51
|
Posttranscriptional regulation of Nrf2 through miRNAs and their role in Alzheimer's disease. Pharmacol Res 2021; 175:106018. [PMID: 34863823 DOI: 10.1016/j.phrs.2021.106018] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 10/27/2021] [Accepted: 11/29/2021] [Indexed: 12/18/2022]
Abstract
The nuclear factor erythroid-derived 2-related factor 2 (NFE2L2/Nrf2) is a pivotal facilitator of cytoprotective responses against the oxidative/electrophilic insults. Upon activation, Nrf2 induces transcription of a wide range of cytoprotective genes having antioxidant response element (ARE) in their promoter region. Dysfunction in Nrf2 signaling has been linked to the pathogenesis of AD and several studies have suggested that boosting Nrf2 expression/activity by genetic or pharmacological approaches is beneficial in AD. Among the diverse mechanisms that regulate the Nrf2 signaling, miRNAs-mediated regulation of Nrf2 has gained much attention in recent years. Several miRNAs have been reported to directly repress the post-transcriptional expression of Nrf2 and thereby negatively regulate the Nrf2-dependent cellular cytoprotective response in AD. Moreover, several Nrf2 targeting miRNAs are misregulated in AD brains. This review is focused on the role of misregulated miRNAs that directly target Nrf2, in AD pathophysiology. Here, alongside a general description of functional interactions between miRNAs and Nrf2, we have reviewed the evidence indicating the possible role of these miRNAs in AD pathogenesis.
Collapse
|
52
|
Liang Y, Wang L. Inflamma-MicroRNAs in Alzheimer's Disease: From Disease Pathogenesis to Therapeutic Potentials. Front Cell Neurosci 2021; 15:785433. [PMID: 34776873 PMCID: PMC8581643 DOI: 10.3389/fncel.2021.785433] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/08/2021] [Indexed: 01/16/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of senile dementia. Although AD research has made important breakthroughs, the pathogenesis of this disease remains unclear, and specific AD diagnostic biomarkers and therapeutic strategies are still lacking. Recent studies have demonstrated that neuroinflammation is involved in AD pathogenesis and is closely related to other health effects. MicroRNAs (miRNAs) are a class of endogenous short sequence non-coding RNAs that indirectly inhibit translation or directly degrade messenger RNA (mRNA) by specifically binding to its 3′ untranslated region (UTR). Several broadly expressed miRNAs including miR-21, miR-146a, and miR-155, have now been shown to regulate microglia/astrocytes activation. Other miRNAs, including miR-126 and miR-132, show a progressive link to the neuroinflammatory signaling. Therefore, further studies on these inflamma-miRNAs may shed light on the pathological mechanisms of AD. The differential expression of inflamma-miRNAs (such as miR-29a, miR-125b, and miR-126-5p) in the peripheral circulation may respond to AD progression, similar to inflammation, and therefore may become potential diagnostic biomarkers for AD. Moreover, inflamma-miRNAs could also be promising therapeutic targets for AD treatment. This review provides insights into the role of inflamma-miRNAs in AD, as well as an overview of general inflamma-miRNA biology, their implications in pathophysiology, and their potential roles as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Yuanyuan Liang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lin Wang
- Department of Emergency Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
53
|
Regional and temporal miRNAs expression profile in a transgenic mouse model of tauopathy: implication for its pathogenesis. Mol Psychiatry 2021; 26:7020-7028. [PMID: 31988432 DOI: 10.1038/s41380-020-0655-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 12/10/2019] [Accepted: 01/16/2020] [Indexed: 11/08/2022]
Abstract
Studies have shown that the expression level of different microRNAs (miRNAs) is altered in neurodegenerative disorders including tauopathies, a group of diseases pathologically defined by accumulation of tau protein in neurons and glia cells. However, despite this evidence we still do not know whether miRNA changes precede their onset, thus potentially contributing to the pathogenesis, or are downstream events secondary to tau pathology. In the current paper, we assessed the miRNA expression profile at different age time points and brain regions in a relevant mouse model of human tauopathy, the hTau mice, in relationship with the development of behavioral deficits and tau neuropathology. Compared with age-matched control, four specific miRNAs (miR-132-3p, miIR-146a-5p, miR-22-3p, and miR-455-5p) were found significantly upregulated in 12-month-old hTau mice. Interestingly, three of them (miR-132-3p, miR-146a-5p, and miR-22-3p) were already increased in 6-month-old mice, an age before the development of tau pathologic phenotype. Investigation of their predicted targets highlighted pathways relevant to neuronal survival and synaptic function. Collectively, our findings support the new hypothesis that in tauopathies the change in the expression level of specific miRNAs is an early event and plays a functional role in the pathogenesis of the diseases by impacting several mechanisms involved in the development of the associated neuropathology.
Collapse
|
54
|
Xiao G, Chen Q, Zhang X. MicroRNA-455-5p/CPEB1 pathway mediates Aβ-related learning and memory deficits in a mouse model of Alzheimer's disease. Brain Res Bull 2021; 177:282-294. [PMID: 34678444 DOI: 10.1016/j.brainresbull.2021.10.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 09/12/2021] [Accepted: 10/12/2021] [Indexed: 12/01/2022]
Abstract
Alzheimer's disease (AD), a common neurodegenerative disease, is the main cause of dementia, with cognitive decline as the core symptom observed during diagnosis. Synaptic loss may be the main cause of early cognitive dysfunction in AD, but the detailed mechanism is still unclear. In this study, we investigated the role of abnormal miR-455-5p/CPEB1 pathway in AD mouse model. We found that miR-455-5p was upregulated, while its downstream target, cytoplasmic polyadenylation element-binding 1 (CPEB1), was downregulated in the hippocampus of APP/PS1 mice at the age of 9 m. Abnormal miR-455-5p/CPEB1 pathway mediated cognitive deficits in APP/PS1 mice through suppressing α-Amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor expressions. And miR-455-5p suppression, CPEB1 overexpression or application of a peptide disrupting the miR-455-5p/CPEB1 interaction in CA1 of APP/PS1 mice rescued AD-like phenotypes in mice, including deficits in synaptic plasticity and memory. In conclusion, our results indicated that miRNA-455-5p/CPEB1 pathway mediated synaptic and memory deficits in Alzheimer's Disease through targeting on AMPARs, providing a potential therapeutic strategy for AD.
Collapse
Affiliation(s)
- Gelei Xiao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Diagnosis and Treatment Center for Hydrocephalus, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Qianwei Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, PR China
| | - Xuewei Zhang
- Department of Health Managent Center, Xiangya hospital, Central South University, Changsha, Hunan 410008, PR China; Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan 410008, PR China.
| |
Collapse
|
55
|
Fonseca Cabral G, Schaan AP, Cavalcante GC, Sena-dos-Santos C, de Souza TP, Souza Port’s NM, dos Santos Pinheiro JA, Ribeiro-dos-Santos Â, Vidal AF. Nuclear and Mitochondrial Genome, Epigenome and Gut Microbiome: Emerging Molecular Biomarkers for Parkinson's Disease. Int J Mol Sci 2021; 22:9839. [PMID: 34576000 PMCID: PMC8471599 DOI: 10.3390/ijms22189839] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/23/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Parkinson's disease (PD) is currently the second most common neurodegenerative disorder, burdening about 10 million elderly individuals worldwide. The multifactorial nature of PD poses a difficult obstacle for understanding the mechanisms involved in its onset and progression. Currently, diagnosis depends on the appearance of clinical signs, some of which are shared among various neurologic disorders, hindering early diagnosis. There are no effective tools to prevent PD onset, detect the disease in early stages or accurately report the risk of disease progression. Hence, there is an increasing demand for biomarkers that may identify disease onset and progression, as treatment-based medicine may not be the best approach for PD. Over the last few decades, the search for molecular markers to predict susceptibility, aid in accurate diagnosis and evaluate the progress of PD have intensified, but strategies aimed to improve individualized patient care have not yet been established. CONCLUSIONS Genomic variation, regulation by epigenomic mechanisms, as well as the influence of the host gut microbiome seem to have a crucial role in the onset and progress of PD, thus are considered potential biomarkers. As such, the human nuclear and mitochondrial genome, epigenome, and the host gut microbiome might be the key elements to the rise of personalized medicine for PD patients.
Collapse
Affiliation(s)
- Gleyce Fonseca Cabral
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
| | - Ana Paula Schaan
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
| | - Giovanna C. Cavalcante
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
| | - Camille Sena-dos-Santos
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
| | - Tatiane Piedade de Souza
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
| | - Natacha M. Souza Port’s
- Laboratório de Neurofarmacologia Molecular, Universidade de São Paulo, São Paulo 05508-000, Brazil;
| | - Jhully Azevedo dos Santos Pinheiro
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
| | - Ândrea Ribeiro-dos-Santos
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará–R. dos Mundurucus, Belém 66073-000, Brazil
- Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil
| | - Amanda F. Vidal
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil; (G.F.C.); (A.P.S.); (G.C.C.); (C.S.-d.-S.); (T.P.d.S.); (J.A.d.S.P.)
- Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Pará, R. Augusto Correa, Belém 66075-110, Brazil
- ITVDS—Instituto Tecnológico Vale Desenvolvimento Sustentável–R. Boaventura da Silva, Belém 66055-090, Brazil
| |
Collapse
|
56
|
Cao J, Huang M, Guo L, Zhu L, Hou J, Zhang L, Pero A, Ng S, El Gaamouch F, Elder G, Sano M, Goate A, Tcw J, Haroutunian V, Zhang B, Cai D. MicroRNA-195 rescues ApoE4-induced cognitive deficits and lysosomal defects in Alzheimer's disease pathogenesis. Mol Psychiatry 2021; 26:4687-4701. [PMID: 32632205 PMCID: PMC7785685 DOI: 10.1038/s41380-020-0824-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 06/03/2020] [Accepted: 06/16/2020] [Indexed: 12/18/2022]
Abstract
Our recent findings link the apolipoprotein E4 (ApoE4)-specific changes in brain phosphoinositol biphosphate (PIP2) homeostasis to the susceptibility of developing Alzheimer's Disease (AD). In the present study, we have identified miR-195 as a top micro-RNA candidate involved in the ApoE/PIP2 pathway using miRNA profiles in human ROSMAP datasets and mouse microarray studies. Further validation studies have demonstrated that levels of miR-195 are significantly lower in human brain tissue of ApoE4+/- patients with clinical diagnosis of mild cognitive impairment (MCI) or early AD when compared to ApoE4-/- subjects. In addition, brain miR-195 levels are reduced along with disease progression from normal aging to early AD, and cerebrospinal fluid (CSF) miR-195 levels of MCI subjects are positively correlated with cognitive performances as measured by mini-mental status examination (MMSE) and negatively correlated with CSF tau levels, suggesting the involvement of miR-195 in early development of AD with a potential impact on cognition. Similar differences in miR-195 levels are seen in ApoE4+/+ mouse hippocampal brain tissue and cultured neurons when compared to ApoE3+/+ counterparts. Over-expressing miR-195 reduces expression levels of its top predicted target synaptojanin 1 (synj1), a brain PIP2-degrading enzyme. Furthermore, elevating miR-195 ameliorates cognitive deficits, amyloid plaque burden, and tau hyper-phosphorylation in ApoE4+/+ mice. In addition, elevating miR-195 rescues AD-related lysosomal defects in inducible pluripotent stem cells (iPSCs)-derived brain cells of ApoE4+/+ AD subjects while inhibiting miR-195 exacerbates these phenotypes. Together, our data uncover a novel regulatory mechanism of miR-195 targeted at ApoE4-associated brain PIP2 dyshomeostasis, cognitive deficits, and AD pathology.
