback regulation between miRNAs and target genes in neurodegenerative diseases, with a focus on Alzheimer's disease and Parkinson's disease.
Collapse
Affiliation(s)
- Lifeng Qiu
- Neural
Stem Cell Research Lab, Research Department, National Neuroscience Institute, 308433, Singapore
| | - Wei Zhang
- Neural
Stem Cell Research Lab, Research Department, National Neuroscience Institute, 308433, Singapore
| | - Eng King Tan
- Department
of Neurology, National Neuroscience Institute, SGH Campus, 169856, Singapore
- Research
Department, National Neuroscience Institute, 308433, Singapore
- Neuroscience & Behavioral Disorders Program, DUKE-NUS Graduate Medical School, 169857, Singapore
| | - Li Zeng
- Neural
Stem Cell Research Lab, Research Department, National Neuroscience Institute, 308433, Singapore
- Neuroscience & Behavioral Disorders Program, DUKE-NUS Graduate Medical School, 169857, Singapore
| |
Collapse
|