Collapse
Affiliation(s)
- Jiqing Cao
- James J Peters VA Medical Center, Research & Development, Bronx, NY, 10468, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Min Huang
- James J Peters VA Medical Center, Research & Development, Bronx, NY, 10468, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lei Guo
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Li Zhu
- James J Peters VA Medical Center, Research & Development, Bronx, NY, 10468, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Jianwei Hou
- James J Peters VA Medical Center, Research & Development, Bronx, NY, 10468, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Larry Zhang
- James J Peters VA Medical Center, Research & Development, Bronx, NY, 10468, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Adriana Pero
- James J Peters VA Medical Center, Research & Development, Bronx, NY, 10468, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sabrina Ng
- James J Peters VA Medical Center, Research & Development, Bronx, NY, 10468, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Cornell University, Ithaca, NY, 14850, USA
| | - Farida El Gaamouch
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Gregory Elder
- James J Peters VA Medical Center, Research & Development, Bronx, NY, 10468, USA
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Mary Sano
- James J Peters VA Medical Center, Research & Development, Bronx, NY, 10468, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Alzheimer Disease Rsearch Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Alison Goate
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Julia Tcw
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Vahram Haroutunian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Alzheimer Disease Rsearch Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- James J Peters VA Medical Center, MIRECC, Bronx, NY, 10468, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Dongming Cai
- James J Peters VA Medical Center, Research & Development, Bronx, NY, 10468, USA.
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Alzheimer Disease Rsearch Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Ronald M. Loeb Center for Alzheimer's disease, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
57
|
García-Fonseca Á, Martin-Jimenez C, Barreto GE, Pachón AFA, González J. The Emerging Role of Long Non-Coding RNAs and MicroRNAs in Neurodegenerative Diseases: A Perspective of Machine Learning. Biomolecules 2021; 11:1132. [PMID: 34439798 PMCID: PMC8391852 DOI: 10.3390/biom11081132] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 12/20/2022] Open
Abstract
Neurodegenerative diseases (NDs) are characterized by progressive neuronal dysfunction and death of brain cells population. As the early manifestations of NDs are similar, their symptoms are difficult to distinguish, making the timely detection and discrimination of each neurodegenerative disorder a priority. Several investigations have revealed the importance of microRNAs and long non-coding RNAs in neurodevelopment, brain function, maturation, and neuronal activity, as well as its dysregulation involved in many types of neurological diseases. Therefore, the expression pattern of these molecules in the different NDs have gained significant attention to improve the diagnostic and treatment at earlier stages. In this sense, we gather the different microRNAs and long non-coding RNAs that have been reported as dysregulated in each disorder. Since there are a vast number of non-coding RNAs altered in NDs, some sort of synthesis, filtering and organization method should be applied to extract the most relevant information. Hence, machine learning is considered as an important tool for this purpose since it can classify expression profiles of non-coding RNAs between healthy and sick people. Therefore, we deepen in this branch of computer science, its different methods, and its meaningful application in the diagnosis of NDs from the dysregulated non-coding RNAs. In addition, we demonstrate the relevance of machine learning in NDs from the description of different investigations that showed an accuracy between 85% to 95% in the detection of the disease with this tool. All of these denote that artificial intelligence could be an excellent alternative to help the clinical diagnosis and facilitate the identification diseases in early stages based on non-coding RNAs.
Collapse
Affiliation(s)
- Ángela García-Fonseca
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (Á.G.-F.); (C.M.-J.); (A.F.A.P.)
| | - Cynthia Martin-Jimenez
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (Á.G.-F.); (C.M.-J.); (A.F.A.P.)
| | - George E. Barreto
- Department of Biological Sciences, University of Limerick, V94 T9PX Limerick, Ireland;
| | - Andres Felipe Aristizábal Pachón
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (Á.G.-F.); (C.M.-J.); (A.F.A.P.)
| | - Janneth González
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá 110231, Colombia; (Á.G.-F.); (C.M.-J.); (A.F.A.P.)
| |
Collapse
|
58
|
Fu CH, Han XY, Tong L, Nie PY, Hu YD, Ji LL. miR-142 downregulation alleviates the impairment of spatial learning and memory, reduces the level of apoptosis, and upregulates the expression of pCaMKII and BAI3 in the hippocampus of APP/PS1 transgenic mice. Behav Brain Res 2021; 414:113485. [PMID: 34302879 DOI: 10.1016/j.bbr.2021.113485] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/20/2021] [Accepted: 07/17/2021] [Indexed: 01/20/2023]
Abstract
MicroRNA-142-5p (miR-142-5p) has been found to be dysregulated in several neurodegenerative disorders. However, little is known about the involvement of miR-142-5p in Alzheimer's disease (AD). Brain angiogenesis inhibitor 3 (BAI3), which belongs to the adhesion-G protein-coupled receptor subgroup, contributes to a variety of neuropsychiatric disorders. Despite its very high expression in neurons, the role of BAI3 in AD remains elusive, and its mechanism at the cellular and molecular levels needs to be further elucidated. The current study sought to investigate whether miR-142-5p influenced BAI3 expression and neuronal synaptotoxicity induced by Aβ, both in APP/PS1 transgenic mice and a cellular model of Alzheimer's disease. Altered expression of miR-142-5p was found in the hippocampus of AD mice. Inhibition of miR-142 could upregulate BAI3 expression, enhance neuronal viability and prevent neurons from undergoing apoptosis. In addition, the reduction of phosphorylation of Synapsin I and calcium/calmodulin-dependent protein kinase II (CaMKII), as well as the expression of PSD-95 in the hippocampus of APP/PS1 transgenic mice, were significantly restored by inhibiting miR-142. Meanwhile, the levels of Aβ1-42, β-APP, BACE-1 and PS-1 in cultured neurons were detected, and the effects of inhibiting miR-142 on spatial learning and memory were also observed. Interestingly, we found that BAI3, an important regulator of excitatory synapses, was a potential target gene of miR-142-5p. Collectively, our findings suggest that miR-142 inhibition can alleviate the impairment of spatial learning and memory, reduce the level of apoptosis, and upregulate the expression of pCaMKII and BAI3 in the hippocampus of APP/PS1 transgenic mice; thus, appropriate interference of miR-142 may provide a potential therapeutic approach to rescue cognitive dysfunction in AD patients.
Collapse
Affiliation(s)
- Chang-Hai Fu
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xue-Yan Han
- Department of Neurology, Seventh People's Hospital of Jinan City, Jinan, China
| | - Lei Tong
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Peng-Yin Nie
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yue-Dong Hu
- Department of Ophthalmology, The First Affiliated Hospital of China Medical University, Shenyang, China.
| | - Li-Li Ji
- Department of Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China.
| |
Collapse
|
59
|
Aslani M, Mortazavi-Jahromi SS, Mirshafiey A. Efficient roles of miR-146a in cellular and molecular mechanisms of neuroinflammatory disorders: An effectual review in neuroimmunology. Immunol Lett 2021; 238:1-20. [PMID: 34293378 DOI: 10.1016/j.imlet.2021.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 07/03/2021] [Accepted: 07/15/2021] [Indexed: 12/16/2022]
Abstract
Known as one of the most sophisticated systems of the human body, the nervous system consists of neural cells and controls all parts of the body. It is closely related to the immune system. The effects of inflammation and immune reactions have been observed in the pathogenesis of some neurological disorders. Defined as the gene expression regulators, miRNAs participate in cellular processes. miR-146a is a mediator in the neuroimmune system, leaving substantial effects on the homeostasis of immune and brain cells, neuronal identities acquisition, and immune responses regulation in the nervous system. Its positive efficiency has been proven in modulating inflammatory reactions, hemorrhagic complications, and pain. Moreover, the miR-146a targets play a key role in the pathogenesis of these illnesses. Based on the performance of its targets, miR-146a can have various effects on the disease progress. The abnormal expression/function of miR-146a has been reported in neuroinflammatory disorders. There is research evidence that this molecule qualifies as a desirable biomarker for some disorders and can even be a therapeutic target. This study aims to provide a meticulous review regarding the roles of miR-146a in the pathogenesis and progression of several neuroinflammatory disorders such as multiple sclerosis, amyotrophic lateral sclerosis, Alzheimer's disease, temporal lobe epilepsy, ischemic stroke, etc. The study also considers its eligibility for use as an ideal biomarker and therapeutic target in these diseases. The awareness of these mechanisms can facilitate the disease management/treatment, lead to patients' amelioration, improve the quality of life, and mitigate the risk of death.
Collapse
Affiliation(s)
- Mona Aslani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Abbas Mirshafiey
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
60
|
Nagaraj S, Want A, Laskowska-Kaszub K, Fesiuk A, Vaz S, Logarinho E, Wojda U. Candidate Alzheimer's Disease Biomarker miR-483-5p Lowers TAU Phosphorylation by Direct ERK1/2 Repression. Int J Mol Sci 2021; 22:ijms22073653. [PMID: 33915734 PMCID: PMC8037306 DOI: 10.3390/ijms22073653] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/24/2021] [Accepted: 03/27/2021] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs have been demonstrated as key regulators of gene expression in the etiology of a range of diseases including Alzheimer's disease (AD). Recently, we identified miR-483-5p as the most upregulated miRNA amongst a panel of miRNAs in blood plasma specific to prodromal, early-stage Alzheimer's disease patients. Here, we investigated the functional role of miR-483-5p in AD pathology. Using TargetScan and miRTarBase, we identified the microtubule-associated protein MAPT, often referred to as TAU, and the extracellular signal-regulated kinases 1 and 2 (ERK1 and ERK2), known to phosphorylate TAU, as predicted direct targets of miR-483-5p. Employing several functional assays, we found that miR-483-5p regulates ERK1 and ERK2 at both mRNA and protein levels, resulting in lower levels of phosphorylated forms of both kinases. Moreover, miR-483-5p-mediated repression of ERK1/2 resulted in reduced phosphorylation of TAU protein at epitopes associated with TAU neurofibrillary pathology in AD. These results indicate that upregulation of miR-483-5p can decrease phosphorylation of TAU via ERK pathway, representing a compensatory neuroprotective mechanism in AD pathology. This miR-483-5p/ERK1/TAU axis thus represents a novel target for intervention in AD.
Collapse
Affiliation(s)
- Siranjeevi Nagaraj
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Pasteur 3, 02-093 Warsaw, Poland; (S.N.); (A.W.); (K.L.-K.); (A.F.)
| | - Andrew Want
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Pasteur 3, 02-093 Warsaw, Poland; (S.N.); (A.W.); (K.L.-K.); (A.F.)
| | - Katarzyna Laskowska-Kaszub
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Pasteur 3, 02-093 Warsaw, Poland; (S.N.); (A.W.); (K.L.-K.); (A.F.)
| | - Aleksandra Fesiuk
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Pasteur 3, 02-093 Warsaw, Poland; (S.N.); (A.W.); (K.L.-K.); (A.F.)
- i3S, Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (S.V.); (E.L.)
| | - Sara Vaz
- i3S, Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (S.V.); (E.L.)
| | - Elsa Logarinho
- i3S, Institute for Research and Innovation in Health, University of Porto, 4200-135 Porto, Portugal; (S.V.); (E.L.)
- Aging and Aneuploidy Laboratory, IBMC, Institute of Molecular and Cellular Biology, University of Porto, 4200-135 Porto, Portugal
| | - Urszula Wojda
- Laboratory of Preclinical Testing of Higher Standard, Nencki Institute of Experimental Biology of Polish Academy of Sciences, Pasteur 3, 02-093 Warsaw, Poland; (S.N.); (A.W.); (K.L.-K.); (A.F.)
- Correspondence: ; Tel.: +48-22-5892578
| |
Collapse
|
61
|
Zhan-Qiang H, Hai-Hua Q, Chi Z, Miao W, Cui Z, Zi-Yin L, Jing H, Yi-Wei W. miR-146a aggravates cognitive impairment and Alzheimer disease-like pathology by triggering oxidative stress through MAPK signaling. Neurologia 2021; 38:S0213-4853(21)00022-0. [PMID: 33715884 DOI: 10.1016/j.nrl.2020.12.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/14/2020] [Accepted: 12/26/2020] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION Mir-146a-5p has been widely recognized as a critical regulatory element in the immune response. However, recent studies have shown that miR-146a-5p may also be involved in the development of Alzheimer disease (AD). Regrettably, the related mechanisms are poorly understood. Here, we investigated the effects of miR-146a in mice models and SH-SY5Y cells treated with amyloid β (Aβ)1-42. METHODS To create a model of AD, SH-SY5Y cells were treated with Aβ1-42 and mice received intracerebroventricular injections of Aβ1-42. Then, the transcriptional levels of miR-146a were estimated by real-time PCR. We transiently transfected the miR-146a-5p mimic/inhibitor into cells and mice to study the role of miR-146a. The role of signaling pathways including p38 and reactive oxygen species (ROS) was studied by using specific inhibitors. Aβ and amyloid-beta precursor protein (APP)levels were measured by immunoblotting. Furthermore, Aβ expression was analyzed by immunofluorescence and histochemical examinations. RESULTS Aβ1-42-stimulated SH-SY5Y cells displayed increased transcriptional levels of miR-146a and APP. Moreover, the p38 MAPK signaling pathway and ROS production were activated upon stimulation with a miR-146a-5p mimic. However, treatment with a miR-146a-5p inhibitor decreased the levels of APP, ROS, and p-p38 MAPK. A similar phenomenon was also observed in the animals treated with Aβ1-42, in which miR-146a upregulation increased the expression of Aβ, p-p38, and ROS, while the inhibition of miR-146a had the opposite effect. This suggests that miR-146a increases Aβ deposition and ROS accumulation via the p-p38 signaling pathway. CONCLUSIONS Our research demonstrates that miR-146a-5pa increases Aβ deposition by triggering oxidative stress through activation of MAPK signaling.
Collapse
Affiliation(s)
- H Zhan-Qiang
- Department of General medicine, Affiliated Hospital of Chengde Medical College, Chengde 067000, China
| | - Q Hai-Hua
- Department of Dermatology, Affiliated Hospital of Chengde Medical College, Chengde 067000, China
| | - Z Chi
- Department of Neurology, Affilicated Hospital of Chengde Medical College, Chengde 067000, China
| | - W Miao
- Department of General medicine, Affiliated Hospital of Chengde Medical College, Chengde 067000, China
| | - Z Cui
- Department of General medicine, Affiliated Hospital of Chengde Medical College, Chengde 067000, China
| | - L Zi-Yin
- Department of General medicine, Affiliated Hospital of Chengde Medical College, Chengde 067000, China
| | - H Jing
- Department of General medicine, Affiliated Hospital of Chengde Medical College, Chengde 067000, China
| | - W Yi-Wei
- Department of General medicine, Affiliated Hospital of Chengde Medical College, Chengde 067000, China.
| |
Collapse
|
62
|
Liu XH, Ning FB, Zhao DP, Chang YY, Wu HM, Zhang WH, Yu AL. Role of miR-211 in a PC12 cell model of Alzheimer's disease via regulation of neurogenin 2. Exp Physiol 2021; 106:1061-1071. [PMID: 33527539 DOI: 10.1113/ep088953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 01/28/2021] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? What is the mechanism of miR-211 in an Alzheimer's disease cell model? What is the main finding and its importance? miR-211 was upregulated in an Alzheimer's disease cell model. It targeted neurogenin 2, reduced the activation of the phosphoinositide 3-kinase-Akt signalling pathway, inhibited the proliferation of the Alzheimer's disease cell model and promoted apoptosis. ABSTRACT MicroRNAs (miRs) are aberrantly expressed in Alzheimer's disease (AD) patients. This study was intended to investigate the effect of miR-211 on an AD cell model and the involvement of neurogenin 2 (Ngn2). The appropriate dose and time for the effect of Aβ1-42 on PC12 cells were determined to establish an AD cell model. An effect of miR-211 expression on cell viability, proliferation and apoptosis was detected after cell transfection. Online prediction and a dual luciferase reporter gene assay were utilized to confirm the binding sequence of miR-211 and Ngn2. qRT-PCR and western blot analysis were applied to measure Ngn2 expression. A gain and loss of function assay of miR-211 and Ngn2 was performed, and activation of the phosphoinositide 3-kinase (PI3K)-Akt signaling pathway was detected. The AD cell model was induced by Aβ1-42 treatment. miR-211 expression was significantly enhanced after miR-211 transfection, leading to suppressed proliferation and promotion of apoptosis in Aβ1-42 -treated PC12 cells. In addition, miR-211 could downregulate Ngn2 mRNA and protein expression, while overexpression of Ngn2 could reverse the effects of miR-211 on Aβ1-42 -treated PC12 cells and significantly enhance the phosphorylated Akt and PI3K protein levels. miR-211 could inhibit growth of PC12 cells by suppressing Ngn2 expression and inactivating the PI3K-Akt signalling pathway.
Collapse
Affiliation(s)
- Xin-Hong Liu
- Department of Cerebral ischemic diseases, Tai'an City Central Hospital, Taian, Shandong, 271000, China
| | - Fang-Bo Ning
- Department of Cerebral ischemic diseases, Tai'an City Central Hospital, Taian, Shandong, 271000, China
| | - Da-Peng Zhao
- Department of Cerebral ischemic diseases, Tai'an City Central Hospital, Taian, Shandong, 271000, China
| | - Yan-Yan Chang
- Department of Cerebral ischemic diseases, Tai'an City Central Hospital, Taian, Shandong, 271000, China
| | - Hua-Min Wu
- Department of Imaging, Tai'an City Central Hospital, Taian, Shandong, 271000, China
| | - Wen-Hiu Zhang
- Department of Cerebral ischemic diseases, Tai'an City Central Hospital, Taian, Shandong, 271000, China
| | - Ai-Ling Yu
- Department of Cerebral ischemic diseases, Tai'an City Central Hospital, Taian, Shandong, 271000, China
| |
Collapse
|
63
|
MicroRNAs as Candidate Biomarkers for Alzheimer's Disease. Noncoding RNA 2021; 7:ncrna7010008. [PMID: 33535543 PMCID: PMC7930943 DOI: 10.3390/ncrna7010008] [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: 11/30/2020] [Revised: 01/14/2021] [Accepted: 01/28/2021] [Indexed: 12/17/2022] Open
Abstract
The neurological damage of Alzheimer’s disease (AD) is thought to be irreversible upon onset of dementia-like symptoms, as it takes years to decades for occult pathologic changes to become symptomatic. It is thus necessary to identify individuals at risk for the development of the disease before symptoms manifest in order to provide early intervention. Surrogate markers are critical for early disease detection, stratification of patients in clinical trials, prediction of disease progression, evaluation of response to treatment, and also insight into pathomechanisms. Here, we review the evidence for a number of microRNAs that may serve as biomarkers with possible mechanistic insights into the AD pathophysiologic processes, years before the clinical manifestation of the disease.
Collapse
|
64
|
Gámez-Valero A, Guisado-Corcoll A, Herrero-Lorenzo M, Solaguren-Beascoa M, Martí E. Non-Coding RNAs as Sensors of Oxidative Stress in Neurodegenerative Diseases. Antioxidants (Basel) 2020; 9:E1095. [PMID: 33171576 PMCID: PMC7695195 DOI: 10.3390/antiox9111095] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/03/2020] [Accepted: 11/06/2020] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress (OS) results from an imbalance between the production of reactive oxygen species and the cellular antioxidant capacity. OS plays a central role in neurodegenerative diseases, where the progressive accumulation of reactive oxygen species induces mitochondrial dysfunction, protein aggregation and inflammation. Regulatory non-protein-coding RNAs (ncRNAs) are essential transcriptional and post-transcriptional gene expression controllers, showing a highly regulated expression in space (cell types), time (developmental and ageing processes) and response to specific stimuli. These dynamic changes shape signaling pathways that are critical for the developmental processes of the nervous system and brain cell homeostasis. Diverse classes of ncRNAs have been involved in the cell response to OS and have been targeted in therapeutic designs. The perturbed expression of ncRNAs has been shown in human neurodegenerative diseases, with these changes contributing to pathogenic mechanisms, including OS and associated toxicity. In the present review, we summarize existing literature linking OS, neurodegeneration and ncRNA function. We provide evidences for the central role of OS in age-related neurodegenerative conditions, recapitulating the main types of regulatory ncRNAs with roles in the normal function of the nervous system and summarizing up-to-date information on ncRNA deregulation with a direct impact on OS associated with major neurodegenerative conditions.
Collapse
Affiliation(s)
- Ana Gámez-Valero
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/Casanova 143, 08036 Barcelona, Spain; (A.G.-V.); (A.G.-C.); (M.H.-L.); (M.S.-B.)
| | - Anna Guisado-Corcoll
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/Casanova 143, 08036 Barcelona, Spain; (A.G.-V.); (A.G.-C.); (M.H.-L.); (M.S.-B.)
| | - Marina Herrero-Lorenzo
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/Casanova 143, 08036 Barcelona, Spain; (A.G.-V.); (A.G.-C.); (M.H.-L.); (M.S.-B.)
| | - Maria Solaguren-Beascoa
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/Casanova 143, 08036 Barcelona, Spain; (A.G.-V.); (A.G.-C.); (M.H.-L.); (M.S.-B.)
| | - Eulàlia Martí
- Department de Biomedicina, Facultat de Medicina i Ciències de la Salut, Institut de Neurociències, Universitat de Barcelona, C/Casanova 143, 08036 Barcelona, Spain; (A.G.-V.); (A.G.-C.); (M.H.-L.); (M.S.-B.)
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Ministerio de Ciencia Innovación y Universidades, 28046 Madrid, Spain
| |
Collapse
|
65
|
Boscher E, Hernandez-Rapp J, Petry S, Keraudren R, Rainone S, Loiselle A, Goupil C, Turgeon A, St-Amour I, Planel E, Hébert SS. Advances and Challenges in Understanding MicroRNA Function in Tauopathies: A Case Study of miR-132/212. Front Neurol 2020; 11:578720. [PMID: 33117266 PMCID: PMC7553085 DOI: 10.3389/fneur.2020.578720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/01/2020] [Indexed: 01/08/2023] Open
Abstract
In the past decade, several groups have reported that microRNAs (miRNAs) can participate in the regulation of tau protein at different levels, including its expression, alternative splicing, phosphorylation, and aggregation. These observations are significant, since the abnormal regulation and deposition of tau is associated with nearly 30 neurodegenerative disorders. Interestingly, miRNA profiles go awry in tauopathies such as Alzheimer's disease, progressive supranuclear palsy, and frontotemporal dementia. Understanding the role and impact of miRNAs on tau biology could therefore provide important insights into disease risk, diagnostics, and perhaps therapeutics. In this Perspective article, we discuss recent advances in miRNA research related to tau. While proof-of-principle studies hold promise, physiological validation remains limited. To help fill this gap, we describe herein a pure tauopathy mouse model deficient for the miR-132/212 cluster. This miRNA family is strongly downregulated in human tauopathies and shown to regulate tau in vitro and in vivo. No significant differences in survival, motor deficits or body weight were observed in PS19 mice lacking miR-132/212. Age-specific effects were seen on tau expression and phosphorylation but not aggregation. Moreover, various miR-132/212 targets previously implicated in tau modulation were unaffected (GSK-3β, Foxo3a, Mapk1, p300) or, unexpectedly, reduced (Mapk3, Foxo1, p300, Calpain 2) in miR-132/212-deficient PS19 mice. These observations highlight the challenges of miRNA research in living models, and current limitations of transgenic tau mouse models lacking functional miRNA binding sites. Based on these findings, we finally recommend different strategies to better understand the role of miRNAs in tau physiology and pathology.
Collapse
Affiliation(s)
- Emmanuelle Boscher
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada
| | - Julia Hernandez-Rapp
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada
| | - Serena Petry
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada
| | - Remi Keraudren
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada
| | - Sara Rainone
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada
| | - Andréanne Loiselle
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada
| | - Claudia Goupil
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada
| | - Andréanne Turgeon
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada
| | - Isabelle St-Amour
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada
| | - Emmanuel Planel
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada
| | - Sébastien S Hébert
- Axe Neurosciences, Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada.,Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada
| |
Collapse
|
66
|
Dong X, Zheng D, Nao J. Circulating Exosome microRNAs as Diagnostic Biomarkers of Dementia. Front Aging Neurosci 2020; 12:580199. [PMID: 33093831 PMCID: PMC7506134 DOI: 10.3389/fnagi.2020.580199] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 08/19/2020] [Indexed: 12/28/2022] Open
Abstract
Dementia is a syndrome of acquired cognitive impairment that leads to a significant decline in a patient’s daily life, ability to learn, and the ability to communicate with others. Dementia occurs in many diseases, including Alzheimer’s disease (AD), dementia with Lewy bodies, frontotemporal dementia, and Parkinson’s disease dementia (PDD). Although the analysis of biomarkers in the cerebrospinal fluid (CSF) and peripheral blood physicochemical analysis can indicate neurological impairment, there are currently no sensitive biomarkers for early clinical diagnosis of dementia or for identifying the cause of dementia. Previous studies have suggested that circulating micro (mi)RNAs may be used as biomarkers for diagnosing neurological disorders. However, miRNAs are susceptible to interference by other components in the peripheral circulation, bringing into question the diagnostic value of circulating miRNAs. Exosomes secreted by most cell types contain proteins, mRNAs, and miRNAs that are closely associated with changes in cellular functions. Exosome miRNAs (ex-miRNAs) are highly stable and resistant to degradation. Therefore, these may serve as useful biomarkers for the early clinical diagnosis of dementia. Here, we review studies of ex-miRNAs that commonly cause clinical dementia and explore whether ex-miRNAs may be used as early diagnostic biomarkers of dementia.
Collapse
Affiliation(s)
- Xiaoyu Dong
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Dongming Zheng
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jianfei Nao
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
67
|
Readhead B, Haure-Mirande JV, Mastroeni D, Audrain M, Fanutza T, Kim SH, Blitzer RD, Gandy S, Dudley JT, Ehrlich ME. miR155 regulation of behavior, neuropathology, and cortical transcriptomics in Alzheimer's disease. Acta Neuropathol 2020; 140:295-315. [PMID: 32666270 PMCID: PMC8414561 DOI: 10.1007/s00401-020-02185-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 06/24/2020] [Indexed: 12/19/2022]
Abstract
MicroRNAs are recognized as important regulators of many facets of physiological brain function while also being implicated in the pathogenesis of several neurological disorders. Dysregulation of miR155 is widely reported across a variety of neurodegenerative conditions, including Alzheimer's disease (AD), Parkinson's disease, amyotrophic lateral sclerosis, and traumatic brain injury. In previous work, we observed that experimentally validated miR155 gene targets were consistently enriched among genes identified as differentially expressed across multiple brain tissue and disease contexts. In particular, we found that human herpesvirus-6A (HHV-6A) suppressed miR155, recapitulating reports of miR155 inhibition by HHV-6A in infected T-cells, thyrocytes, and natural killer cells. In earlier studies, we also reported the effects of constitutive deletion of miR155 on accelerating the accumulation of Aβ deposits in 4-month-old APP/PSEN1 mice. Herein, we complete the cumulative characterization of transcriptomic, electrophysiological, neuropathological, and learning behavior profiles from 4-, 8- and 10-month-old WT and APP/PSEN1 mice in the absence or presence of miR155. We also integrated human post-mortem brain RNA-sequences from four independent AD consortium studies, together comprising 928 samples collected from six brain regions. We report that gene expression perturbations associated with miR155 deletion in mouse cortex are in aggregate observed to be concordant with AD-associated changes across these independent human late-onset AD (LOAD) data sets, supporting the relevance of our findings to human disease. LOAD has recently been formulated as the clinicopathological manifestation of a multiplex of genetic underpinnings and pathophysiological mechanisms. Our accumulated data are consistent with such a formulation, indicating that miR155 may be uniquely positioned at the intersection of at least four components of this LOAD "multiplex": (1) innate immune response pathways; (2) viral response gene networks; (3) synaptic pathology; and (4) proamyloidogenic pathways involving the amyloid β peptide (Aβ).
Collapse
Affiliation(s)
- Ben Readhead
- Arizona State University-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, 85281, USA
- Icahn Institute of Genomic Sciences and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - Diego Mastroeni
- Arizona State University-Banner Neurodegenerative Disease Research Center, Arizona State University, Tempe, AZ, 85281, USA
| | - Mickael Audrain
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Tomas Fanutza
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Soong H Kim
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Robert D Blitzer
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sam Gandy
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Alzheimer's Disease Research Center, Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Mount Sinai Center for Cognitive Health and NFL Neurological Care, Department of Neurology, New York, NY, 10029, USA
- James J. Peters VA Medical Center, 130 West Kingsbridge Road, New York, NY, 10468, USA
| | - Joel T Dudley
- Icahn Institute of Genomic Sciences and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Michelle E Ehrlich
- Icahn Institute of Genomic Sciences and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
68
|
Wei W, Wang ZY, Ma LN, Zhang TT, Cao Y, Li H. MicroRNAs in Alzheimer's Disease: Function and Potential Applications as Diagnostic Biomarkers. Front Mol Neurosci 2020; 13:160. [PMID: 32973449 PMCID: PMC7471745 DOI: 10.3389/fnmol.2020.00160] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/05/2020] [Indexed: 12/14/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia. Although the incidence of AD is high, the rates of diagnosis and treatment are relatively low. Moreover, effective means for the diagnosis and treatment of AD are still lacking. MicroRNAs (miRNAs, miRs) are non-coding RNAs that play regulatory roles by targeting mRNAs. The expression of miRNAs is conserved, temporal, and tissue-specific. Impairment of microRNA function is closely related to AD pathogenesis, including the beta-amyloid and tau hallmarks of AD, and there is evidence that the expression of some microRNAs differs significantly between healthy people and AD patients. These properties of miRNAs endow them with potential diagnostic and therapeutic value in the treatment of this debilitating disease. This review provides comprehensive information about the regulatory function of miRNAs in AD, as well as potential applications as diagnostic biomarkers.
Collapse
Affiliation(s)
- Wei Wei
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China.,Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhi-Yong Wang
- Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li-Na Ma
- Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ting-Ting Zhang
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, China.,Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Cao
- Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hao Li
- Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
69
|
MicroRNA Alterations in a Tg501 Mouse Model of Prion Disease. Biomolecules 2020; 10:biom10060908. [PMID: 32549330 PMCID: PMC7355645 DOI: 10.3390/biom10060908] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/31/2020] [Accepted: 06/12/2020] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) may contribute to the development and pathology of many neurodegenerative diseases, including prion diseases. They are also promising biomarker candidates due to their stability in body fluids. We investigated miRNA alterations in a Tg501 mouse model of prion diseases that expresses a transgene encoding the goat prion protein (PRNP). Tg501 mice intracranially inoculated with mouse-adapted goat scrapie were compared with age-matched, mock inoculated controls in preclinical and clinical stages. Small RNA sequencing from the cervical spinal cord indicated that miR-223-3p, miR-151-3p, and miR-144-5p were dysregulated in scrapie-inoculated animals before the onset of symptoms. In clinical-stage animals, 23 significant miRNA alterations were found. These miRNAs were predicted to modify the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways including prion disease, extracellular matrix interactions, glutaminergic synapse, axon guidance, and transforming growth factor-beta signaling. MicroRNAs miR-146a-5p (up in cervical spinal cord) and miR-342-3p (down in cervical spinal cord, cerebellum and plasma), both indicated in neurodegenerative diseases earlier, were verified by quantitative real-time polymerase chain reaction (qRT-PCR). Minimal changes observed before the disease onset suggests that most miRNA alterations observed here are driven by advanced prion-associated pathology, possibly limiting their use as diagnostic markers. However, the results encourage further mechanistic studies on miRNA-regulated pathways involved in these neurodegenerative conditions.
Collapse
|
70
|
Serpente M, Fenoglio C, D'Anca M, Arcaro M, Sorrentino F, Visconte C, Arighi A, Fumagalli GG, Porretti L, Cattaneo A, Ciani M, Zanardini R, Benussi L, Ghidoni R, Scarpini E, Galimberti D. MiRNA Profiling in Plasma Neural-Derived Small Extracellular Vesicles from Patients with Alzheimer's Disease. Cells 2020; 9:cells9061443. [PMID: 32531989 PMCID: PMC7349735 DOI: 10.3390/cells9061443] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/04/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023] Open
Abstract
Small extracellular vesicles (EVs) are able to pass from the central nervous system (CNS) into peripheral blood and contain molecule markers of their parental origin. The aim of our study was to isolate and characterize total and neural-derived small EVs (NDEVs) and their micro RNA (miRNA) cargo in Alzheimer's disease (AD) patients. Small NDEVs were isolated from plasma in a population consisting of 40 AD patients and 40 healthy subjects (CTRLs) using high throughput Advanced TaqMan miRNA OpenArrays®, which enables the simultaneous determination of 754 miRNAs. MiR-23a-3p, miR-223-3p, miR-100-3p and miR-190-5p showed a significant dysregulation in small NDEVs from AD patients as compared with controls (1.16 ± 0.49 versus 7.54 ± 2.5, p = 0.026; 9.32 ± 2.27 versus 0.66 ± 0.18, p <0.0001; 0.069 ± 0.01 versus 0.5 ± 0.1, p < 0.0001 and 2.9 ± 1.2 versus 1.93 ± 0.9, p < 0.05, respectively). A further validation analysis confirmed that miR-23a-3p, miR-223-3p and miR-190a-5p levels in small NDEVs from AD patients were significantly upregulated as compared with controls (p = 0.008; p = 0.016; p = 0.003, respectively) whereas miR-100-3p levels were significantly downregulated (p = 0.008). This is the first study that carries out the comparison between total plasma small EV population and NDEVs, demonstrating the presence of a specific AD NDEV miRNA signature.
Collapse
Affiliation(s)
- Maria Serpente
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, University of Milan, 20122 Milan, Italy
| | - Chiara Fenoglio
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, University of Milan, 20122 Milan, Italy
| | - Marianna D'Anca
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, University of Milan, 20122 Milan, Italy
| | - Marina Arcaro
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Federica Sorrentino
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, Dino Ferrari Center, CRC Molecular Basis of Neuro-Psycho-Geriatrics Diseases, University of Milan, 20122 Milan, Italy
| | - Caterina Visconte
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Andrea Arighi
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Giorgio G Fumagalli
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Laura Porretti
- Flow Cytometry Service, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Alessandra Cattaneo
- Department of Transfusion Medicine and Haematology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Miriam Ciani
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Roberta Zanardini
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Luisa Benussi
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Roberta Ghidoni
- Molecular Markers Laboratory, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, 25125 Brescia, Italy
| | - Elio Scarpini
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, Dino Ferrari Center, CRC Molecular Basis of Neuro-Psycho-Geriatrics Diseases, University of Milan, 20122 Milan, Italy
| | - Daniela Galimberti
- Neurodegenerative Diseases Unit, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, Dino Ferrari Center, CRC Molecular Basis of Neuro-Psycho-Geriatrics Diseases, University of Milan, 20122 Milan, Italy
| |
Collapse
|
71
|
Xie C, Wang H, Zhang Y, Wei Y. Neuroprotective effects of miR-142-5p downregulation against isoflurane-induced neurological impairment. Diagn Pathol 2020; 15:70. [PMID: 32505188 PMCID: PMC7275573 DOI: 10.1186/s13000-020-00978-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 05/19/2020] [Indexed: 01/02/2023] Open
Abstract
Background Isoflurane can lead to neuron damage to the developing brain, resulting in learning and memory disability. The aim of this study was to investigate the role of miR-142-5p on isoflurane-induced neurological impairment. Methods The Morris water maze (MWM) test was performed to evaluate spatial learning and memory of rats. The expression level of miR-142-5p was measured using qRT-PCR. MTT assay was used to calculate the viability of hippocampal neuronal cells. The cell apoptosis was analyzed using Flow cytometric assay. Results Isoflurane treatment led to the increase of neurological function score and escape latency, and the reduction of time spent in the original quadrant in rats. The expression level of miR-142-5p was increased significantly in isoflurane-treated rats. MiR-142-5p downregulation protected against isoflurane-induced neurological impairment, which was reflected by the decrease of neurological function score and escape latency, and the increase of time spent in the original quadrant. In vitro, downregulation of miR-142-5p alleviated isoflurane-induced neuron cell viability inhibition, and relieved isoflurane-induced cell apoptosis. Conclusions MiR-142-5p downregulation plays a neuroprotective role in protecting against isoflurane-induced neurological impairment through regulating neuron cell viability and apoptosis. It provides a theoretical basis for the investigation of the mechanism underlying the effect on isoflurane-induced neurological impairment.
Collapse
Affiliation(s)
- Cuili Xie
- Department of Anesthesiology, Jining No. 1 People's Hospital, No. 6, Jiankang Road, Jining, Shandong, 272011, People's Republic of China.,Jining Medical University, Jining, Shandong, 272011, People's Republic of China
| | - Hongyue Wang
- Department of Anesthesiology, Jining No. 1 People's Hospital, No. 6, Jiankang Road, Jining, Shandong, 272011, People's Republic of China.,Jining Medical University, Jining, Shandong, 272011, People's Republic of China
| | - Yu Zhang
- Department of Anesthesiology, Jining No. 1 People's Hospital, No. 6, Jiankang Road, Jining, Shandong, 272011, People's Republic of China.,Jining Medical University, Jining, Shandong, 272011, People's Republic of China
| | - Yanhua Wei
- Department of Anesthesiology, Jining No. 1 People's Hospital, No. 6, Jiankang Road, Jining, Shandong, 272011, People's Republic of China. .,Jining Medical University, Jining, Shandong, 272011, People's Republic of China.
| |
Collapse
|
72
|
Ahmadi S, Zobeiri M, Bradburn S. Molecular mechanisms underlying actions of certain long noncoding RNAs in Alzheimer's disease. Metab Brain Dis 2020; 35:681-693. [PMID: 32185592 DOI: 10.1007/s11011-020-00564-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 03/05/2020] [Indexed: 01/08/2023]
Abstract
Long non-coding RNAs (lncRNAs) are a group of non-protein coding RNAs that have more than 200 nucleotides. LncRNAs play an important role in the regulation of protein-coding genes at the transcriptional and post-transcriptional levels. They are found in most organs, with a high prevalence in the central nervous system. Accumulating data suggests that lncRNAs are involved in various neurodegenerative disorders, including the onset and progression of Alzheimer's disease (AD). Recent insights suggest lncRNAs, such as BACE1-AS, 51A, 17A, NDM29 and AS-UCHL1, are dysregulated in AD tissues. Furthermore, there are ongoing efforts to explore the clinical usability of lncRNAs as biomarkers in the disease. In this review, we explore the mechanisms by which aberrant expressions of the most studied lncRNAs contribute to the neuropathologies associated with AD, including amyloid β plaques and neurofibrillary tangles. Understanding the molecular mechanisms of lncRNAs in patients with AD will reveal novel diagnosis strategies and more effective therapeutic targets.
Collapse
Affiliation(s)
- Shamseddin Ahmadi
- Department of Biological Science, Faculty of Science, University of Kurdistan, P.O. Box 416, Sanandaj, Iran.
| | - Mohammad Zobeiri
- Department of Biological Science, Faculty of Science, University of Kurdistan, P.O. Box 416, Sanandaj, Iran
| | - Steven Bradburn
- Bioscience Research Centre, Manchester Metropolitan University, Manchester, UK
| |
Collapse
|
73
|
miR-16-5p is upregulated by amyloid β deposition in Alzheimer's disease models and induces neuronal cell apoptosis through direct targeting and suppression of BCL-2. Exp Gerontol 2020; 136:110954. [PMID: 32320719 DOI: 10.1016/j.exger.2020.110954] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 04/08/2020] [Accepted: 04/15/2020] [Indexed: 01/09/2023]
Abstract
Alzheimer's disease (AD) is the most common form of dementia with irreversible neurodegeneration. Accumulation of amyloid beta (Aβ) in the brain is considered to be a major cause of neuronal cell death in AD, but the neurotoxic mechanism of Aβ is not yet fully understood. Here, we focused on the role of microRNAs (miRNAs) in Aβ-induced neuronal cell death. In microarray and RT-qPCR analysis of plasma miRNAs obtained from 5 familiar AD mutations (5xFAD) and wild-type (WT) mice of various ages, miR-16-5p showed a significant age-related change that was accompanied by neuronal cell death in the brain tissue of 5xFAD mice. In addition, increased miR-16-5p was prominent near Aβ plaque-deposition sites in 5xFAD mouse brains. Aβ treatment induced miR-16-5p upregulation and apoptosis in primary cultured mouse cortical neurons and the SH-SY5Y human neuroblastoma cell line. In silico analysis and reporter gene assays indicated that miR-16-5p directly targets the mRNA encoding the anti-apoptotic factor, B cell lymphoma-2 (BCL-2), in the neuronal cell line. Overexpression of miR-16-5p in SH-SY5Y cells downregulated BCL-2 expression and induced apoptosis. These results collectively suggest that the miR-16-5p/BCL-2 axis plays an important role for neuronal cell apoptosis in AD.
Collapse
|
74
|
Wu YY, Kuo HC. Functional roles and networks of non-coding RNAs in the pathogenesis of neurodegenerative diseases. J Biomed Sci 2020; 27:49. [PMID: 32264890 PMCID: PMC7140545 DOI: 10.1186/s12929-020-00636-z] [Citation(s) in RCA: 150] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 02/26/2020] [Indexed: 12/13/2022] Open
Abstract
Recent transcriptome analyses have revealed that noncoding RNAs (ncRNAs) are broadly expressed in mammalian cells and abundant in the CNS, with tissue and cell type-specific expression patterns. Moreover, ncRNAs have been found to intricately and dynamically regulate various signaling pathways in neurodegeneration. As such, some antisense transcripts and microRNAs are known to directly affect neurodegeneration in disease contexts. The functions of ncRNAs in pathogenesis are unique for each disorder, as are the pertinent networks of ncRNA/miRNA/mRNA that mediate these functions. Thus, further understanding of ncRNA biogenesis and effects might aid the discovery of diagnostic biomarkers or development of effective therapeutics for neurodegenerative disorders. Here, we review the ncRNAs that have so far been identified in major neurodegenerative disease etiology and the mechanisms that link ncRNAs with disease-specific phenotypes, such as HTT aggregation in HD, α-synuclein in PD, and Aβ plaques and hyperphosphorylated Tau in AD. We also summarize the known lncRNA/miRNA/mRNA networks that participate in neurodegenerative diseases, and we discuss ncRNA-related treatments shown to delay disease onset and prolong lifespan in rodent models.
Collapse
Affiliation(s)
- Yi-Ying Wu
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei, 11529, Taiwan
| | - Hung-Chih Kuo
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Sec. 2, Academia Road, Nankang, Taipei, 11529, Taiwan. .,Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
75
|
Sierksma A, Lu A, Mancuso R, Fattorelli N, Thrupp N, Salta E, Zoco J, Blum D, Buée L, De Strooper B, Fiers M. Novel Alzheimer risk genes determine the microglia response to amyloid-β but not to TAU pathology. EMBO Mol Med 2020; 12:e10606. [PMID: 31951107 PMCID: PMC7059012 DOI: 10.15252/emmm.201910606] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 12/20/2019] [Accepted: 12/20/2019] [Indexed: 12/20/2022] Open
Abstract
Polygenic risk scores have identified that genetic variants without genome-wide significance still add to the genetic risk of developing Alzheimer's disease (AD). Whether and how subthreshold risk loci translate into relevant disease pathways is unknown. We investigate here the involvement of AD risk variants in the transcriptional responses of two mouse models: APPswe/PS1L166P and Thy-TAU22. A unique gene expression module, highly enriched for AD risk genes, is specifically responsive to Aβ but not TAU pathology. We identify in this module 7 established AD risk genes (APOE, CLU, INPP5D, CD33, PLCG2, SPI1, and FCER1G) and 11 AD GWAS genes below the genome-wide significance threshold (GPC2, TREML2, SYK, GRN, SLC2A5, SAMSN1, PYDC1, HEXB, RRBP1, LYN, and BLNK), that become significantly upregulated when exposed to Aβ. Single microglia sequencing confirms that Aβ, not TAU, pathology induces marked transcriptional changes in microglia, including increased proportions of activated microglia. We conclude that genetic risk of AD functionally translates into different microglia pathway responses to Aβ pathology, placing AD genetic risk downstream of the amyloid pathway but upstream of TAU pathology.
Collapse
Affiliation(s)
- Annerieke Sierksma
- VIB Center for Brain & Disease ResearchLeuvenBelgium
- Laboratory for the Research of Neurodegenerative DiseasesDepartment of NeurosciencesLeuven Brain Institute (LBI)KU Leuven (University of Leuven)LeuvenBelgium
| | - Ashley Lu
- VIB Center for Brain & Disease ResearchLeuvenBelgium
- Laboratory for the Research of Neurodegenerative DiseasesDepartment of NeurosciencesLeuven Brain Institute (LBI)KU Leuven (University of Leuven)LeuvenBelgium
| | - Renzo Mancuso
- VIB Center for Brain & Disease ResearchLeuvenBelgium
- Laboratory for the Research of Neurodegenerative DiseasesDepartment of NeurosciencesLeuven Brain Institute (LBI)KU Leuven (University of Leuven)LeuvenBelgium
| | - Nicola Fattorelli
- VIB Center for Brain & Disease ResearchLeuvenBelgium
- Laboratory for the Research of Neurodegenerative DiseasesDepartment of NeurosciencesLeuven Brain Institute (LBI)KU Leuven (University of Leuven)LeuvenBelgium
| | - Nicola Thrupp
- VIB Center for Brain & Disease ResearchLeuvenBelgium
- Laboratory for the Research of Neurodegenerative DiseasesDepartment of NeurosciencesLeuven Brain Institute (LBI)KU Leuven (University of Leuven)LeuvenBelgium
| | - Evgenia Salta
- VIB Center for Brain & Disease ResearchLeuvenBelgium
- Laboratory for the Research of Neurodegenerative DiseasesDepartment of NeurosciencesLeuven Brain Institute (LBI)KU Leuven (University of Leuven)LeuvenBelgium
| | - Jesus Zoco
- VIB Center for Brain & Disease ResearchLeuvenBelgium
- Laboratory for the Research of Neurodegenerative DiseasesDepartment of NeurosciencesLeuven Brain Institute (LBI)KU Leuven (University of Leuven)LeuvenBelgium
| | - David Blum
- INSERM, CHU Lille, LabEx DISTALZ, UMR‐S 1172, Alzheimer & TauopathiesUniversité LilleLilleFrance
| | - Luc Buée
- INSERM, CHU Lille, LabEx DISTALZ, UMR‐S 1172, Alzheimer & TauopathiesUniversité LilleLilleFrance
| | - Bart De Strooper
- VIB Center for Brain & Disease ResearchLeuvenBelgium
- Laboratory for the Research of Neurodegenerative DiseasesDepartment of NeurosciencesLeuven Brain Institute (LBI)KU Leuven (University of Leuven)LeuvenBelgium
- UK Dementia Research InstituteUniversity College LondonLondonUK
| | - Mark Fiers
- VIB Center for Brain & Disease ResearchLeuvenBelgium
- Laboratory for the Research of Neurodegenerative DiseasesDepartment of NeurosciencesLeuven Brain Institute (LBI)KU Leuven (University of Leuven)LeuvenBelgium
| |
Collapse
|
76
|
Sandau US, Wiedrick JT, Smith SJ, McFarland TJ, Lusardi TA, Lind B, Harrington CA, Lapidus JA, Galasko DR, Quinn JF, Saugstad JA. Performance of Validated MicroRNA Biomarkers for Alzheimer's Disease in Mild Cognitive Impairment. J Alzheimers Dis 2020; 78:245-263. [PMID: 32955460 PMCID: PMC9262405 DOI: 10.3233/jad-200396] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Cerebrospinal fluid (CSF) microRNA (miRNA) biomarkers of Alzheimer's disease (AD) have been identified, but have not been evaluated in prodromal AD, including mild cognitive impairment (MCI). OBJECTIVE To assess whether a set of validated AD miRNA biomarkers in CSF are also sensitive to early-stage pathology as exemplified by MCI diagnosis. METHODS We measured the expression of 17 miRNA biomarkers for AD in CSF samples from AD, MCI, and cognitively normal controls (NC). We then examined classification performance of the miRNAs individually and in combination. For each miRNA, we assessed median expression in each diagnostic group and classified markers as trending linearly, nonlinearly, or lacking any trend across the three groups. For trending miRNAs, we assessed multimarker classification performance alone and in combination with apolipoprotein E ɛ4 allele (APOEɛ4) genotype and amyloid-β42 to total tau ratio (Aβ42:T-Tau). We identified predicted targets of trending miRNAs using pathway analysis. RESULTS Five miRNAs showed a linear trend of decreasing median expression across the ordered diagnoses (control to MCI to AD). The trending miRNAs jointly predicted AD with area under the curve (AUC) of 0.770, and MCI with AUC of 0.705. Aβ42:T-Tau alone predicted MCI with AUC of 0.758 and the AUC improved to 0.813 (p = 0.051) after adding the trending miRNAs. Multivariate correlation of the five trending miRNAs with Aβ42:T-Tau was weak. CONCLUSION Selected miRNAs combined with Aβ42:T-Tau improved classification performance (relative to protein biomarkers alone) for MCI, despite a weak correlation with Aβ42:T-Tau. Together these data suggest that that these miRNAs carry novel information relevant to AD, even at the MCI stage. Preliminary target prediction analysis suggests novel roles for these biomarkers.
Collapse
Affiliation(s)
- Ursula S. Sandau
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Jack T. Wiedrick
- Biostatistics & Design Program, Oregon Health & Science University, Portland, OR, USA
| | - Sierra J. Smith
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Trevor J. McFarland
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Theresa A. Lusardi
- Knight Cancer Institute Cancer Early Detection Advanced Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Babett Lind
- Department of Neurology, Layton Aging and Alzheimer’s Center, Oregon Health & Science University, Portland, OR, USA
| | | | - Jodi A. Lapidus
- Biostatistics & Design Program, Oregon Health & Science University, Portland, OR, USA
- Oregon Health & Science University–Portland State University School of Public Health, Portland, OR, USA
| | - Douglas R. Galasko
- Department of Neurosciences, University of California San Diego, La Jolla, CA, USA
| | - Joseph F. Quinn
- Parkinson Center and Movement Disorders Program, School of Medicine, Oregon Health & Science University, Portland, OR, USA
- Portland VAMC Parkinson’s Disease Research, Education, and Clinical Center, Portland, OR, USA
| | - Julie A. Saugstad
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
77
|
Meta-Analysis of Gene Expression Changes in the Blood of Patients with Mild Cognitive Impairment and Alzheimer's Disease Dementia. Int J Mol Sci 2019; 20:ijms20215403. [PMID: 31671574 PMCID: PMC6862214 DOI: 10.3390/ijms20215403] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/25/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022] Open
Abstract
Background: Dementia is a major public health concern affecting approximately 47 million people worldwide. Mild cognitive impairment (MCI) is one form of dementia that affects an individual’s memory with or without affecting their daily life. Alzheimer’s disease dementia (ADD) is a more severe form of dementia that usually affects elderly individuals. It remains unclear whether MCI is a distinct disorder from or an early stage of ADD. Methods: Gene expression data from blood were analyzed to identify potential biomarkers that may be useful for distinguishing between these two forms of dementia. Results: A meta-analysis revealed 91 genes dysregulated in individuals with MCI and 387 genes dysregulated in ADD. Pathway analysis identified seven pathways shared between MCI and ADD and nine ADD-specific pathways. Fifteen transcription factors were associated with MCI and ADD, whereas seven transcription factors were specific for ADD. Mir-335-5p was specific for ADD, suggesting that it may be useful as a biomarker. Diseases that are associated with MCI and ADD included developmental delays, cognition impairment, and movement disorders. Conclusion: These results provide a better molecular understanding of peripheral changes that occur in MCI and ADD patients and may be useful in the identification of diagnostic and prognostic biomarkers.
Collapse
|
78
|
Jain G, Stuendl A, Rao P, Berulava T, Pena Centeno T, Kaurani L, Burkhardt S, Delalle I, Kornhuber J, Hüll M, Maier W, Peters O, Esselmann H, Schulte C, Deuschle C, Synofzik M, Wiltfang J, Mollenhauer B, Maetzler W, Schneider A, Fischer A. A combined miRNA-piRNA signature to detect Alzheimer's disease. Transl Psychiatry 2019; 9:250. [PMID: 31591382 PMCID: PMC6779890 DOI: 10.1038/s41398-019-0579-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 06/05/2019] [Accepted: 08/18/2019] [Indexed: 01/03/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder causing huge emotional and economic burden to our societies. An effective therapy has not been implicated yet, which is in part also due to the fact that pathological changes occur years before clinical symptoms manifest. Thus, there is a great need for the development of a translatable biomarker. Recent evidence highlights microRNAs as candidate biomarkers. In this study, we use next-generation sequencing to study the small noncoding RNAome (sncRNAome) in exosomes derived from human cerebrospinal fluid (CSF). We show that the sncRNAome from CSF-derived exosomes is dominated not only by microRNAs (miRNAs) but also by PIWI-interacting RNAs (piRNAs). We define a combined signature consisting of three miRNAs and three piRNAs that are suitable to detect AD with an AUC of 0.83 in a replication cohort and furthermore predict the conversion of mild-cognitive impaired (MCI) patients to AD dementia with an AUC of 0.86 for the piRNA signature. When combining the smallRNA signature with pTau and Aβ 42/40 ratio the AUC reaches 0.98. Our study reports a novel exosomal small noncoding RNA signature to detect AD pathology and provides the first evidence that in addition to miRNAs, piRNAs should also be considered as a candidate biomarker for AD.
Collapse
Affiliation(s)
- Gaurav Jain
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37075 Göttingen, Germany
| | - Anne Stuendl
- 0000 0004 0438 0426grid.424247.3Translational Dementia Research, German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany
| | - Pooja Rao
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37075 Göttingen, Germany
| | - Tea Berulava
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37075 Göttingen, Germany
| | - Tonatiuh Pena Centeno
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37075 Göttingen, Germany ,Bioinformatics Unit, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37075 Goettingen, Germany
| | - Lalit Kaurani
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37075 Göttingen, Germany
| | - Susanne Burkhardt
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37075 Göttingen, Germany
| | - Ivana Delalle
- 0000 0004 0367 5222grid.475010.7Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Johannes Kornhuber
- 0000 0001 2107 3311grid.5330.5Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Michael Hüll
- 0000 0000 9428 7911grid.7708.8Center for Geriatric Medicine and Gerontology, University Medical Center Freiburg, 79106 Freiburg, Germany ,0000 0000 9428 7911grid.7708.8Department of Psychiatry and Psychotherapy, University Medical Centre Freiburg, 79106 Freiburg, Germany
| | - Wolfgang Maier
- 0000 0001 2240 3300grid.10388.32Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, 53127 Bonn, Germany
| | - Oliver Peters
- 0000 0001 2248 7639grid.7468.dDepartment of Psychiatry, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 12200 Berlin, Germany ,0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), 12203 Berlin, Germany ,0000 0001 1014 0849grid.419491.0Memory Clinic and Dementia Prevention Center, Experimental and Clinical Research Center (ECRC), 13125 Berlin, Germany
| | - Hermann Esselmann
- 0000 0001 0482 5331grid.411984.1Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), 37075 Göttingen, Germany
| | - Claudia Schulte
- 0000 0001 2190 1447grid.10392.39Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany ,0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), 72076 Tuebingen, Germany
| | - Christian Deuschle
- 0000 0001 2190 1447grid.10392.39Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany ,0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), 72076 Tuebingen, Germany
| | - Mathis Synofzik
- 0000 0001 2190 1447grid.10392.39Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany ,0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), 72076 Tuebingen, Germany
| | - Jens Wiltfang
- 0000 0001 0482 5331grid.411984.1Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), 37075 Göttingen, Germany ,0000000123236065grid.7311.4iBiMED, Medical Sciences Department, University of Aveiro, Aveiro, Portugal ,German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37075 Göttingen, Germany
| | - Brit Mollenhauer
- 0000 0001 0482 5331grid.411984.1Department of Neurology, University Medical Center Göttingen (UMG), 37075 Göttingen, Germany ,grid.440220.0Paracelsus-Elena-Klinik, 34128 Kassel, Germany
| | - Walter Maetzler
- 0000 0001 2190 1447grid.10392.39Center of Neurology, Department of Neurodegeneration and Hertie-Institute for Clinical Brain Research, University of Tuebingen, 72076 Tuebingen, Germany ,0000 0004 0438 0426grid.424247.3German Center for Neurodegenerative Diseases (DZNE), 72076 Tuebingen, Germany ,0000 0004 0646 2097grid.412468.dDepartment of Neurology, University Hospital Schleswig-Holstein, 24105 Kiel, Germany
| | - Anja Schneider
- Translational Dementia Research, German Center for Neurodegenerative Diseases (DZNE), 53127, Bonn, Germany. .,Department of Neurodegenerative Diseases and Geriatric Psychiatry, University of Bonn, 53127, Bonn, Germany.
| | - Andre Fischer
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases (DZNE) Goettingen, 37075, Göttingen, Germany. .,Department of Psychiatry and Psychotherapy, University Medical Center Göttingen (UMG), 37075, Göttingen, Germany.
| |
Collapse
|
79
|
Weldon Furr J, Morales-Scheihing D, Manwani B, Lee J, McCullough LD. Cerebral Amyloid Angiopathy, Alzheimer's Disease and MicroRNA: miRNA as Diagnostic Biomarkers and Potential Therapeutic Targets. Neuromolecular Med 2019; 21:369-390. [PMID: 31586276 DOI: 10.1007/s12017-019-08568-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 09/04/2019] [Indexed: 12/14/2022]
Abstract
The protein molecules must fold into unique conformations to acquire functional activity. Misfolding, aggregation, and deposition of proteins in diverse organs, the so-called "protein misfolding disorders (PMDs)", represent the conformational diseases with highly ordered assemblies, including oligomers and fibrils that are linked to neurodegeneration in brain illnesses such as cerebral amyloid angiopathy (CAA) and Alzheimer's disease (AD). Recent studies have revealed several aspects of brain pathology in CAA and AD, but both the classification and underlying mechanisms need to be further refined. MicroRNAs (miRNAs) are critical regulators of gene expression at the post-transcriptional level. Increasing evidence with the advent of RNA sequencing technology suggests possible links between miRNAs and these neurodegenerative disorders. To provide insights on the small RNA-mediated regulatory circuitry and the translational significance of miRNAs in PMDs, this review will discuss the characteristics and mechanisms of the diseases and summarize circulating or tissue-resident miRNAs associated with AD and CAA.
Collapse
Affiliation(s)
- J Weldon Furr
- BRAINS Research Laboratory, University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Diego Morales-Scheihing
- BRAINS Research Laboratory, University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Bharti Manwani
- BRAINS Research Laboratory, University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Juneyoung Lee
- BRAINS Research Laboratory, University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Louise D McCullough
- BRAINS Research Laboratory, University of Texas McGovern Medical School, Houston, TX, 77030, USA.
| |
Collapse
|
80
|
Silvestro S, Bramanti P, Mazzon E. Role of miRNAs in Alzheimer's Disease and Possible Fields of Application. Int J Mol Sci 2019; 20:E3979. [PMID: 31443326 PMCID: PMC6720959 DOI: 10.3390/ijms20163979] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/07/2019] [Accepted: 08/14/2019] [Indexed: 01/02/2023] Open
Abstract
miRNAs (or microRNAs) are a class of single-stranded RNA molecules, responsible for post-transcriptional gene silencing through binding to the coding region as well as 3' and 5' untranslated region of target genes. About 70% of experimentally detectable miRNAs are expressed in the brain and some studies suggest that miRNAs are intimately involved in synaptic function and in specific signals during memory formation. More and more evidence demonstrates the possible involvement of miRNAs in Alzheimer's disease (AD). AD is the most common form of senile dementia, a disease that affects memory and cognitive functions. It is a neurodegenerative disorder characterized by loss of synapses, extracellular amyloid plaques composed of the amyloid-β peptide (Aβ), and intracellular aggregates of hyperphosphorylated TAU protein. This review aims to provide an overview of the in vivo studies of the last 5 years in the literature describing the role of the different miRNAs involved in AD. miRNAs hold huge potential as diagnostic and prognostic biomarkers and, at the same time, their modulation could be a potential therapeutic strategy against AD.
Collapse
Affiliation(s)
- Serena Silvestro
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Placido Bramanti
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy
| | - Emanuela Mazzon
- IRCCS Centro Neurolesi "Bonino-Pulejo", Via Provinciale Palermo, Contrada Casazza, 98124 Messina, Italy.
| |
Collapse
|
81
|
Hu YB, Zhang YF, Wang H, Ren RJ, Cui HL, Huang WY, Cheng Q, Chen HZ, Wang G. miR-425 deficiency promotes necroptosis and dopaminergic neurodegeneration in Parkinson's disease. Cell Death Dis 2019; 10:589. [PMID: 31383850 PMCID: PMC6683159 DOI: 10.1038/s41419-019-1809-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 06/15/2019] [Accepted: 07/03/2019] [Indexed: 12/21/2022]
Abstract
A major hallmark of Parkinson’s disease (PD) is the degeneration of dopaminergic neurons in the substantia nigra, and the causative mechanism is thought to be the activation of programmed neuronal death. Necroptosis is a regulated process of cell death triggered by RIPK1. Although the pathophysiology of PD has been studied extensively, the cellular mechanism underlying dopaminergic neuron death remains unclear. In this study, we detected a specific miRNA, miR-425, in response to MPTP toxicity and dopaminergic degeneration. In MPTP-treated mice, we observed necroptosis activation and miR-425 deficiency in the substantia nigra, which is correlated with dopaminergic neuron loss. This miRNA targeted RIPK1 transcripts and promoted the phosphorylation of MLKL and necroptosis. Similarly, in the brains of PD patients, miR-425 deficiency and necroptosis activation were also confirmed in dopaminergic neuron. Furthermore, we found that genetic knockdown of miR-425 aggravated MPTP-induced motor deficits and dopaminergic neurodegeneration via early upregulation of necroptotic genes. Intracerebral miR-425 mimics (AgomiR-425) treatment attenuated necroptosis activation and dopaminergic neuron loss, and improved locomotor behaviors. In conclusion, our study suggests that miR-425 deficiency triggers necroptosis of dopaminergic neurons, and targeting miR-425 in MPTP-treated mice restored dysfunctional dopaminergic neurodegeneration and ameliorated behavioral deficits. These findings identify brain delivery of miR-425 as a potential therapeutic approach for the treatment of PD.
Collapse
Affiliation(s)
- Yong-Bo Hu
- Department of Neurology & Neuroscience Institute, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.,Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Yong-Fang Zhang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Hao Wang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Ru-Jing Ren
- Department of Neurology & Neuroscience Institute, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Hai-Lun Cui
- Department of Neurology & Neuroscience Institute, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Wan-Ying Huang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Qi Cheng
- School of Public health Shanghai Jiao Tong university, 200025, Shanghai, China
| | - Hong-Zhuan Chen
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.,Institute of Interdisciplinary Science, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, China
| | - Gang Wang
- Department of Neurology & Neuroscience Institute, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| |
Collapse
|
82
|
Chen JJ, Yang G, Yan QQ, Zhao J, Li S. Exosome-encapsulated microRNAs as promising biomarkers for Alzheimer’s disease. Rev Neurosci 2019; 31:77-87. [DOI: 10.1515/revneuro-2019-0001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 04/17/2019] [Indexed: 12/13/2022]
Abstract
Abstract
Alzheimer’s disease (AD) is a chronic neurodegenerative disease that locks into long clinical latency and low curative ratio. Therefore, early diagnosis before the clinical phase is quite essential and may be effective for therapeutic prevention. Peripheral blood or cerebrospinal fluid biomarkers symbolizing functional neuronal impairment are gradually applied to diagnose AD in research studies. Exosomes have generated immense interest in the diagnosis field of neurodegenerative disorders after confirmation of their roles as mediators, delivering important proteins and microRNAs (miRNAs) in intercellular communication. Compelling research results reveal that miRNAs released from exosomes modulate expression and function of amyloid precursor proteins and tau proteins. These findings open up possibility that dysfunctional exosomal miRNAs may influence AD progression. In this review, we summarized the existing knowledge of exosomal miRNAs and their involvement in AD, emphasizing their potential to serve as diagnostic biomarkers during the preclinical phase of AD.
Collapse
Affiliation(s)
- Jian-jiao Chen
- Department of General Surgery , Jinhua Hospital of Zhejiang University (Jinhua Municipal Central Hospital) , Jinhua City 321000, Zhejiang Province , P.R. China
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences, Dalian Medical University , Dalian City 116044, Liaoning Province , P.R. China
| | - Guang Yang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan City 430030, Hubei Province , P.R. China
| | - Qing-qing Yan
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences , Dalian Medical University , 9 Western District, Lvshun South Road, Dalian City 116044 , Liaoning Province , P.R. China
| | - Jie Zhao
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences , Dalian Medical University , 9 Western District, Lvshun South Road, Dalian City 116044 , Liaoning Province , P.R. China
- National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases , Dalian Medical University , Dalian 116000 , China
| | - Shao Li
- Liaoning Provincial Key Laboratory of Cerebral Diseases, Department of Physiology, College of Basic Medical Sciences , Dalian Medical University , 9 Western District, Lvshun South Road, Dalian City 116044 , Liaoning Province , P.R. China
- National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases , Dalian Medical University , Dalian 116000 , China
| |
Collapse
|
83
|
Goodall EF, Leach V, Wang C, Cooper-Knock J, Heath PR, Baker D, Drew DR, Saffrey MJ, Simpson JE, Romero IA, Wharton SB. Age-Associated mRNA and miRNA Expression Changes in the Blood-Brain Barrier. Int J Mol Sci 2019; 20:ijms20123097. [PMID: 31242592 PMCID: PMC6627814 DOI: 10.3390/ijms20123097] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 06/21/2019] [Accepted: 06/22/2019] [Indexed: 02/04/2023] Open
Abstract
Functional and structural age-associated changes in the blood-brain barrier (BBB) may affect the neurovascular unit and contribute to the onset and progression of age-associated neurodegenerative pathologies, including Alzheimer’s disease. The current study interrogated the RNA profile of the BBB in an ageing human autopsy brain cohort and an ageing mouse model using combined laser capture microdissection and expression profiling. Only 12 overlapping genes were altered in the same direction in the BBB of both ageing human and mouse cohorts. These included genes with roles in regulating vascular tone, tight junction protein expression and cell adhesion, all processes prone to dysregulation with advancing age. Integrated mRNA and miRNA network and pathway enrichment analysis of the datasets identified 15 overlapping miRNAs that showed altered expression. In addition to targeting genes related to DNA binding and/or autophagy, many of the miRNAs identified play a role in age-relevant processes, including BBB dysfunction and regulating the neuroinflammatory response. Future studies have the potential to develop targeted therapeutic approaches against these candidates to prevent vascular dysfunction in the ageing brain.
Collapse
Affiliation(s)
- Emily F Goodall
- Sheffield Institute for Translational Neuroscience, 385a Glossop Road, University of Sheffield, Sheffield S10 2HQ, UK.
| | - Vicki Leach
- Sheffield Institute for Translational Neuroscience, 385a Glossop Road, University of Sheffield, Sheffield S10 2HQ, UK.
| | - Chunfang Wang
- School of Life Science, Health and Chemical Sciences, Faculty of Science, Technology Engineering and Mathematics, The Open University, Walton Hall, Milton Keynes MK7 6AA, UK.
| | - Johnathan Cooper-Knock
- Sheffield Institute for Translational Neuroscience, 385a Glossop Road, University of Sheffield, Sheffield S10 2HQ, UK.
| | - Paul R Heath
- Sheffield Institute for Translational Neuroscience, 385a Glossop Road, University of Sheffield, Sheffield S10 2HQ, UK.
| | - David Baker
- Sheffield Institute for Translational Neuroscience, 385a Glossop Road, University of Sheffield, Sheffield S10 2HQ, UK.
| | - David R Drew
- Sheffield Institute for Translational Neuroscience, 385a Glossop Road, University of Sheffield, Sheffield S10 2HQ, UK.
| | - M Jill Saffrey
- School of Life Science, Health and Chemical Sciences, Faculty of Science, Technology Engineering and Mathematics, The Open University, Walton Hall, Milton Keynes MK7 6AA, UK.
| | - Julie E Simpson
- Sheffield Institute for Translational Neuroscience, 385a Glossop Road, University of Sheffield, Sheffield S10 2HQ, UK.
| | - Ignacio A Romero
- School of Life Science, Health and Chemical Sciences, Faculty of Science, Technology Engineering and Mathematics, The Open University, Walton Hall, Milton Keynes MK7 6AA, UK.
| | - Stephen B Wharton
- Sheffield Institute for Translational Neuroscience, 385a Glossop Road, University of Sheffield, Sheffield S10 2HQ, UK.
| |
Collapse
|
84
|
Amakiri N, Kubosumi A, Tran J, Reddy PH. Amyloid Beta and MicroRNAs in Alzheimer's Disease. Front Neurosci 2019; 13:430. [PMID: 31130840 PMCID: PMC6510214 DOI: 10.3389/fnins.2019.00430] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 04/15/2019] [Indexed: 11/29/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive mental illness characterized by memory loss and multiple cognitive impairments. In the last several decades, significant progress has been made in understanding basic biology, molecular mechanisms, and development of biomarkers and therapeutic drugs. Multiple cellular changes are implicated in the disease process including amyloid beta and phosphorylation of tau synaptic damage and mitochondrial dysfunction in AD. Among these, amyloid beta is considered a major player in the disease process. Recent advancements in molecular biology revealed that microRNAs (miRNAs) are considered potential biomarkers in AD with a focus on amyloid beta. In this article we discussed several aspects of AD including its prevalence, classifications, risk factors, and amyloid species and their accumulation in subcellular compartments. This article also discusses the discovery and biogenesis of miRNAs and their relevance to AD. Today's research continues to add to the wealth of miRNA data that has been accumulated, however, there still lacks clear-cut understanding of the physiological relevance of miRNAs to AD. MiRNAs appear to regulate translation of gene products in AD and other human diseases. However, the mechanism of how many of these miRNAs regulate both the 5' and 3'UTR of amyloid precursor protein (APP) processing is still being extrapolated. Hence, we still need more research on miRNAs and APP/amyloid beta formation in the progression and pathogenesis of AD.
Collapse
Affiliation(s)
- Nnana Amakiri
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Aaron Kubosumi
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - James Tran
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - P. Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Garrison Institute on Aging, South West Campus, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Speech-Language and Hearing Clinics, Texas Tech University Health Sciences Center, Lubbock, TX, United States
- Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
85
|
Galas MC, Bonnefoy E, Buee L, Lefebvre B. Emerging Connections Between Tau and Nucleic Acids. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1184:135-143. [PMID: 32096035 DOI: 10.1007/978-981-32-9358-8_12] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Connections between tau and nucleic acids have been largely underestimated until recently when several reports highlighted new key roles of tau in relation with DNA and RNA structure, metabolism and integrity, and their implications in the context of tauopathies. Here we focus on recent advances involving tau and nucleic acids in neuronal and non-neuronal cells. Implication of tau and tau pathology in mechanisms regulating genome integrity, chromatin organization and RNA metabolism, highlight the connections between tau and nucleic acid as major mechanisms in neuronal homeostasis and the etiopathology of tauopathies.
Collapse
Affiliation(s)
- Marie-Christine Galas
- University of Lille, INSERM, CHU-Lille, UMR-S 1172, Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France.
| | | | - Luc Buee
- University of Lille, INSERM, CHU-Lille, UMR-S 1172, Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| | - Bruno Lefebvre
- University of Lille, INSERM, CHU-Lille, UMR-S 1172, Alzheimer & Tauopathies, LabEx DISTALZ, Lille, France
| |
Collapse
|